Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Blood ; 137(2): 258-268, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-32735640

RESUMEN

Tissue factor pathway inhibitor (TFPI) inhibits proteases in the blood coagulation cascade that lead to the production of thrombin, including prothrombinase (factor Xa [FXa]/FVa), the catalytic complex that directly generates thrombin. Thus, TFPI and FV are directly linked in regulating the procoagulant response. Studies using knockout mice indicate that TFPI and FV are necessary for embryogenesis, but their contributions to vascular development are unclear. We performed extensive histological analyses of Tfpi-/- and Tfpi-/-F5-/- mouse embryos to investigate the importance of the interplay between TFPI and FV in regulating hemostasis and vascular development during embryogenesis. We observed normal tissue development throughout Tfpi-/- embryos, except in the central nervous system (CNS). The CNS displayed stunted brain growth, delayed development of the meninges, and severe vascular pathology characterized by the formation of glomeruloid bodies surrounding areas of cellular death, fibrin deposition, and hemorrhage. Removing FV from Tfpi-/- embryos completely ameliorated their brain pathology, suggesting that TFPI dampens FV-dependent procoagulant activity in a manner that modulates cerebrovascular development. Thus, we have identified a previously unrecognized role for TFPI activity within the CNS. This TFPI activity likely diminishes an effect of excess thrombin activity on signaling pathways that control cerebral vascular development.


Asunto(s)
Vasos Sanguíneos/embriología , Encéfalo/irrigación sanguínea , Encéfalo/embriología , Desarrollo Embrionario/fisiología , Lipoproteínas/metabolismo , Animales , Factor V/metabolismo , Ratones , Ratones Noqueados
2.
Br J Cancer ; 126(6): 927-936, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34931040

RESUMEN

BACKGROUND: Bone-marrow-derived haematopoietic stem and progenitor cells (HSPCs) are a prominent part of the highly complex tumour microenvironment (TME) where they localise within tumours and maintain haematopoietic potency. Understanding the role HSPCs play in tumour growth and response to radiation therapy (RT) may lead to improved patient treatments and outcomes. METHODS: We used a mouse model of non-small cell lung carcinoma where tumours were exposed to RT regimens alone or in combination with GW2580, a pharmacological inhibitor of colony stimulating factor (CSF)-1 receptor. RT-PCR, western blotting and immunohistochemistry were used to quantify expression levels of factors that affect HSPC differentiation. DsRed+ HSPC intratumoural activity was tracked using flow cytometry and confocal microscopy. RESULTS: We demonstrated that CSF-1 is enhanced in the TME following exposure to RT. CSF-1 signaling induced intratumoural HSPC differentiation into M2 polarised tumour-associated macrophages (TAMs), aiding in post-RT tumour survival and regrowth. In contrast, hyperfractionated/pulsed radiation therapy (PRT) and GW2580 ablated this process resulting in improved tumour killing and mouse survival. CONCLUSIONS: Tumours coopt intratumoural HSPC fate determination via CSF-1 signaling to overcome the effects of RT. Thus, limiting intratumoural HSPC activity represents an attractive strategy for improving the clinical treatment of solid tumours.


Asunto(s)
Células Madre Hematopoyéticas , Neoplasias , Animales , Diferenciación Celular , Humanos , Macrófagos , Ratones , Neoplasias/metabolismo , Microambiente Tumoral
3.
PLoS Genet ; 14(9): e1007658, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30188893

RESUMEN

Although the Factor V Leiden (FVL) gene variant is the most prevalent genetic risk factor for venous thrombosis, only 10% of FVL carriers will experience such an event in their lifetime. To identify potential FVL modifier genes contributing to this incomplete penetrance, we took advantage of a perinatal synthetic lethal thrombosis phenotype in mice homozygous for FVL (F5L/L) and haploinsufficient for tissue factor pathway inhibitor (Tfpi+/-) to perform a sensitized dominant ENU mutagenesis screen. Linkage analysis conducted in the 3 largest pedigrees generated from the surviving F5L/L Tfpi+/- mice ('rescues') using ENU-induced coding variants as genetic markers was unsuccessful in identifying major suppressor loci. Whole exome sequencing was applied to DNA from 107 rescue mice to identify candidate genes enriched for ENU mutations. A total of 3,481 potentially deleterious candidate ENU variants were identified in 2,984 genes. After correcting for gene size and multiple testing, Arl6ip5 was identified as the most enriched gene, though not reaching genome-wide significance. Evaluation of CRISPR/Cas9 induced loss of function in the top 6 genes failed to demonstrate a clear rescue phenotype. However, a maternally inherited (not ENU-induced) de novo mutation (Plcb4R335Q) exhibited significant co-segregation with the rescue phenotype (p = 0.003) in the corresponding pedigree. Thrombosis suppression by heterozygous Plcb4 loss of function was confirmed through analysis of an independent, CRISPR/Cas9-induced Plcb4 mutation (p = 0.01).


Asunto(s)
Factor V/genética , Predisposición Genética a la Enfermedad/genética , Mutagénesis/genética , Fosfolipasa C beta/genética , Tromboembolia Venosa/genética , Animales , Proteínas Portadoras , Modelos Animales de Enfermedad , Etilnitrosourea/toxicidad , Femenino , Proteínas de Choque Térmico , Humanos , Estimación de Kaplan-Meier , Lipoproteínas/genética , Masculino , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis/efectos de los fármacos , Linaje , Penetrancia , Tromboembolia Venosa/mortalidad , Secuenciación del Exoma
4.
Proc Natl Acad Sci U S A ; 114(36): 9659-9664, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28827327

RESUMEN

Factor V Leiden (F5L ) is a common genetic risk factor for venous thromboembolism in humans. We conducted a sensitized N-ethyl-N-nitrosourea (ENU) mutagenesis screen for dominant thrombosuppressor genes based on perinatal lethal thrombosis in mice homozygous for F5L (F5L/L ) and haploinsufficient for tissue factor pathway inhibitor (Tfpi+/- ). F8 deficiency enhanced the survival of F5L/LTfpi+/- mice, demonstrating that F5L/LTfpi+/- lethality is genetically suppressible. ENU-mutagenized F5L/L males and F5L/+Tfpi+/- females were crossed to generate 6,729 progeny, with 98 F5L/LTfpi+/- offspring surviving until weaning. Sixteen lines, referred to as "modifier of Factor 5 Leiden (MF5L1-16)," exhibited transmission of a putative thrombosuppressor to subsequent generations. Linkage analysis in MF5L6 identified a chromosome 3 locus containing the tissue factor gene (F3). Although no ENU-induced F3 mutation was identified, haploinsufficiency for F3 (F3+/- ) suppressed F5L/LTfpi+/- lethality. Whole-exome sequencing in MF5L12 identified an Actr2 gene point mutation (p.R258G) as the sole candidate. Inheritance of this variant is associated with suppression of F5L/LTfpi+/- lethality (P = 1.7 × 10-6), suggesting that Actr2p.R258G is thrombosuppressive. CRISPR/Cas9 experiments to generate an independent Actr2 knockin/knockout demonstrated that Actr2 haploinsufficiency is lethal, supporting a hypomorphic or gain-of-function mechanism of action for Actr2p.R258G Our findings identify F8 and the Tfpi/F3 axis as key regulators in determining thrombosis balance in the setting of F5L and also suggest a role for Actr2 in this process.


Asunto(s)
Factor V/genética , Trombosis/genética , Proteína 2 Relacionada con la Actina/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Modelos Animales de Enfermedad , Etilnitrosourea , Factor VIII/genética , Femenino , Pruebas Genéticas , Haploinsuficiencia , Homocigoto , Humanos , Lipoproteínas/deficiencia , Lipoproteínas/genética , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Mutagénesis , Embarazo , Factores de Riesgo , Trombosis/prevención & control , Secuenciación del Exoma
5.
J Exp Med ; 204(5): 1049-56, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17438064

RESUMEN

We describe a mouse model of fetal loss in factor V Leiden (FvL) mothers in which fetal loss is triggered when the maternal prothrombotic state coincides with fetal gene defects that reduce activation of the protein C anticoagulant pathway within the placenta. Fetal loss is caused by disruption of placental morphogenesis at the stage of labyrinth layer formation and occurs in the absence of overt placental thrombosis, infarction, or perfusion defects. Platelet depletion or elimination of protease-activated receptor 4 (Par4) from the mother allows normal placentation and prevents fetal loss. These findings establish a cause-effect relationship for the observed epidemiologic association between maternal FvL status and fetal loss and identify fetal gene defects as risk modifiers of pregnancy failure in prothrombotic mothers. Pregnancy failure is mediated by Par4-dependent activation of maternal platelets at the fetomaternal interface and likely involves a pathogenic pathway independent of occlusive thrombosis. Our results further demonstrate that the interaction of two given thrombosis risk factors produces markedly disparate consequences on disease manifestation (i.e., thrombosis or pregnancy loss), depending on the vascular bed in which this interaction occurs.


Asunto(s)
Resistencia a la Proteína C Activada/complicaciones , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Factor V/genética , Muerte Fetal/etiología , Enfermedades Fetales/genética , Placenta/patología , Resistencia a la Proteína C Activada/genética , Animales , Femenino , Muerte Fetal/patología , Ratones , Ratones Endogámicos C57BL , Placenta/irrigación sanguínea , Mutación Puntual/genética , Embarazo , Resultado del Embarazo/genética , Receptores de Trombina/metabolismo , Factores de Riesgo , Trombomodulina/genética
6.
Proc Natl Acad Sci U S A ; 107(39): 16904-9, 2010 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-20837540

RESUMEN

In characterizing mice with targeted disruption of the SerpinB2 gene, we observed animals that were small at birth with delayed growth and decreased life expectancy. Although this phenotype cosegregated with homozygosity for the inactive SerpinB2 allele, analysis of homozygous SerpinB2-deficient mice derived from two additional independent embryonic stem (ES) cell clones exhibited no growth abnormalities. Examination of additional progeny from the original SerpinB2-deficient line revealed recombination between the small phenotype (smla) and the SerpinB2 locus. The locus responsible for smla was mapped to a 2.78-Mb interval approximately 30 Mb proximal to SerpinB2, bounded by markers D1Mit382 and D1Mit216. Sequencing of Irs1 identified a nonsense mutation at serine 57 (S57X), resulting in complete loss of IRS1 protein expression. Analysis of ES cell DNA suggests that the S57X Irs1 mutation arose spontaneously in an ES cell subclone during cell culture. Although the smla phenotype is similar to previously reported Irs1 alleles, mice exhibited decreased survival, in contrast to the enhanced longevity reported for IRS1 deficiency generated by gene targeting. This discrepancy could result from differences in strain background, unintended indirect effects of the gene targeting, or the minimal genetic interference of the S57X mutation compared with the conventionally targeted Irs1-KO allele. Spontaneous mutations arising during ES cell culture may be a frequent but underappreciated occurrence. When linked to a targeted allele, such mutations could lead to incorrect assignment of phenotype and may account for a subset of markedly discordant results from experiments independently targeting the same gene.


Asunto(s)
Marcación de Gen , Genes Letales , Proteínas Sustrato del Receptor de Insulina/genética , Inhibidor 2 de Activador Plasminogénico/genética , Recombinación Genética , Alelos , Animales , Secuencia de Bases , Centrómero/genética , Codón sin Sentido , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Femenino , Sitios Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes
7.
J Thromb Haemost ; 21(10): 2917-2928, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37364776

RESUMEN

BACKGROUND: Plasminogen activator inhibitor-1 (PAI-1, Serpine1) is an important circulating fibrinolysis inhibitor. PAI-1 exists in 2 pools, packaged within platelet α-granules and freely circulating in plasma. Elevated plasma PAI-1 levels are associated with cardiovascular disease. However, little is known about the regulation of platelet PAI-1 (pPAI-1). OBJECTIVES: We investigated the genetic control of pPAI-1 levels in mice and humans. METHODS: We measured pPAI-1 antigen levels via enzyme-linked immunosorbent assay in platelets isolated from 10 inbred mouse strains, including LEWES/EiJ (LEWES) and C57BL/6J (B6). LEWES and B6 were crossed to produce the F1 generation, B6LEWESF1. B6LEWESF1 mice were intercrossed to produce B6LEWESF2 mice. These mice were subjected to genome-wide genetic marker genotyping followed by quantitative trait locus analysis to identify pPAI-1 regulatory loci. RESULTS: We identified differences in pPAI-1 between several laboratory strains, with LEWES having pPAI-1 levels more than 10-fold higher than those in B6. Quantitative trait locus analysis of B6LEWESF2 offspring identified a major pPAI-1 regulatory locus on chromosome 5 from 136.1 to 137.6 Mb (logarithm of the odds score, 16.2). Significant pPAI-1 modifier loci on chromosomes 6 and 13 were also identified. CONCLUSION: Identification of pPAI-1 genomic regulatory elements provides insights into platelet/megakaryocyte-specific and cell type-specific gene expression. This information can be used to design more precise therapeutic targets for diseases where PAI-1 plays a role.


Asunto(s)
Plaquetas , Inhibidor 1 de Activador Plasminogénico , Animales , Ratones , Plaquetas/metabolismo , Fibrinólisis , Genómica , Ratones Endogámicos C57BL , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Sitios de Carácter Cuantitativo , Humanos
8.
Genetics ; 222(2)2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36040194

RESUMEN

U12-type or minor introns are found in most multicellular eukaryotes and constitute ∼0.5% of all introns in species with a minor spliceosome. Although the biological significance for the evolutionary conservation of U12-type introns is debated, mutations disrupting U12 splicing cause developmental defects in both plants and animals. In human hematopoietic stem cells, U12 splicing defects disrupt proper differentiation of myeloid lineages and are associated with myelodysplastic syndrome, predisposing individuals to acute myeloid leukemia. Mutants in the maize ortholog of RNA binding motif protein 48 (RBM48) have aberrant U12-type intron splicing. Human RBM48 was recently purified biochemically as part of the minor spliceosome and shown to recognize the 5' end of the U6atac snRNA. In this report, we use CRISPR/Cas9-mediated ablation of RBM48 in human K-562 cells to show the genetic function of RBM48. RNA-seq analysis comparing wild-type and mutant K-562 genotypes found that 48% of minor intron-containing genes have significant U12-type intron retention in RBM48 mutants. Comparing these results to maize rbm48 mutants defined a subset of minor intron-containing genes disrupted in both species. Mutations in the majority of these orthologous minor intron-containing genes have been reported to cause developmental defects in both plants and animals. Our results provide genetic evidence that the primary defect of human RBM48 mutants is aberrant U12-type intron splicing, while a comparison of human and maize RNA-seq data identifies candidate genes likely to mediate mutant phenotypes of U12-type splicing defects.


Asunto(s)
Empalme del ARN , Proteínas de Unión al ARN , Empalmosomas , Humanos , Intrones , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Motivos de Unión al ARN , Proteínas de Unión al ARN/genética , Empalmosomas/genética , Empalmosomas/metabolismo , Zea mays/genética , Zea mays/metabolismo
9.
J Thromb Haemost ; 18(11): 2968-2975, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32780555

RESUMEN

BACKGROUND: Upregulation of the plasminogen activation system, including urokinase plasminogen activator (uPA), has been observed in many malignancies, suggesting that co-opting the PA system is a common method by which tumor cells accomplish extracellular matrix proteolysis. PAI-2, a serine protease inhibitor, produced from the SERPINB2 gene, inhibits circulating and extracellular matrix-tethered uPA. Decreased SERPINB2 expression has been associated with increased tumor invasiveness and metastasis for several types of cancer. PAI-2 deficiency has not been reported in humans and PAI-2-deficient (SerpinB2-/- ) mice exhibit no apparent abnormalities. OBJECTIVES: We investigated the role of PAI-2 deficiency on tumor growth and metastasis. METHODS: To explore the long-term impact of PAI-2 deficiency, a cohort of SerpinB2-/- mice were aged to >18 months, with spontaneous malignancies observed in 4/9 animals, all of apparently vascular origin. To further investigate the role of PAI-2 deficiency in malignancy, SerpinB2-/- and wild-type control mice were injected with either B16 melanoma or Lewis lung carcinoma tumor cells, with markedly accelerated tumor growth observed in SerpinB2-/- mice for both cell lines. To determine the relative contributions of PAI-2 from hematopoietic or nonhematopoietically derived sources, bone marrow transplants between wild-type C57BL/6J and SerpinB2-/- mice were performed. RESULTS AND CONCLUSIONS: Our results suggest that PAI-2 deficiency increases susceptibility to spontaneous tumorigenesis in the mouse, and demonstrate that SerpinB2 expression derived from a nonhematopoietic compartment is a key host factor in the regulation of tumor growth in both the B16 melanoma and Lewis lung carcinoma models.


Asunto(s)
Inhibidor 2 de Activador Plasminogénico , Serpinas , Animales , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Inhibidor 1 de Activador Plasminogénico , Inhibidor 2 de Activador Plasminogénico/genética , Serpinas/genética , Activador de Plasminógeno de Tipo Uroquinasa
10.
J Clin Invest ; 130(10): 5302-5312, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32663190

RESUMEN

Tissue factor (TF) is the primary initiator of blood coagulation in vivo and the only blood coagulation factor for which a human genetic defect has not been described. As there are no routine clinical assays that capture the contribution of endogenous TF to coagulation initiation, the extent to which reduced TF activity contributes to unexplained bleeding is unknown. Using whole genome sequencing, we identified a heterozygous frameshift variant (p.Ser117HisfsTer10) in F3, the gene encoding TF, causing premature termination of TF (TFshort) in a woman with unexplained bleeding. Routine hematological laboratory evaluation of the proposita was normal. CRISPR-edited human induced pluripotent stem cells recapitulating the variant were differentiated into vascular smooth muscle and endothelial cells that demonstrated haploinsufficiency of TF. The variant F3 transcript is eliminated by nonsense-mediated decay. Neither overexpression nor addition of exogenous recombinant TFshort inhibited factor Xa or thrombin generation, excluding a dominant-negative mechanism. F3+/- mice provide an animal model of TF haploinsufficiency and exhibited prolonged bleeding times, impaired thrombus formation, and reduced survival following major injury. Heterozygous TF deficiency is present in at least 1 in 25,000 individuals and could limit coagulation initiation in undiagnosed individuals with abnormal bleeding but a normal routine laboratory evaluation.


Asunto(s)
Trastornos de la Coagulación Sanguínea Heredados/sangre , Trastornos de la Coagulación Sanguínea Heredados/genética , Mutación del Sistema de Lectura , Tromboplastina/deficiencia , Tromboplastina/genética , Animales , Secuencia de Bases , Codón sin Sentido , Modelos Animales de Enfermedad , Femenino , Edición Génica , Haploinsuficiencia , Heterocigoto , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terminación de la Cadena Péptídica Traduccional , Fenotipo
11.
Circ Res ; 101(9): 893-901, 2007 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-17885218

RESUMEN

Heme oxygenase (HO)-1 (encoded by Hmox1) catalyzes the oxidative degradation of heme to biliverdin and carbon monoxide. HO-1 is induced during inflammation and oxidative stress to protect tissues from oxidative damage. Because intravascular thrombosis forms at sites of tissue inflammation, we hypothesized that HO-1 protects against arterial thrombosis during oxidant stress. To investigate the direct function of HO-1 on thrombosis, we used photochemical-induced vascular injury in Hmox1-/- and Hmox1+/+ mice. Hmox1-/- mice developed accelerated, occlusive arterial thrombus compared with Hmox1+/+ mice, and we detected several mechanisms accounting for this antithrombotic effect. First, endothelial cells in Hmox1-/- arteries were more susceptible to apoptosis and denudation, leading to platelet-rich microthrombi in the subendothelium. Second, tissue factor, von Willebrand Factor, and reactive oxygen species were significantly elevated in Hmox1-/- mice, consistent with endothelial cell damage and loss. Third, following transplantation of Hmox1-/- donor bone marrow into Hmox1+/+ recipients and subsequent vascular injury, we observed rapid arterial thrombosis compared with Hmox1+/+ mice receiving Hmox1+/+ bone marrow. Fourth, inhaled carbon monoxide and biliverdin administration rescued the prothrombotic phenotype in Hmox1-/- mice. Fifth, using a transcriptional analysis of arterial tissue, we found that HO-1 determined a transcriptional response to injury, with specific effects on cell cycle regulation, coagulation, thrombosis, and redox homeostasis. These data provide direct genetic evidence for a protective role of HO-1 against thrombosis and reactive oxygen species during vascular damage. Induction of HO-1 may be beneficial in the prevention of thrombosis associated with vascular oxidant stress and inflammation.


Asunto(s)
Monóxido de Carbono/metabolismo , Endotelio Vascular/enzimología , Hemo-Oxigenasa 1/genética , Trombosis/metabolismo , Trombosis/fisiopatología , Administración por Inhalación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Biliverdina/farmacología , Monóxido de Carbono/farmacología , Endotelio Vascular/patología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación Enzimológica de la Expresión Génica , Células Madre Hematopoyéticas/enzimología , Hemo-Oxigenasa 1/deficiencia , Hemostasis/fisiología , Ratones , Ratones Mutantes , Estrés Oxidativo/fisiología , Fenotipo , Tromboplastina/metabolismo , Trombosis/patología , Factor de von Willebrand/metabolismo
12.
Curr Protoc Mouse Biol ; 9(2): e61, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30875463

RESUMEN

The mammalian blood coagulation system was designed to restrict blood loss due to injury as well as keep the blood fluid within the blood vessels of the organism. Blood coagulation activity in inbred mouse strains varies widely among strains, suggesting that many genomic variants affect hemostasis. Some of these molecules have been discovered and characterized; however, many are still unknown. Genetically modified mouse technologies are providing a plethora of new mouse models for investigating the regulation of blood coagulation. Here we provide a protocol for the tail bleeding time as a primary assessment of in vivo blood coagulation, as well as in vitro methods such as the prothrombin time, activated partial thromboplastin time, and thrombin generation assay. We also provide protocols for the assessment of the activities of specific known factors involved in blood coagulation. © 2019 by John Wiley & Sons, Inc.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Pruebas de Coagulación Sanguínea/métodos , Coagulación Sanguínea , Ratones/sangre , Animales , Pruebas de Coagulación Sanguínea/instrumentación , Ratones Endogámicos , Cola (estructura animal) , Factores de Tiempo
13.
Arterioscler Thromb Vasc Biol ; 27(10): 2079-93, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17600224

RESUMEN

Thrombotic complications of vascular disease are the leading cause of morbidity and mortality in most industrialized countries. Despite this, safe and effective drugs targeting these complications are limited, especially in the chronic setting. This is because of the complexity of thrombosis in both arteries and veins, which is becoming increasingly evident as numerous factors are now known to affect the fate of a forming thrombus. To fully characterize thrombus formation in these settings, in vivo models are necessary to study the various components and intricate interactions that are involved. Genetic manipulations in mice are greatly facilitating the dissection of relevant pro- and antithrombotic influences. Standardized models for the study of thrombosis in mice as well as evolving techniques that allow imaging of molecular events during thrombus formation are now available. This review will highlight some of the recent developments in the field of thrombosis using mouse models and how these studies are expanding our knowledge of thrombotic disease.


Asunto(s)
Coagulación Sanguínea , Modelos Animales de Enfermedad , Trombosis , Animales , Aterosclerosis/genética , Vasos Sanguíneos/lesiones , Vasos Sanguíneos/fisiopatología , Cloruros , Colágeno/toxicidad , Epinefrina/toxicidad , Compuestos Férricos/toxicidad , Genotipo , Rayos Láser/efectos adversos , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Microscopía por Video , Fenotipo , Reproducibilidad de los Resultados , Rosa Bengala/toxicidad , Especificidad de la Especie , Trombosis/sangre , Trombosis/inducido químicamente , Trombosis/etiología , Trombosis/genética , Trombosis/fisiopatología
14.
J Clin Invest ; 109(2): 213-9, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11805133

RESUMEN

Heparin cofactor II (HCII) is a plasma protein that inhibits thrombin rapidly in the presence of dermatan sulfate, heparan sulfate, or heparin. HCII has been proposed to regulate coagulation or to participate in processes such as inflammation, atherosclerosis, and wound repair. To investigate the physiologic function of HCII, about 2 kb of the mouse HCII gene, encoding the N-terminal half of the protein, was deleted by homologous recombination in embryonic stem cells. Crosses of F1 HCII(+/-) animals produced HCII(-/-) offspring at the expected mendelian frequency. Biochemical assays confirmed the absence of dermatan sulfate-dependent thrombin inhibition in the plasma of HCII(-/-) animals. Crosses of HCII(-/-) animals produced litters similar in size to those obtained from heterozygous matings. At 1 year of age, HCII-deficient animals were grossly indistinguishable from their wild-type littermates in weight and survival, and they did not appear to have spontaneous thrombosis or other morphologic abnormalities. In comparison with wild-type animals, however, they demonstrated a significantly shorter time to thrombotic occlusion of the carotid artery after photochemically induced endothelial cell injury. This abnormality was corrected by infusion of purified HCII but not ovalbumin. These observations suggest that HCII might inhibit thrombosis in the arterial circulation.


Asunto(s)
Trombosis de las Arterias Carótidas/prevención & control , Cofactor II de Heparina/fisiología , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Endotelio Vascular/efectos de los fármacos , Femenino , Cofactor II de Heparina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pentobarbital/efectos adversos , Recombinación Genética , Flujo Sanguíneo Regional
15.
Circulation ; 111(14): 1822-5, 2005 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-15809370

RESUMEN

BACKGROUND: Activated protein C resistance due to factor V Leiden (FVL) is a common genetic risk factor for venous thrombosis in humans. Although the impact of FVL on the development of venous thrombosis is well established, its effect on arterial thrombosis and atherosclerosis is controversial. METHODS AND RESULTS: To determine the effect of the FVL mutation on arterial thrombosis in the mouse, wild-type (Fv+/+), heterozygous FVL (FvQ/+), and homozygous FVL (FvQ/Q) mice underwent photochemical carotid arterial injury to induce occlusive thrombosis. FvQ/Q mice formed occlusive thromboses 27+/-3 minutes (n=7) after the onset of injury, which was significantly shorter than that observed for Fv+/+ mice (56+/-7 minutes, n=9, P<0.01), whereas FvQ/+ mice (41+/-7 minutes, n=5) were intermediate (P=0.5, compared with Fv+/+). To determine the source of FVL relevant to the enhanced vascular thrombosis, bone marrow transplantation experiments were performed between Fv+/+ and FvQ/Q mice. FvQ/Q mice transplanted with Fv+/+ bone marrow formed occlusive thromboses at 35+/-5 minutes (n=7, P<0.05 compared with Fv+/+ mice), whereas Fv+/+ mice transplanted with FvQ/Q bone marrow occluded at 59+/-7 minutes (n=6, P<0.001 compared with FvQ/Q mice). To assess the effect of the FVL mutation on the development of atherosclerosis, FvQ/Q mice were crossed with the atherosclerosis-prone apolipoprotein E (ApoE)-deficient strain (ApoE-/-) to generate FvQ/Q,ApoE-/- mice. By 52 weeks of age, FvQ/Q,ApoE-/- mice (n=8) had developed more aortic atherosclerosis (40+/-6% lesion area) compared with Fv+/+,ApoE-/- mice (15+/-3% lesion area; n=12, P<0.02). CONCLUSIONS: In conclusion, homozygosity for the FVL mutation in mice leads to enhanced arterial thrombosis and atherosclerosis. The source of the FVL leading to accelerated thrombosis appears to be circulating, non-platelet-derived plasma FVL.


Asunto(s)
Arteriosclerosis/etiología , Factor V/genética , Trombosis/etiología , Animales , Enfermedades de la Aorta/etiología , Trasplante de Médula Ósea , Traumatismos de las Arterias Carótidas , Trombosis de las Arterias Carótidas/etiología , Modelos Animales de Enfermedad , Fibrinógeno/biosíntesis , Homocigoto , Ratones , Ratones Mutantes , Fotoquímica , Trombina/biosíntesis
16.
PLoS One ; 11(3): e0150852, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26950939

RESUMEN

During the analysis of a whole genome ENU mutagenesis screen for thrombosis modifiers, a spontaneous 8 base pair (bp) deletion causing a frameshift in exon 27 of the Nbeal2 gene was identified. Though initially considered as a plausible thrombosis modifier, this Nbeal2 mutation failed to suppress the synthetic lethal thrombosis on which the original ENU screen was based. Mutations in NBEAL2 cause Gray Platelet Syndrome (GPS), an autosomal recessive bleeding disorder characterized by macrothrombocytopenia and gray-appearing platelets due to lack of platelet alpha granules. Mice homozygous for the Nbeal2 8 bp deletion (Nbeal2gps/gps) exhibit a phenotype similar to human GPS, with significantly reduced platelet counts compared to littermate controls (p = 1.63 x 10-7). Nbeal2gps/gps mice also have markedly reduced numbers of platelet alpha granules and an increased level of emperipolesis, consistent with previously characterized mice carrying targeted Nbeal2 null alleles. These findings confirm previous reports, provide an additional mouse model for GPS, and highlight the potentially confounding effect of background spontaneous mutation events in well-characterized mouse strains.


Asunto(s)
Emparejamiento Base/genética , Proteínas Sanguíneas/genética , Mutación del Sistema de Lectura , Síndrome de Plaquetas Grises/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Sanguíneas/química , Médula Ósea/inmunología , Emperipolesis/genética , Exoma/genética , Exones/genética , Femenino , Fertilidad/genética , Síndrome de Plaquetas Grises/complicaciones , Síndrome de Plaquetas Grises/inmunología , Síndrome de Plaquetas Grises/fisiopatología , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Neutropenia/complicaciones , Neutrófilos/citología , Bazo/inmunología , Trombocitopenia/complicaciones
17.
Circulation ; 105(18): 2139-42, 2002 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-11994245

RESUMEN

BACKGROUND: Factor V Leiden (FVL) is a common genetic risk factor for thrombosis in humans. The incomplete penetrance of FVL suggests important contributions from other genetic or environmental modifying factors. Variation in the expression of tissue factor pathway inhibitor (TFPI) has also been proposed as a risk factor for venous thrombosis and has been shown to enhance the prothrombotic effect of FVL in vitro. METHODS AND RESULTS: To examine the potential in vivo interaction between Tfpi and FvL, we analyzed crosses between mice carrying FvL and a deficiency of TFPI. The Fv(Q/Q),Tfpi(+/-) genotype was nearly completely fatal in the early perinatal period. Increased fibrin deposition was observed in multiple organs from the Fv(Q/Q),Tfpi(+/-) fetuses, suggesting disseminated thrombosis. CONCLUSIONS: These observations demonstrate the prothrombotic effect of modest variations in the level of TFPI expression and suggest that TFPI could be an important genetic modifier for the thrombosis associated with FVL in humans.


Asunto(s)
Factor V/fisiología , Lipoproteínas/fisiología , Trombosis/etiología , Animales , Animales Recién Nacidos , Factor V/genética , Fibrina/metabolismo , Heterocigoto , Lipoproteínas/genética , Ratones , Mutación , Fenotipo , Análisis de Supervivencia , Trombosis/metabolismo , Trombosis/patología
18.
Mamm Genome ; 19(1): 32-40, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18188647

RESUMEN

Mvwf1 is a cis-regulatory mutation previously identified in the RIIIS/J mouse strain that causes a unique tissue-specific switch in the expression of an N-acetylgalactosaminyltransferase, B4GALNT2, from intestinal epithelium to vascular endothelium. Vascular B4galnt2 expression results in aberrant glycosylation of von Willebrand Factor (VWF) and accelerated VWF clearance from plasma. We now report that 13 inbred mouse strains share the Mvwf1 tissue-specific switch and low VWF phenotype, including five wild-derived strains. Genomic sequencing identified a highly conserved 97-kb Mvwf1 haplotype block shared by these strains that encompasses a 30-kb region of high nucleotide sequence divergence from C57BL6/J flanking B4galnt2 exon 1. The analysis of a series of bacterial artificial chromosome (BAC) transgenes containing B4galnt2 derived from the RIIIS/J or C57BL6/J inbred mouse strains demonstrates that the corresponding sequences are sufficient to confer the vessel (RIIIS/J) or intestine (C57BL6/J)-specific expression patterns. Taken together, our data suggest that the region responsible for the Mvwf1 regulatory switch lies within an approximately 30-kb genomic interval upstream of the B4galnt2 gene. The observation that Mvwf1 is present in multiple wild-derived strains suggests that this locus may be retained in wild mouse populations due to positive selection. Similar selective pressures could contribute to the high prevalence of von Willebrand disease in humans.


Asunto(s)
Alelos , Endotelio Vascular/metabolismo , Mutación/genética , N-Acetilgalactosaminiltransferasas/genética , Animales , Cromosomas Artificiales Bacterianos , Secuencia Conservada , Endotelio Vascular/enzimología , Genoma , Haplotipos , Intestinos/citología , Intestinos/enzimología , Lectinas/metabolismo , Ratones , Especificidad de Órganos , Fenotipo , Transgenes , Polipéptido N-Acetilgalactosaminiltransferasa
19.
Curr Drug Targets ; 8(9): 966-1002, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17896948

RESUMEN

Thrombotic complications of vascular disease constitute the leading cause of morbidity and mortality in much of the developed world. Current drug therapies available to treat the thrombotic component of arterial and venous vascular complications remain limited. Novel safe and effective treatment strategies to reduce formation of occlusive thrombosis will likely have a major impact on reducing the economic burden of vascular disease on the healthcare system. Enhancing endogenous fibrinolysis by targeting plasminogen activator inhibitor-1 (PAI-1), the primary inhibitor of circulating plasminogen activators, has been shown to be effective in markedly attenuating the formation of arterial and venous occlusive thrombosis in animal models. In addition, animal and human studies of PAI-1 deficiency indicate that spontaneous bleeding complications associated with even complete PAI-1 deficiency would be rare. Patients most likely to benefit from PAI-1 inhibition would be those at high risk for vascular events where PAI-1 is elevated, such as is observed in obesity, diabetes and the metabolic syndrome. Since obesity and metabolic syndrome are now epidemic, and will likely have a major adverse impact on vascular thrombotic events, it may be time to test the clinical effectiveness of PAI-1 inhibition in a patient population at high risk for vascular thrombosis.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/metabolismo , Trombosis/prevención & control , Animales , Plaquetas/metabolismo , Fibrinolisina/metabolismo , Fibrinólisis/fisiología , Humanos , Plasminógeno/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Activadores Plasminogénicos/uso terapéutico , Polimorfismo Genético , Regiones Promotoras Genéticas , Inhibidores de Serina Proteinasa/genética , Inhibidores de Serina Proteinasa/metabolismo , Trombosis/etiología , Vitronectina/metabolismo
20.
Proc Natl Acad Sci U S A ; 104(44): 17518-23, 2007 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-17956977

RESUMEN

The signaling lipid, phosphatidylinositol 3,5-bisphosphate (PI(3,5)P(2)), likely functions in multiple signaling pathways. Here, we report the characterization of a mouse mutant lacking Vac14, a regulator of PI(3,5)P(2) synthesis. The mutant mice exhibit massive neurodegeneration, particularly in the midbrain and in peripheral sensory neurons. Cell bodies of affected neurons are vacuolated, and apparently empty spaces are present in areas where neurons should be present. Similar vacuoles are found in cultured neurons and fibroblasts. Selective membrane trafficking pathways, especially endosome-to-TGN retrograde trafficking, are defective. This report, along with a recent report on a mouse with a null mutation in Fig4, presents the unexpected finding that the housekeeping lipid, PI(3,5)P(2), is critical for the survival of neural cells.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Degeneración Nerviosa/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Transducción de Señal , Animales , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana , Ratones , Ratones Noqueados , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA