Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
Cell Rep ; 40(8): 111249, 2022 08 23.
Article in English | MEDLINE | ID: mdl-36001963

ABSTRACT

The microtubule-associated protein tau is an abundant component of neurons of the central nervous system. In Alzheimer's disease and other neurodegenerative tauopathies, tau is found hyperphosphorylated and aggregated in neurofibrillary tangles. To obtain a better understanding of the cellular perturbations that initiate tau pathogenesis, we performed a CRISPR-Cas9 screen for genetic modifiers that enhance tau aggregation. This initial screen yielded three genes, BANF1, ANKLE2, and PPP2CA, whose inactivation promotes the accumulation of tau in a phosphorylated and insoluble form. In a complementary screen, we identified three additional genes, LEMD2, LEMD3, and CHMP7, that, when overexpressed, provide protection against tau aggregation. The proteins encoded by the identified genes are mechanistically linked and recognized for their roles in the maintenance and repair of the nuclear envelope. These results implicate the disruption of nuclear envelope integrity as a possible initiating event in tauopathies and reveal targets for therapeutic intervention.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Membrane Proteins/metabolism , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Tauopathies/metabolism , tau Proteins/genetics , tau Proteins/metabolism
3.
Nat Commun ; 12(1): 2770, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986266

ABSTRACT

CRISPR-based transcriptional activation is a powerful tool for functional gene interrogation; however, delivery difficulties have limited its applications in vivo. Here, we created a mouse model expressing all components of the CRISPR-Cas9 guide RNA-directed Synergistic Activation Mediator (SAM) from a single transcript that is capable of activating target genes in a tissue-specific manner. We optimized Lipid Nanoparticles and Adeno-Associated Virus guide RNA delivery approaches to achieve expression modulation of one or more genes in vivo. We utilized the SAM mouse model to generate a hypercholesteremia disease state that we could bidirectionally modulate with various guide RNAs. Additionally, we applied SAM to optimize gene expression in a humanized Transthyretin mouse model to recapitulate human expression levels. These results demonstrate that the SAM gene activation platform can facilitate in vivo research and drug discovery.


Subject(s)
CRISPR-Cas Systems/genetics , Hypercholesterolemia/genetics , Liposomes/pharmacology , Prealbumin/metabolism , Transcriptional Activation/genetics , Animals , Cell Line , Gene Expression/genetics , Gene Expression Regulation/genetics , Genetic Engineering/methods , HEK293 Cells , Humans , Hypercholesterolemia/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nanoparticles , Prealbumin/genetics , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
4.
PLoS One ; 10(8): e0136336, 2015.
Article in English | MEDLINE | ID: mdl-26302002

ABSTRACT

Exons are ligated in an ordered manner without the skipping of exons in the constitutive splicing of pre-mRNAs with multiple introns. To identify factors ensuring ordered exon joining in constitutive pre-mRNA splicing, we previously screened for exon skipping mutants in Schizosaccharomyces pombe using a reporter plasmid, and characterized three exon skipping mutants named ods1 (ordered splicing 1), ods2, and ods3, the responsible genes of which encode Prp2/U2AF59, U2AF23, and SF1, respectively. They form an SF1-U2AF59-U2AF23 complex involved in recognition of the branch and 3' splice sites in pre-mRNA. In the present study, we identified a fourth ods mutant, ods4, which was isolated in an exon-skipping screen. The ods4+ gene encodes Cwf16p, which interacts with the NineTeen Complex (NTC), a complex thought to be involved in the first catalytic step of the splicing reaction. We isolated two multi-copy suppressors for the ods4-1 mutation, Srp2p, an SR protein essential for pre-mRNA splicing, and Tif213p, a translation initiation factor, in S. pombe. The overexpression of Srp2p suppressed the exon-skipping phenotype of all ods mutants, whereas Tif213p suppressed only ods4-1, which has a mutation in the translational start codon of the cwf16 gene. We also showed that the decrease in the transcriptional elongation rate induced by drug treatment suppressed exon skipping in ods4-1. We propose that Cwf16p/NTC participates in the early recognition of the branch and 3' splice sites and cooperates with the SF1-U2AF59-U2AF23 complex to maintain ordered exon joining.


Subject(s)
Alternative Splicing/genetics , Multiprotein Complexes/genetics , Schizosaccharomyces pombe Proteins/genetics , Spliceosomes/genetics , Exons/genetics , Introns/genetics , Mutation , RNA Precursors/genetics , RNA Splice Sites/genetics , RNA Splicing/genetics , RNA Splicing Factors , Schizosaccharomyces/genetics
5.
PLoS One ; 10(4): e0125522, 2015.
Article in English | MEDLINE | ID: mdl-25909911

ABSTRACT

In a survey of 20 knockout mouse lines designed to examine the biological functions of large intergenic non-coding RNAs (lincRNAs), we have found a variety of phenotypes, ranging from perinatal lethality to defects associated with premature aging and morphological and functional abnormalities in the lungs, skeleton, and muscle. Each mutant allele carried a lacZ reporter whose expression profile highlighted a wide spectrum of spatiotemporal and tissue-specific transcription patterns in embryos and adults that informed our phenotypic analyses and will serve as a guide for future investigations of these genes. Our study shows that lincRNAs are a new class of encoded molecules that, like proteins, serve essential and important functional roles in embryonic development, physiology, and homeostasis of a broad array of tissues and organs in mammals.


Subject(s)
RNA, Long Noncoding/genetics , Transcription, Genetic/genetics , Transcriptome/genetics , Alleles , Animals , Embryonic Development/genetics , Female , Genes, Reporter/genetics , Male , Mammals/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
6.
Transgenic Res ; 24(1): 19-29, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25087174

ABSTRACT

Known examples of male to female sex reversal in mice are caused by either strain incompatibilities or mutations in genes required for male sex determination. The resultant XY females are often sterile or exhibit very poor fertility. We describe here embryonic stem (ES) cell growth conditions that promote the production of healthy, anatomically normal fertile and fecund female F0 generation mice completely derived from gene-targeted XY male ES cells. The sex reversal is a transient trait that is not transmitted to the F1 progeny. Growth media with low osmolality and reduced sodium bicarbonate, maintained throughout the gene targeting process, enhance the yield of XY females. As a practical application of the induced sex reversal, we demonstrate the generation of homozygous mutant mice ready for phenotypic studies by the breeding of F0 XY females with their isogenic XY male clonal siblings, thereby eliminating one generation of breeding and the associated costs.


Subject(s)
Disorders of Sex Development/genetics , Fertility/genetics , Gonadal Dysgenesis, 46,XY/genetics , Sex Determination Processes , Animals , Embryonic Stem Cells/cytology , Female , Gene Targeting , Male , Mice , Microinjections , Mutation
7.
Proc Natl Acad Sci U S A ; 111(14): 5147-52, 2014 Apr 08.
Article in English | MEDLINE | ID: mdl-24706858

ABSTRACT

Genetic humanization, which involves replacing mouse genes with their human counterparts, can create powerful animal models for the study of human genes and diseases. One important example of genetic humanization involves mice humanized for their Ig genes, allowing for human antibody responses within a mouse background (HumAb mice) and also providing a valuable platform for the generation of fully human antibodies as therapeutics. However, existing HumAb mice do not have fully functional immune systems, perhaps because of the manner in which they were genetically humanized. Heretofore, most genetic humanizations have involved disruption of the endogenous mouse gene with simultaneous introduction of a human transgene at a new and random location (so-called KO-plus-transgenic humanization). More recent efforts have attempted to replace mouse genes with their human counterparts at the same genetic location (in situ humanization), but such efforts involved laborious procedures and were limited in size and precision. We describe a general and efficient method for very large, in situ, and precise genetic humanization using large compound bacterial artificial chromosome-based targeting vectors introduced into mouse ES cells. We applied this method to genetically humanize 3-Mb segments of both the mouse heavy and κ light chain Ig loci, by far the largest genetic humanizations ever described. This paper provides a detailed description of our genetic humanization approach, and the companion paper reports that the humoral immune systems of mice bearing these genetically humanized loci function as efficiently as those of WT mice.


Subject(s)
Genes, Immunoglobulin , Animals , Chromosomes, Artificial, Bacterial , Embryonic Stem Cells/immunology , Homologous Recombination , Humans , Mice , Mice, Knockout , Polymerase Chain Reaction , Transgenes
8.
Breast Cancer Res ; 15(6): R111, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24262428

ABSTRACT

INTRODUCTION: Latent TGFß binding proteins (LTBPs) govern TGFß presentation and activation and are important for elastogenesis. Although TGFß is well-known as a tumor suppressor and metastasis promoter, and LTBP1 is elevated in two distinct breast cancer metastasis signatures, LTBPs have not been studied in the normal mammary gland. METHODS: To address this we have examined Ltbp1 promoter activity throughout mammary development using an Ltbp1L-LacZ reporter as well as expression of both Ltbp1L and 1S mRNA and protein by qRT-PCR, immunofluorescence and flow cytometry. RESULTS: Our data show that Ltbp1L is transcribed coincident with lumen formation, providing a rare marker distinguishing ductal from alveolar luminal lineages. Ltbp1L and Ltbp1S are silent during lactation but robustly induced during involution, peaking at the stage when the remodeling process becomes irreversible. Ltbp1L is also induced within the embryonic mammary mesenchyme and maintained within nipple smooth muscle cells and myofibroblasts. Ltbp1 protein exclusively ensheaths ducts and side branches. CONCLUSIONS: These data show Ltbp1 is transcriptionally regulated in a dynamic manner that is likely to impose significant spatial restriction on TGFß bioavailability during mammary development. We hypothesize that Ltbp1 functions in a mechanosensory capacity to establish and maintain ductal luminal cell fate, support and detect ductal distension, trigger irreversible involution, and facilitate nipple sphincter function.


Subject(s)
Latent TGF-beta Binding Proteins/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/embryology , Mesoderm/cytology , Animals , Cell Lineage , Cell Movement , Female , Gene Expression Regulation, Developmental , Lactation , Latent TGF-beta Binding Proteins/genetics , Mammary Glands, Animal/physiology , Mesoderm/metabolism , Mice, Inbred C57BL , Mice, Mutant Strains , Muscle, Smooth/cytology , Muscle, Smooth/embryology , Pregnancy , Promoter Regions, Genetic , Up-Regulation
9.
Proc Natl Acad Sci U S A ; 110(34): E3179-88, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23918385

ABSTRACT

Conditional mutagenesis is becoming a method of choice for studying gene function, but constructing conditional alleles is often laborious, limited by target gene structure, and at times, prone to incomplete conditional ablation. To address these issues, we developed a technology termed conditionals by inversion (COIN). Before activation, COINs contain an inverted module (COIN module) that lies inertly within the antisense strand of a resident gene. When inverted into the sense strand by a site-specific recombinase, the COIN module causes termination of the target gene's transcription and simultaneously provides a reporter for tracking this event. COIN modules can be inserted into natural introns (intronic COINs) or directly into coding exons as part of an artificial intron (exonic COINs), greatly simplifying allele design and increasing flexibility over previous conditional KO approaches. Detailed analysis of over 20 COIN alleles establishes the reliability of the method and its broad applicability to any gene, regardless of exon-intron structure. Our extensive testing provides rules that help ensure success of this approach and also explains why other currently available conditional approaches often fail to function optimally. Finally, the ability to split exons using the COIN's artificial intron opens up engineering modalities for the generation of multifunctional alleles.


Subject(s)
Alleles , Gene Silencing , Genetic Engineering/methods , Mutagenesis, Insertional/methods , Sequence Inversion/genetics , DNA Nucleotidyltransferases/metabolism
10.
Elife ; 2: e01749, 2013 Dec 31.
Article in English | MEDLINE | ID: mdl-24381249

ABSTRACT

Many studies are uncovering functional roles for long noncoding RNAs (lncRNAs), yet few have been tested for in vivo relevance through genetic ablation in animal models. To investigate the functional relevance of lncRNAs in various physiological conditions, we have developed a collection of 18 lncRNA knockout strains in which the locus is maintained transcriptionally active. Initial characterization revealed peri- and postnatal lethal phenotypes in three mutant strains (Fendrr, Peril, and Mdgt), the latter two exhibiting incomplete penetrance and growth defects in survivors. We also report growth defects for two additional mutant strains (linc-Brn1b and linc-Pint). Further analysis revealed defects in lung, gastrointestinal tract, and heart in Fendrr(-/-) neonates, whereas linc-Brn1b(-/-) mutants displayed distinct abnormalities in the generation of upper layer II-IV neurons in the neocortex. This study demonstrates that lncRNAs play critical roles in vivo and provides a framework and impetus for future larger-scale functional investigation into the roles of lncRNA molecules. DOI: http://dx.doi.org/10.7554/eLife.01749.001.


Subject(s)
Brain/growth & development , RNA, Long Noncoding/physiology , Animals , Mice , Mice, Knockout , RNA, Long Noncoding/genetics
11.
Mamm Genome ; 23(9-10): 580-6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22968824

ABSTRACT

In 2007, the International Knockout Mouse Consortium (IKMC) made the ambitious promise to generate mutations in virtually every protein-coding gene of the mouse genome in a concerted worldwide action. Now, 5 years later, the IKMC members have developed high-throughput gene trapping and, in particular, gene-targeting pipelines and generated more than 17,400 mutant murine embryonic stem (ES) cell clones and more than 1,700 mutant mouse strains, most of them conditional. A common IKMC web portal (www.knockoutmouse.org) has been established, allowing easy access to this unparalleled biological resource. The IKMC materials considerably enhance functional gene annotation of the mammalian genome and will have a major impact on future biomedical research.


Subject(s)
Mice, Knockout/genetics , Animals , Internationality , Internet , Mice
12.
Methods Enzymol ; 476: 285-94, 2010.
Article in English | MEDLINE | ID: mdl-20691872

ABSTRACT

In conventional methods for the generation of genetically modified mice, gene-targeted embryonic stem (ES) cells are injected into blastocyst-stage embryos or are aggregated with morula-stage embryos, which are then transferred to the uterus of a surrogate mother. F0 generation mice born from the embryos are chimeras composed of genetic contributions from both the modified ES cells and the recipient embryos. Obtaining a mouse strain that carries the gene-targeted mutation requires breeding the chimeras to transmit the ES cell genetic component through the germ line to the next (F1) generation (germ line transmission, GLT). To skip the chimera stage, we developed the VelociMouse method, in which injection of genetically modified ES cells into eight-cell embryos followed by maturation to the blastocyst stage and transfer to a surrogate mother produces F0 generation mice that are fully derived from the injected ES cells and exhibit a 100% GLT efficiency. The method is simple and flexible. Both male and female ES cells can be introduced into the eight-cell embryo by any method of injection or aggregation and using all ES cell and host embryo combinations from inbred, hybrid, and outbred genetic backgrounds. The VelociMouse method provides several unique opportunities for shortening project timelines and reducing mouse husbandry costs. First, as VelociMice exhibit 100% GLT, there is no need to test cross chimeras to establish GLT. Second, because the VelociMouse method permits efficient production of ES cell-derived mice from female ES cells, XO ES cell subclones, identified by screening for spontaneous loss of the Y chromosome, can be used to generate F0 females that can be bred with isogenic F0 males derived from the original targeted ES cell clone to obtain homozygous mutant mice in the F1 generation. Third, as VelociMice are genetically identical to the ES cells from which they were derived, the VelociMouse method opens up myriad possibilities for creating mice with complex genotypes in a defined genetic background directly from engineered ES cells without the need for inefficient and lengthy breeding schemes. Examples include creation of F0 knockout mice from ES cells carrying a homozygous null mutation, and creation of a mouse with a tissue-specific gene inactivation by combining null and floxed conditional alleles for the target gene with a transgenic Cre recombinase allele controlled by a tissue-specific promoter. VelociMice with the combinatorial alleles are ready for immediate phenotypic studies, which greatly accelerates gene function assignment and the creation of valuable models of human disease.


Subject(s)
Embryo Culture Techniques/methods , Embryo Transfer/methods , Embryo, Mammalian , Embryonic Stem Cells , Animals , Cell Line , Chimera , Embryo Culture Techniques/instrumentation , Embryo Transfer/instrumentation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Microinjections/methods , Pregnancy
13.
Methods Enzymol ; 476: 295-307, 2010.
Article in English | MEDLINE | ID: mdl-20691873

ABSTRACT

Targeting vectors used to create directed mutations in mouse embryonic stem (ES) cells consist, in their simplest form, of a gene for drug selection flanked by mouse genomic sequences, the so-called homology arms that promote site-directed homologous recombination between the vector and the target gene. The VelociGene method for the creation of targeted mutations in ES cells employs targeting vectors, called BACVecs, that are based on bacterial artificial chromosomes. Compared with conventional short targeting vectors, BacVecs provide two major advantages: (1) their much larger homology arms promote high targeting efficiencies without the need for isogenicity or negative selection strategies; and (2) they enable deletions and insertions of up to 100kb in a single targeting event, making possible gene-ablating definitive null alleles and other large-scale genomic modifications. Because of their large arm sizes, however, BACVecs do not permit screening by conventional assays, such as long-range PCR or Southern blotting, that link the inserted targeting vector to the targeted locus. To exploit the advantages of BACVecs for gene targeting, we inverted the conventional screening logic in developing the loss-of-allele (LOA) assay, which quantifies the number of copies of the native locus to which the mutation was directed. In a correctly targeted ES cell clone, the LOA assay detects one of the two native alleles (for genes not on the X or Y chromosome), the other allele being disrupted by the targeted modification. We apply the same principle in reverse as a gain-of-allele assay to quantify the copy number of the inserted targeting vector. The LOA assay reveals a correctly targeted clone as having lost one copy of the native target gene and gained one copy of the drug resistance gene or other inserted marker. The combination of these quantitative assays makes LOA genotyping unequivocal and amenable to automated scoring. We use the quantitative polymerase chain reaction (qPCR) as our method of allele quantification, but any method that can reliably distinguish the difference between one and two copies of the target gene can be used to develop an LOA assay. We have designed qPCR LOA assays for deletions, insertions, point mutations, domain swaps, conditional, and humanized alleles and have used the insert assays to quantify the copy number of random insertion BAC transgenics. Because of its quantitative precision, specificity, and compatibility with high throughput robotic operations, the LOA assay eliminates bottlenecks in ES cell screening and mouse genotyping and facilitates maximal speed and throughput for knockout mouse production.


Subject(s)
Biological Assay/methods , Embryonic Stem Cells/physiology , Gene Targeting/methods , Genotype , Loss of Heterozygosity , Animals , Biological Assay/instrumentation , Embryonic Stem Cells/cytology , Humans , Mice , Mice, Knockout , Polymerase Chain Reaction/instrumentation , Polymerase Chain Reaction/methods
14.
Methods Mol Biol ; 530: 311-24, 2009.
Article in English | MEDLINE | ID: mdl-19266341

ABSTRACT

With the completion of the human and mouse genome sequences and the development of high-throughput knockout mouse technologies, there is now a need for equally high-throughput methods for the production of mice for phenotypic studies. In response to this challenge, we recently developed a new method termed VelociMouse for the production of F0-generation mice that are fully derived from gene-targeted ES cells. In the version of the VelociMouse method described here, laser ablation of a portion of the zona pellucid (zp) of a normal eight-cell-stage embryo facilitates ES cell injection. Upon gestation in a surrogate mother, the injected embryos produce F0 mice that carry no detectable host embryo contribution (<0.1%). The fully ES cell-derived mice are normal, healthy, and fertile and exhibit 100% germline transmission for optimal breeding efficiency. The VelociMouse method accommodates both inbred or hybrid ES cells and either inbred or outbred eight-cell host embryos. Because the F0 mice produced are suitable for direct phenotyping studies, the VelociMouse method, coupled with high-throughput ES cell targeting technologies, such as VelociGene, offers an accelerated path to new drug target discovery and validation and a revolutionary approach to realize the full value of large-scale functional genomic efforts, such as the NIH Knockout Mouse Project ( 1 ) and the European Conditional Mouse Mutagenesis Project( 9 ).


Subject(s)
Embryo Culture Techniques/methods , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Gene Targeting/methods , Animals , Cell Differentiation , Female , Male , Mice , Mice, Inbred C57BL , Phenotype , Pregnancy
15.
Development ; 134(20): 3723-32, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17804598

ABSTRACT

Latent TGF-beta binding protein 1 (LTBP1) is a member of the LTBP/fibrillin family of extracellular proteins. Due to the usage of different promoters, LTBP1 exists in two major forms, long (L) and short (S), each expressed in a temporally and spatially unique fashion. Both LTBP1 molecules covalently interact with latent TGF-beta and regulate its function, presumably via interaction with the extracellular matrix (ECM). To explore the in vivo role of Ltbp1 in mouse development, at the time when only the L isoform is expressed, we mutated the Ltbp1L locus by gene targeting. Ltbp1L-null animals die shortly after birth from defects in heart development, consisting of the improper septation of the cardiac outflow tract (OFT) and remodeling of the associated vessels. These cardiac anomalies present as persistent truncus arteriosus (PTA) and interrupted aortic arch (IAA), which are associated with the faulty function of cardiac neural crest cells (CNCCs). The lack of Ltbp1L in the ECM of the septating OFT and associated vessels results in altered gene expression and function of CNCCs and decreased Tgf-beta activity in the OFT. This phenotype reveals a crucial role for Ltbp1L and matrix as extracellular regulators of Tgf-beta activity in heart organogenesis.


Subject(s)
Heart Defects, Congenital/genetics , Heart , Latent TGF-beta Binding Proteins/metabolism , Protein Isoforms/metabolism , Animals , Animals, Newborn , Cell Differentiation/physiology , Extracellular Matrix/metabolism , Gene Expression Regulation , Gene Targeting , Heart/anatomy & histology , Heart/embryology , Heart/physiology , Latent TGF-beta Binding Proteins/genetics , Mice , Mice, Knockout , Neural Crest/cytology , Protein Isoforms/genetics , Transforming Growth Factor beta/metabolism
16.
Science ; 316(5824): 604-8, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17463289

ABSTRACT

MicroRNAs are small RNA species involved in biological control at multiple levels. Using genetic deletion and transgenic approaches, we show that the evolutionarily conserved microRNA-155 (miR-155) has an important role in the mammalian immune system, specifically in regulating T helper cell differentiation and the germinal center reaction to produce an optimal T cell-dependent antibody response. miR-155 exerts this control, at least in part, by regulating cytokine production. These results also suggest that individual microRNAs can exert critical control over mammalian differentiation processes in vivo.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , MicroRNAs/physiology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Cells, Cultured , Cytokines/biosynthesis , Immunoglobulin G/analysis , Lymphocyte Activation , Lymphotoxin-alpha/biosynthesis , Lymphotoxin-beta/biosynthesis , Mice , Mice, Knockout , Mice, Transgenic , MicroRNAs/genetics , Nitrophenols/immunology , Peyer's Patches/immunology , Phenylacetates , Somatic Hypermutation, Immunoglobulin , Spleen/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/biosynthesis
17.
J Biol Chem ; 282(4): 2221-8, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17130122

ABSTRACT

To identify genes involved in the mechanism to ensure ordered 5' to 3' exon joining in constitutively spliced pre-mRNAs, we screened for mutants that cause exon skipping in the fission yeast Schizosaccharomyces pombe using a reporter plasmid, which contains the ura4+ gene with the nda3 intron 1-exon 2-intron 2 sequence. The reporter plasmid was designed to produce the functional ura4+ mRNA, when the central nda3 exon is skipped during the splicing reaction. We mutagenized cells harboring the plasmid by UV irradiation and isolated 34 ura+ mutants that grew on minimal medium. Of those, eight mutants were found to be temperature sensitive (ts) for growth. Complementation analyses revealed that the ts mutants belong to three distinct complementation groups named ods (ordered splicing) 1, 2, and 3. RT-PCR analyses showed that products of exon skipping were actually generated in the ods mutants. We cloned the genes responsible for the ods mutations, and found that ods1+, ods2+, and ods3+ encode splicing factors Prp2p/U2AF59, U2AF23, and SF1, respectively, which form a SF1-U2AF59-U2AF23 complex involved in recognition of the branch-point and 3' splice site sequences in a pre-mRNA. We also showed that mutations in the SF1-U2AF59-U2AF23 binding sequences in the reporter plasmid result in exon skipping in wild-type S. pombe cells. In addition, drugs that decrease the rate of transcription elongation were found to suppress the exon skipping in the ods mutants. These results suggest that co-transcriptional recognition of a nascent pre-mRNA by the SF1-U2AF59-U2AF23 complex is essential for ordered exon joining in constitutive splicing in S. pombe.


Subject(s)
RNA Splicing , Schizosaccharomyces/genetics , Base Sequence , Exons , Models, Genetic , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , RNA Precursors/genetics , RNA Splicing Factors , RNA, Fungal/genetics , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoproteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Splicing Factor U2AF
18.
Nat Biotechnol ; 25(1): 91-9, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187059

ABSTRACT

A useful approach for exploring gene function involves generating mutant mice from genetically modified embryonic stem (ES) cells. Recent advances in genetic engineering of ES cells have shifted the bottleneck in this process to the generation of mice. Conventional injections of ES cells into blastocyst hosts produce F0 generation chimeras that are only partially derived from ES cells, requiring additional breeding to obtain mutant mice that can be phenotyped. The tetraploid complementation approach directly yields mice that are almost entirely derived from ES cells, but it is inefficient, works only with certain hybrid ES cell lines and suffers from nonspecific lethality and abnormalities, complicating phenotypic analyses. Here we show that laser-assisted injection of either inbred or hybrid ES cells into eight cell-stage embryos efficiently yields F0 generation mice that are fully ES cell-derived and healthy, exhibit 100% germline transmission and allow immediate phenotypic analysis, greatly accelerating gene function assignment.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Gene Targeting/methods , Laser Therapy/methods , Mice, Transgenic/genetics , Microinjections/methods , Stem Cell Transplantation/methods , Animals , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic/anatomy & histology , Mice, Transgenic/surgery , Microsurgery/methods , Phenotype
19.
Cell ; 124(2): 315-29, 2006 Jan 27.
Article in English | MEDLINE | ID: mdl-16439206

ABSTRACT

The Sir2 histone deacetylase functions as a chromatin silencer to regulate recombination, genomic stability, and aging in budding yeast. Seven mammalian Sir2 homologs have been identified (SIRT1-SIRT7), and it has been speculated that some may have similar functions to Sir2. Here, we demonstrate that SIRT6 is a nuclear, chromatin-associated protein that promotes resistance to DNA damage and suppresses genomic instability in mouse cells, in association with a role in base excision repair (BER). SIRT6-deficient mice are small and at 2-3 weeks of age develop abnormalities that include profound lymphopenia, loss of subcutaneous fat, lordokyphosis, and severe metabolic defects, eventually dying at about 4 weeks. We conclude that one function of SIRT6 is to promote normal DNA repair, and that SIRT6 loss leads to abnormalities in mice that overlap with aging-associated degenerative processes.


Subject(s)
Aging/metabolism , Genetic Diseases, Inborn/genetics , Genomic Instability , Sirtuins/genetics , Sirtuins/physiology , Animals , Cell Proliferation , Chromatin/metabolism , DNA Damage , DNA Repair , Genetic Diseases, Inborn/pathology , Humans , Ki-1 Antigen/metabolism , Lymphocytes/immunology , Mice , Mice, Knockout , Phenotype , Radiation Tolerance , Signal Transduction , Sirtuins/deficiency
20.
Nucleic Acids Res ; 31(24): 7110-6, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14654686

ABSTRACT

Pathways for the maturation of ribosomal RNAs are complex with numerous intermediate cleavage sites that are not always conserved closely in the course of evolution. Both in eukaryotes and bacteria genetic analyses and in vitro studies have strongly implicated RNase III-like enzymes in the processing of rRNA precursors. In Schizosacharomyces pombe, for example, the RNase III-like Pac1 nuclease has been shown to cleave the free 3'ETS at two known intermediate sites but, in the presence of RAC protein, the same RNA also is cleaved at the 3'-end of the 25 S rRNA sequence. In this study normal and mutant 3'ETS sequences were digested with the Pac1 enzyme to further evaluate its role in rRNA processing. Accurate cleavage at the known intermediate processing sites was dependent on the integrity of the helical structure at these sites as well as a more distal upper stem region in the conserved extended hairpin structure of the 3'ETS. The cleavage of mutant 3'ETS sequences also generally correlated with the known effects of these mutations on rRNA production, in vivo. One mutant, however, was efficiently processed in vivo but was not a substrate for the Pac1 nuclease, in vitro. In contrast, in the presence of RAC protein, the same RNA remained susceptible to Pac1 nuclease cleavage at the 3'-end of the 25 rRNA sequence, indicating that the removal of the 3'ETS does not require cleavage at the intermediate sites. These results suggest that basic maturation pathways may be less complex than previously reported raising similar questions about other intermediate processing sites, which have been identified by analyses of termini, and/or processing, in vitro.


Subject(s)
Endoribonucleases/metabolism , Fungal Proteins , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Fungal/metabolism , RNA, Ribosomal/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Base Sequence , Molecular Sequence Data , Mutation/genetics , Nucleic Acid Conformation , RNA Precursors/chemistry , RNA Precursors/genetics , RNA, Fungal/biosynthesis , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Ribosomal/biosynthesis , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Schizosaccharomyces pombe Proteins , Single-Strand Specific DNA and RNA Endonucleases/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...