Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
PLoS Genet ; 18(11): e1010534, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36449521

RÉSUMÉ

Tissue factor (TF) is an evolutionarily conserved protein necessary for initiation of hemostasis. Zebrafish have two copies of the tissue factor gene (f3a and f3b) as the result of an ancestral teleost fish duplication event (so called ohnologs). In vivo physiologic studies of TF function have been difficult given early lethality of TF knockout in the mouse. We used genome editing to produce knockouts of both f3a and f3b in zebrafish. Since ohnologs arose through sub- or neofunctionalization, they can unmask unknown functions of non-teleost genes and could reveal whether mammalian TF has developmental functions distinct from coagulation. Here we show that a single copy of either f3a or f3b is necessary and sufficient for normal lifespan. Complete loss of TF results in lethal hemorrhage by 2-4 months despite normal embryonic and vascular development. Larval vascular endothelial injury reveals predominant roles for TFa in venous circulation and TFb in arterial circulation. Finally, we demonstrate that loss of TF predisposes to a stress-induced cardiac tamponade independent of its role in fibrin formation. Overall, our data suggest partial subfunctionalization of TFa and TFb. This multigenic zebrafish model has the potential to facilitate study of the role of TF in different vascular beds.


Sujet(s)
Duplication de gène , Hémostase , Thromboplastine , Animaux , Souris , Larve , Thromboplastine/génétique , Thromboplastine/physiologie , Danio zébré/génétique , Danio zébré/métabolisme , Protéines de poisson-zèbre/génétique , Hémostase/génétique , Veines/physiologie , Artères/physiologie
2.
Blood Adv ; 4(21): 5480-5491, 2020 11 10.
Article de Anglais | MEDLINE | ID: mdl-33166405

RÉSUMÉ

Plasma fibrinogen molecules comprise 2 copies of Aα, Bß, and γ chains folded into a hexameric protein. A minor fibrinogen isoform with an extended Aα chain (AαE) is more abundant in newborn human blood than in adults. Larval zebrafish produce predominantly AαE-containing fibrinogen, but its functional significance is unclear. In 3-day-old zebrafish, when hemostasis is reliant on fibrinogen and erythrocyte-rich clotting but is largely thrombocyte-independent, we measured the time to occlusion (TTO) in a laser-induced venous thrombosis assay in 3 zebrafish strains (AB, TU, and AB × TL hybrids). AB larvae showed delayed TTO compared with the TU and AB × TL strains. Mating AB with TU or TL produced larvae with a TU-like TTO. In contrast to TU, AB larvae failed to produce fibrinogen AαE, due to a mutation in the AαE-specific coding region of fibrinogen α-chain gene (fga). We investigated whether the lack of AαE explained the delayed AB TTO. Transgenic expression of AαE, but not Aα, shortened the AB TTO to that of TU. AαE rescued venous occlusion in fibrinogen mutants or larvae with morpholino-targeted fibrinogen α-chain messenger RNA, but Aα was less effective. In 5-day-old larvae, circulating thrombocytes contribute to hemostasis, as visualized in Tg(itga2b:EGFP) transgenics. Laser-induced venous thrombocyte adhesion and aggregation is reduced in fibrinogen mutants, but transgenic expression of Aα or AαE restored similar thrombocyte accumulation at the injury site. Our data demonstrate a genetic modifier of venous thrombosis and a role for fibrinogen AαE in early developmental blood coagulation, and suggest a link between differentially expressed fibrinogen isoforms and the cell types available for clotting.


Sujet(s)
Fibrinogène , Hémostatiques , Thrombose veineuse , Animaux , Fibrinogène/génétique , Hémostase , Danio zébré
3.
Hum Mol Genet ; 29(10): 1648-1657, 2020 06 27.
Article de Anglais | MEDLINE | ID: mdl-32277752

RÉSUMÉ

Combined pituitary hormone deficiency (CPHD) is a genetically heterogeneous disorder caused by mutations in over 30 genes. The loss-of-function mutations in many of these genes, including orthodenticle homeobox 2 (OTX2), can present with a broad range of clinical symptoms, which provides a challenge for predicting phenotype from genotype. Another challenge in human genetics is functional evaluation of rare genetic variants that are predicted to be deleterious. Zebrafish are an excellent vertebrate model for evaluating gene function and disease pathogenesis, especially because large numbers of progeny can be obtained, overcoming the challenge of individual variation. To clarify the utility of zebrafish for the analysis of CPHD-related genes, we analyzed the effect of OTX2 loss of function in zebrafish. The otx2b gene is expressed in the developing hypothalamus, and otx2bhu3625/hu3625 fish exhibit multiple defects in the development of head structures and are not viable past 10 days post fertilization (dpf). Otx2bhu3625/hu3625 fish have a small hypothalamus and low expression of pituitary growth hormone and prolactin (prl). The gills of otx2bhu3625/hu3625 fish have weak sodium influx, consistent with the role of prolactin in osmoregulation. The otx2bhu3625/hu3625 eyes are microphthalmic with colobomas, which may underlie the inability of the mutant fish to find food. The small pituitary and eyes are associated with reduced cell proliferation and increased apoptosis evident at 3 and 5 dpf, respectively. These observations establish the zebrafish as a useful tool for the analysis of CPHD genes with variable and complex phenotypes.


Sujet(s)
Hormone de croissance/génétique , Hypopituitarisme/génétique , Facteurs de transcription Otx/génétique , Protéines de poisson-zèbre/génétique , Animaux , Apoptose/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes au cours du développement/génétique , Branchies/métabolisme , Branchies/anatomopathologie , Humains , Hypopituitarisme/anatomopathologie , Hypothalamus/croissance et développement , Hypothalamus/anatomopathologie , Mutation perte de fonction/génétique , Mandibule/anatomopathologie , Prolactine/génétique , Danio zébré/génétique
4.
Blood Adv ; 3(11): 1670-1680, 2019 06 11.
Article de Anglais | MEDLINE | ID: mdl-31167819

RÉSUMÉ

In humans, coagulation factor V (FV) deficiency is a rare, clinically heterogeneous bleeding disorder, suggesting that genetic modifiers may contribute to disease expressivity. Zebrafish possess many distinct advantages including high fecundity, optical clarity, external development, and homology with the mammalian hemostatic system, features that make it ideal for genetic studies. Our aim was to study the role of FV in zebrafish through targeted mutagenesis and apply the model to the study of human F5 variants. CRISPR-mediated genome editing of the zebrafish f5 locus was performed, generating mutants homozygous for a 49 base pair deletion in exon 4. Thrombus formation secondary to vascular endothelial injury was absent in f5 -/- mutant embryos and larvae. Despite this severe hemostatic defect, homozygous mutants survived before succumbing to severe hemorrhage in adulthood. Human F5 variants of uncertain significance from patients with FV deficiency were evaluated, and the causative mutations identified and stratified by their ability to restore thrombus formation in larvae. Analysis of these novel mutations demonstrates variable residual FV function, with minimal activity being required to restore hemostasis in response to laser-induced endothelial injury. This in vivo evaluation may be beneficial for patients whose factor activity levels lack correlation with bleeding symptomatology, although limitations exist. Furthermore, homozygous mutant embryos tolerate what is a severe and lethal defect in mammals, suggesting the possibility of species-specific factors enabling survival, and allowing further study not possible in the mouse. Identification of these factors or other genetic modifiers could lead to novel therapeutic modalities.


Sujet(s)
Proaccélérine/métabolisme , Hémorragie/métabolisme , Hémostase , Thrombose/métabolisme , Danio zébré/métabolisme , Animaux , Animal génétiquement modifié , Modèles animaux de maladie humaine , Proaccélérine/génétique , Hémorragie/génétique , Humains , Thrombose/génétique , Danio zébré/génétique
5.
J Thromb Haemost ; 17(4): 607-617, 2019 04.
Article de Anglais | MEDLINE | ID: mdl-30663848

RÉSUMÉ

Essentials Loss of fibrinogen in zebrafish has been previously shown to result in adult onset hemorrhage Hemostatic defects were discovered in early fga-/- embryos but well tolerated until adulthood Afibrinogenemia and thrombocytopenia results in synthetic lethality in zebrafish. Testing human FGA variants of uncertain significance in zebrafish identified causative mutations SUMMARY: Background Mutations in the alpha chain of fibrinogen (FGA), such as deficiencies in other fibrinogen subunits, lead to rare inherited autosomal recessive hemostatic disorders. These range from asymptomatic to catastrophic life-threatening bleeds and the molecular basis of inherited fibrinogen deficiencies is only partially understood. Zinc finger nucleases have been used to produce mutations in zebrafish fga, resulting in overt adult-onset hemorrhage and reduced survival. Objectives To determine the age of onset of hemostatic defects in afibrinogenemic zebrafish and model human fibrinogen deficiencies. Methods TALEN genome editing (transcription activator-like effector nucleases) was used to generate a zebrafish fga mutant. Hemostatic defects were assessed through survival, gross anatomical and histological observation and laser-induced endothelial injury. Human FGA variants with unknown pathologies were engineered into the orthologous positions in zebrafish fga. Results Loss of Fga decreased survival and resulted in synthetic lethality when combined with thrombocytopenia. Zebrafish fga mutants exhibit a severe hemostatic defect by 3 days of life, but without visible hemorrhage. Induced thrombus formation through venous endothelial injury was completely absent in mutant embryos and larvae. This hemostatic defect was restored by microinjection of wild-type fga cDNA plasmid or purified human fibrinogen. This system was used to determine whether unknown human variants were pathological by engineering them into fga. Conclusions These studies confirm that loss of fibrinogen in zebrafish results in the absence of hemostasis from the embryonic period through adulthood. When combined with thrombocytopenia, zebrafish exhibit synthetic lethality, demonstrating that thrombocytes are necessary for survival in response to hemorrhage.


Sujet(s)
Afibrinogénémie/sang , Afibrinogénémie/métabolisme , Fibrinogène/métabolisme , Hémorragie/sang , Hémostase , Thrombopénie/sang , Protéines de poisson-zèbre/métabolisme , Danio zébré/métabolisme , Animaux , Animal génétiquement modifié , Fibrinogène/génétique , Hémorragie/génétique , Hémostase/génétique , Humains , Sous-unité p45 du facteur de transcription NF-E2/génétique , Sous-unité p45 du facteur de transcription NF-E2/métabolisme , Mutations synthétiques létales , Thrombopénie/génétique , Danio zébré/embryologie , Danio zébré/génétique , Protéines de poisson-zèbre/déficit , Protéines de poisson-zèbre/génétique
6.
Blood Adv ; 2(23): 3418-3427, 2018 12 11.
Article de Anglais | MEDLINE | ID: mdl-30504234

RÉSUMÉ

The NFE2 transcription factor is expressed in multiple hematopoietic lineages with a well-defined role in regulating megakaryocyte biogenesis and platelet production in mammals. Mice deficient in NFE2 develop severe thrombocytopenia with lethality resulting from neonatal hemorrhage. Recent data in mammals reveal potential differences in embryonic and adult thrombopoiesis. Multiple studies in zebrafish have revealed mechanistic insights into hematopoiesis, although thrombopoiesis has been less studied. Rather than platelets, zebrafish possess thrombocytes, which are nucleated cells with similar functional properties. Using transcription activator-like effector nucleases to generate mutations in nfe2, we show that unlike mammals, zebrafish survive to adulthood in the absence of Nfe2. Despite developing severe thrombocytopenia, homozygous mutants do not display overt hemorrhage or reduced survival. Surprisingly, quantification of circulating thrombocytes in mutant 6-day-old larvae revealed no significant differences from wild-type siblings. Both wild-type and nfe2 null larvae formed thrombocyte-rich clots in response to endothelial injury. In addition, ex vivo thrombocytic colony formation was intact in nfe2 mutants, and adult kidney marrow displayed expansion of hematopoietic progenitors. These data suggest that loss of Nfe2 results in a late block in adult thrombopoiesis, with secondary expansion of precursors: features consistent with mammals. Overall, our data suggest parallels with erythropoiesis, including distinct primitive and definitive pathways of development and potential for a previously unknown Nfe2-independent pathway of embryonic thrombopoiesis. Long-term homozygous mutant survival will facilitate in-depth study of Nfe2 deficiency in vivo, and further investigation could lead to alternative methodologies for the enhancement of platelet production.


Sujet(s)
Plaquettes/métabolisme , Facteur de transcription NF-E2/métabolisme , Protéines de poisson-zèbre/métabolisme , Danio zébré/croissance et développement , Séquence d'acides aminés , Animaux , Plaquettes/cytologie , Codon stop , Fibrinogène/métabolisme , Mutation avec décalage du cadre de lecture , Édition de gène , Humains , Larve/métabolisme , Facteur de transcription NF-E2/composition chimique , Facteur de transcription NF-E2/génétique , Alignement de séquences , Thrombopoïèse , Protéines de poisson-zèbre/composition chimique , Protéines de poisson-zèbre/génétique
7.
Blood ; 130(5): 666-676, 2017 08 03.
Article de Anglais | MEDLINE | ID: mdl-28576875

RÉSUMÉ

Deficiency of factor X (F10) in humans is a rare bleeding disorder with a heterogeneous phenotype and limited therapeutic options. Targeted disruption of F10 and other common pathway factors in mice results in embryonic/neonatal lethality with rapid resorption of homozygous mutants, hampering additional studies. Several of these mutants also display yolk sac vascular defects, suggesting a role for thrombin signaling in vessel development. The zebrafish is a vertebrate model that demonstrates conservation of the mammalian hemostatic and vascular systems. We have leveraged these advantages for in-depth study of the role of the coagulation cascade in the developmental regulation of hemostasis and vasculogenesis. In this article, we show that ablation of zebrafish f10 by using genome editing with transcription activator-like effector nucleases results in a major embryonic hemostatic defect. However, widespread hemorrhage and subsequent lethality does not occur until later stages, with absence of any detectable defect in vascular development. We also use f10-/- zebrafish to confirm 5 novel human F10 variants as causative mutations in affected patients, providing a rapid and reliable in vivo model for testing the severity of F10 variants. These findings as well as the prolonged survival of f10-/- mutants will enable us to expand our understanding of the molecular mechanisms of hemostasis, including a platform for screening variants of uncertain significance in patients with F10 deficiency and other coagulation disorders. Further study as to how fish tolerate what is an early lethal mutation in mammals could facilitate improvement of diagnostics and therapeutics for affected patients with bleeding disorders.


Sujet(s)
Coagulation sanguine/génétique , Facteur X , Édition de gène , Mutation , Protéines de poisson-zèbre , Danio zébré , Animaux , Facteur X/génétique , Facteur X/métabolisme , Déficit en facteur X/embryologie , Déficit en facteur X/génétique , Humains , Souris , Danio zébré/embryologie , Danio zébré/génétique , Protéines de poisson-zèbre/génétique , Protéines de poisson-zèbre/métabolisme
8.
Blood ; 124(1): 142-50, 2014 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-24782510

RÉSUMÉ

Pathologic blood clotting is a leading cause of morbidity and mortality in the developed world, underlying deep vein thrombosis, myocardial infarction, and stroke. Genetic predisposition to thrombosis is still poorly understood, and we hypothesize that there are many additional risk alleles and modifying factors remaining to be discovered. Mammalian models have contributed to our understanding of thrombosis, but are low throughput and costly. We have turned to the zebrafish, a tool for high-throughput genetic analysis. Using zinc finger nucleases, we show that disruption of the zebrafish antithrombin III (at3) locus results in spontaneous venous thrombosis in larvae. Although homozygous mutants survive into early adulthood, they eventually succumb to massive intracardiac thrombosis. Characterization of null fish revealed disseminated intravascular coagulation in larvae secondary to unopposed thrombin activity and fibrinogen consumption, which could be rescued by both human and zebrafish at3 complementary DNAs. Mutation of the human AT3-reactive center loop abolished the ability to rescue, but the heparin-binding site was dispensable. These results demonstrate overall conservation of AT3 function in zebrafish, but reveal developmental variances in the ability to tolerate excessive clot formation. The accessibility of early zebrafish development will provide unique methods for dissection of the underlying mechanisms of thrombosis.


Sujet(s)
Déficit en antithrombine III/génétique , Antithrombine-III/génétique , Modèles animaux de maladie humaine , Coagulation intravasculaire disséminée/génétique , Protéines de poisson-zèbre/génétique , Animaux , Animal génétiquement modifié , Humains , Hybridation in situ , Mutagenèse dirigée , RT-PCR , Danio zébré
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...