1
|
Zhang L, Jia R, Palange NJ, Satheka AC, Togo J, An Y, Humphrey M, Ban L, Ji Y, Jin H, Feng X, Zheng Y. Large genomic fragment deletions and insertions in mouse using CRISPR/Cas9. PLoS One 2015; 10:e0120396. [PMID: 25803037 PMCID: PMC4372442 DOI: 10.1371/journal.pone.0120396] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 01/21/2015] [Indexed: 11/18/2022] Open
Abstract
ZFN, TALENs and CRISPR/Cas9 system have been used to generate point mutations and large fragment deletions and insertions in genomic modifications. CRISPR/Cas9 system is the most flexible and fast developing technology that has been extensively used to make mutations in all kinds of organisms. However, the most mutations reported up to date are small insertions and deletions. In this report, CRISPR/Cas9 system was used to make large DNA fragment deletions and insertions, including entire Dip2a gene deletion, about 65kb in size, and β-galactosidase (lacZ) reporter gene insertion of larger than 5kb in mouse. About 11.8% (11/93) are positive for 65kb deletion from transfected and diluted ES clones. High targeting efficiencies in ES cells were also achieved with G418 selection, 46.2% (12/26) and 73.1% (19/26) for left and right arms respectively. Targeted large fragment deletion efficiency is about 21.4% of live pups or 6.0% of injected embryos. Targeted insertion of lacZ reporter with NEO cassette showed 27.1% (13/48) of targeting rate by ES cell transfection and 11.1% (2/18) by direct zygote injection. The procedures have bypassed in vitro transcription by directly co-injection of zygotes or co-transfection of embryonic stem cells with circular plasmid DNA. The methods are technically easy, time saving, and cost effective in generating mouse models and will certainly facilitate gene function studies.
Collapse
Affiliation(s)
- Luqing Zhang
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, China
| | - Ruirui Jia
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Norberto J. Palange
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | | | - Jacques Togo
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yao An
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Mabwi Humphrey
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Luying Ban
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yan Ji
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Honghong Jin
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Xuechao Feng
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, China
- * E-mail: (XCF); (YWZ)
| | - Yaowu Zheng
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Northeast Normal University, Changchun, China
- * E-mail: (XCF); (YWZ)
| |
Collapse
|
2
|
Gama Sosa MA, De Gasperi R, Elder GA. Modeling human neurodegenerative diseases in transgenic systems. Hum Genet 2011; 131:535-63. [PMID: 22167414 DOI: 10.1007/s00439-011-1119-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 11/23/2011] [Indexed: 02/07/2023]
Abstract
Transgenic systems are widely used to study the cellular and molecular basis of human neurodegenerative diseases. A wide variety of model organisms have been utilized, including bacteria (Escherichia coli), plants (Arabidopsis thaliana), nematodes (Caenorhabditis elegans), arthropods (Drosophila melanogaster), fish (zebrafish, Danio rerio), rodents (mouse, Mus musculus and rat, Rattus norvegicus) as well as non-human primates (rhesus monkey, Macaca mulatta). These transgenic systems have enormous value for understanding the pathophysiological basis of these disorders and have, in some cases, been instrumental in the development of therapeutic approaches to treat these conditions. In this review, we discuss the most commonly used model organisms and the methodologies available for the preparation of transgenic organisms. Moreover, we provide selected examples of the use of these technologies for the preparation of transgenic animal models of neurodegenerative diseases, including Alzheimer's disease (AD), frontotemporal lobar degeneration (FTLD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) and Parkinson's disease (PD) and discuss the application of these technologies to AD as an example of how transgenic modeling has affected the study of human neurodegenerative diseases.
Collapse
Affiliation(s)
- Miguel A Gama Sosa
- Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA.
| | | | | |
Collapse
|
3
|
Gene Targeting Vector Design for Embryonic Stem Cell Modifications. SPRINGER PROTOCOLS HANDBOOKS 2011. [DOI: 10.1007/978-3-662-45763-4_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
4
|
Bhattaram P, Penzo-Méndez A, Sock E, Colmenares C, Kaneko KJ, Vassilev A, DePamphilis ML, Wegner M, Lefebvre V. Organogenesis relies on SoxC transcription factors for the survival of neural and mesenchymal progenitors. Nat Commun 2010; 1:9. [PMID: 20596238 PMCID: PMC2892298 DOI: 10.1038/ncomms1008] [Citation(s) in RCA: 174] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Accepted: 03/05/2010] [Indexed: 12/19/2022] Open
Abstract
During organogenesis, neural and mesenchymal progenitor cells give rise to many cell lineages, but their molecular requirements for self-renewal and lineage decisions are incompletely understood. In this study, we show that their survival critically relies on the redundantly acting SoxC transcription factors Sox4, Sox11 and Sox12. The more SoxC alleles that are deleted in mouse embryos, the more severe and widespread organ hypoplasia is. SoxC triple-null embryos die at midgestation unturned and tiny, with normal patterning and lineage specification, but with massively dying neural and mesenchymal progenitor cells. Specific inactivation of SoxC genes in neural and mesenchymal cells leads to selective apoptosis of these cells, suggesting SoxC cell-autonomous roles. Tead2 functionally interacts with SoxC genes in embryonic development, and is a direct target of SoxC proteins. SoxC genes therefore ensure neural and mesenchymal progenitor cell survival, and function in part by activating this transcriptional mediator of the Hippo signalling pathway.
Collapse
Affiliation(s)
- Pallavi Bhattaram
- Department of Cell Biology, and Orthopaedic and Rheumatologic Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, USA
| | - Alfredo Penzo-Méndez
- Department of Cell Biology, and Orthopaedic and Rheumatologic Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, USA
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Clemencia Colmenares
- Department of Cancer Biology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, USA
| | - Kotaro J. Kaneko
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Alex Vassilev
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Melvin L. DePamphilis
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Véronique Lefebvre
- Department of Cell Biology, and Orthopaedic and Rheumatologic Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio 44195, USA
| |
Collapse
|
5
|
Hall B, Limaye A, Kulkarni AB. Overview: generation of gene knockout mice. CURRENT PROTOCOLS IN CELL BIOLOGY 2009; Chapter 19:Unit 19.12 19.12.1-17. [PMID: 19731224 PMCID: PMC2782548 DOI: 10.1002/0471143030.cb1912s44] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The technique of gene targeting allows for the introduction of engineered genetic mutations into a mouse at a determined genomic locus. The process of generating mouse models with targeted mutations was developed through both the discovery of homologous recombination and the isolation of murine embryonic stem cells (ES cells). Homologous recombination is a DNA repair mechanism that is employed in gene targeting to insert a designed mutation into the homologous genetic locus. Targeted homologous recombination can be performed in murine ES cells through electroporation of a targeting construct. These ES cells are totipotent and, when injected into a mouse blastocyst, they can differentiate into all cell types of a chimeric mouse. A chimeric mouse harboring cells derived from the targeted ES cell clone can then generate a whole mouse containing the desired targeted mutation. The initial step for the generation of a mouse with a targeted mutation is the construction of an efficient targeting vector that will be introduced into the ES cells.
Collapse
Affiliation(s)
- Bradford Hall
- Department of Health and Human Services, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
6
|
Kurokawa T, Murashita K. Genomic characterization of multiple leptin genes and a leptin receptor gene in the Japanese medaka, Oryzias latipes. Gen Comp Endocrinol 2009; 161:229-37. [PMID: 19523397 DOI: 10.1016/j.ygcen.2009.01.008] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 12/28/2008] [Accepted: 01/07/2009] [Indexed: 12/22/2022]
Abstract
We comprehensively surveyed leptin (LEP) and leptin receptor (LEPR) genes in medaka, Oryzias latipes and identified two LEP (mLEP-A and mLEP-B) genes and one LEPR (mLEPR) gene. The gene arrangement around both mLEPs in medaka chromosomes 6 and 23 were well conserved with human chromosome 7q31 including LEP. This means that both mLEP-A and mLEP-B are orthologs of human LEP and paralogs derived from whole-genome duplication early in the teleost lineage. The expression of mLEP-A mRNA was relatively high in the liver, and mLEP-B was expressed in the brain and eye. The 3-D modeling of both mLEP-A and mLEP-B protein showed conservation of the four-helix structure that is characteristic in vertebrate leptin. Human LEPR and leptin receptor overlapping the transcript (LEPROT) genes are continuously located on chromosome 1p31. In contrast, medaka LEPR and LEPROT are located on chromosomes 4 and 17, respectively, but both genomic regions showed genomic synteny with the human genome around the LEPR on chromosome 1p31. This result could mean that the medaka chromosome regions around the LEPR and LEPROT are paralogous genomic regions derived from whole-genome duplication, and that the overlapping gene of LEPR and LEPROT was subsequently lost in the medaka genome.
Collapse
Affiliation(s)
- Tadahide Kurokawa
- Tohoku National Fisheries Research Institute, Fisheries Research Agency, Coastal Fisheries and Aquaculture Division, Shinhama Shiogama, Miyagi, Japan.
| | | |
Collapse
|
7
|
Abstract
Sox5 belongs to the Sry-related HMG box gene family, which encodes transcription factors controlling cell fate and differentiation in many lineages. Sox5 produces a long L-Sox5 protein in neuronal, glial, neural crest, cartilage, and other cells, and a short Sox5 protein in spermatids. Sox5(-/-) mice have revealed essential roles for L-Sox5 in development but their neonatal death has prevented postnatal studies. We show here that we have generated mice harboring a conditional null allele for L-Sox5 (Sox5(fl+)) by flanking the fifth coding exon with loxP sites. Cre recombinase-mediated conversion of Sox5(fl+) into Sox5(fl-) abolishes L-Sox5 expression. Expectedly, Sox5(fl+/fl+) mice are indistinguishable from wildtype mice, and Sox5(fl-/fl-) mice from Sox5(-/-) mice. Moreover, the chondrodysplasia of Sox5(fl+/fl+)Sox6(fl+/fl+)Prx1Cre mice demonstrates that the two redundant chondrogenic Sox genes can be efficiently inactivated in a cell type-specific manner. This Sox5 conditional null allele will be valuable in further uncovering the in vivo roles of L-Sox5.
Collapse
Affiliation(s)
- Peter Dy
- Department of Cell Biology and Orthopaedic Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Yu Han
- Department of Cell Biology and Orthopaedic Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Véronique Lefebvre
- Department of Cell Biology and Orthopaedic Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
8
|
Penzo-Méndez A, Dy P, Pallavi B, Lefebvre V. Generation of mice harboring a Sox4 conditional null allele. Genesis 2008; 45:776-80. [PMID: 18064674 DOI: 10.1002/dvg.20358] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Sox4 belongs to the family of Sry-related HMG box transcription factors, which specify cell fate and differentiation in many lineages. Sox4 is widely expressed in the embryo and controls such processes as neuronal tissue, lymphocyte, heart, and bone development. Sox4-null mice die at embryonic day 14 from heart malformation. This early lethality has therefore limited studies on Sox4 functions. We show here that we have generated mice harboring a Sox4 conditional null allele (Sox4fl+) by flanking the entire coding region with loxP sites. Sox4fl+/fl+ mice are indistinguishable from wildtype mice and produce the wildtype Sox4 protein at a normal level. Sox4fl+ is efficiently converted into a null allele (Sox4fl-) by Cre recombinase in somatic and germ-line cells, and Sox4fl-/fl- embryos die from the same heart defects as Sox4-/- mice. This Sox4 conditional null allele will thus be a valuable tool to further uncovering Sox4 functions in various processes in vivo.
Collapse
Affiliation(s)
- Alfredo Penzo-Méndez
- Department of Cell Biology and Orthopaedic Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
9
|
Tao Q, Fujimoto J, Men T, Ye X, Deng J, Lacroix L, Clifford JL, Mao L, Van Pelt CS, Lee JJ, Lotan D, Lotan R. Identification of the retinoic acid-inducible Gprc5a as a new lung tumor suppressor gene. J Natl Cancer Inst 2007; 99:1668-82. [PMID: 18000218 DOI: 10.1093/jnci/djm208] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Lung cancers develop via multiple genetic and epigenetic changes, including inactivation of tumor suppressor genes. We previously cloned human G protein-coupled receptor family C type 5A (GPRC5A), whose expression is suppressed in some human lung carcinoma cells, and its mouse homolog Gprc5a. METHODS We generated Gprc5a knockout mice by homologous recombination and studied their phenotype by macroscopic observation and microscopic histologic analysis of embryos and lungs of 1- to 2-year-old mice. GPRC5A mRNA expression was analyzed by reverse transcription-polymerase chain reaction in surgical specimens of 18 human lung tumors and adjacent normal tissues and by analyzing previously published data from 186 lung tumor tissues of a variety of histologic types and 17 normal lung samples. Human embryonic kidney, human non-small-cell lung cancer, and mouse lung adenocarcinoma cells were transfected with a GPRC5A expression vector or a control vector, and colony formation in semisolid medium was assayed. Statistical tests were two-sided. RESULTS Homozygous knockout mice developed many more lung tumors at 1-2 years of age (incidence: 76% adenomas and 17% adenocarcinomas) than heterozygous (11% adenomas) or wild-type (10% adenomas) mice. Human GPRC5A mRNA levels were lower in most (11 of 18 [61%]) human lung tumors than in adjacent normal tissues. The mean GPRC5A mRNA level in adenocarcinoma (n = 139), squamous cell carcinoma (n = 21), small-cell lung cancer (n = 6), and carcinoid (n = 20) tissues was 46.2% (P = .014), 7.5% (P<.001), 5.3% (P<.001), and 1.8% (P<.001), respectively, that in normal lung tissues (n = 17) GPRC5A transfection suppressed colony formation in semisolid medium of immortalized human embryonic kidney, human non-small-cell lung cancer, and mouse lung adenocarcinoma cells by 91%, 91%, and 68%, respectively, compared with vector controls (all P<.001). CONCLUSIONS Gprc5a functions as a tumor suppressor in mouse lung, and human GPRC5A may share this property. The Gprc5a-deficient mouse is a novel model to study lung carcinogenesis and chemoprevention.
Collapse
Affiliation(s)
- Qingguo Tao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Trejo JL, Carro E, Burks DJ. Experimental models for understanding the role of insulin-like growth factor-I and its receptor during development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 567:27-53. [PMID: 16370135 DOI: 10.1007/0-387-26274-1_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
|
11
|
Dumitriu B, Dy P, Smits P, Lefebvre V. Generation of mice harboring aSox6 conditional null allele. Genesis 2006; 44:219-24. [PMID: 16652367 DOI: 10.1002/dvg.20210] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Sox6 belongs to the family of Sry-related HMG box transcription factors, which determine cell fate and differentiation in various lineages. Sox6 is expressed in several tissues, including cartilage, testis, neuronal, and erythropoietic tissues. Mice lacking Sox6 have revealed critical roles for Sox6 in several of these tissues, but their multiple defects and early lethality has limited studies in specific cell types and in postnatal mice. We show here that we have generated mice harboring a Sox6 conditional null allele (Sox6(fl+)) by flanking the second coding exon with loxP sites. This allele encodes wildtype Sox6 protein, is expressed normally, and is efficiently converted into a null allele (Sox6(fl-)) by Cre-mediated recombination in somatic and germ cells. Sox6(fl+/fl+) mice are indistinguishable from wildtype mice, and Sox6(fl-/fl-) mice from Sox6(-/-) mice. These Sox6 conditional null mice will thus be valuable for further uncovering the roles of Sox6 in various processes in vivo.
Collapse
Affiliation(s)
- Bogdan Dumitriu
- Department of Biomedical Engineering and Orthopaedic Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
12
|
Hamra FK, Chapman KM, Nguyen DM, Williams-Stephens AA, Hammer RE, Garbers DL. Self renewal, expansion, and transfection of rat spermatogonial stem cells in culture. Proc Natl Acad Sci U S A 2005; 102:17430-5. [PMID: 16293688 PMCID: PMC1283987 DOI: 10.1073/pnas.0508780102] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The use of a transgenic line of rats that express enhanced GFP (EGFP) exclusively in the germ line has allowed a separation of feeder layers and contaminating testis somatic cells from germ cells and the identification of a set of spermatogonial stem cell marker transcripts. With these molecular markers as a guide, we have now devised culture conditions where rat spermatogonial stem cells renew and proliferate in culture with a doubling time between 3 and 4 days. The marker transcripts increase in relative abundance as a function of time in culture, and the stem cells retain competency to colonize and develop into spermatids after transplantation to the testes of recipient rats. The cells also remain euploid after at least 12 passages. Cell lines could be isolated and cryopreserved and, upon subsequent thawing, continue to self renew. Transfection of the spermatogonial stem cells with a plasmid containing the neomycin phosphotransferase (neo) selectable marker resulted in selection of G418-resistant cell lines that effectively colonize recipient testes, suggesting that gene targeting is now feasible in the rat.
Collapse
Affiliation(s)
- F Kent Hamra
- The Cecil H. and Ida Green Center for Reproductive Biology Sciences, Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9051, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Sorrell DA, Kolb AF. Targeted modification of mammalian genomes. Biotechnol Adv 2005; 23:431-69. [PMID: 15925473 DOI: 10.1016/j.biotechadv.2005.03.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2004] [Revised: 03/14/2005] [Accepted: 03/14/2005] [Indexed: 12/22/2022]
Abstract
The stable and site-specific modification of mammalian genomes has a variety of applications in biomedicine and biotechnology. Here we outline two alternative approaches that can be employed to achieve this goal: homologous recombination (HR) or site-specific recombination. Homologous recombination relies on sequence similarity (or rather identity) of a piece of DNA that is introduced into a host cell and the host genome. In most cell types, the frequency of homologous recombination is markedly lower than the frequency of random integration. Especially in somatic cells, homologous recombination is an extremely rare event. However, recent strategies involving the introduction of DNA double-strand breaks, triplex forming oligonucleotides or adeno-associated virus can increase the frequency of homologous recombination. Site-specific recombination makes use of enzymes (recombinases, transposases, integrases), which catalyse DNA strand exchange between DNA molecules that have only limited sequence homology. The recognition sites of site-specific recombinases (e.g. Cre, Flp or PhiC31 integrase) are usually 30-50 bp. In contrast, retroviral integrases only require a specific dinucleotide sequence to insert the viral cDNA into the host genome. Depending on the individual enzyme, there are either innumerable or very few potential target sites for a particular integrase/recombinase in a mammalian genome. A number of strategies have been utilised successfully to alter the site-specificity of recombinases. Therefore, site-specific recombinases provide an attractive tool for the targeted modification of mammalian genomes.
Collapse
Affiliation(s)
- David A Sorrell
- Molecular Recognition Group, Hannah Research Institute, Ayr, KA6 5HL, UK
| | | |
Collapse
|
14
|
Valenzuela DM, Murphy AJ, Frendewey D, Gale NW, Economides AN, Auerbach W, Poueymirou WT, Adams NC, Rojas J, Yasenchak J, Chernomorsky R, Boucher M, Elsasser AL, Esau L, Zheng J, Griffiths JA, Wang X, Su H, Xue Y, Dominguez MG, Noguera I, Torres R, Macdonald LE, Stewart AF, DeChiara TM, Yancopoulos GD. High-throughput engineering of the mouse genome coupled with high-resolution expression analysis. Nat Biotechnol 2003; 21:652-9. [PMID: 12730667 DOI: 10.1038/nbt822] [Citation(s) in RCA: 462] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2003] [Accepted: 03/28/2003] [Indexed: 01/22/2023]
Abstract
One of the most effective approaches for determining gene function involves engineering mice with mutations or deletions in endogenous genes of interest. Historically, this approach has been limited by the difficulty and time required to generate such mice. We describe the development of a high-throughput and largely automated process, termed VelociGene, that uses targeting vectors based on bacterial artificial chromosomes (BACs). VelociGene permits genetic alteration with nucleotide precision, is not limited by the size of desired deletions, does not depend on isogenicity or on positive-negative selection, and can precisely replace the gene of interest with a reporter that allows for high-resolution localization of target-gene expression. We describe custom genetic alterations for hundreds of genes, corresponding to about 0.5-1.0% of the entire genome. We also provide dozens of informative expression patterns involving cells in the nervous system, immune system, vasculature, skeleton, fat and other tissues.
Collapse
Affiliation(s)
- David M Valenzuela
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill Road, Tarrytown, New York 10591, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
In the postgenomic era the mouse will be central to the challenge of ascribing a function to the 40,000 or so genes that constitute our genome. In this review, we summarize some of the classic and modern approaches that have fueled the recent dramatic explosion in mouse genetics. Together with the sequencing of the mouse genome, these tools will have a profound effect on our ability to generate new and more accurate mouse models and thus provide a powerful insight into the function of human genes during the processes of both normal development and disease.
Collapse
|
16
|
Hamra FK, Gatlin J, Chapman KM, Grellhesl DM, Garcia JV, Hammer RE, Garbers DL. Production of transgenic rats by lentiviral transduction of male germ-line stem cells. Proc Natl Acad Sci U S A 2002; 99:14931-6. [PMID: 12391306 PMCID: PMC137522 DOI: 10.1073/pnas.222561399] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Primary cultures of rat spermatogenic cells that did not bind to collagen matrices were able to colonize and form mature spermatozoa when transferred to testes of recipient males. Up to 73% of the progeny from matings with recipient males were derived from the transferred spermatogenic cells. Subsequently, two populations of germ cells were obtained by selection on laminin matrices. Both populations expressed the spermatogenic cell marker, DAZL, but not the somatic cell marker, vimentin. The cells that bound to laminin represented approximately 5% of the total population and were greatly enriched in ability to colonize a recipient testis, suggesting an enrichment in germ-line stem cells. The colonization potential was maintained for at least 7 days in culture. These cells were subsequently transduced with a lentiviral enhanced GFP reporter vector and then transferred to WT recipient males. After mating, 26 of 44 pups were derived from the cultured donor germ cells, and 13 pups carried the lentiviral transgene. Based on Southern analysis, the transgene was integrated at a different genetic locus in each animal and was transmitted to approximately 50% of pups in the F(2) generation. Thus, by using these procedures, approximately 30% of pups in the F(1) generation inherited and stably transmitted a lentiviral transgene that integrated at various genomic sites.
Collapse
Affiliation(s)
- F Kent Hamra
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, Dallas, TX 75390, USA
| | | | | | | | | | | | | |
Collapse
|