51
|
Young J, Zastrow-Hayes G, Deschamps S, Svitashev S, Zaremba M, Acharya A, Paulraj S, Peterson-Burch B, Schwartz C, Djukanovic V, Lenderts B, Feigenbutz L, Wang L, Alarcon C, Siksnys V, May G, Chilcoat ND, Kumar S. CRISPR-Cas9 Editing in Maize: Systematic Evaluation of Off-target Activity and Its Relevance in Crop Improvement. Sci Rep 2019; 9:6729. [PMID: 31040331 PMCID: PMC6491584 DOI: 10.1038/s41598-019-43141-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022] Open
Abstract
CRISPR-Cas9 enabled genome engineering has great potential for improving agriculture productivity, but the possibility of unintended off-target edits has evoked some concerns. Here we employ a three-step strategy to investigate Cas9 nuclease specificity in a complex plant genome. Our approach pairs computational prediction with genome-wide biochemical off-target detection followed by validation in maize plants. Our results reveal high frequency (up to 90%) on-target editing with no evidence of off-target cleavage activity when guide RNAs were bioinformatically predicted to be specific. Predictable off-target edits were observed but only with a promiscuous guide RNA intentionally designed to validate our approach. Off-target editing can be minimized by designing guide RNAs that are different from other genomic locations by at least three mismatches in combination with at least one mismatch occurring in the PAM proximal region. With well-designed guides, genetic variation from Cas9 off-target cleavage in plants is negligible, and much less than inherent variation.
Collapse
Affiliation(s)
- Joshua Young
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Gina Zastrow-Hayes
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Stéphane Deschamps
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Sergei Svitashev
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Mindaugas Zaremba
- Institute of Biotechnology, Vilnius University, Vilnius, LT, 10257, Lithuania
| | - Ananta Acharya
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Sushmitha Paulraj
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | | | - Chris Schwartz
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Vesna Djukanovic
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Brian Lenderts
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Lanie Feigenbutz
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Lijuan Wang
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Clara Alarcon
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Virginijus Siksnys
- Institute of Biotechnology, Vilnius University, Vilnius, LT, 10257, Lithuania
| | - Gregory May
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - N Doane Chilcoat
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA
| | - Sandeep Kumar
- Corteva Agriscience™, Agriculture Division of DowDuPont™, Johnston, IA, 50131, USA.
| |
Collapse
|
52
|
MtGA2ox10 encoding C20-GA2-oxidase regulates rhizobial infection and nodule development in Medicago truncatula. Sci Rep 2019; 9:5952. [PMID: 30976084 PMCID: PMC6459840 DOI: 10.1038/s41598-019-42407-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/01/2019] [Indexed: 01/03/2023] Open
Abstract
Gibberellin (GA) plays a controversial role in the legume-rhizobium symbiosis. Recent studies have shown that the GA level in legumes must be precisely controlled for successful rhizobial infection and nodule organogenesis. However, regulation of the GA level via catabolism in legume roots has not been reported to date. Here, we investigate a novel GA inactivating C20-GA2-oxidase gene MtGA2ox10 in Medicago truncatula. RNA sequencing analysis and quantitative polymerase chain reaction revealed that MtGA2ox10 was induced as early as 6 h post-inoculation (hpi) of rhizobia and reached peak transcript abundance at 12 hpi. Promoter::β-glucuronidase fusion showed that the promoter activity was localized in the root infection/differentiation zone during the early stage of rhizobial infection and in the vascular bundle of the mature nodule. The CRISPR/Cas9-mediated deletion mutation of MtGA2ox10 suppressed infection thread formation, which resulted in reduced development and retarded growth of nodules on the Agrobacterium rhizogenes-transformed roots. Over-expression of MtGA2ox10 in the stable transgenic plants caused dwarfism, which was rescued by GA3 application, and increased infection thread formation but inhibition of nodule development. We conclude that MtGA2ox10 plays an important role in the rhizobial infection and the development of root nodules through fine catabolic tuning of GA in M. truncatula.
Collapse
|
53
|
Dyikanov DT, Vasiluev PA, Rysenkova KD, Aleksandrushkina NA, Tyurin-Kuzmin PA, Kulebyakin KY, Rubtsov YP, Shmakova AA, Evseeva MN, Balatskiy AV, Semina EV, Rostovtseva AI, Makarevich PI, Karagyaur MN. Optimization of CRISPR/Cas9 Technology to Knock Out Genes of Interest in Aneuploid Cell Lines. Tissue Eng Part C Methods 2019; 25:168-175. [DOI: 10.1089/ten.tec.2018.0365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Daniyar T. Dyikanov
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Petr A. Vasiluev
- FGBI of Science “Institute of Gene Biology” of the Russian Academy of Sciences, Moscow, Russia
- FGBI “Medical Genetics Research Center,” Moscow, Russia
| | - Karina D. Rysenkova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- FGBI “National Medical Research Center of Cardiology” of the Ministry of Health of Russia, Moscow, Russia
| | - Natalia A. Aleksandrushkina
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | | | - Konstantin Y. Kulebyakin
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Yury P. Rubtsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- National Research University Higher School of Economics, Moscow, Russia
| | - Anna A. Shmakova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maria N. Evseeva
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Alexander V. Balatskiy
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina V. Semina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- FGBI “National Medical Research Center of Cardiology” of the Ministry of Health of Russia, Moscow, Russia
| | | | - Pavel I. Makarevich
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim N. Karagyaur
- Institute of Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
54
|
Chang L, Wu S, Tian L. Effective genome editing and identification of a regiospecific gallic acid 4- O-glycosyltransferase in pomegranate ( Punica granatum L.). HORTICULTURE RESEARCH 2019; 6:123. [PMID: 31728198 PMCID: PMC6838055 DOI: 10.1038/s41438-019-0206-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/27/2019] [Accepted: 10/01/2019] [Indexed: 05/10/2023]
Abstract
Pomegranate (Punica granatum L.) trees are woody perennials that bear colorful and nutritious fruits rich in phenolic metabolites, e.g., hydrolyzable tannins (HTs) and flavonoids. We here report genome editing and gene discovery in pomegranate hairy roots using Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) (CRISPR/Cas9), coupled with transcriptome and biochemical analyses. Single guide RNAs (sgRNAs) were designed to target two UDP-dependent glycosyltransferases (UGTs), PgUGT84A23 and PgUGT84A24, which possess overlapping activities in β-glucogallin (a galloylglucose ester; biosynthetic precursor of HTs) biosynthesis. A unique accumulation of gallic acid 3-O- and 4-O-glucosides (galloylglucose ethers) was observed in the PgUGT84A23 and PgUGT84A24 dual CRISPR/Cas9-edited lines (i.e., ugt84a23 ugt84a24) but not the control (empty vector) or PgUGT84A23/PgUGT84A24 single edited lines (ugt84a23 or ugt84a24). Transcriptome and real-time qPCR analyses identified 11 UGTs with increased expression in the ugt84a23 ugt84a24 hairy roots compared to the controls. Of the 11 candidate UGTs, only PgUGT72BD1 used gallic acid as substrate and produced a regiospecific product gallic acid 4-O-glucoside. This work demonstrates that the CRISPR/Cas9 method can facilitate functional genomics studies in pomegranate and shows promise for capitalizing on the metabolic potential of pomegranate for germplasm improvement.
Collapse
Affiliation(s)
- Lijing Chang
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, 201602 Shanghai, China
- Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, 201602 Shanghai, China
| | - Sheng Wu
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, 201602 Shanghai, China
- Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, 201602 Shanghai, China
| | - Li Tian
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, 201602 Shanghai, China
- Shanghai Chenshan Plant Science Research Center, Chinese Academy of Sciences, 201602 Shanghai, China
- Department of Plant Sciences, University of California, Davis, CA 95616 USA
| |
Collapse
|
55
|
Abstract
Precise genome engineering can be efficiently made using the revolutionary tool named CRISPR/Cas (clustered regularly interspaced short palindromic repeat/CRISPR-associated protein) systems. Adapted from the bacterial immune system, CRISPR/Cas systems can generate highly specific double-strand breaks (DSBs) at the target site, and desired sequence modifications can be introduced during the DSB repair process, such as nonhomologous end-joining (NHEJ) or homology-directed repair (HDR) pathways. CRISPR/Cas9 is the most widely used genome editing tool for targeted mutagenesis, precise sequence modification, transcriptional reprogramming, epigenome editing, disease treatment, and many more. The ease of use and high specificity make CRISPR/Cas9 a great tool not only for basic researches but also for crop trait improvements, such as higher grain yield, better tolerance to abiotic stresses, enhanced disease resistance, and better nutritional contents. In this protocol, we present a step-by-step guide to the CRISPR/Cas9-mediated targeted mutagenesis in maize Hi II genotype. Detailed procedures will guide through the essential steps including gRNA design, CRISPR/Cas9 vector construction, Agrobacterium-mediated maize immature embryo transformation, and molecular analysis of the transgenic plants to identify desired mutant lines.
Collapse
|
56
|
Hwang GH, Park J, Lim K, Kim S, Yu J, Yu E, Kim ST, Eils R, Kim JS, Bae S. Web-based design and analysis tools for CRISPR base editing. BMC Bioinformatics 2018; 19:542. [PMID: 30587106 PMCID: PMC6307267 DOI: 10.1186/s12859-018-2585-4] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 12/14/2018] [Indexed: 12/31/2022] Open
Abstract
Background As a result of its simplicity and high efficiency, the CRISPR-Cas system has been widely used as a genome editing tool. Recently, CRISPR base editors, which consist of deactivated Cas9 (dCas9) or Cas9 nickase (nCas9) linked with a cytidine or a guanine deaminase, have been developed. Base editing tools will be very useful for gene correction because they can produce highly specific DNA substitutions without the introduction of any donor DNA, but dedicated web-based tools to facilitate the use of such tools have not yet been developed. Results We present two web tools for base editors, named BE-Designer and BE-Analyzer. BE-Designer provides all possible base editor target sequences in a given input DNA sequence with useful information including potential off-target sites. BE-Analyzer, a tool for assessing base editing outcomes from next generation sequencing (NGS) data, provides information about mutations in a table and interactive graphs. Furthermore, because the tool runs client-side, large amounts of targeted deep sequencing data (< 1 GB) do not need to be uploaded to a server, substantially reducing running time and increasing data security. BE-Designer and BE-Analyzer can be freely accessed at http://www.rgenome.net/be-designer/ and http://www.rgenome.net/be-analyzer/, respectively. Conclusion We develop two useful web tools to design target sequence (BE-Designer) and to analyze NGS data from experimental results (BE-Analyzer) for CRISPR base editors. Electronic supplementary material The online version of this article (10.1186/s12859-018-2585-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gue-Ho Hwang
- Department of Chemistry, Hanyang University, Seoul, South Korea
| | - Jeongbin Park
- Center for Digital Health, Berlin Institute of Health and Charité Universitätsmedizin Berlin, Berlin, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Kayeong Lim
- Department of Chemistry, Seoul National University, Seoul, South Korea.,Center for Genome Engineering, Institute for Basic Science, Seoul, South Korea
| | - Sunghyun Kim
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Jihyeon Yu
- Department of Chemistry, Hanyang University, Seoul, South Korea.,Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, South Korea
| | - Eunchong Yu
- Department of Chemistry, Hanyang University, Seoul, South Korea
| | - Sang-Tae Kim
- Center for Genome Engineering, Institute for Basic Science, Daejeon, South Korea
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health and Charité Universitätsmedizin Berlin, Berlin, Germany.,Health Data Science Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Jin-Soo Kim
- Department of Chemistry, Seoul National University, Seoul, South Korea.,Center for Genome Engineering, Institute for Basic Science, Seoul, South Korea
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul, South Korea. .,Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, South Korea.
| |
Collapse
|
57
|
Zhou W, Sun W, Yung MMH, Dai S, Cai Y, Chen CW, Meng Y, Lee JB, Braisted JC, Xu Y, Southall NT, Shinn P, Huang X, Song Z, Chen X, Kai Y, Cai X, Li Z, Hao Q, Cheung ANY, Ngan HYS, Liu SS, Barak S, Hao J, Dai Z, Tzatsos A, Peng W, Pei H, Han Z, Chan DW, Zheng W, Zhu W. Autocrine activation of JAK2 by IL-11 promotes platinum drug resistance. Oncogene 2018; 37:3981-3997. [PMID: 29662190 PMCID: PMC6054535 DOI: 10.1038/s41388-018-0238-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/20/2018] [Accepted: 03/02/2018] [Indexed: 02/08/2023]
Abstract
Antineoplastic platinum agents are used in first-line treatment of ovarian cancer, but treatment failure frequently results from platinum drug resistance. Emerging observations suggest a role of reactive oxygen species (ROS) in the resistance of cancer drugs including platinum drugs. However, the molecular link between ROS and cellular survival pathway is poorly understood. Using quantitative high-throughput combinational screen (qHTCS) and genomic sequencing, we show that in platinum-resistant ovarian cancer elevated ROS levels sustain high level of IL-11 by stimulating FRA1-mediated IL-11 expression and increased IL-11 causes resistance to platinum drugs by constitutively activating JAK2-STAT5 via an autocrine mechanism. Inhibition of JAK2 by LY2784544 or IL-11 by anti-IL-11 antibody overcomes the platinum resistance in vitro or in vivo. Significantly, clinic studies also confirm the activated IL-11-JAK2 pathway in platinum-resistant ovarian cancer patients, which highly correlates with poor prognosis. These findings not only identify a novel ROS-IL-11-JAK2-mediated platinum resistance mechanism but also provide a new strategy for using LY2784544- or IL-11-mediated immunotherapy to treat platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mingo M H Yung
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sheng Dai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yihua Cai
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Chi-Wei Chen
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Yunxiao Meng
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Jennifer B Lee
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - John C Braisted
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yinghua Xu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Noel T Southall
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Paul Shinn
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Xiulei Chen
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Yan Kai
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
- Department of Physics, The George Washington University Columbian College of Arts & Sciences, Washington, DC, 20052, USA
| | - Xin Cai
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Zongzhu Li
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Qiang Hao
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Annie N Y Cheung
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hextan Y S Ngan
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephanie S Liu
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Stephanie Barak
- Department of Pathology, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Jing Hao
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Zhijun Dai
- Department of Oncology, the Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Alexandros Tzatsos
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
- Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - Weiqun Peng
- Department of Physics, The George Washington University Columbian College of Arts & Sciences, Washington, DC, 20052, USA
| | - Huadong Pei
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA
| | - Zhiyong Han
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA
| | - David W Chan
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, 20037, USA.
- GW Cancer Centre, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
58
|
Chuai G, Ma H, Yan J, Chen M, Hong N, Xue D, Zhou C, Zhu C, Chen K, Duan B, Gu F, Qu S, Huang D, Wei J, Liu Q. DeepCRISPR: optimized CRISPR guide RNA design by deep learning. Genome Biol 2018; 19:80. [PMID: 29945655 PMCID: PMC6020378 DOI: 10.1186/s13059-018-1459-4] [Citation(s) in RCA: 260] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/28/2018] [Indexed: 12/22/2022] Open
Abstract
A major challenge for effective application of CRISPR systems is to accurately predict the single guide RNA (sgRNA) on-target knockout efficacy and off-target profile, which would facilitate the optimized design of sgRNAs with high sensitivity and specificity. Here we present DeepCRISPR, a comprehensive computational platform to unify sgRNA on-target and off-target site prediction into one framework with deep learning, surpassing available state-of-the-art in silico tools. In addition, DeepCRISPR fully automates the identification of sequence and epigenetic features that may affect sgRNA knockout efficacy in a data-driven manner. DeepCRISPR is available at http://www.deepcrispr.net/ .
Collapse
Affiliation(s)
- Guohui Chuai
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Hanhui Ma
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jifang Yan
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Ming Chen
- R&D Information, Innovation Center China, AstraZeneca, 199 Liangjing Road, Shanghai, 201203, China
| | - Nanfang Hong
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Dongyu Xue
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Chi Zhou
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Chenyu Zhu
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Ke Chen
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Bin Duan
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Feng Gu
- State Key Laboratory Cultivation Base and Key Laboratory of Vision Science, Ministry of Health and Zhejiang Provincial Key Laboratory of Ophthalmology and Optometry, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Sheng Qu
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China
| | - Deshuang Huang
- Machine Learning & Systems Biology Lab, School of Electronics and Information Engineering, Tongji University, Shanghai, 201804, China.
| | - Jia Wei
- R&D Information, Innovation Center China, AstraZeneca, 199 Liangjing Road, Shanghai, 201203, China.
| | - Qi Liu
- Department of Endocrinology & Metabolism, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 20009, China.
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 20009, China.
| |
Collapse
|
59
|
Al Abdallah Q, Souza ACO, Martin-Vicente A, Ge W, Fortwendel JR. Whole-genome sequencing reveals highly specific gene targeting by in vitro assembled Cas9-ribonucleoprotein complexes in Aspergillus fumigatus. Fungal Biol Biotechnol 2018; 5:11. [PMID: 29992034 PMCID: PMC5987418 DOI: 10.1186/s40694-018-0057-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022] Open
Abstract
Background CRISPR/Cas9-based genome editing is quickly becoming a powerful tool within the field of fungal genetics. Adaptation of CRISPR/Cas9 systems are allowing for rapid and highly efficient gene targeting within fungi. We recently reported the adaptation of a simple CRISPR/Cas9 system for gene deletion that is effective across multiple genetic backgrounds of Aspergillus fumigatus. This system employs in vitro assembly of Cas9 ribonucleoproteins (RNPs) coupled with micro-homology repair templates for gene deletion. Although highly efficient at gene targeting in wild type genetic backgrounds of A. fumigatus, the potential for our system to produce unwanted off-target mutations has not been addressed. Results Next-generation Illumina sequencing was used to identify genome mutations among transformants isolated from standard (no Cas9) and Cas9-mediated integration of a hygromycin deletion cassette. Two different concentrations of Cas9 were utilized to examine the association of Cas9 concentration with total numbers and types of genomic mutations. For each of the three test groups (zero, low, and high Cas9), three transformants were sequenced and compared to the parent strain. Bioinformatics analyses revealed the average number of total mutations to be similar among all three test groups. A. fumigatus transformation using standard, non-Cas9-mediated methods resulted in an average of 373 ± 28 mutations. In comparison, transformation with in vitro assembled Cas9-RNPs using either high (1 µg/µl) or low (0.5 µg/µl) levels of Cas9 resulted in an average of 326 ± 19 and 395 ± 69 mutations, respectively. In all cases, the vast majority of mutations identified were intergenic. No correlation between the amount of Cas9 utilized for transformation and the overall number of mutations was found. Finally, the specific type of mutation introduced during the transformation process was not Cas9-dependent, as both single-nucleotide polymorphisms and insertion/deletion events were not significantly different between the experimental groups. Conclusions CRISPR/Cas9-based genome editing in A. fumigatus using in vitro assembled RNPs coupled with microhomology templates is a reliable method of gene targeting. This system is highly efficient and is not associated with increased off-target mutations caused by introduction of the Cas9 nuclease.
Collapse
Affiliation(s)
- Qusai Al Abdallah
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN USA
| | - Ana Camila Oliveira Souza
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN USA
| | - Adela Martin-Vicente
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN USA
| | - Wenbo Ge
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN USA
| | - Jarrod R Fortwendel
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN USA
| |
Collapse
|
60
|
Karagyaur MN, Rubtsov YP, Vasiliev PA, Tkachuk VA. Practical Recommendations for Improving Efficiency and Accuracy of the CRISPR/Cas9 Genome Editing System. BIOCHEMISTRY. BIOKHIMIIA 2018; 83:629-642. [PMID: 30195321 DOI: 10.1134/s0006297918060020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Indexed: 12/26/2022]
Abstract
CRISPR/Cas9 genome-editing system is a powerful, fairly accurate, and efficient tool for modifying genomic DNA. Despite obvious advantages, it is not devoid of certain drawbacks, such as propensity for introduction of additional nonspecific DNA breaks, insufficient activity against aneuploid genomes, and relative difficulty in delivering its components to cells. In this review, we focus on the difficulties that can limit the use of CRISPR/Cas9 and suggest a number of practical recommendations and information sources that will make it easier for the beginners to work with this outstanding technological achievement of the XXI century.
Collapse
Affiliation(s)
- M N Karagyaur
- Lomonosov Moscow State University, Institute of Regenerative Medicine, Moscow, 119192, Russia.
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, 119192, Russia
| | - Y P Rubtsov
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, 119192, Russia
| | - P A Vasiliev
- Research Center of Medical Genetics, Russian Academy of Medical Sciences, Moscow, 115478, Russia
| | - V A Tkachuk
- Lomonosov Moscow State University, Institute of Regenerative Medicine, Moscow, 119192, Russia
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, 119192, Russia
| |
Collapse
|
61
|
Sekine R, Kawata T, Muramoto T. CRISPR/Cas9 mediated targeting of multiple genes in Dictyostelium. Sci Rep 2018; 8:8471. [PMID: 29855514 PMCID: PMC5981456 DOI: 10.1038/s41598-018-26756-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/18/2018] [Indexed: 01/17/2023] Open
Abstract
CRISPR/Cas9 has emerged in various organisms as a powerful technology for targeted gene knockout; however, no reports of editing the Dictyostelium genome efficiently using this system are available. We describe here the application of CRISPR/Cas9-mediated gene modification in Dictyostelium. The endogenous tRNA-processing system for expressing sgRNA was approximately 10 times more effective than the commonly used U6 promoter. The resulting sgRNA affected the sub-nuclear localisation of Cas9, indicating that the expression level of sgRNA was sufficiently high to form Cas9 and sgRNA complexes within the nucleus. The all-in-one vector containing Cas9 and sgRNA was transiently expressed to generate mutants in five PI3K genes. Mutation detective PCR revealed the mutagenesis frequency of the individual genes to be between 72.9% and 100%. We confirmed that all five targeting loci in the four independent clones had insertion/deletion mutations in their target sites. Thus, we show that the CRISPR/Cas9 system can be used in Dictyostelium cells to enable efficient genome editing of multiple genes. Since this system utilises transient expression of the all-in-one vector, it has the advantage that the drug resistance cassette is not integrated into the genome and simple vector construction, involving annealing two oligo-DNAs.
Collapse
Affiliation(s)
- Ryoya Sekine
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Takefumi Kawata
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan
| | - Tetsuya Muramoto
- Department of Biology, Faculty of Science, Toho University, 2-2-1 Miyama, Funabashi, Chiba, 274-8510, Japan.
| |
Collapse
|
62
|
Bhatia S, Sharma J, Bukkapatnam S, Oweida A, Lennon S, Phan A, Milner D, Uyanga N, Jimeno A, Raben D, Somerset H, Heasley L, Karam SD. Inhibition of EphB4-Ephrin-B2 Signaling Enhances Response to Cetuximab-Radiation Therapy in Head and Neck Cancers. Clin Cancer Res 2018; 24:4539-4550. [PMID: 29848571 DOI: 10.1158/1078-0432.ccr-18-0327] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/23/2018] [Accepted: 05/25/2018] [Indexed: 01/30/2023]
Abstract
Purpose: The clinical success of targeted therapies such as cetuximab and radiotherapy (RT) is hampered by the low response rates and development of therapeutic resistance. In the current study, we investigated the involvement of EphB4-ephrin-B2 protumorigenic signaling in mediating resistance to EGFR inhibition and RT in head and neck cancers.Experimental Design: We used patient-derived xenograft (PDX) models of head and neck squamous cell carcinoma (HNSCC) and HNSCC cell lines to test our hypothesis. Tumor tissues were subjected to PhosphoRTK array, and Western blotting to detect changes in EphB4-ephrin-B2 targets. mRNA sequencing and microarray data analysis were performed on PDX tumors and HNSCC cell lines, respectively, to determine differences in gene expression of molecules involved in tumor cell growth, proliferation, and survival pathways. Effects on cell growth were determined by MTT assay on HNSCC cells downregulated for EphB4/ephrin-B2 expression, with and without EGFR inhibitor and radiation.Results: Our data from locally advanced HNSCC patients treated with standard-of-care definitive chemo-RT show elevated EphB4 and ephrin-B2 levels after failure of treatment. We observed significant response toward cetuximab and RT following EphB4-ephrin-B2 inhibition, resulting in improved survival in tumor-bearing mice. Tumor growth inhibition was accompanied by a decrease in the levels of proliferation and prosurvival molecules and increased apoptosis.Conclusions: Our findings underscore the importance of adopting rational drug combinations to enhance therapeutic effect. Our study documenting enhanced response of HNSCC to cetuximab-RT with EphB4-ephrin-B2 blockade has the potential to translate into the clinic to benefit this patient population. Clin Cancer Res; 24(18); 4539-50. ©2018 AACR.
Collapse
Affiliation(s)
- Shilpa Bhatia
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Jaspreet Sharma
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Sanjana Bukkapatnam
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Ayman Oweida
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Shelby Lennon
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Andy Phan
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Dallin Milner
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Nomin Uyanga
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Antonio Jimeno
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - David Raben
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Hilary Somerset
- Department of Pathology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Lynn Heasley
- Department of Craniofacial Biology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
63
|
Abstract
The utility of human induced pluripotent stem cells (iPSCs) is enhanced by an ability to precisely modify a chosen locus with minimal impact on the remaining genome. However, the derivation of gene-edited iPSCs typically involves multiple steps requiring lengthy culture periods and several clonal events. Here, we describe a one-step protocol for reliable generation of clonally derived gene-edited iPSC lines from human fibroblasts in the absence of drug selection or FACS enrichment. Using enhanced episomal-based reprogramming and CRISPR/Cas9 systems, gene-edited and passage-matched unmodified iPSC lines are obtained following a single electroporation of human fibroblasts. To minimize unwanted mutations within the target locus, we use a Cas9 variant that is associated with decreased nonhomologous end-joining (NHEJ) activity. This protocol outlines in detail how this streamlined approach can be used for both monoallelic and biallelic introduction of specific base changes or transgene cassettes in a manner that is efficient, rapid (∼6-8 weeks), and cost-effective.
Collapse
|
64
|
Sun J, Wang Q, Jiang Y, Wen Z, Yang L, Wu J, Yang S. Genome editing and transcriptional repression in Pseudomonas putida KT2440 via the type II CRISPR system. Microb Cell Fact 2018. [PMID: 29534717 PMCID: PMC5851096 DOI: 10.1186/s12934-018-0887-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background The soil bacterium Pseudomonas putida KT2440 is a “generally recognized as safe”-certified strain with robust property and versatile metabolism. Thus, it is an ideal candidate for synthetic biology, biodegradation, and other biotechnology applications. The known genome editing approaches of Pseudomonas are suboptimal; thus, it is necessary to develop a high efficiency genome editing tool. Results In this study, we established a fast and convenient CRISPR–Cas9 method in P. putida KT2440. Gene deletion, gene insertion and gene replacement could be achieved within 5 days, and the mutation efficiency reached > 70%. Single nucleotide replacement could be realized, overcoming the limitations of protospacer adjacent motif sequences. We also applied nuclease-deficient Cas9 binding at three locations upstream of enhanced green fluorescent protein (eGFP) for transcriptional inhibition, and the expression intensity of eGFP reduced to 28.5, 29.4, and 72.1% of the control level, respectively. Furthermore, based on this CRISPR–Cas9 system, we also constructed a CRISPR–Cpf1 system, which we validated for genome editing in P. putida KT2440. Conclusions In this research, we established CRISPR based genome editing and regulation control systems in P. putida KT2440. These fast and efficient approaches will greatly facilitate the application of P. putida KT2440.![]() Electronic supplementary material The online version of this article (10.1186/s12934-018-0887-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jun Sun
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qingzhuo Wang
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yu Jiang
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032, China.,Shanghai Research and Development Center of Industrial Biotechnology, Shanghai, 201206, China
| | - Zhiqiang Wen
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Lirong Yang
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianping Wu
- Institute of Bioengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Sheng Yang
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200032, China. .,Shanghai Research and Development Center of Industrial Biotechnology, Shanghai, 201206, China. .,Jiangsu National Synergetic Innovation Center for Advanced Materials, SICAM, Nanjing, 210009, China.
| |
Collapse
|
65
|
Zhang H, Zhou Y, Wang Y, Zhao Y, Qiu Y, Zhang X, Yue D, Zhou Z, Wei W. A surrogate reporter system for multiplexable evaluation of CRISPR/Cas9 in targeted mutagenesis. Sci Rep 2018; 8:1042. [PMID: 29348585 PMCID: PMC5773543 DOI: 10.1038/s41598-018-19317-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/27/2017] [Indexed: 12/17/2022] Open
Abstract
Engineered nucleases in genome editing manifest diverse efficiencies at different targeted loci. There is therefore a constant need to evaluate the mutation rates at given loci. T7 endonuclease 1 (T7E1) and Surveyor mismatch cleavage assays are the most widely used methods, but they are labour and time consuming, especially when one must address multiple samples in parallel. Here, we report a surrogate system, called UDAR (Universal Donor As Reporter), to evaluate the efficiency of CRISPR/Cas9 in targeted mutagenesis. Based on the non-homologous end-joining (NHEJ)-mediated knock-in strategy, the UDAR-based assay allows us to rapidly evaluate the targeting efficiencies of sgRNAs. With one-step transfection and fluorescence-activated cell sorting (FACS) analysis, the UDAR assay can be completed on a large scale within three days. For detecting mutations generated by the CRISPR/Cas9 system, a significant positive correlation was observed between the results from the UDAR and T7E1 assays. Consistently, the UDAR assay could quantitatively assess bleomycin- or ICRF193-induced double-strand breaks (DSBs), which suggests that this novel strategy is broadly applicable to assessing the DSB-inducing capability of various agents. With the increasing impact of genome editing in biomedical studies, the UDAR method can significantly benefit the evaluation of targeted mutagenesis, especially for high-throughput purposes.
Collapse
Affiliation(s)
- Hongmin Zhang
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yuexin Zhou
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yinan Wang
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Yige Zhao
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yeting Qiu
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xinyi Zhang
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Di Yue
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Zhuo Zhou
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China.
| | - Wensheng Wei
- Beijing Advanced Innovation Center for Genomics, Biodynamic Optical Imaging Center (BIOPIC), Peking-Tsinghua Center for Life Sciences, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
66
|
Khan S, Fagerholm R, Kadalayil L, Tapper W, Aittomäki K, Liu J, Blomqvist C, Eccles D, Nevanlinna H. Meta-analysis of three genome-wide association studies identifies two loci that predict survival and treatment outcome in breast cancer. Oncotarget 2018; 9:4249-4257. [PMID: 29423119 PMCID: PMC5790536 DOI: 10.18632/oncotarget.22747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/09/2017] [Indexed: 11/25/2022] Open
Abstract
The majority of breast cancers are driven by the female hormone oestrogen via oestrogen receptor (ER) alpha. ER-positive patients are commonly treated with adjuvant endocrine therapy, however, resistance is a common occurrence and aside from ER-status, no unequivocal predictive biomarkers are currently in clinical use. In this study, we aimed to identify constitutional genetic variants influencing breast cancer survival among ER-positive patients and specifically, among endocrine-treated patients. We conducted a meta-analysis of three genome-wide association studies comprising in total 3,136 patients with ER-positive breast cancer of which 2,751 had received adjuvant endocrine therapy. We identified a novel locus (rs992531 at 8p21.2) associated with reduced survival among the patients with ER-positive breast cancer (P = 3.77 × 10-8). Another locus (rs7701292 at 5q21.3) was associated with reduced survival among the endocrine-treated patients (P = 2.13 × 10-8). Interaction analysis indicated that the survival association of rs7701292 is treatment-specific and independent of conventional prognostic markers. In silico functional studies suggest plausible biological mechanisms for the observed survival associations and a functional link between the putative target genes of the rs992531 and rs7701292 (RHOBTB2 and RAB9P1, respectively). We further explored the genetic interaction between rs992531 and rs7701292 and found a significant, treatment-specific interactive effect on survival among ER-positive, endocrine-treated patients (hazard ratio = 6.97; 95% confidence interval, 1.79-27.08, Pinteraction= 0.036). This is the first study to identify a genetic interaction that specifically predicts treatment outcome. These findings may provide predictive biomarkers based on germ line genotype informing more personalized treatment selection.
Collapse
Affiliation(s)
- Sofia Khan
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Biomedicum, Helsinki, Finland
| | - Rainer Fagerholm
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Biomedicum, Helsinki, Finland
| | - Latha Kadalayil
- Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - William Tapper
- Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| | - Kristiina Aittomäki
- Department of Clinical Genetics, Helsinki University Hospital and Genome Scale Biology Research Program, University of Helsinki, Helsinki, Finland
| | - Jianjun Liu
- Human Genetics, Genome Institute of Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Carl Blomqvist
- Department of Oncology, Helsinki University Hospital, Helsinki, Finland
- Department of Oncology, University of Örebro, Örebro, Sweden
| | - Diana Eccles
- Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, UK
| | - Heli Nevanlinna
- Department of Obstetrics and Gynaecology, University of Helsinki and Helsinki University Hospital, Biomedicum, Helsinki, Finland
| |
Collapse
|
67
|
Sonmez AB, Can T. Comparison of tissue/disease specific integrated networks using directed graphlet signatures. BMC Bioinformatics 2017; 18:135. [PMID: 28361704 PMCID: PMC5374550 DOI: 10.1186/s12859-017-1525-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Analysis of integrated genome-scale networks is a challenging problem due to heterogeneity of high-throughput data. There are several topological measures, such as graphlet counts, for characterization of biological networks. Results In this paper, we present methods for counting small sub-graph patterns in integrated genome-scale networks which are modeled as labeled multidigraphs. We have obtained physical, regulatory, and metabolic interactions between H. sapiens proteins from the Pathway Commons database. The integrated network is filtered for tissue/disease specific proteins by using a large-scale human transcriptional profiling study, resulting in several tissue and disease specific sub-networks. We have applied and extended the idea of graphlet counting in undirected protein-protein interaction (PPI) networks to directed multi-labeled networks and represented each network as a vector of graphlet counts. Graphlet counts are assessed for statistical significance by comparison against a set of randomized networks. We present our results on analysis of differential graphlets between different conditions and on the utility of graphlet count vectors for clustering multiple condition specific networks. Conclusions Our results show that there are numerous statistically significant graphlets in integrated biological networks and the graphlet signature vector can be used as an effective representation of a multi-labeled network for clustering and systems level analysis of tissue/disease specific networks.
Collapse
Affiliation(s)
- Arzu Burcak Sonmez
- Department of Medical Informatics, Informatics Institute, Middle East Technical University, Dumlupinar Bulvari No:1, Ankara, 06800, Turkey
| | - Tolga Can
- Department of Computer Egineering, Middle East Technical University, Dumlupinar Bulvari No:1, Ankara, 06800, Turkey.
| |
Collapse
|
68
|
Genome editing using FACS enrichment of nuclease-expressing cells and indel detection by amplicon analysis. Nat Protoc 2017; 12:581-603. [PMID: 28207001 DOI: 10.1038/nprot.2016.165] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This protocol describes methods for increasing and evaluating the efficiency of genome editing based on the CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-CRISPR-associated 9) system, transcription activator-like effector nucleases (TALENs) or zinc-finger nucleases (ZFNs). First, Indel Detection by Amplicon Analysis (IDAA) determines the size and frequency of insertions and deletions elicited by nucleases in cells, tissues or embryos through analysis of fluorophore-labeled PCR amplicons covering the nuclease target site by capillary electrophoresis in a sequenator. Second, FACS enrichment of cells expressing nucleases linked to fluorescent proteins can be used to maximize knockout or knock-in editing efficiencies or to balance editing efficiency and toxic/off-target effects. The two methods can be combined to form a pipeline for cell-line editing that facilitates the testing of new nuclease reagents and the generation of edited cell pools or clonal cell lines, reducing the number of clones that need to be generated and increasing the ease with which they are screened. The pipeline shortens the time line, but it most prominently reduces the workload of cell-line editing, which may be completed within 4 weeks.
Collapse
|
69
|
Kim H, Kim ST, Ryu J, Kang BC, Kim JS, Kim SG. CRISPR/Cpf1-mediated DNA-free plant genome editing. Nat Commun 2017; 8:14406. [PMID: 28205546 PMCID: PMC5316869 DOI: 10.1038/ncomms14406] [Citation(s) in RCA: 278] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/22/2016] [Indexed: 12/17/2022] Open
Abstract
Cpf1, a type V CRISPR effector, recognizes a thymidine-rich protospacer-adjacent motif and induces cohesive double-stranded breaks at the target site guided by a single CRISPR RNA (crRNA). Here we show that Cpf1 can be used as a tool for DNA-free editing of plant genomes. We describe the delivery of recombinant Cpf1 proteins with in vitro transcribed or chemically synthesized target-specific crRNAs into protoplasts isolated from soybean and wild tobacco. Designed crRNAs are unique and do not have similar sequences (≤3 mismatches) in the entire soybean reference genome. Targeted deep sequencing analyses show that mutations are successfully induced in FAD2 paralogues in soybean and AOC in wild tobacco. Unlike SpCas9, Cpf1 mainly induces various nucleotide deletions at target sites. No significant mutations are detected at potential off-target sites in the soybean genome. These results demonstrate that Cpf1-crRNA complex is an effective DNA-free genome-editing tool for plant genome editing.
Collapse
Affiliation(s)
- Hyeran Kim
- Center for Genome Engineering, Institute for Basic Science, 70, Yuseong-daero 1689-gil, Yuseong-gu, Daejeon 34047, South Korea
| | - Sang-Tae Kim
- Center for Genome Engineering, Institute for Basic Science, 70, Yuseong-daero 1689-gil, Yuseong-gu, Daejeon 34047, South Korea
| | - Jahee Ryu
- Center for Genome Engineering, Institute for Basic Science, 70, Yuseong-daero 1689-gil, Yuseong-gu, Daejeon 34047, South Korea
| | - Beum-Chang Kang
- Center for Genome Engineering, Institute for Basic Science, 70, Yuseong-daero 1689-gil, Yuseong-gu, Daejeon 34047, South Korea
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, 70, Yuseong-daero 1689-gil, Yuseong-gu, Daejeon 34047, South Korea
- Department of Chemistry, Seoul National University, Seoul 08826, South Korea
| | - Sang-Gyu Kim
- Center for Genome Engineering, Institute for Basic Science, 70, Yuseong-daero 1689-gil, Yuseong-gu, Daejeon 34047, South Korea
| |
Collapse
|
70
|
Abstract
Post-translational modifications (PTMs) are an important source of protein regulation; they fine-tune the function, localization, and interaction with other molecules of the majority of proteins and are partially responsible for their multifunctionality. Usually, proteins have several potential modification sites, and their patterns of occupancy are associated with certain functional states. These patterns imply cross talk among PTMs within and between proteins, the majority of which are still to be discovered. Several methods detect associations between PTMs; these have recently combined into a global resource, the PTMcode database, which contains already known and predicted functional associations between pairs of PTMs from more than 45,000 proteins in 19 eukaryotic species.
Collapse
Affiliation(s)
- Pablo Minguez
- Department of Genetics and Genomics, Instituto de Investigacion Sanitaria-University Hospital Fundacion Jimenez Diaz (IIS-FJD), Avda. Reyes Católicos 2, 28040, Madrid, Spain.
| | - Peer Bork
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, 69117, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, 13125, Berlin, Germany
| |
Collapse
|
71
|
Baek K, Kim DH, Jeong J, Sim SJ, Melis A, Kim JS, Jin E, Bae S. DNA-free two-gene knockout in Chlamydomonas reinhardtii via CRISPR-Cas9 ribonucleoproteins. Sci Rep 2016; 6:30620. [PMID: 27466170 PMCID: PMC4964356 DOI: 10.1038/srep30620] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 07/06/2016] [Indexed: 12/14/2022] Open
Abstract
Microalgae are versatile organisms capable of converting CO2, H2O, and sunlight into fuel and chemicals for domestic and industrial consumption. Thus, genetic modifications of microalgae for enhancing photosynthetic productivity, and biomass and bio-products generation are crucial for both academic and industrial applications. However, targeted mutagenesis in microalgae with CRISPR-Cas9 is limited. Here we report, a one-step transformation of Chlamydomonas reinhardtii by the DNA-free CRISPR-Cas9 method rather than plasmids that encode Cas9 and guide RNAs. Outcome was the sequential CpFTSY and ZEP two-gene knockout and the generation of a strain constitutively producing zeaxanthin and showing improved photosynthetic productivity.
Collapse
Affiliation(s)
- Kwangryul Baek
- Department of Life Science, Hanyang University, Seoul, South Korea
| | - Duk Hyoung Kim
- Department of Chemistry, Hanyang University, Seoul, South Korea
| | - Jooyeon Jeong
- Department of Life Science, Hanyang University, Seoul, South Korea
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, Seoul, South Korea
| | - Anastasios Melis
- Department of Plant and Microbial Biology, University of California, Berkeley, CA 94720-3102, USA
| | - Jin-Soo Kim
- Center for Genome Engineering, Institute for Basic Science, Seoul, South Korea.,Department of Chemistry, Seoul National University, Seoul, South Korea
| | - EonSeon Jin
- Department of Life Science, Hanyang University, Seoul, South Korea
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul, South Korea
| |
Collapse
|
72
|
Subburaj S, Chung SJ, Lee C, Ryu SM, Kim DH, Kim JS, Bae S, Lee GJ. Site-directed mutagenesis in Petunia × hybrida protoplast system using direct delivery of purified recombinant Cas9 ribonucleoproteins. PLANT CELL REPORTS 2016; 35:1535-44. [PMID: 26825596 DOI: 10.1007/s00299-016-1937-7] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/15/2016] [Indexed: 05/21/2023]
Abstract
Site-directed mutagenesis of nitrate reductase genes using direct delivery of purified Cas9 protein preassembled with guide RNA produces mutations efficiently in Petunia × hybrida protoplast system. The clustered, regularly interspaced, short palindromic repeat (CRISPR)-CRISPR associated endonuclease 9 (CRISPR/Cas9) system has been recently announced as a powerful molecular breeding tool for site-directed mutagenesis in higher plants. Here, we report a site-directed mutagenesis method targeting Petunia nitrate reductase (NR) gene locus. This method could create mutations efficiently using direct delivery of purified Cas9 protein and single guide RNA (sgRNA) into protoplast cells. After transient introduction of RNA-guided endonuclease (RGEN) ribonucleoproteins (RNPs) with different sgRNAs targeting NR genes, mutagenesis at the targeted loci was detected by T7E1 assay and confirmed by targeted deep sequencing. T7E1 assay showed that RGEN RNPs induced site-specific mutations at frequencies ranging from 2.4 to 21 % at four different sites (NR1, 2, 4 and 6) in the PhNR gene locus with average mutation efficiency of 14.9 ± 2.2 %. Targeted deep DNA sequencing revealed mutation rates of 5.3-17.8 % with average mutation rate of 11.5 ± 2 % at the same NR gene target sites in DNA fragments of analyzed protoplast transfectants. Further analysis from targeted deep sequencing showed that the average ratio of deletion to insertion produced collectively by the four NR-RGEN target sites (NR1, 2, 4, and 6) was about 63:37. Our results demonstrated that direct delivery of RGEN RNPs into protoplast cells of Petunia can be exploited as an efficient tool for site-directed mutagenesis of genes or genome editing in plant systems.
Collapse
Affiliation(s)
- Saminathan Subburaj
- Department of Horticultural Science, Chungnam National University, Daejeon, 305-764, South Korea
| | - Sung Jin Chung
- Department of Horticultural Science, Chungnam National University, Daejeon, 305-764, South Korea
| | - Choongil Lee
- Department of Chemistry, Seoul National University, Seoul, 151-742, South Korea
- Center for Genome Engineering, Institute for Basic Science, Seoul, 151-742, South Korea
| | - Seuk-Min Ryu
- Department of Chemistry, Seoul National University, Seoul, 151-742, South Korea
- Center for Genome Engineering, Institute for Basic Science, Seoul, 151-742, South Korea
| | - Duk Hyoung Kim
- Department of Chemistry, Hanyang University, Seoul, 133-791, South Korea
| | - Jin-Soo Kim
- Department of Chemistry, Seoul National University, Seoul, 151-742, South Korea
- Center for Genome Engineering, Institute for Basic Science, Seoul, 151-742, South Korea
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul, 133-791, South Korea.
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul, 133-791, South Korea.
| | - Geung-Joo Lee
- Department of Horticultural Science, Chungnam National University, Daejeon, 305-764, South Korea.
| |
Collapse
|
73
|
Abstract
“Big Data” has surpassed “systems biology” and “omics” as the hottest buzzword in the biological sciences, but is there any substance behind the hype? Certainly, we have learned about various aspects of cell and molecular biology from the many individual high-throughput data sets that have been published in the past 15–20 years. These data, although useful as individual data sets, can provide much more knowledge when interrogated with Big Data approaches, such as applying integrative methods that leverage the heterogeneous data compendia in their entirety. Here we discuss the benefits and challenges of such Big Data approaches in biology and how cell and molecular biologists can best take advantage of them.
Collapse
Affiliation(s)
- Kara Dolinski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540
| | - Olga G Troyanskaya
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08540 Department of Computer Science, Princeton University, Princeton, NJ 08540 Simons Center for Data Analysis, Simons Foundation, New York, NY 10010
| |
Collapse
|
74
|
Chasman D, Fotuhi Siahpirani A, Roy S. Network-based approaches for analysis of complex biological systems. Curr Opin Biotechnol 2016; 39:157-166. [PMID: 27115495 DOI: 10.1016/j.copbio.2016.04.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 04/04/2016] [Accepted: 04/05/2016] [Indexed: 12/22/2022]
Abstract
Cells function and respond to changes in their environment by the coordinated activity of their molecular components, including mRNAs, proteins and metabolites. At the heart of proper cellular function are molecular networks connecting these components to process extra-cellular environmental signals and drive dynamic, context-specific cellular responses. Network-based computational approaches aim to systematically integrate measurements from high-throughput experiments to gain a global understanding of cellular function under changing environmental conditions. We provide an overview of recent methodological developments toward solving two major computational problems within this field in the past two years (2013-2015): network reconstruction and network-based interpretation. Looking forward, we envision development of methods that can predict phenotypes with high accuracy as well as provide biologically plausible mechanistic hypotheses.
Collapse
Affiliation(s)
- Deborah Chasman
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Alireza Fotuhi Siahpirani
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI 53706, United States; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, United States; Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53792, United States
| | - Sushmita Roy
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, United States; Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53792, United States; Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI 53706, United States.
| |
Collapse
|
75
|
Vojta A, Samaržija I, Bočkor L, Zoldoš V. Glyco-genes change expression in cancer through aberrant methylation. Biochim Biophys Acta Gen Subj 2016; 1860:1776-85. [PMID: 26794090 DOI: 10.1016/j.bbagen.2016.01.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 01/08/2016] [Accepted: 01/09/2016] [Indexed: 01/25/2023]
Abstract
BACKGROUND Most eukaryotic proteins are modified by covalent addition of glycan molecules that considerably influence their function. Aberrant glycosylation is profoundly involved in malignant transformation, tumor progression and metastasis. Some glycan structures are tumor-specific and reflect disturbed glycan biosynthesis pathways. METHODS We analyzed DNA methylation and expression of 86 glyco-genes in melanoma, hepatocellular, breast and cervical cancers using data from publicly available databases. We also analyzed methylation datasets without the available matching expression data for glyco-genes in lung cancer, and progression of melanoma into lymph node and brain metastases. RESULTS Ten glyco-genes (GALNT3, GALNT6, GALNT7, GALNT14, MGAT3, MAN1A1, MAN1C1, ST3GAL2, ST6GAL1, ST8SIA3) showing changes in both methylation and expression in the same type of cancer belong to GalNAc transferases, GlcNAc transferases, mannosidases and sialyltransferases, which is in line with changes in glycan structures already reported in the same type of tumors. Some of those genes were additionally identified as potentially valuable for disease prognosis. The MGAT5B gene, so far identified as specifically expressed in brain, emerged as a novel candidate gene that is epigenetically dysregulated in different cancers other than brain cancer. We also report for the first time aberrant expression of the GALNT and MAN genes in cancer by aberrant promoter methylation. CONCLUSIONS Aberrant expression of glyco-genes due to aberrant promoter methylation could be a way leading to characteristic glycosylation profiles commonly described in cancer. GENERAL SIGNIFICANCE Methylation status in promoters of candidate glyco-genes might serve as prognostic markers for specific tumors and point to potential novel targets for epigenetic drugs. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.
Collapse
Affiliation(s)
- Aleksandar Vojta
- University of Zagreb Faculty of Science, Department of Biology, Division of Molecular Biology, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Ivana Samaržija
- University of Zagreb Faculty of Science, Department of Biology, Division of Molecular Biology, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Luka Bočkor
- University of Zagreb Faculty of Science, Department of Biology, Division of Molecular Biology, Horvatovac 102a, HR-10000 Zagreb, Croatia
| | - Vlatka Zoldoš
- University of Zagreb Faculty of Science, Department of Biology, Division of Molecular Biology, Horvatovac 102a, HR-10000 Zagreb, Croatia.
| |
Collapse
|
76
|
Gorenshteyn D, Zaslavsky E, Fribourg M, Park CY, Wong AK, Tadych A, Hartmann BM, Albrecht RA, García-Sastre A, Kleinstein SH, Troyanskaya OG, Sealfon SC. Interactive Big Data Resource to Elucidate Human Immune Pathways and Diseases. Immunity 2015; 43:605-14. [PMID: 26362267 DOI: 10.1016/j.immuni.2015.08.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 04/24/2015] [Accepted: 06/25/2015] [Indexed: 12/21/2022]
Abstract
Many functionally important interactions between genes and proteins involved in immunological diseases and processes are unknown. The exponential growth in public high-throughput data offers an opportunity to expand this knowledge. To unlock human-immunology-relevant insight contained in the global biomedical research effort, including all public high-throughput datasets, we performed immunological-pathway-focused Bayesian integration of a comprehensive, heterogeneous compendium comprising 38,088 genome-scale experiments. The distillation of this knowledge into immunological networks of functional relationships between molecular entities (ImmuNet), and tools to mine this resource, are accessible to the public at http://immunet.princeton.edu. The predictive capacity of ImmuNet, established by rigorous statistical validation, is easily accessed by experimentalists to generate data-driven hypotheses. We demonstrate the power of this approach through the identification of unique host-virus interaction responses, and we show how ImmuNet complements genetic studies by predicting disease-associated genes. ImmuNet should be widely beneficial for investigating the mechanisms of the human immune system and immunological diseases.
Collapse
Affiliation(s)
- Dmitriy Gorenshteyn
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Elena Zaslavsky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miguel Fribourg
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher Y Park
- New York Genome Center, 101 Avenue of the Americas, New York, NY 10013, USA
| | - Aaron K Wong
- Simons Center for Data Analysis, Simons Foundation, New York, NY 10010, USA
| | - Alicja Tadych
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Boris M Hartmann
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Randy A Albrecht
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Steven H Kleinstein
- Departments of Pathology and Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06511, USA
| | - Olga G Troyanskaya
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Simons Center for Data Analysis, Simons Foundation, New York, NY 10010, USA; Department of Computer Science, Princeton University, Princeton, NJ 08540, USA.
| | - Stuart C Sealfon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|