Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Dis Model Mech ; 14(2)2021 02 05.
Article in English | MEDLINE | ID: mdl-33729986

ABSTRACT

Gene trapping is a high-throughput approach that has been used to introduce insertional mutations into the genome of mouse embryonic stem (ES) cells. It is performed with generic gene trap vectors that simultaneously mutate and report the expression of the endogenous gene at the site of insertion and provide a DNA sequence tag for the rapid identification of the disrupted gene. Large-scale international efforts assembled a gene trap library of 566,554 ES cell lines with single gene trap integrations distributed throughout the genome. Here, we re-investigated this unique library and identified mutations in 2202 non-coding RNA (ncRNA) genes, in addition to mutations in 12,078 distinct protein-coding genes. Moreover, we found certain types of gene trap vectors preferentially integrating into genes expressing specific long non-coding RNA (lncRNA) biotypes. Together with all other gene-trapped ES cell lines, lncRNA gene-trapped ES cell lines are readily available for functional in vitro and in vivo studies.


Subject(s)
Embryonic Stem Cells/metabolism , Genetic Techniques , Mutation , RNA, Untranslated , Animals , Cell Line , Disease Models, Animal , Embryonic Stem Cells/cytology , Exons , Gene Library , Genetic Vectors , Genome , In Vitro Techniques , Mice , Mouse Embryonic Stem Cells , Mutagenesis, Site-Directed , Phenotype , RNA, Long Noncoding/metabolism , RNA, Untranslated/metabolism , Software
3.
Methods ; 164-165: 59-66, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30797903

ABSTRACT

Systematic protein localization and protein-protein interaction studies to characterize specific protein functions are most effectively performed using tag-based assays. Ideally, protein tags are introduced into a gene of interest by homologous recombination to ensure expression from endogenous control elements. However, inefficient homologous recombination makes this approach difficult in mammalian cells. Although gene targeting efficiency by homologous recombination increased dramatically with the development of designer endonuclease systems such as CRISPR/Cas9 capable of inducing DNA double-strand breaks with unprecedented accuracy, the strategies still require synthesis or cloning of homology templates for every single gene. Recent developments have shown that endogenous protein tagging can be achieved efficiently in a homology independent manner. Hence, combinations between CRISPR/Cas9 and generic tag-donor plasmids have been used successfully for targeted gene modifications in mammalian cells. Here, we developed a tool kit comprising a CRISPR/Cas9 expression vector with several EGFP encoding plasmids that should enable tagging of almost every protein expressed in mammalian cells. By performing protein-protein interaction and subcellular localization studies of mTORC1 signal transduction pathway-related proteins expressed in HEK293T cells, we show that tagged proteins faithfully reflect the behavior of their native counterparts under physiological conditions.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing/methods , Gene Targeting/methods , Protein Interaction Mapping/methods , Recombinant Fusion Proteins/genetics , Chromatography, Affinity/instrumentation , Chromatography, Affinity/methods , Gene Editing/instrumentation , Gene Targeting/instrumentation , Genes, Reporter/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/isolation & purification , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/isolation & purification , Mechanistic Target of Rapamycin Complex 1/metabolism , Microscopy, Confocal/instrumentation , Microscopy, Confocal/methods , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Plasmids/genetics , Protein Interaction Mapping/instrumentation , Proteomics/methods , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics , Transfection/instrumentation , Transfection/methods
4.
Drug Discov Today Technol ; 28: 33-39, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30205879

ABSTRACT

The development of genome editing tools capable of modifying specific genomic sequences with unprecedented accuracy has opened up a wide range of new possibilities in targeted gene manipulation. In particular, the CRISPR/Cas9 system, a repurposed prokaryotic adaptive immune system, has been widely adopted because of its unmatched simplicity and flexibility. In this review we discuss achievements and current limitations of CRISPR/Cas9 genome editing in hematopoietic cells with special emphasis on its potential use in ex vivo gene therapy of monogenic blood disorders, HIV and cancer.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Genetic Therapy/methods , Acquired Immunodeficiency Syndrome/genetics , Acquired Immunodeficiency Syndrome/therapy , Animals , Gene Targeting/methods , Hematologic Diseases/genetics , Hematologic Diseases/therapy , Humans , Neoplasms/genetics , Neoplasms/therapy
5.
Mol Ther Nucleic Acids ; 10: 1-8, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29499925

ABSTRACT

The CRISPR/Cas9 prokaryotic adaptive immune system and its swift repurposing for genome editing enables modification of any prespecified genomic sequence with unprecedented accuracy and efficiency, including targeted gene repair. We used the CRISPR/Cas9 system for targeted repair of patient-specific point mutations in the Cytochrome b-245 heavy chain gene (CYBB), whose inactivation causes chronic granulomatous disease (XCGD)-a life-threatening immunodeficiency disorder characterized by the inability of neutrophils and macrophages to produce microbicidal reactive oxygen species (ROS). We show that frameshift mutations can be effectively repaired in hematopoietic cells by non-integrating lentiviral vectors carrying RNA-guided Cas9 endonucleases (RGNs). Because about 25% of most inherited blood disorders are caused by frameshift mutations, our results suggest that up to a quarter of all patients suffering from monogenic blood disorders could benefit from gene therapy employing personalized, donor template-free RGNs.

6.
Nat Commun ; 9(1): 1281, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29599503

ABSTRACT

Nitric oxide (NO) synthesis is a late event during differentiation of mouse embryonic stem cells (mESC) and occurs after release from serum and leukemia inhibitory factor (LIF). Here we show that after release from pluripotency, a subpopulation of mESC, kept in the naive state by 2i/LIF, expresses endothelial nitric oxide synthase (eNOS) and endogenously synthesizes NO. This eNOS/NO-positive subpopulation (ESNO+) expresses mesendodermal markers and is more efficient in the generation of cardiovascular precursors than eNOS/NO-negative cells. Mechanistically, production of endogenous NO triggers rapid Hdac2 S-nitrosylation, which reduces association of Hdac2 with the transcriptional repression factor Zeb1, allowing mesendodermal gene expression. In conclusion, our results suggest that the interaction between Zeb1, Hdac2, and eNOS is required for early mesendodermal differentiation of naive mESC.


Subject(s)
Histone Deacetylase 2/metabolism , Mouse Embryonic Stem Cells/cytology , Myocardium/cytology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/biosynthesis , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Cell Differentiation/physiology , Cell Line, Tumor , HeLa Cells , Humans , Leukemia Inhibitory Factor/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Myocardium/metabolism
7.
Circ Res ; 122(1): 31-46, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29158345

ABSTRACT

RATIONALE: Human cardiac mesenchymal cells (CMSCs) are a therapeutically relevant primary cell population. Diabetes mellitus compromises CMSC function as consequence of metabolic alterations and incorporation of stable epigenetic changes. OBJECTIVE: To investigate the role of α-ketoglutarate (αKG) in the epimetabolic control of DNA demethylation in CMSCs. METHODS AND RESULTS: Quantitative global analysis, methylated and hydroxymethylated DNA sequencing, and gene-specific GC methylation detection revealed an accumulation of 5-methylcytosine, 5-hydroxymethylcytosine, and 5-formylcytosine in the genomic DNA of human CMSCs isolated from diabetic donors. Whole heart genomic DNA analysis revealed iterative oxidative cytosine modification accumulation in mice exposed to high-fat diet (HFD), injected with streptozotocin, or both in combination (streptozotocin/HFD). In this context, untargeted and targeted metabolomics indicated an intracellular reduction of αKG synthesis in diabetic CMSCs and in the whole heart of HFD mice. This observation was paralleled by a compromised TDG (thymine DNA glycosylase) and TET1 (ten-eleven translocation protein 1) association and function with TET1 relocating out of the nucleus. Molecular dynamics and mutational analyses showed that αKG binds TDG on Arg275 providing an enzymatic allosteric activation. As a consequence, the enzyme significantly increased its capacity to remove G/T nucleotide mismatches or 5-formylcytosine. Accordingly, an exogenous source of αKG restored the DNA demethylation cycle by promoting TDG function, TET1 nuclear localization, and TET/TDG association. TDG inactivation by CRISPR/Cas9 knockout or TET/TDG siRNA knockdown induced 5-formylcytosine accumulation, thus partially mimicking the diabetic epigenetic landscape in cells of nondiabetic origin. The novel compound (S)-2-[(2,6-dichlorobenzoyl)amino]succinic acid (AA6), identified as an inhibitor of αKG dehydrogenase, increased the αKG level in diabetic CMSCs and in the heart of HFD and streptozotocin mice eliciting, in HFD, DNA demethylation, glucose uptake, and insulin response. CONCLUSIONS: Restoring the epimetabolic control of DNA demethylation cycle promises beneficial effects on cells compromised by environmental metabolic changes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Ketoglutaric Acids/metabolism , Mesenchymal Stem Cells/metabolism , Mixed Function Oxygenases/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins/metabolism , Thymine DNA Glycosylase/metabolism , Animals , Cells, Cultured , Cytosine/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Enzyme Inhibitors/pharmacology , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Ketoglutaric Acids/antagonists & inhibitors , Male , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Oxidation-Reduction/drug effects
8.
Dis Model Mech ; 10(7): 909-922, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28424158

ABSTRACT

Gene trap mutagenesis is a powerful tool to create loss-of-function mutations in mice and other model organisms. Modifications of traditional gene trap cassettes, including addition of conditional features in the form of Flip-excision (FlEx) arrays to enable directional gene trap cassette inversions by Cre and Flpe site-specific recombinases, greatly enhanced their experimental potential. By taking advantage of these conditional gene trap cassettes, we developed a generic strategy for generating conditional mutations and validated this strategy in mice carrying a multipurpose allele of the Prdm16 transcription factor gene. We demonstrate that the gene trap insertion creates a null mutation replicating the Pierre Robin sequence-type cleft palate phenotype of other Prdm16 mutant mice. Consecutive breeding to Flpe and Emx1IREScre deleter mice spatially restricted Prdm16 loss to regions of the forebrain expressing the homeobox gene Emx1, demonstrating the utility of the technology for the analysis of tissue-specific gene functions.


Subject(s)
Alleles , DNA-Binding Proteins/genetics , Gene Targeting , Transcription Factors/genetics , Animals , Brain/metabolism , Breeding , Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Gene Deletion , Genes, Reporter , Genetic Vectors/metabolism , Head/embryology , Mice , Mutation/genetics , Organ Specificity , Phenotype
9.
Sci Rep ; 7: 43714, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28252045

ABSTRACT

Microfibrils are exracellular matrix components necessary for elastic fiber assembly and for suspending lenses. We previously reported that latent TGF-ß binding protein 2 (LTBP-2), a microfibril-associated protein, is required for forming stable microfibril bundles in ciliary zonules. However, it was not understood why Ltbp2 null mice only showed an eye-specific phenotype, whereas LTBP-2 is abundantly expressed in other tissues containing microfibrils in wild type mice. Here, we show that LTBP-4, another microfibril-associated protein, compensates for the loss of LTBP-2 in microfibril formation. Ltbp2/4S double knockout (DKO) mice showed increased lethality due to emphysema, which was much more severe than that found in Ltbp4S null mice. Elastic fibers in the lungs of Ltbp2/4S DKO mice were severely disorganized and fragmented. Cultured mouse embryonic fibroblasts (MEFs) from Ltbp2/4S DKO embryos developed reduced microfibril meshwork in serum-free conditions, whereas the microfibril formation was restored by the addition of either recombinant LTBP-2 or -4. Finally, ectopic expression of LTBP-4 in the whole body restored ciliary zonule microfibril bundles in the eyes of Ltbp2 null mice. These data suggest that LTBP-2 and -4 have critical overlapping functions in forming the robust structure of microfibrils in vitro and in vivo.


Subject(s)
Latent TGF-beta Binding Proteins/metabolism , Microfibrils/metabolism , Animals , Cilia/metabolism , Emphysema/genetics , Emphysema/metabolism , Emphysema/pathology , Emphysema/physiopathology , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Gene Expression , Genotype , Latent TGF-beta Binding Proteins/genetics , Lung/metabolism , Lung/pathology , Lung/ultrastructure , Mice , Mice, Knockout , Mutation , Phenotype , Protein Binding , RNA, Messenger/genetics
10.
Matrix Biol ; 59: 109-120, 2017 05.
Article in English | MEDLINE | ID: mdl-27645114

ABSTRACT

Latent transforming growth factor beta binding protein 4 (LTBP4) belongs to the fibrillin/LTBP family of proteins and plays an important role as a structural component of extracellular matrix (ECM) and local regulator of TGFß signaling. We have previously reported that Ltbp4S knock out mice (Ltbp4S-/-) develop centrilobular emphysema reminiscent of late stage COPD, which could be partially rescued by inactivating the antioxidant protein Sestrin 2 (Sesn2). More recent studies showed that Sesn2 knock out mice upregulate Pdgfrß-controlled alveolar maintenance programs that protect against cigarette smoke induced pulmonary emphysema. Based on this, we hypothesized that the emphysema of Ltbp4S-/- mice is primarily caused by defective Pdgfrß signaling. Here we show that LTBP4 induces Pdgfrß signaling by inhibiting the antioxidant Nrf2/Keap1 pathway in a TGFß-dependent manner. Overall, our data identified Ltbp4 as a major player in lung remodeling and injury repair.


Subject(s)
Extracellular Matrix/metabolism , Latent TGF-beta Binding Proteins/genetics , NF-E2-Related Factor 2/genetics , Pulmonary Emphysema/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Latent TGF-beta Binding Proteins/deficiency , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Mink , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peroxidases , Plasmids/chemistry , Plasmids/metabolism , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Tropoelastin/deficiency , Tropoelastin/genetics
11.
Dis Model Mech ; 9(11): 1367-1374, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27585882

ABSTRACT

LTBP-4L and LTBP-4S are two isoforms of the extracellular matrix protein latent-transforming growth factor beta-binding protein 4 (LTBP-4). The mutational inactivation of both isoforms causes autosomal recessive cutis laxa type 1C (ARCL1C) in humans and an ARCL1C-like phenotype in Ltbp4-/- mice, both characterized by high postnatal mortality and severely affected elastogenesis. However, genetic data in mice suggest isoform-specific functions for Ltbp-4 because Ltbp4S-/- mice, solely expressing Ltbp-4L, survive to adulthood. This clearly suggests a requirement of Ltbp-4L for postnatal survival. A major difference between Ltbp4S-/- and Ltbp4-/- mice is the matrix incorporation of fibulin-4 (a key factor for elastogenesis; encoded by the Efemp2 gene), which is normal in Ltbp4S-/- mice, whereas it is defective in Ltbp4-/- mice, suggesting that the presence of Ltbp-4L might be required for this process. To investigate the existence of a functional interaction between Ltbp-4L and fibulin-4, we studied the consequences of fibulin-4 deficiency in mice only expressing Ltbp-4L. Resulting Ltbp4S-/-;Fibulin-4R/R mice showed a dramatically reduced lifespan compared to Ltbp4S-/- or Fibulin-4R/R mice, which survive to adulthood. This dramatic reduction in survival of Ltbp4S-/-;Fibulin-4R/R mice correlates with severely impaired elastogenesis resulting in defective alveolar septation and distal airspace enlargement in lung, and increased aortic wall thickness with severely fragmented elastic lamellae. Additionally, Ltbp4S-/-;Fibulin-4R/R mice suffer from aortic aneurysm formation combined with aortic tortuosity, in contrast to Ltbp4S-/- or Fibulin-4R/R mice. Together, in accordance with our previous biochemical findings of a physical interaction between Ltbp-4L and fibulin-4, these novel in vivo data clearly establish a functional link between Ltbp-4L and fibulin-4 as a crucial molecular requirement for survival and elastogenesis in mice.


Subject(s)
Elastin/metabolism , Extracellular Matrix Proteins/metabolism , Latent TGF-beta Binding Proteins/metabolism , Animals , Mice , Models, Biological , Protein Binding , Survival Analysis
12.
Cell Rep ; 11(12): 1847-55, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26095368

ABSTRACT

The ability of hematopoietic stem cells (HSCs) to self-renew is a prerequisite for the establishment of definitive hematopoiesis and life-long blood regeneration. Here, we report the single-stranded DNA-binding transcriptional regulator far upstream element (FUSE)-binding protein 1 (FUBP1) as an essential factor of HSC self-renewal. Functional inactivation of FUBP1 in two different mouse models resulted in embryonic lethal anemia at around E15.5 caused by severely diminished HSCs. Fetal and adult HSCs lacking FUBP1 revealed an HSC-intrinsic defect in their maintenance, expansion, and long-term blood reconstitution, but could differentiate into all hematopoietic lineages. FUBP1-deficient adult HSCs exhibit significant transcriptional changes, including upregulation of the cell-cycle inhibitor p21 and the pro-apoptotic Noxa molecule. These changes caused an increase in generation time and death of HSCs as determined by video-microscopy-based tracking. Our data establish FUBP1 and its recognition of single-stranded genomic DNA as an important element in the transcriptional regulation of HSC self-renewal.


Subject(s)
Cell Self Renewal/genetics , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Fetal Stem Cells/cytology , Fetal Stem Cells/metabolism , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Mice , Signal Transduction/genetics
13.
J Biol Chem ; 290(15): 9738-52, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25716320

ABSTRACT

We recently identified the antioxidant protein Sestrin 2 (Sesn2) as a suppressor of platelet-derived growth factor receptor ß (Pdgfrß) signaling and Pdgfrß signaling as an inducer of lung regeneration and injury repair. Here, we identified Sesn2 and the antioxidant gene inducer nuclear factor erythroid 2-related factor 2 (Nrf2) as positive regulators of proteasomal function. Inactivation of Sesn2 or Nrf2 induced reactive oxygen species-mediated proteasomal inhibition and Pdgfrß accumulation. Using bacterial artificial chromosome (BAC) transgenic HeLa and mouse embryonic stem cells stably expressing enhanced green fluorescent protein-tagged Sesn2 at nearly endogenous levels, we also showed that Sesn2 physically interacts with 2-Cys peroxiredoxins and Nrf2 albeit under different reductive conditions. Overall, we characterized a novel, redox-sensitive Sesn2/Pdgfrß suppressor pathway that negatively interferes with lung regeneration and is up-regulated in the emphysematous lungs of patients with chronic obstructive pulmonary disease (COPD).


Subject(s)
NF-E2-Related Factor 2/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Adult , Animals , Blotting, Western , Cell Line , Cells, Cultured , Female , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Humans , Lung/metabolism , Lung/pathology , Male , Mice, Knockout , Microscopy, Confocal , Middle Aged , NF-E2-Related Factor 2/genetics , Nuclear Proteins/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction/genetics , Young Adult
14.
Dis Model Mech ; 8(4): 403-15, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25713297

ABSTRACT

Recent studies have revealed an important role for LTBP-4 in elastogenesis. Its mutational inactivation in humans causes autosomal recessive cutis laxa type 1C (ARCL1C), which is a severe disorder caused by defects of the elastic fiber network. Although the human gene involved in ARCL1C has been discovered based on similar elastic fiber abnormalities exhibited by mice lacking the short Ltbp-4 isoform (Ltbp4S(-/-)), the murine phenotype does not replicate ARCL1C. We therefore inactivated both Ltbp-4 isoforms in the mouse germline to model ARCL1C. Comparative analysis of Ltbp4S(-/-) and Ltbp4-null (Ltbp4(-/-)) mice identified Ltbp-4L as an important factor for elastogenesis and postnatal survival, and showed that it has distinct tissue expression patterns and specific molecular functions. We identified fibulin-4 as a previously unknown interaction partner of both Ltbp-4 isoforms and demonstrated that at least Ltbp-4L expression is essential for incorporation of fibulin-4 into the extracellular matrix (ECM). Overall, our results contribute to the current understanding of elastogenesis and provide an animal model of ARCL1C.


Subject(s)
Cutis Laxa/genetics , Cutis Laxa/pathology , Genes, Recessive , Latent TGF-beta Binding Proteins/genetics , Animals , Animals, Newborn , Aorta/abnormalities , Aorta/pathology , Cardiomegaly/complications , Cardiomegaly/pathology , Elastic Tissue/metabolism , Elastin/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gene Silencing , Glycosylation , Heart Ventricles/pathology , Humans , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/deficiency , Latent TGF-beta Binding Proteins/metabolism , Lung/abnormalities , Lung/pathology , Mice, Inbred C57BL , Models, Biological , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Skin/pathology , Weight Loss
15.
Methods Enzymol ; 533: 133-55, 2013.
Article in English | MEDLINE | ID: mdl-24182921

ABSTRACT

This is a protocol that describes the generation of targeted embryonic stem (ES) cell clones. The targeted cells can be used for generating a mouse either by injection into blastocysts or by morula aggregation. Alternatively, the ES cells can be used for targeting the second allele and thus creating an in-vitro knockout model. In the latter case, the phenotype of the mutation can be analyzed either in the undifferentiated state or after differentiation of the cells into the three germ layers (endoderm, mesoderm, and ectoderm). This protocol describes only a part of the pipeline for generating a conditional knockout mouse. The whole procedure includes (1) design and generation of the targeting construct, (2) generation of targeted ES clones, and (3) generation of the knockout mouse. Detailed protocols for preparing DNA, culturing ES cells, and screening the transfected ES clones for correct targeted events by long-range PCR or Southern blotting can be found elsewhere (see Isolation of Genomic DNA from Mammalian Cells and Analysis of DNA by Southern Blotting). Here, we describe only the protocol used for transfecting the targeting construct into ES cells and for removing antibiotic selection cassettes or other DNA fragments flanked by site-specific recombination target sites using transient transfection of recombinase expression vectors. In addition, we describe a short protocol for screening the clones that underwent complete recombination. A protocol to prepare DNA from 96-, 48-, and 24-well plates is also described.


Subject(s)
Embryonic Stem Cells , Gene Targeting/methods , Recombination, Genetic , Animals , Anti-Bacterial Agents/pharmacology , Blotting, Southern , Cells, Cultured , DNA/isolation & purification , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Gene Targeting/instrumentation , Genetic Vectors , Mice , Mice, Knockout , Polymerase Chain Reaction , Recombinases/genetics , Selection, Genetic , Transfection/methods
16.
Dis Model Mech ; 6(6): 1378-87, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24046361

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide. COPD is caused by chronic exposure to cigarette smoke and/or other environmental pollutants that are believed to induce reactive oxygen species (ROS) that gradually disrupt signalling pathways responsible for maintaining lung integrity. Here we identify the antioxidant protein sestrin-2 (SESN2) as a repressor of PDGFRß signalling, and PDGFRß signalling as an upstream regulator of alveolar maintenance programmes. In mice, the mutational inactivation of Sesn2 prevents the development of cigarette-smoke-induced pulmonary emphysema by upregulating PDGFRß expression via a selective accumulation of intracellular superoxide anions (O2(-)). We also show that SESN2 is overexpressed and PDGFRß downregulated in the emphysematous lungs of individuals with COPD and to a lesser extent in human lungs of habitual smokers without COPD, implicating a negative SESN2-PDGFRß interrelationship in the pathogenesis of COPD. Taken together, our results imply that SESN2 could serve as both a biomarker and as a drug target in the clinical management of COPD.


Subject(s)
Nuclear Proteins/physiology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Emphysema/etiology , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Signal Transduction/physiology , Smoke , Up-Regulation , Animals , Humans , Lung/metabolism , Mice , Mice, Knockout , Nuclear Proteins/genetics , Peroxidases , Receptor, Platelet-Derived Growth Factor beta/metabolism , Superoxides/metabolism
17.
Genes Dev ; 27(8): 859-72, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23599346

ABSTRACT

Cohesin plays an important role in chromatid cohesion and has additional functions in higher-order chromatin organization and in transcriptional regulation. The binding of cohesin to euchromatic regions is largely mediated by CTCF or the mediator complex. However, it is currently unknown how cohesin is recruited to pericentric heterochromatin in mammalian cells. Here we define the histone methyltransferase Suv4-20h2 as a major structural constituent of heterochromatin that mediates chromatin compaction and cohesin recruitment. Suv4-20h2 stably associates with pericentric heterochromatin through synergistic interactions with multiple heterochromatin protein 1 (HP1) molecules, resulting in compaction of heterochromatic regions. Suv4-20h mutant cells display an overall reduced chromatin compaction and an altered chromocenter organization in interphase referred to as "chromocenter scattering." We found that Suv4-20h-deficient cells display chromosome segregation defects during mitosis that coincide with reduced sister chromatid cohesion. Notably, cohesin subunits interact with Suv4-20h2 both in vitro and in vivo. This interaction is necessary for cohesin binding to heterochromatin, as Suv4-20h mutant cells display substantially reduced cohesin levels at pericentric heterochromatin. This defect is most prominent in G0-phase cells, where cohesin is virtually lost from heterochromatin, suggesting that Suv4-20h2 is involved in the initial loading or maintenance of cohesion subunits. In summary, our data provide the first compelling evidence that Suv4-20h2 plays essential roles in regulating nuclear architecture and ensuring proper chromosome segregation.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Animals , Cell Line , Chromosome Segregation/physiology , Histone-Lysine N-Methyltransferase/genetics , Mice , Mutation , Protein Structure, Tertiary , Protein Transport , Cohesins
18.
Antioxid Redox Signal ; 19(17): 2013-23, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-23495831

ABSTRACT

AIMS: Neuropathic pain is a chronic debilitating disease that is often unresponsive to currently available treatments. Emerging lines of evidence indicate that reactive oxygen species (ROS) are required for the development and maintenance of neuropathic pain. However, little is known about endogenous mechanisms that neutralize the pain-relevant effects of ROS. In the present study, we tested whether the stress-responsive antioxidant protein Sestrin 2 (Sesn2) blocks the ROS-induced neuropathic pain processing in vivo. RESULTS: We observed that Sesn2 mRNA and protein expression was up-regulated in peripheral nerves after spared nerve injury, a well-characterized model of neuropathic pain. Sesn2 knockout (Sesn2(-/-)) mice exhibited considerably increased late-phase neuropathic pain behavior, while their behavior in acute nociceptive and in inflammatory pain models remained unaffected. The exacerbated neuropathic pain behavior of Sesn2(-/-) mice was associated with elevated ROS levels and an enhanced activating transcription factor 3 up-regulation in sensory neurons, and it was reversed by the ROS scavenger N-tert-Butyl-α-phenylnitrone. In contrast, administration of the ROS donor tert-butyl hydroperoxide induced a prolonged pain behavior in naive Sesn2(-/-) mice. INNOVATION: We show that the antioxidant function of Sesn2 limits neuropathic pain processing in vivo. CONCLUSION: Sesn2 controls ROS-dependent neuropathic pain signaling after peripheral nerve injury and may, thus, provide a potential new target for the clinical management of chronic neuropathic pain conditions.


Subject(s)
Neuralgia/metabolism , Nuclear Proteins/physiology , Peripheral Nerve Injuries/metabolism , Activating Transcription Factor 3/metabolism , Animals , Antioxidants/physiology , Female , Ganglia, Spinal/metabolism , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Nociception , Peripheral Nerve Injuries/pathology , Peroxidases , Reactive Oxygen Species/metabolism , Sensory Receptor Cells/metabolism , Up-Regulation , Zymosan
19.
Proc Natl Acad Sci U S A ; 110(8): 2852-7, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23382201

ABSTRACT

Elastic fiber assembly requires deposition of elastin monomers onto microfibrils, the mechanism of which is incompletely understood. Here we show that latent TGF-ß binding protein 4 (LTBP-4) potentiates formation of elastic fibers through interacting with fibulin-5, a tropoelastin-binding protein necessary for elastogenesis. Decreased expression of LTBP-4 in human dermal fibroblast cells by siRNA treatment abolished the linear deposition of fibulin-5 and tropoelastin on microfibrils. It is notable that the addition of recombinant LTBP-4 to cell culture medium promoted elastin deposition on microfibrils without changing the expression of elastic fiber components. This elastogenic property of LTBP-4 is independent of bound TGF-ß because TGF-ß-free recombinant LTBP-4 was as potent an elastogenic inducer as TGF-ß-bound recombinant LTBP-4. Without LTBP-4, fibulin-5 and tropoelastin deposition was discontinuous and punctate in vitro and in vivo. These data suggest a unique function for LTBP-4 during elastic fibrogenesis, making it a potential therapeutic target for elastic fiber regeneration.


Subject(s)
Extracellular Matrix Proteins/metabolism , Latent TGF-beta Binding Proteins/physiology , Recombinant Proteins/metabolism , Animals , HEK293 Cells , Humans , Latent TGF-beta Binding Proteins/metabolism , Mice , Mice, Knockout , Protein Binding , RNA Interference
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL