Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Transgenic Res ; 26(2): 237-246, 2017 04.
Article in English | MEDLINE | ID: mdl-27873161

ABSTRACT

In spite of the emergence of genome editing tools, ES cell mediated transgenesis remains the most controllable way of creating genetically modified animals. Although tetraploid (4N) complementation of 4N host embryos and ES cells, is the only method guaranteeing that offspring are entirely ES cell derived, this technique is challenging, not always successful and difficult to implement in some laboratory settings. The current study shows that pretreatment of host blastocysts with FGF4 prior to ES cell injection can provide an alternative method for the generation of animals displaying high rates of chimaerism. Chimaerism assessment in E11 fetuses and born pups shows that a large percentage of resulting conceptuses show a high ES cell contribution from implantation onwards and that developing pups do not necessitate c-section for delivery.


Subject(s)
Animals, Genetically Modified/genetics , Embryonic Development/genetics , Embryonic Stem Cells , Fibroblast Growth Factor 4/genetics , Animals , Animals, Genetically Modified/growth & development , Blastocyst/metabolism , Chimera/genetics , Chimera/growth & development , Cloning, Organism , Fibroblast Growth Factor 4/metabolism , Gene Editing/methods , Gene Transfer Techniques , Mice
2.
PLoS One ; 7(12): e51272, 2012.
Article in English | MEDLINE | ID: mdl-23272095

ABSTRACT

The human ß-globin locus contains the ß-like globin genes (i.e. fetal γ-globin and adult ß-globin), which heterotetramerize with α-globin subunits to form fetal or adult hemoglobin. Thalassemia is one of the commonest inherited disorders in the world, which results in quantitative defects of the globins, based on a number of genome variations found in the globin gene clusters. Hereditary persistence of fetal hemoglobin (HPFH) also caused by similar types of genomic alterations can compensate for the loss of adult hemoglobin. Understanding the regulation of the human γ-globin gene expression is a challenge for the treatment of thalassemia. A mouse model that facilitates high-throughput assays would simplify such studies. We have generated a transgenic dual reporter mouse model by tagging the γ- and ß-globin genes with GFP and DsRed fluorescent proteins respectively in the endogenous human ß-globin locus. Erythroid cell lines derived from this mouse model were tested for their capacity to reactivate the γ-globin gene. Here, we discuss the applications and limitations of this fluorescent reporter model to study the genetic basis of red blood cell disorders and the potential use of such model systems in high-throughput screens for hemoglobinopathies therapeutics.


Subject(s)
beta-Globins/genetics , gamma-Globins/genetics , Animals , Cell Line , Disease Models, Animal , Erythrocytes/cytology , Fetal Hemoglobin/metabolism , Fluorescent Dyes/pharmacology , Genes, Reporter , Genes, p53 , Green Fluorescent Proteins/metabolism , Hemoglobinopathies/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Multigene Family
3.
BMC Dev Biol ; 12: 18, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22690876

ABSTRACT

BACKGROUND: To facilitate the in vivo study of esophageal (stem) cell biology in homeostasis and cancer, novel mouse models are necessary to elicit expression of candidate genes in a tissue-specific and inducible fashion. To this aim, we developed and studied a mouse model to allow labeling of esophageal cells with the histone 2B-GFP (H2B-GFP) fusion protein. RESULTS: First, we generated a transgenic mouse model expressing the reverse tetracycline transactivator rtTA2-M2 under control of the promoter (ED-L2) of the Epstein-Barr virus (EBV) gene encoding the latent membrane protein-1 (LMP-1). The newly generated ED-L2-rtTA2-M2 (ED-L2-rtTA) mice were then bred with the previously developed tetO-HIST1H2BJ/GFP (tetO-H2B-GFP) model to assess inducibility and tissue-specificity. Expression of the H2B-GFP fusion protein was observed upon doxycycline induction but was restricted to the terminally differentiated cells above the basal cell layer. To achieve expression in the basal compartment of the esophagus, we subsequently employed a different transgenic model expressing the reverse transactivator rtTA2S-M2 under the control of the ubiquitous, methylation-free CpG island of the human hnRNPA2B1-CBX3 gene (hnRNP-rtTA). Upon doxycycline administration to the compound hnRNP-rtTA/tetO-H2B-GFP mice, near-complete labeling of all esophageal cells was achieved. Pulse-chase experiments confirmed that complete turnover of the esophageal epithelium in the adult mouse is achieved within 7-10 days. CONCLUSIONS: We show that the esophagus-specific promoter ED-L2 is expressed only in the differentiated cells above the basal layer. Moreover, we confirmed that esophageal turn-over in the adult mouse does not exceed 7-10 days.


Subject(s)
Esophagus/metabolism , Genetic Engineering , Transcriptional Activation , Animals , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/genetics , Epithelium/metabolism , Esophagus/cytology , Female , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Histones/biosynthesis , Histones/genetics , Humans , Keratins/metabolism , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Trans-Activators/genetics
4.
PLoS Genet ; 6(11): e1001203, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21085687

ABSTRACT

SUMO modification of transcription factors is linked to repression of transcription. The physiological significance of SUMO attachment to a particular transcriptional regulator, however, is largely unknown. We have employed the ubiquitously expressed murine transcription factor Sp3 to analyze the role of SUMOylation in vivo. We generated mice and mouse embryonic fibroblasts (MEFs) carrying a subtle point mutation in the SUMO attachment sequence of Sp3 (IKEE(553)D mutation). The E(553)D mutation impedes SUMOylation of Sp3 at K(551)in vivo, without affecting Sp3 protein levels. Expression profiling revealed that spermatocyte-specific genes, such as Dmc1 and Dnahc8, and neuronal genes, including Paqr6, Rims3, and Robo3, are de-repressed in non-testicular and extra-neuronal mouse tissues and in mouse embryonic fibroblasts expressing the SUMOylation-deficient Sp3E(553)D mutant protein. Chromatin immunoprecipitation experiments show that transcriptional de-repression of these genes is accompanied by the loss of repressive heterochromatic marks such as H3K9 and H4K20 tri-methylation and impaired recruitment of repressive chromatin-modifying enzymes. Finally, analysis of the DNA methylation state of the Dmc1, Paqr6, and Rims3 promoters by bisulfite sequencing revealed that these genes are highly methylated in Sp3wt MEFs but are unmethylated in Sp3E(553)D MEFs linking SUMOylation of Sp3 to tissue-specific CpG methylation. Our results establish SUMO conjugation to Sp3 as a molecular beacon for the assembly of repression machineries to maintain tissue-specific transcriptional gene silencing.


Subject(s)
Gene Silencing , Neurons/metabolism , Sp3 Transcription Factor/genetics , Spermatocytes/metabolism , Sumoylation/genetics , Animals , Chromatin/metabolism , DNA Methylation/genetics , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Targeting , Male , Meiosis/genetics , Mice , Mice, Mutant Strains , Neurons/cytology , Organ Specificity/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sp3 Transcription Factor/metabolism
5.
Blood ; 110(13): 4188-97, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17823307

ABSTRACT

The transcription factor Runx1/AML1 is an important regulator of hematopoiesis and is critically required for the generation of the first definitive hematopoietic stem cells (HSCs) in the major vasculature of the mouse embryo. As a pivotal factor in HSC ontogeny, its transcriptional regulation is of high interest but is largely undefined. In this study, we used a combination of comparative genomics and chromatin analysis to identify a highly conserved 531-bp enhancer located at position + 23.5 in the first intron of the 224-kb mouse Runx1 gene. We show that this enhancer contributes to the early hematopoietic expression of Runx1. Transcription factor binding in vivo and analysis of the mutated enhancer in transient transgenic mouse embryos implicate Gata2 and Ets proteins as critical factors for its function. We also show that the SCL/Lmo2/Ldb-1 complex is recruited to the enhancer in vivo. Importantly, transplantation experiments demonstrate that the intronic Runx1 enhancer targets all definitive HSCs in the mouse embryo, suggesting that it functions as a crucial cis-regulatory element that integrates the Gata, Ets, and SCL transcriptional networks to initiate HSC generation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/physiology , GATA2 Transcription Factor/physiology , Hematopoietic Stem Cells/cytology , Proto-Oncogene Protein c-ets-1/physiology , Proto-Oncogene Proteins/physiology , Transcription, Genetic , Adaptor Proteins, Signal Transducing , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Enhancer Elements, Genetic/physiology , GATA2 Transcription Factor/metabolism , LIM Domain Proteins , Metalloproteins/metabolism , Mice , Multiprotein Complexes/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1
6.
Exp Hematol ; 32(2): 224-33, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15102485

ABSTRACT

OBJECTIVE: Persistent expression of the human fetal gamma-globin genes in the adult stage is often associated with naturally occurring deletions in the human beta-globin locus. The mapping of the 5' breakpoints of these deletions within the Agamma- to delta-globin intergenic region has suggested that regulatory elements involved in the silencing of the gamma-globin genes in the adult may be present. We previously identified two elements in this region, termed Enh and F, located 3' to the Agamma-globin gene acting as silencers in transient transfection assays. Here, we tested directly the in vivo significance of these elements in the developmental regulation of the human beta-like globin genes. MATERIALS AND METHODS. We selectively deleted both Enh and F elements in the context of a 185-kb human beta-globin locus PAC (P1 phage artificial chromosome) and tested the effects of this deletion on the expression of the human P-like globin genes in transgenic mice. RESULTS: The Enh/F deletion resulted in an increase in epsilon- and gamma-globin mRNA levels in the embryonic yolk sac stage of erythropoiesis, which appears to be due to an increase in the rate of transcription rather than to an increase in the number of cells transcribing the human globin locus. However, the human developmental switching from fetal gamma-globin to adult beta-globin gene expression in transgenic mice was not affected by this deletion. CONCLUSION: These results identify Enh and F as locus-wide regulatory elements capable of down-regulating transcription of the human beta-globin locus in an embryonic-specific manner.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Globins/genetics , Repressor Proteins/physiology , Animals , Chromosome Mapping , Escherichia coli/genetics , Gene Deletion , Humans , In Situ Hybridization , Mice , Mice, Transgenic , Recombination, Genetic
7.
Br J Haematol ; 116(2): 401-8, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11841445

ABSTRACT

The Sca-1 surface glycoprotein is used routinely as a marker for haematopoietic stem cell enrichment. Two allelic genes, Ly-6A and Ly-6E, encode this marker and appear to be differentially regulated in haematopoietic cells and haematopoietic stem cells. The Sca-1 protein has been shown to be expressed at a greater frequency in these cells from Ly-6A strains of mice. To study the specific expression pattern and haematopoietic regulation of the Ly-6A gene, we constructed a 14 kb cassette from a genomic Ly-6A fragment, inserted a lacZ reporter gene and created transgenic mice. We found that the Ly-6A lacZ transgene was expressed in the haematopoietic tissues and predominantly in the T-lymphoid lineage. Some expression was also found in the B-lymphoid and myeloid lineages. We demonstrated functional haematopoietic stem cell enrichment by sorting for beta-galactosidase-expressing cells from the bone marrow. In addition, we found an interesting embryonic expression pattern in the AGM region, the site of the first haematopoietic stem cell generation. Surprisingly, when compared with data from Ly-6E lacZ transgenic mice, our results suggest that the Ly-6A cassette does not improve lacZ marker gene expression in haematopoietic cells.


Subject(s)
Antigens, Ly/genetics , Embryo, Mammalian/metabolism , Hematopoietic Stem Cells/metabolism , Membrane Proteins/genetics , T-Lymphocytes/metabolism , Animals , Blotting, Northern/methods , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Female , Flow Cytometry , Gene Expression , Genetic Engineering , Genetic Markers , Lac Operon , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...