1
|
Chen Y, Jiang Y, Sarvanantharajah N, Apirakkan O, Yang M, Milcova A, Topinka J, Abbate V, Arlt VM, Stürzenbaum SR. Genome-modified Caenorhabditis elegans expressing the human cytochrome P450 (CYP1A1 and CYP1A2) pathway: An experimental model for environmental carcinogenesis and pharmacological research. ENVIRONMENT INTERNATIONAL 2024; 194:109187. [PMID: 39671827 DOI: 10.1016/j.envint.2024.109187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs), including the Group 1 human carcinogen benzo[a]pyrene (BaP), are produced by the incomplete combustion of organic matter and thus are present in tobacco smoke, charbroiled food and diesel exhaust. The nematode Caenorhabditis elegans is an established model organism, however it lacks the genetic components of the classical mammalian cytochrome P450 (CYP)-mediated BaP-diol-epoxide metabolism pathway. We therefore introduced human CYP1A1 or CYP1A2 together with human epoxide hydrolase (EPHX) into the worm genome by Mos1-mediated Single Copy Insertion (MosSCI) and evaluated their response to BaP exposure via toxicological endpoints. Compared to wild-type control, CYP-humanised worms were characterised by an increase in pharyngeal pumping rate and a decrease in volumetric surface area. Furthermore, BaP exposure reduced reproductive performance, as reflected in smaller brood size, which coincided with the downregulation of the nematode-specific major sperm protein as determined by transcriptomics (RNAseq). BaP-mediated reproductive toxicity was exacerbated in CYP-humanised worms at higher exposure levels. Collagen-related genes were downregulated in BaP-exposed animals, which correlate with the reduction in volumetric size. Whole genome DNA sequencing revealed a higher frequency of T > G (A > C) base substitution mutations in worms expressing human CYP1A1;EPHX which aligned with an increase in DNA adducts identified via an ELISA method (but not classical 32P-postlabelling). Overall, the CYP-humanised worms provided new insights into the value of genome-optimised invertebrate models by identifying the benefits and limitations within the context of the (3Rs) concept which aims to replace, reduce and refine the use of animals in research.
Collapse
Affiliation(s)
- Yuzhi Chen
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Yang Jiang
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Utrecht, Netherlands
| | - Nirujah Sarvanantharajah
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Orapan Apirakkan
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Mengqi Yang
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Alena Milcova
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
| | - Jan Topinka
- Department of Toxicology and Molecular Epidemiology, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic
| | - Vincenzo Abbate
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK; Toxicology Department, GAB Consulting GmbH, 69126 Heidelberg, Germany
| | - Stephen R Stürzenbaum
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
2
|
Hatanaka R, Tamagawa K, Haruta N, Sugimoto A. The impact of differential transposition activities of autonomous and nonautonomous hAT transposable elements on genome architecture and gene expression in Caenorhabditis inopinata. Genetics 2024; 227:iyae052. [PMID: 38577765 PMCID: PMC11492494 DOI: 10.1093/genetics/iyae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/08/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
Transposable elements are DNA sequences capable of moving within genomes and significantly influence genomic evolution. The nematode Caenorhabditis inopinata exhibits a much higher transposable element copy number than its sister species, Caenorhabditis elegans. In this study, we identified a novel autonomous transposable element belonging to the hAT superfamily from a spontaneous transposable element-insertion mutant in C. inopinata and named this transposon Ci-hAT1. Further bioinformatic analyses uncovered 3 additional autonomous hAT elements-Ci-hAT2, Ci-hAT3, and Ci-hAT4-along with over 1,000 copies of 2 nonautonomous miniature inverted-repeat transposable elements, mCi-hAT1 and mCi-hAT4, likely derived from Ci-hAT1 and Ci-hAT4 through internal deletion. We tracked at least 3 sequential transpositions of Ci-hAT1 over several years. However, the transposition rates of the other 3 autonomous hAT elements were lower, suggesting varying activity levels. Notably, the distribution patterns of the 2 miniature inverted-repeat transposable element families differed significantly: mCi-hAT1 was primarily located in the chromosome arms, a pattern observed in the transposable elements of other Caenorhabditis species, whereas mCi-hAT4 was more evenly distributed across chromosomes. Additionally, interspecific transcriptome analysis indicated that C. inopinata genes with upstream or intronic these miniature inverted-repeat transposable element insertions tend to be more highly expressed than their orthologous genes in C. elegans. These findings highlight the significant role of de-silenced transposable elements in driving the evolution of genomes and transcriptomes, leading to species-specific genetic diversity.
Collapse
Affiliation(s)
- Ryuhei Hatanaka
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Katsunori Tamagawa
- Laboratory of Evolutionary Genomics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Nami Haruta
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Asako Sugimoto
- Laboratory of Developmental Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| |
Collapse
|
3
|
Peng D, Jackson D, Palicha B, Kernfeld E, Laughner N, Shoemaker A, Celniker SE, Loganathan R, Cahan P, Andrew DJ. Organogenetic transcriptomes of the Drosophila embryo at single cell resolution. Development 2024; 151:dev202097. [PMID: 38174902 PMCID: PMC10820837 DOI: 10.1242/dev.202097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
To gain insight into the transcription programs activated during the formation of Drosophila larval structures, we carried out single cell RNA sequencing during two periods of Drosophila embryogenesis: stages 10-12, when most organs are first specified and initiate morphological and physiological specialization; and stages 13-16, when organs achieve their final mature architectures and begin to function. Our data confirm previous findings with regards to functional specialization of some organs - the salivary gland and trachea - and clarify the embryonic functions of another - the plasmatocytes. We also identify two early developmental trajectories in germ cells and uncover a potential role for proteolysis during germline stem cell specialization. We identify the likely cell type of origin for key components of the Drosophila matrisome and several commonly used Drosophila embryonic cell culture lines. Finally, we compare our findings with other recent related studies and with other modalities for identifying tissue-specific gene expression patterns. These data provide a useful community resource for identifying many new players in tissue-specific morphogenesis and functional specialization of developing organs.
Collapse
Affiliation(s)
- Da Peng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dorian Jackson
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bianca Palicha
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Eric Kernfeld
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nathaniel Laughner
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ashleigh Shoemaker
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Susan E. Celniker
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Rajprasad Loganathan
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Deborah J. Andrew
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
4
|
Wang W, Flury AG, Rodriguez AT, Garrison JL, Brem RB. A role for worm cutl-24 in background- and parent-of-origin-dependent ER stress resistance. BMC Genomics 2022; 23:842. [PMID: 36539699 PMCID: PMC9764823 DOI: 10.1186/s12864-022-09063-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Organisms in the wild can acquire disease- and stress-resistance traits that outstrip the programs endogenous to humans. Finding the molecular basis of such natural resistance characters is a key goal of evolutionary genetics. Standard statistical-genetic methods toward this end can perform poorly in organismal systems that lack high rates of meiotic recombination, like Caenorhabditis worms. RESULTS Here we discovered unique ER stress resistance in a wild Kenyan C. elegans isolate, which in inter-strain crosses was passed by hermaphrodite mothers to hybrid offspring. We developed an unbiased version of the reciprocal hemizygosity test, RH-seq, to explore the genetics of this parent-of-origin-dependent phenotype. Among top-scoring gene candidates from a partial-coverage RH-seq screen, we focused on the neuronally-expressed, cuticlin-like gene cutl-24 for validation. In gene-disruption and controlled crossing experiments, we found that cutl-24 was required in Kenyan hermaphrodite mothers for ER stress tolerance in their inter-strain hybrid offspring; cutl-24 was also a contributor to the trait in purebred backgrounds. CONCLUSIONS These data establish the Kenyan strain allele of cutl-24 as a determinant of a natural stress-resistant state, and they set a precedent for the dissection of natural trait diversity in invertebrate animals without the need for a panel of meiotic recombinants.
Collapse
Affiliation(s)
- Wenke Wang
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Plant and Microbial Biology, UC Berkeley, Berkeley, CA, United States
| | - Anna G Flury
- Buck Institute for Research on Aging, Novato, CA, United States
- Department of Plant and Microbial Biology, UC Berkeley, Berkeley, CA, United States
| | - Andrew T Rodriguez
- Buck Institute for Research on Aging, Novato, CA, United States
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Jennifer L Garrison
- Buck Institute for Research on Aging, Novato, CA, United States.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States.
- Department of Cellular and Molecular Pharmacology, UC San Francisco, San Francisco, CA, United States.
- Global Consortium for Reproductive Longevity & Equality, Novato, CA, United States.
| | - Rachel B Brem
- Buck Institute for Research on Aging, Novato, CA, United States.
- Department of Plant and Microbial Biology, UC Berkeley, Berkeley, CA, United States.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
5
|
El Mouridi S, Alkhaldi F, Frøkjær-Jensen C. Modular safe-harbor transgene insertion for targeted single-copy and extrachromosomal array integration in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2022; 12:jkac184. [PMID: 35900171 PMCID: PMC9434227 DOI: 10.1093/g3journal/jkac184] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/29/2022] [Indexed: 12/02/2022]
Abstract
Efficient and reproducible transgenesis facilitates and accelerates research using genetic model organisms. Here, we describe a modular safe-harbor transgene insertion (MosTI) for use in Caenorhabditis elegans which improves targeted insertion of single-copy transgenes by homology directed repair and targeted integration of extrachromosomal arrays by nonhomologous end-joining. MosTI allows easy conversion between selection markers at insertion site and a collection of universal targeting vectors with commonly used promoters and fluorophores. Insertions are targeted at three permissive safe-harbor intergenic locations and transgenes are reproducibly expressed in somatic and germ cells. Chromosomal integration is mediated by CRISPR/Cas9, and positive selection is based on a set of split markers (unc-119, hygroR, and gfp) where only animals with chromosomal insertions are rescued, resistant to antibiotics, or fluorescent, respectively. Single-copy insertion is efficient using either constitutive or heat-shock inducible Cas9 expression (25-75%) and insertions can be generated from a multiplexed injection mix. Extrachromosomal array integration is also efficient (7-44%) at modular safe-harbor transgene insertion landing sites or at the endogenous unc-119 locus. We use short-read sequencing to estimate the plasmid copy numbers for 8 integrated arrays (6-37 copies) and long-read Nanopore sequencing to determine the structure and size (5.4 Mb) of 1 array. Using universal targeting vectors, standardized insertion strains, and optimized protocols, it is possible to construct complex transgenic strains which should facilitate the study of increasingly complex biological problems in C. elegans.
Collapse
Affiliation(s)
- Sonia El Mouridi
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Faisal Alkhaldi
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Christian Frøkjær-Jensen
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
6
|
Jiang L, Li R, Han Z, Zhao X, Cao D, Ow DW. Target lines for recombinase-mediated gene stacking in soybean. TAG. THEORETICAL AND APPLIED GENETICS. THEORETISCHE UND ANGEWANDTE GENETIK 2022; 135:1163-1175. [PMID: 35325256 DOI: 10.1007/s00122-021-04015-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/06/2021] [Indexed: 05/10/2023]
Abstract
KEY MESSAGE Five soybean target lines with recombinase sites at suitable genomic positions were obtained and tested for site-specific gene stacking. For introgression of new transgenic traits to field cultivars, adding new DNA to an existing transgene locus would reduce the number of segregating loci to reassemble back into a breeding line. We described previously an in planta transgene stacking system using the Bxb1 integrase to direct new DNA into a genomic target, but for this system to operate, the target locus must have a preexisting recombination site for Bxb1-mediated integration. Here, we describe 5 soybean target lines from the screening of 118 Agrobacterium-mediated transgenic plants that were positive for gus expression. Each of the 5 target lines has a single copy of the transgenic DNA with precise DNA sequences of the recombinase recognition sites, located at least 1 kb away from the nearest coding region, not close to the centromere, and showed good expression of the reporter gene. We tested Bxb1 integrase-mediated integration of a gfp-containing plasmid into each of these lines and showed precise site-specific integration in bombarded calluses. For plant regeneration, we used embryonic axes of mature soybean seeds to conduct a new set of biolistic transformation with a DsRed-containing plasmid. Three integration events were regenerated into whole plants, demonstrating the principle that target lines can serve as foundation lines for the stacking of DNA to predefined locations in the soybean genome.
Collapse
Affiliation(s)
- Li Jiang
- Plant Gene Engineering Center; Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Guangdong Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, China
- University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China
| | - Ruyu Li
- Plant Gene Engineering Center; Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Guangdong Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, China.
| | - Zhiguo Han
- Plant Gene Engineering Center; Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Guangdong Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, China
| | - Xiaohui Zhao
- The Innovative Academy of Seed Design, Key Laboratory of Soybean Molecular Design Breeding, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Harbin, 150081, China
| | - Dong Cao
- The Innovative Academy of Seed Design, Key Laboratory of Soybean Molecular Design Breeding, Northeast Institute of Geography and Agroecology, Chinese Academy of Sciences, Harbin, 150081, China
| | - David W Ow
- Plant Gene Engineering Center; Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Guangdong Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, 510650, China.
| |
Collapse
|
7
|
Li Y, Li R, Han Z, Wang H, Zhou S, Li Y, Wang Y, Qi J, Ow DW. Recombinase-mediated gene stacking in cotton. PLANT PHYSIOLOGY 2022; 188:1852-1865. [PMID: 35088863 PMCID: PMC8968315 DOI: 10.1093/plphys/kiac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/08/2021] [Indexed: 05/24/2023]
Abstract
Site-specific gene stacking could reduce the number of segregating loci and expedite the introgression of transgenes from experimental lines to field lines. Recombinase-mediated site-specific gene stacking provides a flexible and efficient solution, but this approach requires a recombinase recognition site in the genome. Here, we describe several cotton (Gossypium hirsutum cv. Coker 312) target lines suitable for Mycobacteriophage Bxb1 recombinase-mediated gene stacking. Obtained through the empirical screening of random insertion events, each of these target lines contains a single intact copy of the target construct with precise sequences of RS2, lox, and attP sites that is not inserted within or close to a known gene or near a centromere and shows good expression of the reporter gene gfp. Gene stacking was tested with insertion of different combinations of three candidate genes for resistance to verticillium wilt into three cotton target lines: CTS1, CTS3, and CTS4. Nine site-specific integration events were recovered from 95 independently transformed embryogenic calluses. Southern and DNA sequence analyses of regenerated plants confirmed precise site-specific integration, and resistance to verticillium wilt was observed for plant CTS1i3, which has a single precise copy of site-specifically integrated DNA. These cotton target lines can serve as foundation lines for recombinase-mediated gene stacking to facilitate precise DNA integration and introgression to field cultivars.
Collapse
Affiliation(s)
- Yamei Li
- Plant Gene Engineering Center, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruyu Li
- Plant Gene Engineering Center, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Zhiguo Han
- Plant Gene Engineering Center, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Haitang Wang
- Plant Gene Engineering Center, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Sixian Zhou
- Plant Gene Engineering Center, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yongqing Li
- Plant Gene Engineering Center, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Yumei Wang
- Plant Gene Engineering Center, Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junsheng Qi
- Department of Plant Science, State Key Laboratory of Plant Physiology and Biochemistry, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | | |
Collapse
|
8
|
Quinzo MJ, Perteguer MJ, Brindley PJ, Loukas A, Sotillo J. Transgenesis in parasitic helminths: a brief history and prospects for the future. Parasit Vectors 2022; 15:110. [PMID: 35346328 PMCID: PMC8962113 DOI: 10.1186/s13071-022-05211-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/18/2022] [Indexed: 12/15/2022] Open
Abstract
Helminth infections impact the health of hundreds of millions of persons globally and also cause important economic losses in livestock farming. Methodological limitations as well as the low attention given to the study of helminths have impacted biological research and, thus, the procurement of accurate diagnosis and effective treatments. Understanding the biology of helminths using genomic and proteomic approaches could contribute to advances in understanding host-helminth interactions and lead to new vaccines, drugs and diagnostics. Despite the significant advances in genomics in the last decade, the lack of methodological adaptation of current transgenesis techniques has hampered the progression of post-genomic research in helminthology. However, the application of new techniques, such as CRISPR, to the study of trematodes and nematodes has opened new avenues for genome editing-powered functional genomics for these pathogens. This review summarises the historical advances in functional genomics in parasitic helminths and highlights pending limitations that will need to be overcome to deploy transgenesis tools.
Collapse
Affiliation(s)
- M J Quinzo
- Parasitology Reference and Research Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
- Escuela Internacional de Doctorado, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - M J Perteguer
- Parasitology Reference and Research Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - P J Brindley
- Department of Microbiology, Immunology and Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine and Health Sciences, George Washington University, Washington, DC, 20037, USA
| | - A Loukas
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - J Sotillo
- Parasitology Reference and Research Laboratory, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain.
| |
Collapse
|
9
|
El Mouridi S, Frøkjær-Jensen C. Targeted and Random Transposon-Assisted Single-Copy Transgene Insertion in C. elegans. Methods Mol Biol 2022; 2468:239-256. [PMID: 35320568 DOI: 10.1007/978-1-0716-2181-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Transgenesis in model organisms is an essential tool for determining the function of protein-coding genes and non-coding regulatory regions. In Caenorhabditis elegans, injected DNA can be propagated as multicopy extra-chromosomal arrays, but transgenes in arrays are frequently mosaic, over-expressed in some tissues, and silenced in the germline. Here, we describe methods to insert single-copy transgenes into specific genomic locations (MosSCI) or random locations (miniMos) using Mos1 transposons. Single-copy insertions allow expression at endogenous levels, expression in the germline, and identification of active and repressed regions of the genome.
Collapse
Affiliation(s)
- Sonia El Mouridi
- Biological and Environmental Science and Engineering Division (BESE), KAUST Environmental Epigenetics Program (KEEP), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Christian Frøkjær-Jensen
- Biological and Environmental Science and Engineering Division (BESE), KAUST Environmental Epigenetics Program (KEEP), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.
| |
Collapse
|
10
|
Powell G, Pavlovic Djuranovic S, Djuranovic S. Gene dosage effects of poly(A) track-engineered hypomorphs. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 26:865-878. [PMID: 34729253 PMCID: PMC8536507 DOI: 10.1016/j.omtn.2021.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/29/2021] [Accepted: 10/01/2021] [Indexed: 11/18/2022]
Abstract
Manipulation of gene activity through creation of hypomorphic mutants has been a long-standing tool in examining gene function. Our previous studies have indicated that hypomorphic mutants could be created by inserting cis-regulatory sequences composed of consecutive adenosine nucleotides called poly(A) tracks. Here we use poly(A) tracks to create hypomorphic mutants and functional characterization of membrane, secretory, and endogenous proteins. Insertion of poly(A) tracks into the sequences of interleukin-2 and membrane protein CD20 results in a programmable reduction of mRNA stability and attenuation of protein expression regardless of the presence of a signaling sequence. Likewise, CRISPR-Cas9 targeted insertion of poly(A) tracks into the coding sequence of the endogenous human genes AUF1 and TP53 results in a programmable reduction of targeted protein and mRNA levels. Functional analyses of AUF1-engineered hypomorphs indicate a direct correlation between AUF1 gene levels and the stability of AUF1-regulated mRNAs. Hypomorphs of TP53 affect expression of the target genes differentially depending on the severity of the hypomorphic mutation. Finally, decreases in TP53 protein affect the same cellular pathways in poly(A) track-engineered cells as in cancer cells, indicating these variants’ biological relevance. These results highlight this technology’s power to create predictable, stable hypomorphs in recombinant or endogenous genes in combination with CRISPR-Cas9 engineering tools.
Collapse
Affiliation(s)
- Geralle Powell
- Department of Cell Biology and Physiology, Washington University School of Medicine, 600 South Euclid Avenue, Campus Box 8228, St. Louis, MO 63110, USA
| | - Slavica Pavlovic Djuranovic
- Department of Cell Biology and Physiology, Washington University School of Medicine, 600 South Euclid Avenue, Campus Box 8228, St. Louis, MO 63110, USA
| | - Sergej Djuranovic
- Department of Cell Biology and Physiology, Washington University School of Medicine, 600 South Euclid Avenue, Campus Box 8228, St. Louis, MO 63110, USA
- Correspondence: Sergej Djuranovic, Department of Cell Biology and Physiology, Washington University School of Medicine, 600 South Euclid Avenue, Campus Box 8228, St. Louis, MO 63110, USA.
| |
Collapse
|
11
|
Campos TL, Korhonen PK, Hofmann A, Gasser RB, Young ND. Harnessing model organism genomics to underpin the machine learning-based prediction of essential genes in eukaryotes - Biotechnological implications. Biotechnol Adv 2021; 54:107822. [PMID: 34461202 DOI: 10.1016/j.biotechadv.2021.107822] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/17/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022]
Abstract
The availability of high-quality genomes and advances in functional genomics have enabled large-scale studies of essential genes in model eukaryotes, including the 'elegant worm' (Caenorhabditis elegans; Nematoda) and the 'vinegar fly' (Drosophila melanogaster; Arthropoda). However, this is not the case for other, much less-studied organisms, such as socioeconomically important parasites, for which functional genomic platforms usually do not exist. Thus, there is a need to develop innovative techniques or approaches for the prediction, identification and investigation of essential genes. A key approach that could enable the prediction of such genes is machine learning (ML). Here, we undertake an historical review of experimental and computational approaches employed for the characterisation of essential genes in eukaryotes, with a particular focus on model ecdysozoans (C. elegans and D. melanogaster), and discuss the possible applicability of ML-approaches to organisms such as socioeconomically important parasites. We highlight some recent results showing that high-performance ML, combined with feature engineering, allows a reliable prediction of essential genes from extensive, publicly available 'omic data sets, with major potential to prioritise such genes (with statistical confidence) for subsequent functional genomic validation. These findings could 'open the door' to fundamental and applied research areas. Evidence of some commonality in the essential gene-complement between these two organisms indicates that an ML-engineering approach could find broader applicability to ecdysozoans such as parasitic nematodes or arthropods, provided that suitably large and informative data sets become/are available for proper feature engineering, and for the robust training and validation of algorithms. This area warrants detailed exploration to, for example, facilitate the identification and characterisation of essential molecules as novel targets for drugs and vaccines against parasitic diseases. This focus is particularly important, given the substantial impact that such diseases have worldwide, and the current challenges associated with their prevention and control and with drug resistance in parasite populations.
Collapse
Affiliation(s)
- Tulio L Campos
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia; Bioinformatics Core Facility, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz (IAM-Fiocruz), Recife, Pernambuco, Brazil
| | - Pasi K Korhonen
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Hofmann
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
12
|
Ustyantsev K, Wudarski J, Sukhikh I, Reinoite F, Mouton S, Berezikov E. Proof of principle for piggyBac-mediated transgenesis in the flatworm Macrostomum lignano. Genetics 2021; 218:6276877. [PMID: 33999134 PMCID: PMC8717057 DOI: 10.1093/genetics/iyab076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/06/2021] [Indexed: 12/03/2022] Open
Abstract
Regeneration-capable flatworms are informative research models to study the mechanisms of stem cell regulation, regeneration, and tissue patterning. The free-living flatworm Macrostomum lignano is currently the only flatworm where stable transgenesis is available, and as such it offers a powerful experimental platform to address questions that were previously difficult to answer. The published transgenesis approach relies on random integration of DNA constructs into the genome. Despite its efficiency, there is room and need for further improvement and diversification of transgenesis methods in M. lignano. Transposon-mediated transgenesis is an alternative approach, enabling easy mapping of the integration sites and the possibility of insertional mutagenesis studies. Here, we report for the first time that transposon-mediated transgenesis using piggyBac can be performed in M. lignano to create stable transgenic lines with single-copy transgene insertions.
Collapse
Affiliation(s)
- Kirill Ustyantsev
- Institute of Cytology and Genetics SB RAS, Novosibirsk, 630090, Russia
| | - Jakub Wudarski
- Laboratory of Biological Diversity, National Institute for Basic Biology, Okazaki 444-8585 Aichi, Japan
| | - Igor Sukhikh
- Institute of Cytology and Genetics SB RAS, Novosibirsk, 630090, Russia
| | - Filipa Reinoite
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, 9700AD, The Netherlands
| | - Stijn Mouton
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, 9700AD, The Netherlands
| | - Eugene Berezikov
- Institute of Cytology and Genetics SB RAS, Novosibirsk, 630090, Russia.,European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, 9700AD, The Netherlands
| |
Collapse
|
13
|
Abstract
Developments in genome editing offer potential solutions to challenges in agriculture, industry, medicine, and the environment. However, many technologies remain unexploited due to limitations in the use of genetically altered organisms. In this study, we use B. subtilis spores to explore the possibility of bioengineering organisms while leaving their genome intact. Taking advantage of the differential expression between the mother cell and the fore-spore compartments during sporulation, we created plasmids programmed to modify the spore phenotype from the mother cell compartment, but to "self-digest" in the fore-spore. At the end of sporulation, the mother cell undergoes lysis and releases the phenotypically engineered, genetically unaltered spores. Using this approach, we demonstrated the potential to express foreign proteins in B. subtilis spores without genome alterations by producing spores expressing GFP in their protective coats, where approximately 90% of the spore population had no detectable plasmid or chromosome alterations. In a separate demonstration, we programmed KinA overexpression during vegetative growth to artificially induce sporulation, and also obtained spores with nearly 90% of them free of detectable plasmid. Artificial induction of sporulation could potentially simplify the bioprocess for industrial spore production, as it reduces the number of steps involved. Overall, these findings demonstrate the potential to create genetically intact bioengineered organisms.
Collapse
Affiliation(s)
- Juan F. Quijano
- Department of Biological Sciences, Columbia University, New York, 10027, United States
- Department of Biological Sciences and Department of Physics, Columbia University, New York, 10027, United States
| | - Ozgur Sahin
- Department of Biological Sciences, Columbia University, New York, 10027, United States
- Department of Biological Sciences and Department of Physics, Columbia University, New York, 10027, United States
| |
Collapse
|
14
|
Thurman M, Sun H, Kubica S, Praitis V. The slo-1 BK potassium channel interacts genetically with pmr-1 secretory pathway calcium ATPase during C. elegans embryonic cell migration. MICROPUBLICATION BIOLOGY 2021; 2021. [PMID: 33474530 PMCID: PMC7812384 DOI: 10.17912/micropub.biology.000351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
| | - Haonan Sun
- Grinnell College, Grinnell IA, 50112 USA
| | - Sam Kubica
- Grinnell College, Grinnell IA, 50112 USA
| | | |
Collapse
|
15
|
PIG-1 MELK-dependent phosphorylation of nonmuscle myosin II promotes apoptosis through CES-1 Snail partitioning. PLoS Genet 2020; 16:e1008912. [PMID: 32946434 PMCID: PMC7527206 DOI: 10.1371/journal.pgen.1008912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/30/2020] [Accepted: 07/29/2020] [Indexed: 01/06/2023] Open
Abstract
The mechanism(s) through which mammalian kinase MELK promotes tumorigenesis is not understood. We find that the C. elegans orthologue of MELK, PIG-1, promotes apoptosis by partitioning an anti-apoptotic factor. The C. elegans NSM neuroblast divides to produce a larger cell that differentiates into a neuron and a smaller cell that dies. We find that in this context, PIG-1 MELK is required for partitioning of CES-1 Snail, a transcriptional repressor of the pro-apoptotic gene egl-1 BH3-only. pig-1 MELK is controlled by both a ces-1 Snail- and par-4 LKB1-dependent pathway, and may act through phosphorylation and cortical enrichment of nonmuscle myosin II prior to neuroblast division. We propose that pig-1 MELK-induced local contractility of the actomyosin network plays a conserved role in the acquisition of the apoptotic fate. Our work also uncovers an auto-regulatory loop through which ces-1 Snail controls its own activity through the formation of a gradient of CES-1 Snail protein. Apoptosis is critical for the elimination of ‘unwanted’ cells. What distinguishes wanted from unwanted cells in developing animals is poorly understood. We report that in the C. elegans NSM neuroblast lineage, the level of CES-1, a Snail-family member and transcriptional repressor of the pro-apoptotic gene egl-1, contributes to this process. In addition, we demonstrate that C. elegans PIG-1, the orthologue of mammalian proto-oncoprotein MELK, plays a critical role in controlling CES-1Snail levels. Specifically, during NSM neuroblast division, PIG-1MELK controls partitioning of CES-1Snail into one but not the other daughter cell thereby promoting the making of one wanted and one unwanted cell. Furthermore, we present evidence that PIG-1MELK acts prior to NSM neuroblast division by locally activating the actomyosin network.
Collapse
|
16
|
Altendorfer E, Láscarez-Lagunas LI, Nadarajan S, Mathieson I, Colaiácovo MP. Crossover Position Drives Chromosome Remodeling for Accurate Meiotic Chromosome Segregation. Curr Biol 2020; 30:1329-1338.e7. [PMID: 32142707 PMCID: PMC7162695 DOI: 10.1016/j.cub.2020.01.079] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/16/2019] [Accepted: 01/28/2020] [Indexed: 10/24/2022]
Abstract
Interhomolog crossovers (COs) are a prerequisite for achieving accurate chromosome segregation during meiosis [1, 2]. COs are not randomly positioned, occurring at distinct genomic intervals during meiosis in all species examined [3-10]. The role of CO position as a major determinant of accurate chromosome segregation has not been previously directly analyzed in a metazoan. Here, we use spo-11 mutants, which lack endogenous DNA double-strand breaks (DSBs), to induce a single DSB by Mos1 transposon excision at defined chromosomal locations in the C. elegans germline and show that the position of the resulting CO directly affects the formation of distinct chromosome subdomains during meiotic chromosome remodeling. CO formation in the typically CO-deprived center region of autosomes leads to premature loss of sister chromatid cohesion and chromosome missegregation, whereas COs at an off-centered position, as in wild type, can result in normal remodeling and accurate segregation. Ionizing radiation (IR)-induced DSBs lead to the same outcomes, and modeling of IR dose-response reveals that the CO-unfavorable center region encompasses up to 6% of the total chromosome length. DSBs proximal to telomeres rarely form COs, likely because of formation of unstable recombination intermediates that cannot be sustained as chiasmata until late prophase. Our work supports a model in which regulation of CO position early in meiotic prophase is required for proper designation of chromosome subdomains and normal chromosome remodeling in late meiotic prophase I, resulting in accurate chromosome segregation and providing a mechanism to prevent aneuploid gamete formation.
Collapse
Affiliation(s)
- Elisabeth Altendorfer
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Laura I Láscarez-Lagunas
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Saravanapriah Nadarajan
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Iain Mathieson
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Monica P Colaiácovo
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
17
|
Wei J, Dong Z, Ow DW. Spontaneous reactivation of a site-specifically placed transgene independent of copy number or DNA methylation. JOURNAL OF EXPERIMENTAL BOTANY 2020; 71:1574-1584. [PMID: 31740977 DOI: 10.1093/jxb/erz514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 11/14/2019] [Indexed: 06/10/2023]
Abstract
As millions of seeds are produced from a breeding line, the long-term stability of transgene expression is vital for commercial-scale production of seeds with transgenic traits. Transgenes can be silenced by epigenetic mechanisms, but reactivation of expression can occur as a result of treatment with chromatin modification inhibitors such as 5-azacytidine, from stress such as heat or UV-B, or in mutants that have acquired a defect in gene silencing. Previously, we targeted a gfp reporter gene into the tobacco (Nicotiana tabacum) genome by site-specific recombination but still found some silenced lines among independent integration events. One such line also had a second random copy and both copies showed DNA hypermethylation. To test whether removing the second copy would reactivate gfp expression, two T1 plants were backcrossed to the wild type. Whereas the silenced status was maintained in the progenies from one backcross, spontaneous partial reactivation of gfp expression was found among progenies from a second backcross. However, this reactivation did not correlate with loss of the second random copy or with a significant change in the pattern or amount of DNA hypermethylation. This finding supports the suggestion that gene reactivation does not necessarily involve loss of DNA homology or methylation.
Collapse
Affiliation(s)
- Junjie Wei
- Plant Gene Engineering Center; Chinese Academy of Sciences Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement; Guangdong Key Laboratory of Applied Botany. South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhicheng Dong
- Plant Gene Engineering Center; Chinese Academy of Sciences Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement; Guangdong Key Laboratory of Applied Botany. South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - David W Ow
- Plant Gene Engineering Center; Chinese Academy of Sciences Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement; Guangdong Key Laboratory of Applied Botany. South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
18
|
Nance J, Frøkjær-Jensen C. The Caenorhabditis elegans Transgenic Toolbox. Genetics 2019; 212:959-990. [PMID: 31405997 PMCID: PMC6707460 DOI: 10.1534/genetics.119.301506] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/01/2019] [Indexed: 12/30/2022] Open
Abstract
The power of any genetic model organism is derived, in part, from the ease with which gene expression can be manipulated. The short generation time and invariant developmental lineage have made Caenorhabditis elegans very useful for understanding, e.g., developmental programs, basic cell biology, neurobiology, and aging. Over the last decade, the C. elegans transgenic toolbox has expanded considerably, with the addition of a variety of methods to control expression and modify genes with unprecedented resolution. Here, we provide a comprehensive overview of transgenic methods in C. elegans, with an emphasis on recent advances in transposon-mediated transgenesis, CRISPR/Cas9 gene editing, conditional gene and protein inactivation, and bipartite systems for temporal and spatial control of expression.
Collapse
Affiliation(s)
- Jeremy Nance
- Helen L. and Martin S. Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, Department of Cell Biology, New York University School of Medicine, New York 10016
| | - Christian Frøkjær-Jensen
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), KAUST Environmental Epigenetics Program (KEEP), Thuwal 23955-6900, Saudi Arabia
| |
Collapse
|
19
|
Quijano MR, Zach C, Miller FS, Lee AR, Imani AS, Künzler M, Freeman MF. Distinct Autocatalytic α-N-Methylating Precursors Expand the Borosin RiPP Family of Peptide Natural Products. J Am Chem Soc 2019; 141:9637-9644. [DOI: 10.1021/jacs.9b03690] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Marissa R. Quijano
- Department of Biochemistry, Molecular Biology, and Biophysics and BioTechnology Institute, University of Minnesota−Twin Cities, St. Paul, Minnesota 55108, United States
| | - Christina Zach
- Department of Biology, Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zürich, Switzerland
| | - Fredarla S. Miller
- Department of Biochemistry, Molecular Biology, and Biophysics and BioTechnology Institute, University of Minnesota−Twin Cities, St. Paul, Minnesota 55108, United States
| | - Aileen R. Lee
- Department of Biochemistry, Molecular Biology, and Biophysics and BioTechnology Institute, University of Minnesota−Twin Cities, St. Paul, Minnesota 55108, United States
| | - Aman S. Imani
- Department of Biochemistry, Molecular Biology, and Biophysics and BioTechnology Institute, University of Minnesota−Twin Cities, St. Paul, Minnesota 55108, United States
| | - Markus Künzler
- Department of Biology, Institute of Microbiology, Eidgenössische Technische Hochschule (ETH) Zürich, 8093 Zürich, Switzerland
| | - Michael F. Freeman
- Department of Biochemistry, Molecular Biology, and Biophysics and BioTechnology Institute, University of Minnesota−Twin Cities, St. Paul, Minnesota 55108, United States
| |
Collapse
|
20
|
D'Alessandro M, Richard M, Stigloher C, Gache V, Boulin T, Richmond JE, Bessereau JL. CRELD1 is an evolutionarily-conserved maturational enhancer of ionotropic acetylcholine receptors. eLife 2018; 7:39649. [PMID: 30407909 PMCID: PMC6245729 DOI: 10.7554/elife.39649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/05/2018] [Indexed: 12/22/2022] Open
Abstract
The assembly of neurotransmitter receptors in the endoplasmic reticulum limits the number of receptors delivered to the plasma membrane, ultimately controlling neurotransmitter sensitivity and synaptic transfer function. In a forward genetic screen conducted in the nematode C. elegans, we identified crld-1 as a gene required for the synaptic expression of ionotropic acetylcholine receptors (AChR). We demonstrated that the CRLD-1A isoform is a membrane-associated ER-resident protein disulfide isomerase (PDI). It physically interacts with AChRs and promotes the assembly of AChR subunits in the ER. Mutations of Creld1, the human ortholog of crld-1a, are responsible for developmental cardiac defects. We showed that Creld1 knockdown in mouse muscle cells decreased surface expression of AChRs and that expression of mouse Creld1 in C. elegans rescued crld-1a mutant phenotypes. Altogether these results identify a novel and evolutionarily-conserved maturational enhancer of AChR biogenesis, which controls the abundance of functional receptors at the cell surface.
Collapse
Affiliation(s)
- Manuela D'Alessandro
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| | - Magali Richard
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| | - Christian Stigloher
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| | - Vincent Gache
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| | - Thomas Boulin
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| | - Janet E Richmond
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, United States
| | - Jean-Louis Bessereau
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| |
Collapse
|
21
|
|
22
|
Trubitsyna M, Michlewski G, Finnegan DJ, Elfick A, Rosser SJ, Richardson JM, French CE. Use of mariner transposases for one-step delivery and integration of DNA in prokaryotes and eukaryotes by transfection. Nucleic Acids Res 2017; 45:e89. [PMID: 28204586 PMCID: PMC5449632 DOI: 10.1093/nar/gkx113] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 02/06/2017] [Indexed: 11/20/2022] Open
Abstract
Delivery of DNA to cells and its subsequent integration into the host genome is a fundamental task in molecular biology, biotechnology and gene therapy. Here we describe an IP-free one-step method that enables stable genome integration into either prokaryotic or eukaryotic cells. A synthetic mariner transposon is generated by flanking a DNA sequence with short inverted repeats. When purified recombinant Mos1 or Mboumar-9 transposase is co-transfected with transposon-containing plasmid DNA, it penetrates prokaryotic or eukaryotic cells and integrates the target DNA into the genome. In vivo integrations by purified transposase can be achieved by electroporation, chemical transfection or Lipofection of the transposase:DNA mixture, in contrast to other published transposon-based protocols which require electroporation or microinjection. As in other transposome systems, no helper plasmids are required since transposases are not expressed inside the host cells, thus leading to generation of stable cell lines. Since it does not require electroporation or microinjection, this tool has the potential to be applied for automated high-throughput creation of libraries of random integrants for purposes including gene knock-out libraries, screening for optimal integration positions or safe genome locations in different organisms, selection of the highest production of valuable compounds for biotechnology, and sequencing.
Collapse
Affiliation(s)
- Maryia Trubitsyna
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FF, UK
| | - Gracjan Michlewski
- Institute of Cell Biology, School of Biological Sciences, Wellcome Trust Centre for Cell Biology, University of Edinburgh, Edinburgh EH9 3BF, UK
| | - David J Finnegan
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FF, UK
| | - Alistair Elfick
- Institute of BioEngineering, School of Engineering, University of Edinburgh, Edinburgh EH9 3JL, UK
| | - Susan J Rosser
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, UK Centre for Mammalian Synthetic Biology, University of Edinburgh, Edinburgh EH9 3FF, UK
| | - Julia M Richardson
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FF, UK
| | - Christopher E French
- Institute of Quantitative Biology, Biochemistry and Biotechnology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FF, UK
| |
Collapse
|
23
|
Zhou MB, Hu H, Miskey C, Lazarow K, Ivics Z, Kunze R, Yang G, Izsvák Z, Tang DQ. Transposition of the bamboo Mariner-like element Ppmar1 in yeast. Mol Phylogenet Evol 2017; 109:367-374. [PMID: 28189615 DOI: 10.1016/j.ympev.2017.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 01/26/2017] [Accepted: 02/03/2017] [Indexed: 12/30/2022]
Abstract
The moso bamboo genome contains the two structurally intact and thus potentially functional mariner-like elements Ppmar1 and Ppmar2. Both elements contain perfect terminal inverted repeats (TIRs) and a full-length intact transposase gene. Here we investigated whether Ppmar1 is functional in yeast (Saccharomyces cerevisiae). We have designed a two-component system consisting of a transposase expression cassette and a non-autonomous transposon on two separate plasmids. We demonstrate that the Ppmar1 transposase Pptpase1 catalyses excision of the non-autonomous Ppmar1NA element from the plasmid and reintegration at TA dinucleotide sequences in the yeast chromosomes. In addition, we generated 14 hyperactive Ppmar1 transposase variants by systematic single amino acid substitutions. The most active transposase variant, S171A, induces 10-fold more frequent Ppmar1NA excisions in yeast than the wild type transposase. The Ppmar1 transposon is a promising tool for insertion mutagenesis in moso bamboo and may be used in other plants as an alternative to the established transposon tagging systems.
Collapse
Affiliation(s)
- Ming-Bing Zhou
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, LinAn, China
| | - Hui Hu
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, LinAn, China
| | - Csaba Miskey
- Paul Ehrlich Institute, Paul Ehrlich Str. 51-59, 63225 Langen, Germany
| | - Katina Lazarow
- Institute of Biology, Dahlem Centre of Plant Sciences, Freie Universität Berlin, 14195 Berlin, Germany
| | - Zoltán Ivics
- Paul Ehrlich Institute, Paul Ehrlich Str. 51-59, 63225 Langen, Germany
| | - Reinhard Kunze
- Institute of Biology, Dahlem Centre of Plant Sciences, Freie Universität Berlin, 14195 Berlin, Germany
| | - Guojun Yang
- Department of Biology, University of Toronto, Mississauga, ON, Canada
| | - Zsuzsanna Izsvák
- Max Delbrück Center for Molecular Medicine in the Helmholtz Society, Berlin, Germany.
| | - Ding-Qin Tang
- The Nurturing Station for the State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, LinAn, China.
| |
Collapse
|
24
|
Jeennor S, Cheawchanlertfa P, Suttiwattanakul S, Panchanawaporn S, Chutrakul C, Laoteng K. The codon-optimized Δ(6)-desaturase gene of Pythium sp. as an empowering tool for engineering n3/n6 polyunsaturated fatty acid biosynthesis. BMC Biotechnol 2015; 15:82. [PMID: 26369666 PMCID: PMC4570148 DOI: 10.1186/s12896-015-0200-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 08/29/2015] [Indexed: 11/10/2022] Open
Abstract
Background The ∆6-desaturase gene, encoding a key enzyme in the biosynthesis of polyunsaturated fatty acids, has potential in pharmaceutical and nutraceutical applications. Results The ∆6-desaturase gene has been isolated from a selected strain of Oomycetes, Pythium sp. BCC53698. The cloned gene (PyDes6) contained an open reading frame (ORF) of 1401 bp encoding 466 amino acid residues. The deduced amino acid sequence shared a high similarity to those of other ∆6-desaturases that contained the signature features of a membrane-bound ∆6-desaturase, including a cytochrome b5 and three histidine-rich motifs and membrane-spanning regions. Heterologous expression in Saccharomyces cerevisiae showed that monoene, diene and triene fatty acids having ∆9-double bond were substrates for PyDes6. No distinct preference between the n-3 and n-6 polyunsaturated fatty acyl substrates was found. The ∆6-desaturated products were markedly increased by codon optimization of PyDes6. Conclusion The codon-optimized ∆6-desaturase gene generated in this study is a promising tool for further reconstitution of the fatty acid profile, in a host system of choice, for the production of economically important fatty acids, particularly the n-3 and n-6 polyunsaturated fatty acids. Electronic supplementary material The online version of this article (doi:10.1186/s12896-015-0200-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sukanya Jeennor
- Bioprocess Technology Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand
| | - Pattsarun Cheawchanlertfa
- Bioprocess Technology Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand
| | - Sarinya Suttiwattanakul
- Bioprocess Technology Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand
| | - Sarocha Panchanawaporn
- Bioassay Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand
| | - Chanikul Chutrakul
- Bioassay Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand
| | - Kobkul Laoteng
- Bioprocess Technology Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand.
| |
Collapse
|
25
|
Frokjaer-Jensen C, Davis MW, Sarov M, Taylor J, Flibotte S, LaBella M, Pozniakovski A, Moerman DG, Jorgensen EM. Random and targeted transgene insertion in Caenorhabditis elegans using a modified Mos1 transposon. Nat Methods 2014; 11:529-34. [PMID: 24820376 PMCID: PMC4126194 DOI: 10.1038/nmeth.2889] [Citation(s) in RCA: 241] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 01/29/2014] [Indexed: 12/22/2022]
Abstract
We have generated a recombinant Mos1 transposon that can insert up to 45-kb transgenes into the Caenorhabditis elegans genome. The minimal Mos1 transposon (miniMos) is 550 bp long and inserts DNA into the genome at high frequency (~60% of injected animals). Genetic and antibiotic markers can be used for selection, and the transposon is active in C. elegans isolates and Caenorhabditis briggsae. We used the miniMos transposon to generate six universal Mos1-mediated single-copy insertion (mosSCI) landing sites that allow targeted transgene insertion with a single targeting vector into permissive expression sites on all autosomes. We also generated two collections of strains: a set of bright fluorescent insertions that are useful as dominant, genetic balancers and a set of lacO insertions to track genome position.
Collapse
Affiliation(s)
- Christian Frokjaer-Jensen
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Biology, University of Utah, Salt Lake City, Utah, USA
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | - M Wayne Davis
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Mihail Sarov
- TransgeneOmics, Max Planck Institute of Molecular Cell Biology and Genetics, D-01307 Dresden, Germany
| | - Jon Taylor
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephane Flibotte
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew LaBella
- Department of Biology, University of Utah, Salt Lake City, Utah, USA
| | - Andrei Pozniakovski
- TransgeneOmics, Max Planck Institute of Molecular Cell Biology and Genetics, D-01307 Dresden, Germany
| | - Donald G Moerman
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erik M Jorgensen
- Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, USA
- Department of Biology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
26
|
Abstract
Mutagenesis drives natural selection. In the lab, mutations allow gene function to be deciphered. C. elegans is highly amendable to functional genetics because of its short generation time, ease of use, and wealth of available gene-alteration techniques. Here we provide an overview of historical and contemporary methods for mutagenesis in C. elegans, and discuss principles and strategies for forward (genome-wide mutagenesis) and reverse (target-selected and gene-specific mutagenesis) genetic studies in this animal.
Collapse
Affiliation(s)
- Lena M Kutscher
- Laboratory of Developmental Genetics, The Rockefeller University, New York NY 10065, USA.
| | | |
Collapse
|
27
|
Bouchet N, Jaillet J, Gabant G, Brillet B, Briseño-Roa L, Cadene M, Augé-Gouillou C. cAMP protein kinase phosphorylates the Mos1 transposase and regulates its activity: evidences from mass spectrometry and biochemical analyses. Nucleic Acids Res 2014; 42:1117-28. [PMID: 24081583 PMCID: PMC3902898 DOI: 10.1093/nar/gkt874] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/05/2013] [Accepted: 09/06/2013] [Indexed: 12/19/2022] Open
Abstract
Genomic plasticity mediated by transposable elements can have a dramatic impact on genome integrity. To minimize its genotoxic effects, it is tightly regulated either by intrinsic mechanisms (linked to the element itself) or by host-mediated mechanisms. Using mass spectrometry, we show here for the first time that MOS1, the transposase driving the mobility of the mariner Mos1 element, is phosphorylated. We also show that the transposition activity of MOS1 is downregulated by protein kinase AMP cyclic-dependent phosphorylation at S170, which renders the transposase unable to promote Mos1 transposition. One step in the transposition cycle, the assembly of the paired-end complex, is specifically inhibited. At the cellular level, we provide evidence that phosphorylation at S170 prevents the active transport of the transposase into the nucleus. Our data suggest that protein kinase AMP cyclic-dependent phosphorylation may play a double role in the early stages of genome invasion by mariner elements.
Collapse
Affiliation(s)
- Nicolas Bouchet
- Innovation Moléculaire Thérapeutique, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Université François Rabelais, 37200 Tours, France, Centre de Biophysique Moléculaire, UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans, France, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IUT de Quimper, Université de Bretagne Occidentale, 6 rue de l’Université, 29000 Quimper, France and Biologie Cellulaire de la Synapse, INSERM U789, Ecole Normale Supérieure, 46 rue d'Ulm, 75005 Paris, France
| | - Jérôme Jaillet
- Innovation Moléculaire Thérapeutique, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Université François Rabelais, 37200 Tours, France, Centre de Biophysique Moléculaire, UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans, France, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IUT de Quimper, Université de Bretagne Occidentale, 6 rue de l’Université, 29000 Quimper, France and Biologie Cellulaire de la Synapse, INSERM U789, Ecole Normale Supérieure, 46 rue d'Ulm, 75005 Paris, France
| | - Guillaume Gabant
- Innovation Moléculaire Thérapeutique, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Université François Rabelais, 37200 Tours, France, Centre de Biophysique Moléculaire, UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans, France, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IUT de Quimper, Université de Bretagne Occidentale, 6 rue de l’Université, 29000 Quimper, France and Biologie Cellulaire de la Synapse, INSERM U789, Ecole Normale Supérieure, 46 rue d'Ulm, 75005 Paris, France
| | - Benjamin Brillet
- Innovation Moléculaire Thérapeutique, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Université François Rabelais, 37200 Tours, France, Centre de Biophysique Moléculaire, UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans, France, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IUT de Quimper, Université de Bretagne Occidentale, 6 rue de l’Université, 29000 Quimper, France and Biologie Cellulaire de la Synapse, INSERM U789, Ecole Normale Supérieure, 46 rue d'Ulm, 75005 Paris, France
| | - Luis Briseño-Roa
- Innovation Moléculaire Thérapeutique, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Université François Rabelais, 37200 Tours, France, Centre de Biophysique Moléculaire, UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans, France, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IUT de Quimper, Université de Bretagne Occidentale, 6 rue de l’Université, 29000 Quimper, France and Biologie Cellulaire de la Synapse, INSERM U789, Ecole Normale Supérieure, 46 rue d'Ulm, 75005 Paris, France
| | - Martine Cadene
- Innovation Moléculaire Thérapeutique, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Université François Rabelais, 37200 Tours, France, Centre de Biophysique Moléculaire, UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans, France, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IUT de Quimper, Université de Bretagne Occidentale, 6 rue de l’Université, 29000 Quimper, France and Biologie Cellulaire de la Synapse, INSERM U789, Ecole Normale Supérieure, 46 rue d'Ulm, 75005 Paris, France
| | - Corinne Augé-Gouillou
- Innovation Moléculaire Thérapeutique, EA 6306, UFR Sciences Pharmaceutiques, Parc Grandmont, Université François Rabelais, 37200 Tours, France, Centre de Biophysique Moléculaire, UPR 4301 CNRS, Rue Charles Sadron, 45071 Orléans, France, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, IUT de Quimper, Université de Bretagne Occidentale, 6 rue de l’Université, 29000 Quimper, France and Biologie Cellulaire de la Synapse, INSERM U789, Ecole Normale Supérieure, 46 rue d'Ulm, 75005 Paris, France
| |
Collapse
|
28
|
Cao Y, Rui B, Wellems DL, Li M, Chen B, Zhang D, Pan W. Identification of piggyBac-mediated insertions in Plasmodium berghei by next generation sequencing. Malar J 2013; 12:287. [PMID: 23961915 PMCID: PMC3765144 DOI: 10.1186/1475-2875-12-287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/18/2013] [Indexed: 01/24/2023] Open
Abstract
Background The piggyBac transposon system provides a powerful forward genetics tool to study gene function in Plasmodium parasites via random insertion mutagenesis and phenotypic screening. The identification of genotype of piggyBac mutants in the Plasmodium genome is thus an indispensable step in forward genetic analysis. Several PCR-based approaches have been used to identify the piggyBac insertion sites in Plasmodium falciparum and Plasmodium berghei, but all are tedious and inefficient. Next generation sequencing can produce large amounts of sequence data and is particularly suitable for genome-wide association studies. In this study, the Next generation sequencing technology was employed to efficiently identify piggyBac insertion sites in the genome of P. berghei. Methods Plasmodium berghei parasites were co-transfected with piggyBac donor and helper plasmids. Initially, the classical inverse PCR method was used to identify the existence of piggyBac insertions in the P. berghei genome. The whole genome of post-transfection parasites was subsequently sequenced with a PCR-free paired-end module using the Illumina HiSeq sequencing system. The two distinct methods (‘BLAST method’ and ‘SOAP method’) were employed to identify piggyBac insertion sites in the P. berghei genome with Illumina sequencing data. All the identified piggyBac insertions were further tested by half-nested PCR. Results The inverse PCR method resulted in a very low yield of ten individual insertions identified. Conversely, 47 piggyBac insertions were identified from about 1 Gb of Illumina sequencing data via the two distinct analysis methods. The majority of identified piggyBac insertions were confirmed by half-nested PCR. In addition, 1,850 single nucleotide polymorphisms were identified through alignment of the Illumina sequencing data of the P. berghei ANKA strain used in this study with the reference genome sequences. Conclusion This study demonstrates that a high-throughput genome sequencing approach is an efficient tool for the identification of piggyBac-mediated insertions in Plasmodium parasites.
Collapse
Affiliation(s)
- Yi Cao
- Department of Tropical Infectious Diseases, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
29
|
Inoue A, Sawatari E, Hisamoto N, Kitazono T, Teramoto T, Fujiwara M, Matsumoto K, Ishihara T. Forgetting in C. elegans Is Accelerated by Neuronal Communication via the TIR-1/JNK-1 Pathway. Cell Rep 2013; 3:808-19. [DOI: 10.1016/j.celrep.2013.02.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 09/21/2012] [Accepted: 02/15/2013] [Indexed: 01/13/2023] Open
|
30
|
Gürel G, Gustafson MA, Pepper JS, Horvitz HR, Koelle MR. Receptors and other signaling proteins required for serotonin control of locomotion in Caenorhabditis elegans. Genetics 2012; 192:1359-71. [PMID: 23023001 PMCID: PMC3512144 DOI: 10.1534/genetics.112.142125] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 09/15/2012] [Indexed: 01/05/2023] Open
Abstract
A better understanding of the molecular mechanisms of signaling by the neurotransmitter serotonin is required to assess the hypothesis that defects in serotonin signaling underlie depression in humans. Caenorhabditis elegans uses serotonin as a neurotransmitter to regulate locomotion, providing a genetic system to analyze serotonin signaling. From large-scale genetic screens we identified 36 mutants of C. elegans in which serotonin fails to have its normal effect of slowing locomotion, and we molecularly identified eight genes affected by 19 of the mutations. Two of the genes encode the serotonin-gated ion channel MOD-1 and the G-protein-coupled serotonin receptor SER-4. mod-1 is expressed in the neurons and muscles that directly control locomotion, while ser-4 is expressed in an almost entirely non-overlapping set of sensory and interneurons. The cells expressing the two receptors are largely not direct postsynaptic targets of serotonergic neurons. We analyzed animals lacking or overexpressing the receptors in various combinations using several assays for serotonin response. We found that the two receptors act in parallel to affect locomotion. Our results show that serotonin functions as an extrasynaptic signal that independently activates multiple receptors at a distance from its release sites and identify at least six additional proteins that appear to act with serotonin receptors to mediate serotonin response.
Collapse
Affiliation(s)
- Güliz Gürel
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Megan A. Gustafson
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Judy S. Pepper
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520
| | - H. Robert Horvitz
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Michael R. Koelle
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
31
|
Positive modulation of a Cys-loop acetylcholine receptor by an auxiliary transmembrane subunit. Nat Neurosci 2012; 15:1374-81. [DOI: 10.1038/nn.3197] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/30/2012] [Indexed: 02/07/2023]
|
32
|
Robert VJP. Engineering the Caenorhabditis elegans genome by Mos1-induced transgene-instructed gene conversion. Methods Mol Biol 2012; 859:189-201. [PMID: 22367873 DOI: 10.1007/978-1-61779-603-6_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Mos1-induced transgene-instructed gene conversion (MosTIC) is a technique of choice to engineer the genome of the nematode Caenorhabditis elegans. MosTIC is initiated by the excision of Mos1, a DNA transposon of the Tc1/Mariner super family that can be mobilized in the germ line of C. elegans. Mos1 excision creates a DNA double-strand break that is repaired by several cellular mechanisms, including transgene-instructed gene conversion. For MosTIC, the transgenic repair template used by the gene conversion machinery is made of sequences that share DNA homologies with the genomic region to engineer and carries the modifications to be introduced in the genome. In this chapter, we present two MosTIC protocols routinely used.
Collapse
Affiliation(s)
- Valérie J P Robert
- Laboratory of Molecular and Cellular Biology, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
33
|
Vallin E, Gallagher J, Granger L, Martin E, Belougne J, Maurizio J, Duverger Y, Scaglione S, Borrel C, Cortier E, Abouzid K, Carre-Pierrat M, Gieseler K, Ségalat L, Kuwabara PE, Ewbank JJ. A genome-wide collection of Mos1 transposon insertion mutants for the C. elegans research community. PLoS One 2012; 7:e30482. [PMID: 22347378 PMCID: PMC3275553 DOI: 10.1371/journal.pone.0030482] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 12/16/2011] [Indexed: 11/24/2022] Open
Abstract
Methods that use homologous recombination to engineer the genome of C. elegans commonly use strains carrying specific insertions of the heterologous transposon Mos1. A large collection of known Mos1 insertion alleles would therefore be of general interest to the C. elegans research community. We describe here the optimization of a semi-automated methodology for the construction of a substantial collection of Mos1 insertion mutant strains. At peak production, more than 5,000 strains were generated per month. These strains were then subject to molecular analysis, and more than 13,300 Mos1 insertions characterized. In addition to targeting directly more than 4,700 genes, these alleles represent the potential starting point for the engineered deletion of essentially all C. elegans genes and the modification of more than 40% of them. This collection of mutants, generated under the auspices of the European NEMAGENETAG consortium, is publicly available and represents an important research resource.
Collapse
Affiliation(s)
- Elodie Vallin
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Joseph Gallagher
- School of Biochemistry, University of Bristol, Bristol, United Kingdom
| | - Laure Granger
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Edwige Martin
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Jérôme Belougne
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, Marseille, France
- INSERM, U1104, Marseille, France
- CNRS, UMR7280, Marseille, France
| | - Julien Maurizio
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, Marseille, France
- INSERM, U1104, Marseille, France
- CNRS, UMR7280, Marseille, France
| | - Yohann Duverger
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, Marseille, France
- INSERM, U1104, Marseille, France
- CNRS, UMR7280, Marseille, France
| | - Sarah Scaglione
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, Marseille, France
- INSERM, U1104, Marseille, France
- CNRS, UMR7280, Marseille, France
| | - Caroline Borrel
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Elisabeth Cortier
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Karima Abouzid
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Maité Carre-Pierrat
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
- Plateforme “Biologie de Caenorhabditis elegans”, CNRS UMS3421, Campus de la Doua, Villeurbanne, France
| | - Kathrin Gieseler
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Laurent Ségalat
- Centre de Génétique et de Physiologie Moléculaires et Cellulaires, CNRS UMR 5534, Campus de la Doua, Villeurbanne, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
| | | | - Jonathan J. Ewbank
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, Marseille, France
- INSERM, U1104, Marseille, France
- CNRS, UMR7280, Marseille, France
| |
Collapse
|
34
|
Huang L, Szymanska K, Jensen V, Janecke A, Innes A, Davis E, Frosk P, Li C, Willer J, Chodirker B, Greenberg C, McLeod D, Bernier F, Chudley A, Müller T, Shboul M, Logan C, Loucks C, Beaulieu C, Bowie R, Bell S, Adkins J, Zuniga F, Ross K, Wang J, Ban M, Becker C, Nürnberg P, Douglas S, Craft C, Akimenko MA, Hegele R, Ober C, Utermann G, Bolz H, Bulman D, Katsanis N, Blacque O, Doherty D, Parboosingh J, Leroux M, Johnson C, Boycott K. TMEM237 is mutated in individuals with a Joubert syndrome related disorder and expands the role of the TMEM family at the ciliary transition zone. Am J Hum Genet 2011; 89:713-30. [PMID: 22152675 PMCID: PMC3234373 DOI: 10.1016/j.ajhg.2011.11.005] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Revised: 10/25/2011] [Accepted: 11/08/2011] [Indexed: 12/23/2022] Open
Abstract
Joubert syndrome related disorders (JSRDs) have broad but variable phenotypic overlap with other ciliopathies. The molecular etiology of this overlap is unclear but probably arises from disrupting common functional module components within primary cilia. To identify additional module elements associated with JSRDs, we performed homozygosity mapping followed by next-generation sequencing (NGS) and uncovered mutations in TMEM237 (previously known as ALS2CR4). We show that loss of the mammalian TMEM237, which localizes to the ciliary transition zone (TZ), results in defective ciliogenesis and deregulation of Wnt signaling. Furthermore, disruption of Danio rerio (zebrafish) tmem237 expression produces gastrulation defects consistent with ciliary dysfunction, and Caenorhabditis elegans jbts-14 genetically interacts with nphp-4, encoding another TZ protein, to control basal body-TZ anchoring to the membrane and ciliogenesis. Both mammalian and C. elegans TMEM237/JBTS-14 require RPGRIP1L/MKS5 for proper TZ localization, and we demonstrate additional functional interactions between C. elegans JBTS-14 and MKS-2/TMEM216, MKSR-1/B9D1, and MKSR-2/B9D2. Collectively, our findings integrate TMEM237/JBTS-14 in a complex interaction network of TZ-associated proteins and reveal a growing contribution of a TZ functional module to the spectrum of ciliopathy phenotypes.
Collapse
Affiliation(s)
- Lijia Huang
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Katarzyna Szymanska
- Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Victor L. Jensen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Andreas R. Janecke
- Department of Pediatrics II, Innsbruck Medical University, Innsbruck 6020, Austria
- Division of Human Genetics, Innsbruck Medical University, Innsbruck 6020, Austria
| | - A. Micheil Innes
- Department of Medical Genetics, University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Erica E. Davis
- Center for Human Disease Modeling, Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Patrick Frosk
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3R 0J9, Canada
| | - Chunmei Li
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Jason R. Willer
- Center for Human Disease Modeling, Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Bernard N. Chodirker
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3R 0J9, Canada
| | - Cheryl R. Greenberg
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3R 0J9, Canada
| | - D. Ross McLeod
- Department of Medical Genetics, University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Francois P. Bernier
- Department of Medical Genetics, University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Albert E. Chudley
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3R 0J9, Canada
| | - Thomas Müller
- Department of Pediatrics II, Innsbruck Medical University, Innsbruck 6020, Austria
| | - Mohammad Shboul
- Institute of Medical Biology: Human Embryology, 8A Biomedical Grove, #05-40 Immunos, Singapore 138648, Singapore
| | - Clare V. Logan
- Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Catrina M. Loucks
- Department of Medical Genetics, University of Calgary, Calgary, AB T3B 6A8, Canada
| | - Chandree L. Beaulieu
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Rachel V. Bowie
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sandra M. Bell
- Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Jonathan Adkins
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | - Freddi I. Zuniga
- Mary D. Allen Laboratory in Vision Research, Doheny Eye Institute, Departments of Ophthalmology and Cell and Neurobiology, Los Angeles, CA 90033-9224, USA
| | - Kevin D. Ross
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Jian Wang
- Robarts Research Institute and University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Matthew R. Ban
- Robarts Research Institute and University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Christian Becker
- Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Stuart Douglas
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Cheryl M. Craft
- Mary D. Allen Laboratory in Vision Research, Doheny Eye Institute, Departments of Ophthalmology and Cell and Neurobiology, Los Angeles, CA 90033-9224, USA
| | | | - Robert A. Hegele
- Robarts Research Institute and University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Gerd Utermann
- Division of Human Genetics, Innsbruck Medical University, Innsbruck 6020, Austria
| | - Hanno J. Bolz
- Center for Human Genetics, Bioscientia, 55218 Ingelheim, Germany
- Institute of Human Genetics, University Hospital of Cologne, 50931 Cologne, Germany
| | - Dennis E. Bulman
- Ottawa Hospital Research Institute and University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Oliver E. Blacque
- School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dan Doherty
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
| | | | - Michel R. Leroux
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Colin A. Johnson
- Section of Ophthalmology and Neurosciences, Leeds Institute of Molecular Medicine, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Kym M. Boycott
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
35
|
Boulin T, Hobert O. From genes to function: the C. elegans genetic toolbox. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2011; 1:114-37. [PMID: 23801671 DOI: 10.1002/wdev.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
This review aims to provide an overview of the technologies which make the nematode Caenorhabditis elegans an attractive genetic model system. We describe transgenesis techniques and forward and reverse genetic approaches to isolate mutants and clone genes. In addition, we discuss the new possibilities offered by genome engineering strategies and next-generation genome analysis tools.
Collapse
Affiliation(s)
- Thomas Boulin
- Department of Biology, Institut de Biologie de l'École Normale Supérieure, Paris, France.
| | | |
Collapse
|
36
|
Checchi PM, Engebrecht J. Caenorhabditis elegans histone methyltransferase MET-2 shields the male X chromosome from checkpoint machinery and mediates meiotic sex chromosome inactivation. PLoS Genet 2011; 7:e1002267. [PMID: 21909284 PMCID: PMC3164706 DOI: 10.1371/journal.pgen.1002267] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 07/12/2011] [Indexed: 11/23/2022] Open
Abstract
Meiosis is a specialized form of cellular division that results in the precise halving of the genome to produce gametes for sexual reproduction. Checkpoints function during meiosis to detect errors and subsequently to activate a signaling cascade that prevents the formation of aneuploid gametes. Indeed, asynapsis of a homologous chromosome pair elicits a checkpoint response that can in turn trigger germline apoptosis. In a heterogametic germ line, however, sex chromosomes proceed through meiosis with unsynapsed regions and are not recognized by checkpoint machinery. We conducted a directed RNAi screen in Caenorhabditis elegans to identify regulatory factors that prevent recognition of heteromorphic sex chromosomes as unpaired and uncovered a role for the SET domain histone H3 lysine 9 histone methyltransferase (HMTase) MET-2 and two additional HMTases in shielding the male X from checkpoint machinery. We found that MET-2 also mediates the transcriptional silencing program of meiotic sex chromosome inactivation (MSCI) but not meiotic silencing of unsynapsed chromatin (MSUC), suggesting that these processes are distinct. Further, MSCI and checkpoint shielding can be uncoupled, as double-strand breaks targeted to an unpaired, transcriptionally silenced extra-chromosomal array induce checkpoint activation in germ lines depleted for met-2. In summary, our data uncover a mechanism by which repressive chromatin architecture enables checkpoint proteins to distinguish between the partnerless male X chromosome and asynapsed chromosomes thereby shielding the lone X from inappropriate activation of an apoptotic program. Meiosis results in the generation of non-identical haploid gametes and maintenance of chromosome number during sexual reproduction. Precise meiotic chromosome segregation is essential for life, and in humans errors in this process contribute to aneuploidy or failure in meiosis, which manifests as spontaneous abortions or infertility. Cellular surveillance pathways monitor the steps of meiosis; and, if homologous chromosomes fail to pair and recombine, checkpoint machinery responds by eliciting signals to induce apoptosis. However, in many species males possess a single X chromosome that is transcriptionally silenced, accumulates repressive histone marks, and is not recognized as partnerless by meiotic checkpoints. Here, we used C. elegans to investigate how the male X is precluded from checkpoint signaling and uncovered a role for conserved chromatin-remodeling proteins that block checkpoints and mediate meiotic silencing. Our data elucidate the molecular mechanisms by which chromatin architecture influences both transcriptional silencing and checkpoint response to breaks on unpaired sex chromosomes, and we propose a model by which repressive chromatin modifiers directly block meiotic checkpoints from accessing the male X chromosome.
Collapse
Affiliation(s)
- Paula M. Checchi
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - JoAnne Engebrecht
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
37
|
Behavioral choice between conflicting alternatives is regulated by a receptor guanylyl cyclase, GCY-28, and a receptor tyrosine kinase, SCD-2, in AIA interneurons of Caenorhabditis elegans. J Neurosci 2011; 31:3007-15. [PMID: 21414922 DOI: 10.1523/jneurosci.4691-10.2011] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Animals facing conflicting sensory cues make a behavioral choice between competing alternatives through integration of the sensory cues. Here, we performed a genetic screen to identify genes important for the sensory integration of two conflicting cues, the attractive odorant diacetyl and the aversive stimulus Cu(2+), and found that the membrane-bound guanylyl cyclase GCY-28 and the receptor tyrosine kinase SCD-2 regulate the behavioral choice between these alternatives in Caenorhabditis elegans. The gcy-28 mutants and scd-2 mutants show an abnormal bias in the behavioral choice between the cues, although their responses to each individual cue are similar to those in wild-type animals. Mutants in a gene encoding a cyclic nucleotide gated ion channel, cng-1, also exhibit the defect in sensory integration. Molecular genetic analyses suggested that GCY-28 and SCD-2 regulate sensory integration in AIA interneurons, where the conflicting sensory cues may converge. Genetic ablation or hyperpolarization of AIA interneurons showed nearly the same phenotype as gcy-28 or scd-2 mutants in the sensory integration, although this did not affect the sensory response to each individual cue. In gcy-28 or scd-2 mutants, activation of AIA interneurons is sufficient to restore normal sensory integration. These results suggest that the activity of AIA interneurons regulates the behavioral choice between the alternatives. We propose that GCY-28 and SCD-2 regulate sensory integration by modulating the activity of AIA interneurons.
Collapse
|
38
|
Williams CL, Li C, Kida K, Inglis PN, Mohan S, Semenec L, Bialas NJ, Stupay RM, Chen N, Blacque OE, Yoder BK, Leroux MR. MKS and NPHP modules cooperate to establish basal body/transition zone membrane associations and ciliary gate function during ciliogenesis. ACTA ACUST UNITED AC 2011; 192:1023-41. [PMID: 21422230 PMCID: PMC3063147 DOI: 10.1083/jcb.201012116] [Citation(s) in RCA: 366] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Eight proteins, defects in which are associated with Meckel-Gruber syndrome and nephronophthisis ciliopathies, work together as two functional modules at the transition zone to establish basal body/transition zone connections with the membrane and barricade entry of non-ciliary components into this organelle. Meckel-Gruber syndrome (MKS), nephronophthisis (NPHP), and related ciliopathies present with overlapping phenotypes and display considerable allelism between at least twelve different genes of largely unexplained function. We demonstrate that the conserved C. elegans B9 domain (MKS-1, MKSR-1, and MKSR-2), MKS-3/TMEM67, MKS-5/RPGRIP1L, MKS-6/CC2D2A, NPHP-1, and NPHP-4 proteins exhibit essential, collective functions at the transition zone (TZ), an underappreciated region at the base of all cilia characterized by Y-shaped assemblages that link axoneme microtubules to surrounding membrane. These TZ proteins functionally interact as members of two distinct modules, which together contribute to an early ciliogenic event. Specifically, MKS/MKSR/NPHP proteins establish basal body/TZ membrane attachments before or coinciding with intraflagellar transport–dependent axoneme extension and subsequently restrict accumulation of nonciliary components within the ciliary compartment. Together, our findings uncover a unified role for eight TZ-localized proteins in basal body anchoring and establishing a ciliary gate during ciliogenesis, and suggest that disrupting ciliary gate function contributes to phenotypic features of the MKS/NPHP disease spectrum.
Collapse
Affiliation(s)
- Corey L Williams
- Department of Cell Biology, University of Alabama, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Robert VJP, Bessereau JL. Genome engineering by transgene-instructed gene conversion in C. elegans. Methods Cell Biol 2011; 106:65-88. [PMID: 22118274 DOI: 10.1016/b978-0-12-544172-8.00003-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The nematode Caenorhabditis elegans is an anatomically simple metazoan that has been used over the last 40 years to address an extremely wide range of biological questions. One major advantage of the C. elegans system is the possibility to conduct large-scale genetic screens on randomly mutagenized animals, either looking for a phenotype of interest and subsequently relate the mutated gene to the biological process under study ("forward genetics"), or screening for molecular lesions impairing the function of a specific gene and later analyze the phenotype of the mutant ("reverse genetics"). However, the nature of the genomic lesion is not controlled in either strategy. Here we describe a technique to engineer customized mutations in the C. elegans genome by homologous recombination. This technique, called MosTIC (for Mos1 excision induced transgene-instructed gene conversion), requires a C. elegans strain containing an insertion of the Drosophila transposon Mos1 within the locus to modify. Expression of the Mos transposase in the germ line triggers Mos1 excision, which causes a DNA double strand break (DSB) in the chromosome at the excision site. The DSB locally stimulates DNA repair by homologous recombination, which can sometimes occur between the chromosome and a transgene containing sequence homologous to the broken locus. In that case, sequence variations contained in the repair template will be copied by gene conversion into the genome. Here we provide a detailed protocol of the MosTIC technique, which can be used to introduce point mutations and generate knockout and knock-in alleles.
Collapse
Affiliation(s)
- Valérie J P Robert
- Ecole Normale Supérieure, Institut de Biologie de l'ENS, IBENS, Paris, France
| | | |
Collapse
|
40
|
Abstract
The ability to manipulate the genome of organisms at will is perhaps the single most useful ability for the study of biological systems. Techniques for the generation of transgenics in the nematode Caenorhabditis elegans became available in the late 1980s. Since then, improvements to the original approach have been made to address specific limitations with transgene expression, expand on the repertoire of the types of biological information that transgenes can provide, and begin to develop methods to target transgenes to defined chromosomal locations. Many recent, detailed protocols have been published, and hence in this chapter, we will review various approaches to making C. elegans transgenics, discuss their applications, and consider their relative advantages and disadvantages. Comments will also be made on anticipated future developments and on the application of these methods to other nematodes.
Collapse
Affiliation(s)
- Vida Praitis
- Biology Department, Grinnell College, Grinnell, Iowa, USA
| | | |
Collapse
|
41
|
Thomas X, Hedhili S, Beuf L, Demattéi MV, Laparra H, Khong GN, Breitler JC, Montandon F, Carnus E, Norre F, Burtin D, Gantet P, Bigot Y, Renault S. The mariner Mos1 transposase produced in tobacco is active in vitro. Genetica 2010; 138:519-30. [PMID: 19847655 DOI: 10.1007/s10709-009-9414-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 10/05/2009] [Indexed: 11/25/2022]
Abstract
The mariner-like transposon Mos1 is used for insertional mutagenesis and transgenesis in different animals (insects, nematodes), but has never been used in plants. In this paper, the transposition activity of Mos1 was tested in Nicotiana tabacum, but no transposition event was detected. In an attempt to understand the absence of in planta transposition, Mos1 transposase (MOS1) was produced and purified from transgenic tobacco (HMNtMOS1). HMNtMOS1 was able to perform all transposition reaction steps in vitro: binding to ITR, excision and integration of the same pseudo-transposon used in in planta transposition assays. The in vitro transposition reaction was not inhibited by tobacco nuclear proteins, and did not depend on the temperature used for plant growth. Several hypotheses are proposed that could explain the inhibition of HMNtMOS1 activity in planta.
Collapse
Affiliation(s)
- Xavier Thomas
- Université François Rabelais de Tours, GICC, Parc de Grandmont, 37200 Tours, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Butschi A, Titz A, Wälti MA, Olieric V, Paschinger K, Nöbauer K, Guo X, Seeberger PH, Wilson IBH, Aebi M, Hengartner MO, Künzler M. Caenorhabditis elegans N-glycan core beta-galactoside confers sensitivity towards nematotoxic fungal galectin CGL2. PLoS Pathog 2010; 6:e1000717. [PMID: 20062796 PMCID: PMC2798750 DOI: 10.1371/journal.ppat.1000717] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 12/04/2009] [Indexed: 12/01/2022] Open
Abstract
The physiological role of fungal galectins has remained elusive. Here, we show that feeding of a mushroom galectin, Coprinopsis cinerea CGL2, to Caenorhabditis elegans inhibited development and reproduction and ultimately resulted in killing of this nematode. The lack of toxicity of a carbohydrate-binding defective CGL2 variant and the resistance of a C. elegans mutant defective in GDP-fucose biosynthesis suggested that CGL2-mediated nematotoxicity depends on the interaction between the galectin and a fucose-containing glycoconjugate. A screen for CGL2-resistant worm mutants identified this glycoconjugate as a Galbeta1,4Fucalpha1,6 modification of C. elegans N-glycan cores. Analysis of N-glycan structures in wild type and CGL2-resistant nematodes confirmed this finding and allowed the identification of a novel putative glycosyltransferase required for the biosynthesis of this glycoepitope. The X-ray crystal structure of a complex between CGL2 and the Galbeta1,4Fucalpha1,6GlcNAc trisaccharide at 1.5 A resolution revealed the biophysical basis for this interaction. Our results suggest that fungal galectins play a role in the defense of fungi against predators by binding to specific glycoconjugates of these organisms.
Collapse
Affiliation(s)
- Alex Butschi
- Institute of Molecular Biology, University of Zürich, Zürich, Switzerland
| | - Alexander Titz
- Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Martin A. Wälti
- Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | - Vincent Olieric
- Swiss Light Source (SLS), Paul-Scherrer-Institute (PSI), Villigen, Switzerland
| | - Katharina Paschinger
- Department of Chemistry, University of Natural Resources and Applied Life Sciences (BOKU), Vienna, Austria
| | - Katharina Nöbauer
- VetOMICS Core Facility for Proteomics & Metabolomics Studies, University of Veterinary Medicine, Vienna, Austria
| | - Xiaoqiang Guo
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Peter H. Seeberger
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Iain B. H. Wilson
- Department of Chemistry, University of Natural Resources and Applied Life Sciences (BOKU), Vienna, Austria
| | - Markus Aebi
- Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| | | | - Markus Künzler
- Institute of Microbiology, Swiss Federal Institute of Technology (ETH) Zürich, Zürich, Switzerland
| |
Collapse
|
43
|
Hao L, Acar S, Evans J, Ou G, Scholey JM. Analysis of intraflagellar transport in C. elegans sensory cilia. Methods Cell Biol 2009; 93:235-66. [PMID: 20409821 DOI: 10.1016/s0091-679x(08)93013-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cilia are assembled and maintained by intraflagellar transport (IFT), the motor-dependent, bidirectional movement of multiprotein complexes, called IFT particles, along the axoneme. The sensory cilia of Caenorhabditis elegans represent very useful objects for studying IFT because of the availability of in vivo time-lapse fluorescence microscopy assays of IFT and multiple ciliary mutants. In this system there are 60 sensory neurons, each having sensory cilia on the endings of their dendrites, and most components of the IFT machinery operating in these structures have been identified using forward and reverse genetic approaches. By analyzing the rate of IFT along cilia within living wild-type and mutant animals, two anterograde and one retrograde IFT motors were identified, the functional coordination of the two anterograde kinesin-2 motors was established and the transport properties of all the known IFT particle components have been characterized. The anterograde kinesin motors have been heterologously expressed and purified, and their biochemical properties have been characterized using MT gliding and single molecule motility assays. In this chapter, we summarize how the tools of genetics, cell biology, electron microscopy, and biochemistry are being used to dissect the composition and mechanism of action of IFT motors and IFT particles in C. elegans.
Collapse
Affiliation(s)
- Limin Hao
- Molecular and Cell Biology, University of California, Davis, California 95616, USA
| | | | | | | | | |
Collapse
|
44
|
Physical properties of DNA components affecting the transposition efficiency of the mariner Mos1 element. Mol Genet Genomics 2009; 282:531-46. [PMID: 19774400 DOI: 10.1007/s00438-009-0484-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
Abstract
Previous studies have shown that the transposase and the inverted terminal repeat (ITR) of the Mos1 mariner elements are suboptimal for transposition; and that hyperactive transposases and transposon with more efficient ITR configurations can be obtained by rational molecular engineering. In an attempt to determine the extent to which this element is suboptimal for transposition, we investigate here the impact of the three main DNA components on its transposition efficiency in bacteria and in vitro. We found that combinations of natural and synthetic ITRs obtained by systematic evolution of ligands by exponential enrichment did increase the transposition rate. We observed that when untranslated terminal regions were associated with their respective natural ITRs, they acted as transposition enhancers, probably via the early transposition steps. Finally, we demonstrated that the integrity of the Mos1 inner region was essential for transposition. These findings allowed us to propose prototypes of optimized Mos1 vectors, and to define the best sequence features of their associated marker cassettes. These vector prototypes were assayed in HeLa cells, in which Mos1 vectors had so far been found to be inactive. The results obtained revealed that using these prototypes does not circumvent this problem. However, such vectors can be expected to provide new tools for the use in genome engineering in systems such as Caenorhabditis elegans in which Mos1 is very active.
Collapse
|
45
|
Lampe DJ. Bacterial genetic methods to explore the biology of mariner transposons. Genetica 2009; 138:499-508. [PMID: 19711186 DOI: 10.1007/s10709-009-9401-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Accepted: 08/10/2009] [Indexed: 10/20/2022]
Abstract
Mariners are small DNA mediated transposons of eukaryotes that fortuitously function in bacteria. Using bacterial genetics, it is possible to study a variety of properties of mariners, including transpositional ability, dominant-negative regulation, overexpresson inhibition, and the function of cis-acting sequences like the inverted terminal repeats. In conjunction with biochemical techniques, the structure of the transposase can be elucidated and the activity of the elements can be improved for genetic tool use. Finally, it is possible to uncover functional transposase genes directly from genomes given a suitable bacterial genetic screen.
Collapse
Affiliation(s)
- David J Lampe
- Department of Biological Sciences, Duquesne University, 600 Forbes Ave., Pittsburgh, PA 15116, USA.
| |
Collapse
|
46
|
Park JM, Evertts AG, Levin HL. The Hermes transposon of Musca domestica and its use as a mutagen of Schizosaccharomyces pombe. Methods 2009; 49:243-7. [PMID: 19450689 DOI: 10.1016/j.ymeth.2009.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/06/2009] [Accepted: 05/08/2009] [Indexed: 11/26/2022] Open
Abstract
Transposon mutagenesis allows for the discovery and characterization of genes by creating mutations that can be easily mapped and sequenced. Moreover, this method allows for a relatively unbiased approach to isolating genes of interest. Recently, a system of transposon based mutagenesis for Schizosaccharomyces pombe became available. This mutagenesis relies on Hermes, a DNA transposon from the house fly that readily integrates into the chromosomes of S. pombe. The Hermes system is distinct from the retrotransposons of S. pombe because it efficiently integrates into open reading frames. To mutagenize S. pombe, cells are transformed with a plasmid that contains a drug resistance marker flanked by the terminal inverted repeats of Hermes. The Hermes transposase expressed from a second plasmid excises the resistance marker with the inverted repeats and inserts this DNA into chromosomal sites. After S. pombe with these two plasmids grow 25 generations, approximately 2% of the cells contain insertions. Of the cells with insertions, 68% contain single integration events. The protocols listed here provide the detailed information necessary to mutagenize a strain of interest, screen for specific phenotypes, and sequence the positions of insertion.
Collapse
Affiliation(s)
- Jung M Park
- Section on Eukaryotic Transposable Elements, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
47
|
Manipulating the Caenorhabditis elegans genome using mariner transposons. Genetica 2009; 138:541-9. [PMID: 19347589 DOI: 10.1007/s10709-009-9362-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 03/20/2009] [Indexed: 12/16/2022]
Abstract
Tc1, one of the founding members of the Tc1/mariner transposon superfamily, was identified in the nematode Caenorhabditis elegans more than 25 years ago. Over the years, Tc1 and other endogenous mariner transposons became valuable tools for mutagenesis and targeted gene inactivation in C. elegans. However, transposition is naturally repressed in the C. elegans germline by an RNAi-like mechanism, necessitating the use of mutant strains in which transposition was globally derepressed, which causes drawbacks such as uncontrolled proliferation of the transposons in the genome and accumulation of background mutations. The more recent mobilization of the Drosophila mariner transposon Mos1 in the C. elegans germline circumvented the problems inherent to endogenous transposons. Mos1 transposition strictly depends on the expression of the Mos transposase, which can be controlled in the germline using inducible promoters. First, Mos1 can be used for insertional mutagenesis. The mobilization of Mos1 copies present on an extrachromosomal array results in the generation of a small number of Mos1 genomic insertions that can be rapidly cloned by inverse PCR. Second, Mos1 insertions can be used for genome engineering. Triggering the excision of a genomic Mos1 insertion causes a chromosomal break, which can be repaired by transgene-instructed gene conversion. This process is used to introduce specific changes in a given gene, such as point mutations, deletions or insertions of a tag, and to create single-copy transgenes.
Collapse
|
48
|
Bazopoulou D, Tavernarakis N. The NemaGENETAG initiative: large scale transposon insertion gene-tagging in Caenorhabditis elegans. Genetica 2009; 137:39-46. [PMID: 19343510 DOI: 10.1007/s10709-009-9361-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 03/20/2009] [Indexed: 12/01/2022]
Abstract
The nematode Caenorhabditis elegans is a widely appreciated, powerful platform in which to study important biological mechanisms related to human health. More than 65% of human disease genes have homologues in the C. elegans genome, and essential aspects of mammalian cell biology, neurobiology and development are faithfully recapitulated in this organism. The EU-funded NemaGENETAG project was initiated with the aim to develop cutting-edge tools and resources that will facilitate modelling of human pathologies in C. elegans, and advance our understanding of animal development and physiology. The main objective of the project involves the generation and evaluation of a large collection of transposon-tagged mutants. In the process of achieving this objective the NemaGENETAG consortium also endeavours to optimize and automate existing transposon-mediated mutagenesis methodologies based on the Mos1 transposable element, in addition to developing alternatives using other transposon systems. The final product of this initiative-a comprehensive collection of transposon-tagged alleles-together with the acquisition of efficient transposon-based tools for mutagenesis and transgenesis in C. elegans, should yield a wealth of information on gene function, immediately relevant to key biological processes and to pharmaceutical research and development.
Collapse
Affiliation(s)
- Daphne Bazopoulou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, N. Plastira 100, Vassilika Vouton, 70013, Heraklion, Crete, Greece
| | | |
Collapse
|
49
|
Automated screening for mutants affecting dopaminergic-neuron specification in C. elegans. Nat Methods 2008; 5:869-72. [PMID: 18758453 PMCID: PMC2693092 DOI: 10.1038/nmeth.1250] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 07/29/2008] [Indexed: 12/11/2022]
Abstract
We describe an automated method to isolate mutant C. elegans animals, which fail to appropriately execute a cellular differentiation program that we monitor with gfp-based reporter gene technology. A fluorescence activated sorting mechanism implemented in the COPAS Biosort machine is able to isolate mutants with subtle alterations in the cellular specificity of gfp expression. This methodology is significantly more efficient than comparable manual screens and enabled us to isolate mutants in which dopamine neurons fail to differentiate appropriately.
Collapse
|
50
|
Moerman DG, Barstead RJ. Towards a mutation in every gene in Caenorhabditis elegans. BRIEFINGS IN FUNCTIONAL GENOMICS AND PROTEOMICS 2008; 7:195-204. [PMID: 18417533 DOI: 10.1093/bfgp/eln016] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
The combined efforts of the Caenorhabditis elegans Knockout Consortium and individuals within the worm community are moving us closer to the goal of identifying mutations in every gene in the nematode C. elegans. At present, we count about 7000 deletion alleles that fall within 5500 genes. The principal method used to detect deletion mutations in the nematode utilizes polymerase chain reaction (PCR). More recently, the Moerman group has incorporated array comparative genome hybridization (aCGH) to detect deletions across the entire coding genome. Other methods used to detect mutant alleles in C. elegans include targeting induced local lesion in genomes (TILLING), transposon tagging, using either Tc1 or Mos1 and resequencing. These combined strategies have improved the overall throughput of the gene-knockout labs, and have broadened the types of mutations that we, and others, can identify. In this review, we will discuss these different approaches.
Collapse
Affiliation(s)
- Donald G Moerman
- Department of Zoology, University of British Columbia, Life Sciences Centre, 2350 Health Sciences Mall, Vancouver B.C. V6T 1Z3 Canada.
| | | |
Collapse
|