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.
Clin Exp Pharmacol Physiol ; 42(12): 1275-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26743678

ABSTRACT

Hemangioblast, including primitive hematopoietic progenitor cells, play an important role in hematopoietic development, however, the underlying mechanism for the propagation of hematopoietic progenitor cells remains elusive. A variety of regulatory molecules activated in early embryonic development play a critical role in the maintenance of function of hematopoietic progenitor cells. Homeobox transcription factors are an important class of early embryonic developmental regulators determining hematopoietic development. However, the effect of homeobox protein Hox-B4 (HOXB4) ectopic expression on the development of hemangioblasts has not been fully addressed. This study aimed to investigate the role of Hoxb4a, an ortholog gene of HOXB4 in zebrafish, in the hematopoietic development in zebrafish. A transgenic zebrafish line was established with Cre-loxP system that stably overexpressed enhanced green fluorescent protein (EGFP)-tagged Hoxb4a protein under the control of hemangioblast-specific lmo2 promoter. Overexpression of Hoxb4a in the development of hemangioblasts resulted in a considerable increase in the number of stem cell leukemia (scl) and lmo2-positive primitive hematopoietic progenitor cells occurring in the posterior intermediate cell mass (ICM). Interestingly, Hoxb4a overexpression also disrupted the development of myelomonocytes in the anterior yolk sac and the posterior ICM, without affecting erythropoiesis in the posterior ICM. Taken together, these results indicate that Hoxb4a favours the development of hematopoietic progenitor cells originated from hemangioblasts in vivo.


Subject(s)
Ectopic Gene Expression , Embryonic Development/genetics , Hemangioblasts/metabolism , Hematopoiesis/genetics , Homeodomain Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Animals, Genetically Modified , Base Sequence , Gene Expression Regulation, Developmental , LIM Domain Proteins/genetics , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Recombination, Genetic , Transcription Factors/genetics
2.
Blood ; 115(23): 4715-24, 2010 Jun 10.
Article in English | MEDLINE | ID: mdl-20371743

ABSTRACT

Loss of function of tumor suppressor genes, such as PTEN, CEBPAlpha, and CTNNA1 (encoding the alpha-catenin protein), has been found to play an essential role in leukemogenesis. However, whether these genes genetically interact remains largely unknown. Here, we show that PTEN-mammalian target of rapamycin signaling acts upstream to dictate the ratio of wild-type p42 C/EBPalpha to its dominant-negative p30 isoform, which critically determines whether p30 C/EBPalpha (lower p42/p30 ratio) or p42 C/EBPalpha (higher p42/p30 ratio) binds to the proximal promoter of the retained CTNNA1 allele. Binding of p30 C/EBPalpha recruits the polycomb repressive complex 2 to suppress CTNNA1 transcription through repressive H3K27me3 modification, whereas binding of p42 C/EBPalpha relieves this repression and promotes CTNNA1 expression through activating H3K4me3 modification. Loss of Pten function in mice and zebrafish induces myelodysplasia with abnormal invasiveness of myeloid progenitors accompanied by significant reductions in both wild-type C/EBPalpha and alpha-catenin protein. Importantly, frame-shift mutations in either PTEN or CEBPA were detected exclusively in the primary LICs with low CTNNA1 expression. This study uncovers a novel molecular pathway, PTEN-C/EBPalpha-CTNNA1, which is evolutionarily conserved and might be therapeutically targeted to eradicate LICs with low CTNNA1 expression.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Leukemia/metabolism , Myelopoiesis , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , alpha Catenin/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Transformation, Neoplastic/genetics , Frameshift Mutation , Gene Expression Regulation, Leukemic/genetics , HL-60 Cells , Humans , Leukemia/genetics , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Polycomb-Group Proteins , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , Protein Processing, Post-Translational/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics , Transcription, Genetic/genetics , Zebrafish , alpha Catenin/genetics
3.
PLoS One ; 4(7): e6125, 2009 Jul 03.
Article in English | MEDLINE | ID: mdl-19582161

ABSTRACT

BACKGROUND: Reduced expression of developmentally important genes and tumor suppressors due to haploinsufficiency or epigenetic suppression has been shown to contribute to the pathogenesis of various malignancies. However, methodology that allows spatio-temporally knockdown of gene expression in various model organisms such as zebrafish has not been well established, which largely limits the potential of zebrafish as a vertebrate model of human malignant disorders. PRINCIPAL FINDING: Here, we report that multiple copies of small hairpin RNA (shRNA) are expressed from a single transcript that mimics the natural microRNA-30e precursor (mir-shRNA). The mir-shRNA, when microinjected into zebrafish embryos, induced an efficient knockdown of two developmentally essential genes chordin and alpha-catenin in a dose-controllable fashion. Furthermore, we designed a novel cassette vector to simultaneously express an intronic mir-shRNA and a chimeric red fluorescent protein driven by lineage-specific promoter, which efficiently reduced the expression of a chromosomally integrated reporter gene and an endogenously expressed gata-1 gene in the developing erythroid progenitors and hemangioblasts, respectively. SIGNIFICANCE: This methodology provides an invaluable tool to knockdown developmental important genes in a tissue-specific manner or to establish animal models, in which the gene dosage is critically important in the pathogenesis of human disorders. The strategy should be also applicable to other model organisms.


Subject(s)
Gene Knockdown Techniques , Zebrafish/embryology , Animals , DNA Polymerase II/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Promoter Regions, Genetic , RNA/genetics , Zebrafish/genetics
4.
Biochem Biophys Res Commun ; 381(4): 688-93, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19254693

ABSTRACT

MicroRNAs (miRNAs) are endogenous small non-protein coding RNAs that play important regulatory roles in animals and plants by binding to target transcripts for cleavage or translational repression. Despite increasing number of genes has been predicted to be miRNA targets by bioinformatics analysis and luciferase-based reporter assay in vitro (RA-In Vitro), few of them have been experimentally validated in physiological context. Using transient reporter assay in vivo (TRA-In Vivo), we identified hydroxymethylbilane synthase b (hmbsb) and Krüppel-like transcription factor d (klfd) as potential target gene for miR-451 and miR-144, respectively. Although hmbsb, miR-451, klfd and miR-144 are all co-expressed in the developing erythroid progenitors during zebrafish erythropoiesis, only klfd can be validated as a bona fide physiological target of miR-144 using a transgene-based physiological reporter assay in vivo (PRA-In Vivo). Thus, failure to verify hmbsb as miR-451 target in physiological context raises a crucial question as to how to determine bona fide target of miRNA. The results address the importance of using multiple approaches combined with Western blot analysis to validate the physiological target of a given miRNA.


Subject(s)
Erythropoiesis/genetics , Hydroxymethylbilane Synthase/genetics , Kruppel-Like Transcription Factors/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Erythroid Cells/enzymology , Evolution, Molecular , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Genes, Reporter , Heme/biosynthesis , Zebrafish/genetics , Zebrafish/physiology
5.
Blood ; 113(6): 1340-9, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-18941117

ABSTRACT

Precise transcriptional control of developmental stage-specific expression and switching of alpha- and beta-globin genes is significantly important to understand the general principles controlling gene expression and the pathogenesis of thalassemia. Although transcription factors regulating beta-globin genes have been identified, little is known about the microRNAs and trans-acting mechanism controlling alpha-globin genes transcription. Here, we show that an erythroid lineage-specific microRNA gene, miR-144, expressed at specific developmental stages during zebrafish embryogenesis, negatively regulates the embryonic alpha-globin, but not embryonic beta-globin, gene expression, through physiologically targeting klfd, an erythroid-specific Krüppel-like transcription factor. Klfd selectively binds to the CACCC boxes in the promoters of both alpha-globin and miR-144 genes to activate their transcriptions, thus forming a negative feedback circuitry to fine-tune the expression of embryonic alpha-globin gene. The selective effect of the miR-144-Klfd pathway on globin gene regulation may thereby constitute a novel therapeutic target for improving the clinical outcome of patients with thalassemia.


Subject(s)
Embryo, Nonmammalian/metabolism , Erythropoiesis/physiology , Gene Expression Regulation, Developmental , MicroRNAs/genetics , alpha-Globins/genetics , Animals , Animals, Genetically Modified , Apoptosis , Blotting, Northern , Blotting, Western , Computational Biology , Embryo, Nonmammalian/cytology , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoenzyme Techniques , In Situ Hybridization , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/physiology , MicroRNAs/metabolism , Oligonucleotides/pharmacology , Promoter Regions, Genetic/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/physiology , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/metabolism
6.
J Immunol ; 181(3): 2155-64, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641354

ABSTRACT

Interstitial cell migration through extracellular matrix is a hallmark of the inflammation response, tumor invasion, and metastasis. We have established a stable zebrafish transgenic line expressing enhanced GFP under the lysozyme C promoter for visualizing and measuring primitive macrophage migration in vivo. We show that tissue-resident primitive macrophages migrate rapidly through extracellular matrix to the site of acute injury induced by tail transection. Mechanistically, the specific inhibition of JNK, but not p38 and ERK, dramatically abolished the chemotactic migration in a dose-dependent manner, suppressing the trauma-induced recruitment of phosphorylated C-Jun transcription factor to proximal AP-1 sites in the promoter of matrix metalloproteinase 13 (mmp13), a gene specifically expressed in primitive macrophages during embryogenesis and required for the interstitial migration. Furthermore, dexamethasone suppressed the trauma-induced JNK phosphorylation and macrophage migration accompanied by simultaneous up-regulation of mkp-1, a well-known phosphatase capable of inactivating phosphorylated JNK. The results indicate that the JNK-Mmp13 signaling pathway plays an essential role in regulating the innate immune cell migration in response to severe injury in vivo.


Subject(s)
Intestines/cytology , Intestines/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/cytology , Macrophages/enzymology , Matrix Metalloproteinase 13/metabolism , Signal Transduction , Acute Disease , Animals , Animals, Genetically Modified , Cell Movement/drug effects , Dual Specificity Phosphatase 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucocorticoids/pharmacology , Intestines/embryology , Intestines/injuries , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Macrophages/drug effects , Matrix Metalloproteinase 13/genetics , Molecular Structure , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Transcriptional Activation/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Dev Dyn ; 237(8): 2139-46, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18627109

ABSTRACT

Cre/loxP system is a powerful tool to manipulate the genome. Transgenic animals expressing Cre recombinase in specific tissues or cells have been widely used for conditional gene targeting, lineage tracing, and other genetic analyses. In zebrafish, the transgenic line with stable expression of Cre in specific tissues and cell subtypes has not been generated and its functional activity remains to be defined. Here we report the establishment of a stable transgenic fish Tg(zlmo2:Cre), which specifically expresses Cre in the primitive hematopoietic progenitors and vascular endothelial cells, under the control of lmo2 promoter. Our result shows that the Cre expression pattern recapitulates the endogenous lmo2 expression pattern during embryogenesis. Crossing of the Tg(zlmo2:Cre) line with another established transgenic reporter line Tg(zlmo2:loxP-DsRed-loxP-EGFP), induces a robust recombination activity in hematopoietic progenitors and vascular endothelial cells. Thus, the Tg(zlmo2:Cre) transgenic line provides an invaluable tool to dissect genetic pathways in hematopoietic development and diseases.


Subject(s)
Animals, Genetically Modified/genetics , DNA-Binding Proteins/genetics , Integrases/genetics , Metalloproteins/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Base Sequence , Embryonic Development/physiology , Gene Expression Regulation, Developmental , Genomics , Hematopoietic Stem Cells/physiology , Kidney/embryology , Kidney/physiology , LIM Domain Proteins , Models, Animal , Molecular Sequence Data , Transcription Factors , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...