Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(10): e2315083121, 2024 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-38408253

RESUMO

Tissue plasminogen activator (tPA) is the only FDA-approved treatment for ischemic stroke but carries significant risks, including major hemorrhage. Additional options are needed, especially in small vessel thrombi which account for ~25% of ischemic strokes. We have previously shown that tPA-functionalized colloidal microparticles can be assembled into microwheels (µwheels) and manipulated under the control of applied magnetic fields to enable rapid thrombolysis of fibrin gels in microfluidic models of thrombosis. Transparent zebrafish larvae have a highly conserved coagulation cascade that enables studies of hemostasis and thrombosis in the context of intact vasculature, clotting factors, and blood cells. Here, we show that tPA-functionalized µwheels can perform rapid and targeted recanalization in vivo. This effect requires both tPA and µwheels, as minimal to no recanalization is achieved with tPA alone, µwheels alone, or tPA-functionalized microparticles in the absence of a magnetic field. We evaluated tPA-functionalized µwheels in CRISPR-generated plasminogen (plg) heterozygous and homozygous mutants and confirmed that tPA-functionalized µwheels are dose-dependent on plasminogen for lysis. We have found that magnetically powered µwheels as a targeted tPA delivery system are dramatically more efficient at plasmin-mediated thrombolysis than systemic delivery in vivo. Further development of this system in fish and mammalian models could enable a less invasive strategy for alleviating ischemia that is safer than directed thrombectomy or systemic infusion of tPA.


Assuntos
Acidente Vascular Cerebral , Trombose , Animais , Ativador de Plasminogênio Tecidual/farmacologia , Ativador de Plasminogênio Tecidual/uso terapêutico , Peixe-Zebra , Plasminogênio , Trombose/terapia , Terapia Trombolítica , Mamíferos
2.
PLoS Genet ; 18(11): e1010534, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36449521

RESUMO

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.


Assuntos
Duplicação Gênica , Hemostasia , Tromboplastina , Animais , Camundongos , Larva , Tromboplastina/genética , Tromboplastina/fisiologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Hemostasia/genética , Veias/fisiologia , Artérias/fisiologia
3.
Hum Mol Genet ; 29(10): 1648-1657, 2020 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-32277752

RESUMO

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.


Assuntos
Hormônio do Crescimento/genética , Hipopituitarismo/genética , Fatores de Transcrição Otx/genética , Proteínas de Peixe-Zebra/genética , Animais , Apoptose/genética , Proliferação de Células/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Brânquias/metabolismo , Brânquias/patologia , Humanos , Hipopituitarismo/patologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/patologia , Mutação com Perda de Função/genética , Mandíbula/patologia , Prolactina/genética , Peixe-Zebra/genética
4.
Pediatr Blood Cancer ; 68(5): e28934, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33577709

RESUMO

Hemophilia A and B are inherited hemorrhagic disorders that result from alterations in the coagulation cascade. Aside from spontaneous bleeding, the main complication of hemophilia is hemarthrosis. Progress over the last three decades, specifically prophylaxis using recombinant factor, has prevented hemarthrosis and lengthened patient life expectancies. However, many treatments require frequent dosing up to three times a week, and alloantibodies (inhibitors) against replacement factor continues to be an issue. These problems call for novel treatments for patients with hemophilia. Although there has been progress in extended half-life factors and mimetics of factor VIII, an alternative treatment methodology is to rebalance the activities of pro- and anticoagulant factors through inhibition of the natural anticoagulants: antithrombin, tissue factor pathway inhibitor, protein C, and protein S. This review will explore the efficacy of targeting these inhibitory pathways from preclinical development through clinical trials, and delve into concerns of thrombotic risk.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Hemofilia A/tratamento farmacológico , Animais , Coagulantes/uso terapêutico , Humanos
5.
Proc Natl Acad Sci U S A ; 115(33): E7748-E7757, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30065114

RESUMO

Approximately one-third of the mammalian proteome is transported from the endoplasmic reticulum-to-Golgi via COPII-coated vesicles. SEC23, a core component of coat protein-complex II (COPII), is encoded by two paralogous genes in vertebrates (Sec23a and Sec23b). In humans, SEC23B deficiency results in congenital dyserythropoietic anemia type-II (CDAII), while SEC23A deficiency results in a skeletal phenotype (with normal red blood cells). These distinct clinical disorders, together with previous biochemical studies, suggest unique functions for SEC23A and SEC23B. Here we show indistinguishable intracellular protein interactomes for human SEC23A and SEC23B, complementation of yeast Sec23 by both human and murine SEC23A/B, and rescue of the lethality of sec23b deficiency in zebrafish by a sec23a-expressing transgene. We next demonstrate that a Sec23a coding sequence inserted into the murine Sec23b locus completely rescues the lethal SEC23B-deficient pancreatic phenotype. We show that SEC23B is the predominantly expressed paralog in human bone marrow, but not in the mouse, with the reciprocal pattern observed in the pancreas. Taken together, these data demonstrate an equivalent function for SEC23A/B, with evolutionary shifts in the transcription program likely accounting for the distinct phenotypes of SEC23A/B deficiency within and across species, a paradigm potentially applicable to other sets of paralogous genes. These findings also suggest that enhanced erythroid expression of the normal SEC23A gene could offer an effective therapeutic approach for CDAII patients.


Assuntos
Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Eritrócitos/metabolismo , Complexos Multiproteicos/biossíntese , Proteínas de Transporte Vesicular/biossíntese , Anemia Diseritropoética Congênita/genética , Anemia Diseritropoética Congênita/metabolismo , Medula Óssea/metabolismo , Medula Óssea/patologia , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/genética , Eritrócitos/patologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Complexos Multiproteicos/genética , Especificidade da Espécie , Proteínas de Transporte Vesicular/genética
6.
J Infect Dis ; 220(3): 524-534, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-30877311

RESUMO

BACKGROUND: Infection-induced thrombocytosis is a clinically important complication of tuberculosis infection. Recent studies have highlighted the utility of aspirin as a host-directed therapy modulating the inflammatory response to infection but have not investigated the possibility that the effect of aspirin is related to an antiplatelet mode of action. METHODS: In this study, we utilize the zebrafish-Mycobacterium marinum model to show mycobacteria drive host hemostasis through the formation of granulomas. Treatment of infected zebrafish with aspirin markedly reduced mycobacterial burden. This effect is reproduced by treatment with platelet-specific glycoprotein IIb/IIIa inhibitors demonstrating a detrimental role for infection-induced thrombocyte activation. RESULTS: We find that the reduction in mycobacterial burden is dependent on macrophages and granuloma formation, providing the first in vivo experimental evidence that infection-induced platelet activation compromises protective host immunity to mycobacterial infection. CONCLUSIONS: Our study illuminates platelet activation as an efficacious target of aspirin, a widely available and affordable host-directed therapy candidate for tuberculosis.


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/imunologia , Infecções por Mycobacterium não Tuberculosas/tratamento farmacológico , Infecções por Mycobacterium não Tuberculosas/imunologia , Mycobacterium marinum/imunologia , Peixe-Zebra/imunologia , Peixe-Zebra/microbiologia , Animais , Proteínas de Bactérias/imunologia , Modelos Animais de Doenças , Granuloma/tratamento farmacológico , Granuloma/imunologia , Granuloma/microbiologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Infecções por Mycobacterium não Tuberculosas/microbiologia , Mycobacterium marinum/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Tuberculose/tratamento farmacológico , Tuberculose/imunologia , Tuberculose/microbiologia
7.
Br J Haematol ; 186(4): 574-579, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31119735

RESUMO

Hereditary thrombocytopenias can be subclassified based on mode of inheritance and platelet size. Here we report a family with autosomal dominant (AD) thrombocytopenia with normal platelet size. Linkage analysis and whole exome sequencing identified the R1026W substitution in ITGA2B as the causative defect. The same mutation has been previously reported in 7 Japanese families/patients with AD thrombocytopenia, but all of these patients had macrothrombocytopenia. This is the first report of a family with AD thrombocytopenia with normal platelet size resulting from mutation in ITGA2B. ITGA2B mutations should therefore be included in the differential diagnosis of this latter disorder.


Assuntos
Sequenciamento do Exoma , Ligação Genética , Integrina alfa2/genética , Mutação , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Plaquetas/metabolismo , Medula Óssea/patologia , Feminino , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Masculino , Repetições de Microssatélites , Linhagem , Contagem de Plaquetas , Análise de Sequência de DNA
8.
Blood ; 130(5): 666-676, 2017 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-28576875

RESUMO

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.


Assuntos
Coagulação Sanguínea/genética , Fator X , Edição de Genes , Mutação , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Fator X/genética , Fator X/metabolismo , Deficiência do Fator X/embriologia , Deficiência do Fator X/genética , Humanos , Camundongos , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
9.
J Biol Chem ; 291(40): 21283-21295, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27528607

RESUMO

The yeast Set2 histone methyltransferase is a critical enzyme that plays a number of key roles in gene transcription and DNA repair. Recently, the human homologue, SETD2, was found to be recurrently mutated in a significant percentage of renal cell carcinomas, raising the possibility that the activity of SETD2 is tumor-suppressive. Using budding yeast and human cell line model systems, we examined the functional significance of two evolutionarily conserved residues in SETD2 that are recurrently mutated in human cancers. Whereas one of these mutations (R2510H), located in the Set2 Rpb1 interaction domain, did not result in an observable defect in SETD2 enzymatic function, a second mutation in the catalytic domain of this enzyme (R1625C) resulted in a complete loss of histone H3 Lys-36 trimethylation (H3K36me3). This mutant showed unchanged thermal stability as compared with the wild type protein but diminished binding to the histone H3 tail. Surprisingly, mutation of the conserved residue in Set2 (R195C) similarly resulted in a complete loss of H3K36me3 but did not affect dimethylated histone H3 Lys-36 (H3K36me2) or functions associated with H3K36me2 in yeast. Collectively, these data imply a critical role for Arg-1625 in maintaining the protein interaction with H3 and specific H3K36me3 function of this enzyme, which is conserved from yeast to humans. They also may provide a refined biochemical explanation for how H3K36me3 loss leads to genomic instability and cancer.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metiltransferases/metabolismo , Mutação , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Estabilidade Enzimática/genética , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Humanos , Metilação , Metiltransferases/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Relação Estrutura-Atividade
10.
FASEB J ; 30(5): 1798-810, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26839379

RESUMO

Protoporphyria is a metabolic disease that causes excess production of protoporphyrin IX (PP-IX), the final biosynthetic precursor to heme. Hepatic PP-IX accumulation may lead to end-stage liver disease. We tested the hypothesis that systemic administration of porphyrin precursors to zebrafish larvae results in protoporphyrin accumulation and a reproducible nongenetic porphyria model. Retro-orbital infusion of PP-IX or the iron chelator deferoxamine mesylate (DFO), with the first committed heme precursor α-aminolevulinic acid (ALA), generates high levels of PP-IX in zebrafish larvae. Exogenously infused or endogenously produced PP-IX accumulates preferentially in the liver of zebrafish larvae and peaks 1 to 3 d after infusion. Similar to patients with protoporphyria, PP-IX is excreted through the biliary system. Porphyrin accumulation in zebrafish liver causes multiorganelle protein aggregation as determined by mass spectrometry and immunoblotting. Endoplasmic reticulum stress and induction of autophagy were noted in zebrafish larvae and corroborated in 2 mouse models of protoporphyria. Furthermore, electron microscopy of zebrafish livers from larvae administered ALA + DFO showed hepatocyte autophagosomes, nuclear membrane ruffling, and porphyrin-containing vacuoles with endoplasmic reticulum distortion. In conclusion, systemic administration of the heme precursors PP-IX or ALA + DFO into zebrafish larvae provides a new model of acute protoporphyria with consequent hepatocyte protein aggregation and proteotoxic multiorganelle alterations and stress.-Elenbaas, J. S., Maitra, D., Liu, Y., Lentz, S. I., Nelson, B., Hoenerhoff, M. J., Shavit, J. A., Omary, M. B. A precursor-inducible zebrafish model of acute protoporphyria with hepatic protein aggregation and multiorganelle stress.


Assuntos
Modelos Animais de Doenças , Agregação Patológica de Proteínas/patologia , Protoporfiria Eritropoética/genética , Protoporfiria Eritropoética/patologia , Estresse Fisiológico , Peixe-Zebra , Ácido Aminolevulínico/farmacologia , Animais , Desferroxamina/farmacologia , Predisposição Genética para Doença , Larva/metabolismo , Fígado/metabolismo , Fígado/patologia , Camundongos , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas/genética , Protoporfirinas/metabolismo , Sideróforos/farmacologia
11.
Blood ; 124(1): 142-50, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24782510

RESUMO

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.


Assuntos
Deficiência de Antitrombina III/genética , Antitrombina III/genética , Modelos Animais de Doenças , Coagulação Intravascular Disseminada/genética , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Humanos , Hibridização In Situ , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra
12.
Pediatr Blood Cancer ; 63(3): 561-3, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26505894

RESUMO

Arthrogryposis, renal dysfunction, and cholestasis (ARC) syndrome is a rare disorder associated with platelet abnormalities resembling gray platelet syndrome. Affected patients have normal platelet numbers but abnormal morphology and function. Bleeding symptomatology ranges from postprocedural to spontaneous life-threatening hemorrhage. We report a patient with ARC syndrome and compound heterozygous mutations in VPS33B (vacuolar protein sorting 33B) who presented with significant bleeding requiring numerous admissions and transfusions. She was treated with prophylactic platelet transfusions and ε-aminocaproic acid. This was well-tolerated and significantly decreased transfusion requirements and admissions for bleeding. Our experience provides support for consideration of prophylactic measures in these patients as well as the possibility of using prophylaxis in related disorders.


Assuntos
Ácido Aminocaproico/uso terapêutico , Artrogripose/complicações , Artrogripose/terapia , Plaquetas/fisiologia , Colestase/complicações , Colestase/terapia , Hemorragia/complicações , Transfusão de Plaquetas , Insuficiência Renal/complicações , Insuficiência Renal/terapia , Artrogripose/genética , Plaquetas/patologia , Colestase/genética , Feminino , Heterozigoto , Humanos , Lactente , Mutação , Insuficiência Renal/genética , Proteínas de Transporte Vesicular/genética
13.
Proc Natl Acad Sci U S A ; 110(2): 588-93, 2013 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-23267103

RESUMO

The plasma glycoprotein von Willebrand factor (VWF) exhibits fivefold antigen level variation across the normal human population determined by both genetic and environmental factors. Low levels of VWF are associated with bleeding and elevated levels with increased risk for thrombosis, myocardial infarction, and stroke. To identify additional genetic determinants of VWF antigen levels and to minimize the impact of age and illness-related environmental factors, we performed genome-wide association analysis in two young and healthy cohorts (n = 1,152 and n = 2,310) and identified signals at ABO (P < 7.9E-139) and VWF (P < 5.5E-16), consistent with previous reports. Additionally, linkage analysis based on sibling structure within the cohorts, identified significant signals at chromosome 2q12-2p13 (LOD score 5.3) and at the ABO locus on chromosome 9q34 (LOD score 2.9) that explained 19.2% and 24.5% of the variance in VWF levels, respectively. Given its strong effect, the linkage region on chromosome 2 could harbor a potentially important determinant of bleeding and thrombosis risk. The absence of a chromosome 2 association signal in this or previous association studies suggests a causative gene harboring many genetic variants that are individually rare, but in aggregate common. These results raise the possibility that similar loci could explain a significant portion of the "missing heritability" for other complex genetic traits.


Assuntos
Cromossomos Humanos Par 2/genética , Cromossomos Humanos Par 9/genética , Ligação Genética/genética , Locos de Características Quantitativas/genética , Fator de von Willebrand/genética , Sistema ABO de Grupos Sanguíneos/genética , Adolescente , Adulto , Fatores Etários , Biologia Computacional , Frequência do Gene , Estudo de Associação Genômica Ampla , Genótipo , Haplótipos/genética , Humanos , Escore Lod , Polimorfismo de Nucleotídeo Único/genética , Análise de Componente Principal , Fatores Sexuais , Fator de von Willebrand/metabolismo
14.
Curr Opin Hematol ; 21(5): 418-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25054910

RESUMO

PURPOSE OF REVIEW: Although the zebrafish has been established as a research tool over the past two to three decades, in hematology it has primarily been used to investigate areas distinct from coagulation. The advantages of this vertebrate model include high fecundity, rapid and external development, and conservation of virtually all clotting factors in the zebrafish genomic sequence. Here, we summarize the growing application of this technology to the study of hemostasis and thrombosis. RECENT FINDINGS: Loss of function studies have demonstrated conservation of function for a number of zebrafish coagulation factors. These include positive and negative regulators of coagulation, as well as key components of the thrombus itself, such as von Willebrand factor, fibrinogen, and thrombocytes. Such analyses have also been leveraged to aid in the understanding of human variation and disease, as well as to perform in-vivo structure/function experiments. SUMMARY: The zebrafish is an organism that lends itself to a number of unique and powerful approaches not possible in mammals. This review demonstrates that there is a high degree of genetic and functional conservation of coagulation, portending future insights into hemostasis and thrombosis through the use of this model.


Assuntos
Hemostasia , Trombose , Peixe-Zebra/fisiologia , Animais , Plaquetas/metabolismo , Fibrinogênio/metabolismo , Humanos , Trombose/metabolismo , Fator de von Willebrand/metabolismo
16.
J Thromb Haemost ; 22(2): 558-564, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37866516

RESUMO

BACKGROUND: Leukocyte adhesion deficiency III (LAD-III) is a rare autosomal recessive syndrome characterized by functional deficiencies of platelets and leukocytes that occurs due to mutations in the FERMT3 gene encoding kindlin-3. Kindlin-3 is a FERM domain-containing adaptor protein that is essential in integrin activation. We have previously demonstrated that the FERM domain of kindlin-3 is structurally compact and plays an important role in supporting integrin activation in a mouse model. The impact of destabilizing the compact FERM domain in kindlin-3 on the development of LAD-III in humans remains uncertain. OBJECTIVES: To use primary cells from a patient with LAD-III to validate the role of the compact FERM domain in kindlin-3 function in platelets and leukocytes. METHODS: The patient is a 4-year-old girl who since infancy has displayed clinical features of LAD-III. Patient platelets and leukocytes were functionally analyzed, and structural analysis of the kindlin-3 variant was conducted. RESULTS: We identified a novel homozygous missense mutation in the FERMT3 (c.412G>A, p.E138K) FERM domain. Substantially reduced levels of kindlin-3 were detected in the proband's platelets and leukocytes. Functional evaluation verified that integrin αIIbß3-mediated platelet activation, spreading, and aggregation and ß2-integrin-mediated neutrophil adhesion and spreading were significantly compromised. Structural analysis revealed that this newly identified E138K substitution in kindlin-3 destabilizes the compacted FERM domain, resulting in poor expression of kindlin-3 in blood cells and subsequent LAD-III. CONCLUSION: We have identified a novel missense mutation and verified the functional significance of the compact kindlin-3 FERM domain in supporting integrin functions in platelets and leukocytes.


Assuntos
Domínios FERM , Síndrome da Aderência Leucocítica Deficitária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Animais , Pré-Escolar , Feminino , Humanos , Camundongos , Adesão Celular/genética , Proteínas do Citoesqueleto/metabolismo , Síndrome da Aderência Leucocítica Deficitária/genética , Leucócitos/metabolismo , Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo
17.
J Thromb Haemost ; 22(4): 1056-1068, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38160724

RESUMO

BACKGROUND: Blood clots are primarily composed of red blood cells (RBCs), platelets/thrombocytes, and fibrin. Despite the similarities observed between mammals and zebrafish, the composition of fish thrombi is not as well known. OBJECTIVES: To analyze the formation of zebrafish blood clots ex vivo and arterial and venous thrombi in vivo. METHODS: Transgenic zebrafish lines and laser-mediated endothelial injury were used to determine the relative ratio of RBCs and thrombocytes in clots. Scanning electron and confocal microscopy provided high-resolution images of the structure of adult and larval clots. Adult and larval thrombocyte spreading on fibrinogen was evaluated ex vivo. RESULTS: RBCs were present in arterial and venous thrombi, making up the majority of cells in both circulations. However, bloodless mutant fish demonstrated that fibrin clots can form in vivo in the absence of blood cells. Scanning electron and confocal microscopy showed that larval and adult zebrafish thrombi and mammalian thrombi look surprisingly similar externally and internally, even though the former have nucleated RBCs and thrombocytes. Although adult thrombocytes spread on fibrinogen, we found that larval cells do not fully activate without the addition of plasma from adult fish, suggesting a developmental deficiency of a plasma activating factor. Finally, mutants lacking αIIbß3 demonstrated that this integrin mediates thrombocyte spreading on fibrinogen. CONCLUSION: Our data showed strong conservation of arterial and venous and clot/thrombus formation across species, including developmental regulation of thrombocyte function. This correlation supports the possibility that mammals also do not absolutely require circulating cells to form fibrin clots in vivo.


Assuntos
Hemostáticos , Tromboembolia , Trombose , Animais , Peixe-Zebra , Trombose/genética , Plaquetas , Fibrina/química , Fibrinogênio/genética , Mamíferos
18.
bioRxiv ; 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37090609

RESUMO

Defects in blood development frequently occur among syndromic congenital anomalies. Thrombocytopenia-Absent Radius (TAR) syndrome is a rare congenital condition with reduced platelets (hypomegakaryocytic thrombocytopenia) and forelimb anomalies, concurrent with more variable heart and kidney defects. TAR syndrome associates with hypomorphic gene function for RBM8A/Y14 that encodes a component of the exon junction complex involved in mRNA splicing, transport, and nonsense-mediated decay. How perturbing a general mRNA-processing factor causes the selective TAR Syndrome phenotypes remains unknown. Here, we connect zebrafish rbm8a perturbation to early hematopoietic defects via attenuated non-canonical Wnt/Planar Cell Polarity (PCP) signaling that controls developmental cell re-arrangements. In hypomorphic rbm8a zebrafish, we observe a significant reduction of cd41-positive thrombocytes. rbm8a-mutant zebrafish embryos accumulate mRNAs with individual retained introns, a hallmark of defective nonsense-mediated decay; affected mRNAs include transcripts for non-canonical Wnt/PCP pathway components. We establish that rbm8a-mutant embryos show convergent extension defects and that reduced rbm8a function interacts with perturbations in non-canonical Wnt/PCP pathway genes wnt5b, wnt11f2, fzd7a, and vangl2. Using live-imaging, we found reduced rbm8a function impairs the architecture of the lateral plate mesoderm (LPM) that forms hematopoietic, cardiovascular, kidney, and forelimb skeleton progenitors as affected in TAR Syndrome. Both mutants for rbm8a and for the PCP gene vangl2 feature impaired expression of early hematopoietic/endothelial genes including runx1 and the megakaryocyte regulator gfi1aa. Together, our data propose aberrant LPM patterning and hematopoietic defects as consequence of attenuated non-canonical Wnt/PCP signaling upon reduced rbm8a function. These results also link TAR Syndrome to a potential LPM origin and a developmental mechanism.

19.
Methods Mol Biol ; 2631: 371-380, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36995678

RESUMO

The CRISPR/Cas9 system is a powerful tool for genome editing in zebrafish. This workflow takes advantage of the genetic tractability of zebrafish and will allow users to edit genomic sites and produce mutant lines using selective breeding. Established lines may then be employed by researchers for downstream genetic and phenotypic analyses.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Animais , Sistemas CRISPR-Cas/genética , Peixe-Zebra/genética , Genoma
20.
J Thromb Haemost ; 21(4): 983-994, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36696210

RESUMO

BACKGROUND: Fibrin-rich clot formation in thrombo-occlusive pathologies is currently treated by systemic administration of plasminogen activators (e.g. tPA), to convert fibrin-associated plasminogen to plasmin for fibrinolytic action. However, this conversion is not restricted to clot site only but also occurs on circulating plasminogen, causing systemic fibrinogenolysis and bleeding risks. To address this, past research has explored tPA delivery using clot-targeted nanoparticles. OBJECTIVES: We designed a nanomedicine system that can (1) target clots via binding to activated platelets and fibrin, (2) package plasmin instead of tPA as a direct fibrinolytic agent, and (3) release this plasmin triggered by thrombin for clot-localized action. METHODS: Clot-targeted thrombin-cleavable nanoparticles (CTNPs) were manufactured using self-assembly of peptide-lipid conjugates. Plasmin loading and its thrombin-triggered release from CTNPs were characterized by UV-visible spectroscopy. CTNP-targeting to clots under flow was studied using microfluidics. Fibrinolytic effect of CTNP-delivered plasmin was studied in vitro using BioFlux imaging and D-dimer analysis and in vivo in a zebrafish thrombosis model. RESULTS: Plasmin-loaded CTNPs significantly bound to clots under shear flow and showed thrombin-triggered enhanced release of plasmin. BioFlux studies confirmed that thrombin-triggered plasmin released from CTNPs rendered fibrinolysis similar to free plasmin, further corroborated by D-dimer analysis. In the zebrafish model, CTNP-delivered plasmin accelerated time-to-recanalization, or completely prevented occlusion when infused before thrombus formation. CONCLUSION: Considering that the very short circulation half-life (<1 second) of plasmin prevents its systemic use but also makes it safer without off-target drug effects, clot-targeted delivery of plasmin using CTNPs can enable safer and more efficacious fibrinolytic therapy.


Assuntos
Nanopartículas , Trombose , Animais , Fibrinolisina/metabolismo , Trombina/química , Peixe-Zebra/metabolismo , Fibrinólise , Terapia Trombolítica , Trombose/tratamento farmacológico , Fibrina/química , Plasminogênio , Ativador de Plasminogênio Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA