Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Biol ; 373(2): 431-41, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-22960038

ABSTRACT

Growth Factor Independence (Gfi) transcription factors play essential roles in hematopoiesis, differentially activating and repressing transcriptional programs required for hematopoietic stem/progenitor cell (HSPC) development and lineage specification. In mammals, Gfi1a regulates hematopoietic stem cells (HSC), myeloid and lymphoid populations, while its paralog, Gfi1b, regulates HSC, megakaryocyte and erythroid development. In zebrafish, gfi1aa is essential for primitive hematopoiesis; however, little is known about the role of gfi1aa in definitive hematopoiesis or about additional gfi factors in zebrafish. Here, we report the isolation and characterization of an additional hematopoietic gfi factor, gfi1b. We show that gfi1aa and gfi1b are expressed in the primitive and definitive sites of hematopoiesis in zebrafish. Our functional analyses demonstrate that gfi1aa and gfi1b have distinct roles in regulating primitive and definitive hematopoietic progenitors, respectively. Loss of gfi1aa silences markers of early primitive progenitors, scl and gata1. Conversely, loss of gfi1b silences runx-1, c-myb, ikaros and cd41, indicating that gfi1b is required for definitive hematopoiesis. We determine the epistatic relationships between the gfi factors and key hematopoietic transcription factors, demonstrating that gfi1aa and gfi1b join lmo2, scl, runx-1 and c-myb as critical regulators of teleost HSPC. Our studies establish a comparative paradigm for the regulation of hematopoietic lineages by gfi transcription factors.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Conserved Sequence/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian/metabolism , Epistasis, Genetic , Erythropoiesis/genetics , Evolution, Molecular , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hematopoietic System/embryology , Hematopoietic System/metabolism , Models, Biological , Molecular Sequence Data , Zebrafish/embryology , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
2.
Blood ; 114(21): 4654-63, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19729519

ABSTRACT

The nuclear protein FOG-1 binds transcription factor GATA-1 to facilitate erythroid and megakaryocytic maturation. However, little is known about the function of FOG-1 during myeloid and lymphoid development or how FOG-1 expression is regulated in any tissue. We used in situ hybridization, gain- and loss-of-function studies in zebrafish to address these problems. Zebrafish FOG-1 is expressed in early hematopoietic cells, as well as heart, viscera, and paraspinal neurons, suggesting that it has multifaceted functions in organogenesis. We found that FOG-1 is dispensable for endoderm specification but is required for endoderm patterning affecting the expression of late-stage T-cell markers, independent of GATA-1. The suppression of FOG-1, in the presence of normal GATA-1 levels, induces severe anemia and thrombocytopenia and expands myeloid-progenitor cells, indicating that FOG-1 is required during erythroid/myeloid commitment. To functionally interrogate whether GATA-1 regulates FOG-1 in vivo, we used bioinformatics combined with transgenic assays. Thus, we identified 2 cis-regulatory elements that control the tissue-specific gene expression of FOG-1. One of these enhancers contains functional GATA-binding sites, indicating the potential for a regulatory loop in which GATA factors control the expression of their partner protein FOG-1.


Subject(s)
Embryonic Development/physiology , Gene Expression Regulation, Developmental , Nuclear Proteins , Zebrafish Proteins , Zebrafish/embryology , Animals , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Hematopoiesis/physiology , In Situ Hybridization , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Regulatory Elements, Transcriptional/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
3.
Exp Hematol ; 37(8): 901-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19460416

ABSTRACT

OBJECTIVE: A missense mutation in the microtubule-associated serine/threonine-like kinase gene (MASTL, FLJ14813) on human chromosome 10 was previously linked to a novel form of autosomal dominant inherited thrombocytopenia in a single pedigree. The mutation results in an amino acid change from glutamic acid at position 167 to aspartic acid and segregates perfectly with thrombocytopenic individuals within this extended family. The phenotype is characterized by mild thrombocytopenia with an average platelet count of 60,000 platelets per microliter of blood. We wanted to determine the expression and localization of MASTL, as well as its role in developing thrombocytes using an in vivo model system. MATERIALS AND METHODS: Northern blot analysis allowed us to examine expression patterns. Morpholino knockdown assays in zebrafish (Danio rerio) were employed to determine in vivo contribution to thrombocyte development. Transient expression in baby hamster kidney cells resulted in localization of both the wild-type and E167D mutant forms of MASTL kinase to the nucleus. RESULTS: Northern blot analysis indicates that MASTL messenger RNA is restricted in its expression to hematopoietic and cancer cell lines. A transient knockdown of MASTL in zebrafish results in deficiency of circulating thrombocytes. Transient expression of recombinant MASTL kinase in vitro demonstrates localization to the nucleus. CONCLUSIONS: Functional studies presented here demonstrate a direct relationship between transient knockdown of MASTL kinase gene expression and reduction of circulating thrombocytes in zebrafish. This transient knockdown of MASTL in zebrafish correlates with a decrease in the expression of the thrombopoietin receptor, c-mpl, and the CD41 platelet adhesion protein, GpIIb, but has no effect on essential housekeeping zebrafish gene, EF1alpha.


Subject(s)
Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Thrombocytopenia/etiology , Animals , Blood Platelets/enzymology , Blood Platelets/metabolism , Blood Platelets/pathology , Cell Count , Cell Lineage , Enzyme Activation , Gene Expression Profiling , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Mutation, Missense , Platelet Membrane Glycoprotein IIb/genetics , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/physiology , RNA, Messenger/analysis , Receptors, Thrombopoietin/genetics , Thrombocytopenia/enzymology , Thrombocytopenia/genetics , Zebrafish
4.
Article in English | MEDLINE | ID: mdl-15533782

ABSTRACT

We used zebrafish to screen and identify small molecules that affect the process of vertebrate hematopoietic development. Zebrafish embryos were exposed to a library of 5000 synthetic compounds and screened for defects in primitive erythropoiesis. Here, we present the characterization of hemolytic anemia induced in zebrafish by the small molecule 5115318 (3-[5-methyl-furan 2-yl]-propionic acid N'-phenyl-hydrazide). This compound is capable of generating hemoglobin aggregates and Heinz bodies in red cells in vivo only. The induced anemia is reversible and treated fish recover in about 4 days. This study shows the feasibility of using zebrafish to screen for small molecules that can modulate the specific process of erythropoiesis.


Subject(s)
Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/pathology , Drug Evaluation, Preclinical/methods , Furans/pharmacology , Propionates/pharmacology , Zebrafish/physiology , Anemia, Hemolytic/drug therapy , Animals , Disease Models, Animal , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/pathology , Erythroid Cells/drug effects , Erythroid Cells/pathology , Erythropoiesis/drug effects , Furans/chemistry , Molecular Structure , Phenylhydrazines , Propionates/chemistry
5.
Curr Opin Hematol ; 11(4): 255-61, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15314525

ABSTRACT

PURPOSE OF REVIEW: This review summarizes the status of zebrafish as a genetic model to study human hematological disorders. Much of our current understanding of the function of genes modulating the process of hematopoietic stem cell generation, specification, and differentiation has come from mutant analysis. Because of the transparency of zebrafish embryos that allows for direct visualization of circulating erythroid cells, mutations affecting zebrafish erythropoiesis were among the first characterized mutants through positional cloning and candidate gene strategies. RECENT FINDINGS: New technologies have evolved that allow for generation, detection, and characterization of lineage specific alterations in the hematopoietic system. We will also briefly discuss the applications of several of these technologies such as targeted gene knockdown using antisense morpholinos, small molecule screen, transgenesis, and cell transplantation as related to blood disorders and hematopoietic development. SUMMARY: The combination of phenotype-driven forward genetic analyses and innovative technical advances has conferred zebrafish as a powerful genetic model to further dissect the function of hematopoietic genes. Through the use of available resources, the identification of novel genes or novel function for known hematopoietic genes will have important implications for our understanding of human disease pathogenesis, treatment, and prevention.


Subject(s)
Disease Models, Animal , Erythropoiesis/genetics , Hematologic Diseases , Hematopoietic System/physiology , Zebrafish , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Erythropoiesis/physiology , Hematologic Diseases/genetics , Humans , Mutation , Zebrafish/genetics
6.
Development ; 129(18): 4359-70, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183387

ABSTRACT

The red blood cell membrane skeleton is an elaborate and organized network of structural proteins that interacts with the lipid bilayer and transmembrane proteins to maintain red blood cell morphology, membrane deformability and mechanical stability. A crucial component of red blood cell membrane skeleton is the erythroid specific protein 4.1R, which anchors the spectrin-actin based cytoskeleton to the plasma membrane. Qualitative and quantitative defects in protein 4.1R result in congenital red cell membrane disorders characterized by reduced cellular deformability and abnormal cell morphology. The zebrafish mutants merlot (mot) and chablis (cha) exhibit severe hemolytic anemia characterized by abnormal cell morphology and increased osmotic fragility. The phenotypic analysis of merlot indicates severe hemolysis of mutant red blood cells, consistent with the observed cardiomegaly, splenomegaly, elevated bilirubin levels and erythroid hyperplasia in the kidneys. The result of electron microscopic analysis demonstrates that mot red blood cells have membrane abnormalities and exhibit a severe loss of cortical membrane organization. Using positional cloning techniques and a candidate gene approach, we demonstrate that merlot and chablis are allelic and encode the zebrafish erythroid specific protein 4.1R. We show that mutant cDNAs from both alleles harbor nonsense point mutations, resulting in premature stop codons. This work presents merlot/chablis as the first characterized non-mammalian vertebrate models of hereditary anemia due to a defect in protein 4.1R integrity.


Subject(s)
Anemia, Hemolytic/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mutation , Neuropeptides , Zebrafish/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , Codon, Nonsense , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA Primers , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Disease Models, Animal , Erythrocyte Membrane/physiology , Erythrocyte Membrane/ultrastructure , Genetic Linkage , Membrane Proteins/metabolism , Polymerase Chain Reaction , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Zebrafish/embryology
8.
Front Biosci ; 7: d1439-53, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12045008

ABSTRACT

Much of our current understanding of the function of genes modulating the normal process of embryonic development has come from mutant analysis. The availability of thousands of mutant lines in zebrafish that allows for identification of novel genes regulating various aspects of embryogenesis has been instrumental in establishing zebrafish as a robust and reliable genetic system. With the advances in genomic sequencing, the construction of several genetic maps, and cloning of hundreds of ESTs, positional cloning experiments in zebrafish have become more approachable. An increasing number of mutant genes have been cloned. Several zebrafish mutants are representative of known forms of human genetic diseases. The success of morpholino antisense technology in zebrafish potentially opens the door for modeling nearly any inherited developmental defect. This review highlights the strengths and limitations of using the zebrafish as an organism for elucidation of the genetic etiology of human disease. Additionally a survey of current and future zebrafish models of human disease is presented.


Subject(s)
Disease Models, Animal , Zebrafish/genetics , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...