Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Blood ; 139(9): 1374-1388, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34905618

RESUMEN

Genetic variants within the fibrinogen Aα chain encoding the αC-region commonly result in hypodysfibrinogenemia in patients. However, the (patho)physiological consequences and underlying mechanisms of such mutations remain undefined. Here, we generated Fga270 mice carrying a premature termination codon within the Fga gene at residue 271. The Fga270 mutation was compatible with Mendelian inheritance for offspring of heterozygous crosses. Adult Fga270/270 mice were hypofibrinogenemic with ∼10% plasma fibrinogen levels relative to FgaWT/WT mice, linked to 90% reduction in hepatic Fga messenger RNA (mRNA) because of nonsense-mediated decay of the mutant mRNA. Fga270/270 mice had preserved hemostatic potential in vitro and in vivo in models of tail bleeding and laser-induced saphenous vein injury, whereas Fga-/- mice had continuous bleeding. Platelets from FgaWT/WT and Fga270/270 mice displayed comparable initial aggregation following adenosine 5'-diphosphate stimulation, but Fga270/270 platelets quickly disaggregated. Despite ∼10% plasma fibrinogen, the fibrinogen level in Fga270/270 platelets was ∼30% of FgaWT/WT platelets with a compensatory increase in fibronectin. Notably, Fga270/270 mice showed complete protection from thrombosis in the inferior vena cava stasis model. In a model of Staphylococcus aureus peritonitis, Fga270/270 mice supported local, fibrinogen-mediated bacterial clearance and host survival comparable to FgaWT/WT, unlike Fga-/- mice. Decreasing the normal fibrinogen levels to ∼10% with small interfering RNA in mice also provided significant protection from venous thrombosis without compromising hemostatic potential and antimicrobial function. These findings both reveal novel molecular mechanisms underpinning fibrinogen αC-region truncation mutations and highlight the concept that selective fibrinogen reduction may be efficacious for limiting thrombosis while preserving hemostatic and immune protective functions.


Asunto(s)
Afibrinogenemia , Plaquetas/metabolismo , Fibrinógeno , Hemostasis/genética , Mutación , Agregación Plaquetaria/genética , Trombosis , Afibrinogenemia/genética , Afibrinogenemia/metabolismo , Animales , Fibrinógeno/genética , Fibrinógeno/metabolismo , Ratones , Ratones Noqueados , Trombosis/genética , Trombosis/metabolismo
2.
Blood ; 139(9): 1302-1311, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-34958662

RESUMEN

Fibrinogen plays a pathologic role in multiple diseases. It contributes to thrombosis and modifies inflammatory and immune responses, supported by studies in mice expressing fibrinogen variants with altered function or with a germline fibrinogen deficiency. However, therapeutic strategies to safely and effectively tailor plasma fibrinogen concentration are lacking. Here, we developed a strategy to tune fibrinogen expression by administering lipid nanoparticle (LNP)-encapsulated small interfering RNA (siRNA) targeting the fibrinogen α chain (siFga). Three distinct LNP-siFga reagents reduced both hepatic Fga messenger RNA and fibrinogen levels in platelets and plasma, with plasma levels decreased to 42%, 16%, and 4% of normal within 1 week of administration. Using the most potent siFga, circulating fibrinogen was controllably decreased to 32%, 14%, and 5% of baseline with 0.5, 1.0, and 2.0 mg/kg doses, respectively. Whole blood from mice treated with siFga formed clots with significantly decreased clot strength ex vivo, but siFga treatment did not compromise hemostasis following saphenous vein puncture or tail transection. In an endotoxemia model, siFga suppressed the acute phase response and decreased plasma fibrinogen, D-dimer, and proinflammatory cytokine levels. In a sterile peritonitis model, siFga restored normal macrophage migration in plasminogen-deficient mice. Finally, treatment of mice with siFga decreased the metastatic potential of tumor cells in a manner comparable to that observed in fibrinogen-deficient mice. The results indicate that siFga causes robust and controllable depletion of fibrinogen and provides the proof-of-concept that this strategy can modulate the pleiotropic effects of fibrinogen in relevant disease models.


Asunto(s)
Afibrinogenemia/metabolismo , Fibrina/biosíntesis , Fibrinógeno/biosíntesis , Técnicas de Silenciamiento del Gen , Liposomas/farmacología , ARN Interferente Pequeño , Afibrinogenemia/genética , Animales , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrina/genética , Fibrinógeno/genética , Humanos , Masculino , Ratones , Nanopartículas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
3.
Semin Thromb Hemost ; 48(8): 889-903, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35073585

RESUMEN

Fibrinogen is a complex protein that plays a key role in the blood clotting process. It is a hexamer composed of two copies of three distinct chains: Aα, Bß, and γ encoded by three genes, FGA, FGB, and FGG, clustered on the long arm of chromosome 4. Congenital fibrinogen disorders (CFDs) are divided into qualitative deficiencies (dysfibrinogenemia, hypodysfibrinogenemia) in which the mutant fibrinogen molecule is present in the circulation and quantitative deficiencies (afibrinogenemia, hypofibrinogenemia) with no mutant molecule present in the bloodstream. Phenotypic manifestations are variable, patients may be asymptomatic, or suffer from bleeding or thrombosis. Causative mutations can occur in any of the three fibrinogen genes and can affect one or both alleles. Given the large number of studies reporting on novel causative mutations for CFDs since the review on the same topic published in 2016, we performed an extensive search of the literature and list here 120 additional mutations described in both quantitative and qualitative disorders. The visualization of causative single nucleotide variations placed on the coding sequences of FGA, FGB, and FGG reveals important structure function insight for several domains of the fibrinogen molecule.


Asunto(s)
Afibrinogenemia , Hemostáticos , Humanos , Fibrinógeno/genética , Fibrinógeno/metabolismo , Afibrinogenemia/genética , Afibrinogenemia/metabolismo , Mutación , Hemorragia/genética , Exones
4.
Int J Mol Sci ; 22(11)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071368

RESUMEN

Alpha-1-antitrypsin (AAT) and fibrinogen are secretory acute phase reactant proteins. Circulating AAT and fibrinogen are synthesized exclusively in the liver. Mutations in the encoding genes result in conformational abnormalities of the two molecules that aggregate within the rough endoplasmic reticulum (RER) instead of being regularly exported. That results in AAT-deficiency (AATD) and in hereditary hypofibrinogenemia with hepatic storage (HHHS). The association of plasma deficiency and liver storage identifies a new group of pathologies: endoplasmic reticulum storage disease (ERSD).


Asunto(s)
Afibrinogenemia/metabolismo , Retículo Endoplásmico/metabolismo , Hígado/metabolismo , Deficiencia de alfa 1-Antitripsina/metabolismo , Afibrinogenemia/genética , Hepatocitos/citología , Hepatocitos/metabolismo , Hepatocitos/ultraestructura , Humanos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/ultraestructura , Hígado/citología , Microscopía Electrónica de Transmisión , Mutación , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/genética
5.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830348

RESUMEN

Dysfunction of cellular homeostasis can lead to misfolding of proteins thus acquiring conformations prone to polymerization into pathological aggregates. This process is associated with several disorders, including neurodegenerative diseases, such as Parkinson's disease (PD), and endoplasmic reticulum storage disorders (ERSDs), like alpha-1-antitrypsin deficiency (AATD) and hereditary hypofibrinogenemia with hepatic storage (HHHS). Given the shared pathophysiological mechanisms involved in such conditions, it is necessary to deepen our understanding of the basic principles of misfolding and aggregation akin to these diseases which, although heterogeneous in symptomatology, present similarities that could lead to potential mutual treatments. Here, we review: (i) the pathological bases leading to misfolding and aggregation of proteins involved in PD, AATD, and HHHS: alpha-synuclein, alpha-1-antitrypsin, and fibrinogen, respectively, (ii) the evidence linking each protein aggregation to the stress mechanisms occurring in the endoplasmic reticulum (ER) of each pathology, (iii) a comparison of the mechanisms related to dysfunction of proteostasis and regulation of homeostasis between the diseases (such as the unfolded protein response and/or autophagy), (iv) and clinical perspectives regarding possible common treatments focused on improving the defensive responses to protein aggregation for diseases as different as PD, and ERSDs.


Asunto(s)
Afibrinogenemia/genética , Fibrinógeno/química , Enfermedad de Parkinson/genética , Deficiencia de alfa 1-Antitripsina/genética , alfa 1-Antitripsina/química , alfa-Sinucleína/química , Afibrinogenemia/tratamiento farmacológico , Afibrinogenemia/metabolismo , Afibrinogenemia/patología , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Coagulantes/uso terapéutico , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Fibrinógeno/genética , Fibrinógeno/metabolismo , Regulación de la Expresión Génica , Humanos , Hígado/metabolismo , Hígado/patología , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Inhibidores de Proteasas/uso terapéutico , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de los fármacos , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico , Deficiencia de alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/patología , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo
6.
Chin Med Sci J ; 35(2): 151-156, 2020 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-32684235

RESUMEN

Objective Hemocoagulase injection based on the venom of Agkistrodon halys Pallas is widely used in the treatment of hemorrhagic disorders. This study aimed to characterize the clinical laboratory findings of hemocoagulase-induced hypofibrinogenemia as the associated adverse reaction of hemocoagulase injection.Methods We retrospectively enrolled 27 in-patients who were treated with hemocoagulase injection for hemoptysis and developed hypofibrinogenemia during the period of January 1, 2015 to March 31, 2018. Clinical data were collected and investigated, including clinical manifestations, hemostatic and fibrinolytic parameters, dosage of hemocoagulase, the medication time, and the cryoprecipitate blood product infusion. Differences in fibrinogen, D-dimer, and fibrin/fibrinogen degradation products (FDP) before, during, and after the application of hemocoagulase injection were analyzed statistically.Results Plasma fibrinogen level during medication of hemocoagulase injection decreased significantly compared to that before the treatment (F=1.80, P<0.001), with the average decrease of 2.28 g/L (0.63-3.9 g/L). After withdrawal, fibrinogen level increased significantly compared to that during the medication (F=-1.20, P<0.001), but was still lower than that before the medication (F=0.59, P=0.03). The D-dimer level and the FDP level after withdrawal decreased significantly compared to the levels during the medication (F=0.83, P=0.002; Wilcoxon-test, Z=-4.54, P<0.001). Spearman's correlation analyses did not find either fibrinogen change during-before the administration or FDP change after-during the administration was associated with the dosage of hemocoagulase (r=-0.17, P=0.40; r=-0.28, P=0.15; respectively) and the time of recovery from hypofibrinogenemia (r=-0.45, P=0.05; r=0.13, P=0.61; respectively).Conclusion Monitoring both clotting and fibrinolysis parameters is essential in the management of hemoptysis patients treated with hemocoagulase injection. Clinicians should be aware of hypofibrinogenemia and consider discontinuation of the administration of hemocoagulase whenever necessary.


Asunto(s)
Afibrinogenemia/inducido químicamente , Batroxobina/efectos adversos , Fibrinógeno/metabolismo , Afibrinogenemia/sangre , Afibrinogenemia/metabolismo , Batroxobina/administración & dosificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
7.
Pediatr Blood Cancer ; 66(9): e27832, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31131962

RESUMEN

Hypodysfibrinogenemia, the least frequently reported congenital fibrinogen disorder is characterized by low circulating levels of a dysfunctional protein, and is associated with phenotypic features of both hypo- and dysfibrinogenemia. Herein, we report an adolescent male with unprovoked venous thromboembolism and hypodysfibrinogenemia. Patient had recurrent, progressive thrombosis despite therapeutic anticoagulation with both low molecular weight heparin and warfarin. He had clinical and radiological improvement after transition to a direct thrombin inhibitor. Sequencing of the FGG gene identified a novel heterozygous mutation, c.1075G>T. Structural visualization of the identified variant was pursued and suggested that the mutation likely destabilizes the Ca2+ -binding site of fibrinogen resulting in pathogenicity.


Asunto(s)
Afibrinogenemia , Fibrinógenos Anormales , Heterocigoto , Mutación Puntual , Trombosis de la Vena , Adolescente , Afibrinogenemia/genética , Afibrinogenemia/metabolismo , Sitios de Unión , Calcio/química , Calcio/metabolismo , Fibrinógenos Anormales/química , Fibrinógenos Anormales/genética , Fibrinógenos Anormales/metabolismo , Humanos , Masculino , Trombosis de la Vena/genética , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología
8.
Haemophilia ; 24(6): e410-e416, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30345592

RESUMEN

Fibrinogen is a glycoprotein with a crucial role in blood coagulation. Upon enzymatic cleavage by thrombin, fibrinogen is converted from its soluble form to insoluble fibrin which is key structural protein of a clot. It also participates in platelet aggregation by binding to GPIIb/IIIa. Genetic alterations can lead to either complete or partial, quantitative or qualitative defects of fibrinogen. Inherited afibrinogenemia is a rare bleeding disorder with autosomal recessive inheritance due to a complete absence of fibrinogen. The primary aim of this study was to determine whether 70 mg/kg of human fibrinogen concentrate (HFC) is an adequate dose in subjects with inherited afibrinogenemia to reach normal levels of plasmatic fibrinogen (1.5-2 g/L). Secondary aims included assessing changes in thromboelastography (TEG) and thrombin generation assay (TGA) before and after a dose of HFC. Four patients were included, and each underwent pre-and post (one time-point) HFC dose laboratory testing. Two patients needed dose adjustments to reach a normal post-dose fibrinogen level. In addition, we noted that the TEG parameter maximum amplitude (MA) improved in accordance with correction of the fibrinogen levels. TGA results were normal in all subjects. Our results suggest that individualized dosing based on fibrinogen levels may be necessary.


Asunto(s)
Afibrinogenemia/diagnóstico , Afibrinogenemia/genética , Tromboelastografía , Trombina/biosíntesis , Adolescente , Afibrinogenemia/metabolismo , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Adulto Joven
9.
Int J Mol Sci ; 19(1)2018 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-29316703

RESUMEN

The study of inherited fibrinogen disorders, characterized by extensive allelic heterogeneity, allows the association of defined mutations with specific defects providing significant insight into the location of functionally important sites in fibrinogen and fibrin. Since the identification of the first causative mutation for congenital afibrinogenemia, studies have elucidated the underlying molecular pathophysiology of numerous causative mutations leading to fibrinogen deficiency, developed cell-based and animal models to study human fibrinogen disorders, and further explored the clinical consequences of absent, low, or dysfunctional fibrinogen. Since qualitative disorders are addressed by another review in this special issue, this review will focus on quantitative disorders and will discuss their diagnosis, clinical features, molecular bases, and introduce new models to study the phenotypic consequences of fibrinogen deficiency.


Asunto(s)
Afibrinogenemia/patología , Fibrinógeno/genética , Afibrinogenemia/genética , Afibrinogenemia/metabolismo , Animales , Modelos Animales de Enfermedad , Fibrinógeno/química , Fibrinógeno/metabolismo , Humanos , Fenotipo
10.
Int J Mol Sci ; 19(6)2018 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-29844251

RESUMEN

Congenital fibrinogen disorders can be quantitative (afibrinogenemia, hypofibrinogenemia) or functional (dysfibrinognemia). To date, several genetic variants have been identified in individuals with fibrinogen disorders. The complexity of the fibrinogen molecules, formed by three non-identical chains and with a trinodal organization, renders the identification of molecular causes and of clinical and biochemical phenotypes very challenging. However, the acknowledgement of the type of molecular defect is crucial for a safer therapy, which is going to improve the clinical management of these patients. In this review, some aspects concerning molecular and clinical findings available on congenital fibrinogen disorders will be discussed.


Asunto(s)
Afibrinogenemia , Fibrinógeno , Afibrinogenemia/genética , Afibrinogenemia/metabolismo , Afibrinogenemia/terapia , Fibrinógeno/genética , Fibrinógeno/metabolismo , Humanos
11.
J Hepatol ; 66(4): 787-797, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27965156

RESUMEN

BACKGROUND & AIMS: Acetaminophen (APAP)-induced liver injury is coupled with activation of the blood coagulation cascade and fibrin(ogen) accumulation within APAP-injured livers of experimental mice. We sought to define the role of fibrin(ogen) deposition in APAP-induced liver injury and repair. METHODS: Wild-type, fibrinogen-deficient mice, mutant mice with fibrin(ogen) incapable of binding leukocyte αMß2 integrin (Fibγ390-396A mice) and matrix metalloproteinase 12 (Mmp12)-deficient mice were fasted, injected with 300mg/kg APAP i.p. and evaluated at a range of time-points. Plasma and liver tissue were analyzed. Rescue of Fibγ390-396A mice was carried out with exogenous Mmp12. To examine the effect of the allosteric leukocyte integrin αMß2 activator leukadherin-1 (LA-1), APAP-treated mice were injected with LA-1. RESULTS: In wild-type mice, APAP overdose increased intrahepatic levels of high molecular weight cross-linked fibrin(ogen). Anticoagulation reduced early APAP hepatotoxicity (6h), but increased hepatic injury at 24h, implying a protective role for coagulation at the onset of repair. Complete fibrin(ogen) deficiency delayed liver repair after APAP overdose, evidenced by a reduction of proliferating hepatocytes (24h) and unresolved hepatocellular necrosis (48 and 72h). Fibγ390-396A mice had decreased hepatocyte proliferation and increased multiple indices of liver injury, suggesting a mechanism related to fibrin(ogen)-leukocyte interaction. Induction of Mmp12, was dramatically reduced in APAP-treated Fibγ390-396A mice. Mice lacking Mmp12 displayed exacerbated APAP-induced liver injury, resembling Fibγ390-396A mice. In contrast, administration of LA-1 enhanced hepatic Mmp12 mRNA and reduced necrosis in APAP-treated mice. Further, administration of recombinant Mmp12 protein to APAP-treated Fibγ390-396A mice restored hepatocyte proliferation. CONCLUSIONS: These studies highlight a novel pathway of liver repair after APAP overdose, mediated by fibrin(ogen)-αMß2 integrin engagement, and demonstrate a protective role of Mmp12 expression after APAP overdose. LAY SUMMARY: Acetaminophen overdose leads to activation of coagulation cascade and deposition of high molecular weight cross-linked fibrin(ogen) species in the liver. Fibrin(ogen) is required for stimulating liver repair after acetaminophen overdose. The mechanism whereby fibrin(ogen) drives liver repair after acetaminophen overdose requires engagement of leukocyte αMß2 integrin and subsequent induction of matrix metalloproteinase 12.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Fibrina/metabolismo , Fibrinógeno/metabolismo , Antígeno de Macrófago-1/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , Acetaminofén/toxicidad , Afibrinogenemia/genética , Afibrinogenemia/metabolismo , Animales , Antitrombinas/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Dabigatrán/farmacología , Femenino , Fibrina/deficiencia , Fibrina/genética , Fibrinógeno/genética , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Regeneración Hepática/efectos de los fármacos , Regeneración Hepática/fisiología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinasa 12 de la Matriz/deficiencia , Metaloproteinasa 12 de la Matriz/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes
12.
Int J Mol Sci ; 18(12)2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29240685

RESUMEN

Fibrinogen is a highly pleiotropic protein that is involved in the final step of the coagulation cascade, wound healing, inflammation, and angiogenesis. Heterozygous mutations in Aα, Bß, or γ fibrinogen-chain genes (FGA, FGB, FGG) have been described as being responsible for fibrinogen deficiencies (hypofibrinogenemia, hypo-dysfibrinogenemia, dysfibrinogenemia) and for more rare conditions, such as fibrinogen storage disease and hereditary renal amyloidosis. Instead, biallelic mutations have been associated with afibrinogenemia/severe hypofibrinogenemia, i.e., the severest forms of fibrinogen deficiency, affecting approximately 1-2 cases per million people. However, the "true" prevalence for these conditions on a global scale is currently not available. Here, we defined the mutational burden of the FGA, FGB, and FGG genes, and estimated the prevalence of inherited fibrinogen disorders through a systematic analysis of exome/genome data from ~140,000 individuals belonging to the genome Aggregation Database. Our analysis showed that the world-wide prevalence for recessively-inherited fibrinogen deficiencies could be 10-fold higher than that reported so far (prevalence rates vary from 1 in 106 in East Asians to 24.5 in 106 in non-Finnish Europeans). The global prevalence for autosomal-dominant fibrinogen disorders was estimated to be ~11 in 1000 individuals, with heterozygous carriers present at a frequency varying from 3 every 1000 individuals in Finns, to 1-2 every 100 individuals among non-Finnish Europeans and Africans/African Americans. Our analysis also allowed for the identification of recurrent (i.e., FGG-p.Ala108Gly, FGG-Thr47Ile) or ethnic-specific mutations (e.g., FGB-p.Gly103Arg in Admixed Americans, FGG-p.Ser245Phe in Africans/African Americans).


Asunto(s)
Afibrinogenemia/genética , Fibrinógeno/genética , Pleiotropía Genética/genética , Mutación , Afibrinogenemia/etnología , Afibrinogenemia/metabolismo , Pueblo Asiatico/genética , Población Negra/genética , Bases de Datos Genéticas , Fibrinógeno/metabolismo , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Genotipo , Humanos , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Población Blanca/genética
13.
Int J Mol Sci ; 19(1)2017 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-29286337

RESUMEN

Congenital fibrinogen disorders are caused by mutations in one of the three fibrinogen genes that affect the synthesis, assembly, intracellular processing, stability or secretion of fibrinogen. Functional studies of mutant Bß-chains revealed the importance of individual residues as well as three-dimensional structures for fibrinogen assembly and secretion. This study describes two novel homozygous fibrinogen Bß chain mutations in two Slovak families with afibrinogenemia and hypofibrinogenemia. Peripheral blood samples were collected from all subjects with the aim of identifying the causative mutation. Coagulation-related tests and rotational thromboelastometry were performed. All exons and exon-intron boundaries of the fibrinogen genes (FGA, FGB and FGG) were amplified by PCR followed by direct sequencing. Sequence analysis of the three fibrinogen genes allowed us to identify two novel homozygous mutations in the FGB gene. A novel Bß chain truncation (BßGln180Stop) was detected in a 28-year-old afibrinogenemic man with bleeding episodes including repeated haemorrhaging into muscles, joints, and soft tissues, and mucocutaneous bleeding and a novel Bß missense mutation (BßTyr368His) was found in a 62-year-old hypofibrinogenemic man with recurrent deep and superficial venous thromboses of the lower extremities. The novel missense mutation was confirmed by molecular modelling. Both studying the molecular anomalies and the modelling of fibrinogenic mutants help us to understand the extremely complex machinery of fibrinogen biosynthesis and finally better assess its correlation with the patient's clinical course.


Asunto(s)
Afibrinogenemia/genética , Fibrinógeno/genética , Hemorragia/genética , Mutación , Trombosis de la Vena/genética , Adulto , Afibrinogenemia/metabolismo , Afibrinogenemia/fisiopatología , Secuencia de Bases , Pruebas de Coagulación Sanguínea , Familia , Fibrinógeno/química , Fibrinógeno/metabolismo , Fibrinógenos Anormales/genética , Fibrinógenos Anormales/metabolismo , Expresión Génica , Hemorragia/metabolismo , Hemorragia/fisiopatología , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Linaje , Estructura Secundaria de Proteína , Índice de Severidad de la Enfermedad , Trombosis de la Vena/metabolismo , Trombosis de la Vena/fisiopatología
14.
Semin Thromb Hemost ; 42(4): 356-65, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27019463

RESUMEN

Congenital fibrinogen disorders are classified into two types of plasma fibrinogen defects: type I (quantitative fibrinogen deficiencies), that is, hypofibrinogenemia or afibrinogenemia, in which there are low or absent plasma fibrinogen antigen levels, respectively, and type II (qualitative fibrinogen deficiencies), that is, dysfibrinogenemia or hypodysfibrinogenemia, in which there are normal or reduced antigen levels associated with disproportionately low functional activity. These disorders are caused by mutations in the three fibrinogen-encoding genes FGA, FGB, and FGG. Afibrinogenemia is associated with mild to severe bleeding, whereas hypofibrinogenemia is often asymptomatic. For these quantitative disorders, the majority of mutations prevent protein production. However, in some cases, missense or late-truncating nonsense mutations allow synthesis of the mutant fibrinogen chain, but intracellular fibrinogen assembly and/or secretion are impaired. Qualitative fibrinogen disorders are associated with bleeding, thrombosis, or both thrombosis and bleeding, but many dysfibrinogenemias are asymptomatic. The majority of cases are caused by heterozygous missense mutations. Here, we review the laboratory and genetic diagnosis of fibrinogen gene anomalies with an updated discussion of causative mutations identified.


Asunto(s)
Afibrinogenemia , Fibrinógeno , Hemorragia , Mutación Missense , Trombosis , Afibrinogenemia/genética , Afibrinogenemia/metabolismo , Animales , Fibrinógeno/genética , Fibrinógeno/metabolismo , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Trombosis/genética , Trombosis/metabolismo
15.
Clin Exp Pharmacol Physiol ; 43(2): 149-56, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26510121

RESUMEN

Congenital (hypo)dysfibrinogenemia patients may have obstetric complications during their pregnancies. This study aimed to evaluate thromboelastography (TEG) as a potential tool for assessing the tendency for obstetric complications in those patients in a non-pregnant state. A total of 22 female subjects with congenital (hypo)dysfibrinogenemia were recruited. Nine subjects had histories of obstetric complications and the other 13 subjects had at least one uneventful pregnancy without obstetric complications as yet. Detailed clinical investigation and phenotype/genotype detection were carried out, and both kaolin-activated TEG and functional fibrinogen TEG (FF-TEG) were applied in all subjects. Significant differences were identified in all TEG parameters except for R and angle between these two groups (P < 0.05) by covariance analysis. Receiver operating characteristic (ROC) analysis of discrimination between these two groups of patients was performed for TEG parameters. Significantly high odds ratio (OR) of obstetric complications occurrence were demonstrated in K ≥ 3.8 min, maximum amplitude (MA) ≤ 54.2 mm, comprehensive index (CI) ≤ -3 (11.67, 95% CI 1.527-89.121, P < 0.05 in all), and MA-CFF ≤ 12.1 mm (20.00, 95% confidence interval (95% CI) 1.967-203.322, P = 0.002). Moreover, MA-CFF had better prognostic performance, with a corresponding area under the receiver operating curve of 0.923 (range 0.815-1.031, P = 0.001). This study suggests that (hypo)dysfibrinogenemia patients with values outside of the cut-off values of TEG assays under non-pregnant state may have a higher risk of obstetric complications occurring when they are pregnant. No parameters under non-pregnant state in clinical laboratory have ever been reported to be risk factors for obstetric complication occurrence in (hypo)dysfibrinogenemia patients. This study explored such parameters in TEG assays and found that parameters of TEG assays under non-pregnant status might predict the occurrence of obstetric complications, which could provide physicians with important information about whether fibrinogen replacement therapy is required, so as to prevent the occurrence of obstetric complications, especially for patients who are asymptomatic in daily life.


Asunto(s)
Afibrinogenemia/diagnóstico , Complicaciones del Embarazo/diagnóstico , Tromboelastografía , Adulto , Afibrinogenemia/complicaciones , Afibrinogenemia/metabolismo , Femenino , Fibrinógeno/metabolismo , Humanos , Caolín/farmacología , Embarazo , Complicaciones del Embarazo/metabolismo , Riesgo
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 33(2): 160-3, 2016 Apr.
Artículo en Zh | MEDLINE | ID: mdl-27060305

RESUMEN

OBJECTIVE: To explore the clinical phenotype of a family affected with congenital dysfibrinogenemia and potential mutations underlying the disease. METHODS: Coagulation testing and hepatorenal function testing were conducted on 18 individuals from three generations. Plasma fibrinogen was extracted and analyzed with SDS-PAGE electrophoresis. All of the exons and flanking sequences of fibrinogen FGA, FGB, FGG genes were analyzed by PCR, and the products were subjected to Sanger sequencing. RESULTS: Hepatorenal function, prothrombin time and activated partial thromboplastin time of the proband were all normal. However, his thrombin time was significantly prolonged. Fibrinogen activity was decreased, while the concentration of antigen was in the normal range. The results of his mother, brother, and nephew were similar. DNA sequencing has confirmed that the proband, his mother, brother, and nephew have all carried a g.5877G>A mutation in the exon 8 of the FGG gene, which resulted in replacement of arginine (Arg) by histidine (His) at position 275. CONCLUSION: The Arg275His mutation of the fibrinogen gamma chain probably underlies the pathogenesis of congenital dysfibrinogenemia in this family.


Asunto(s)
Afibrinogenemia/genética , Fibrinógeno/genética , Mutación Missense , Adulto , Afibrinogenemia/metabolismo , Pueblo Asiatico/genética , Secuencia de Bases , China , Femenino , Fibrinógeno/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Mutación Puntual
17.
Liver Int ; 35(12): 2501-5, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26176881

RESUMEN

BACKGROUND AND AIMS: Fibrinogen gene mutations can rarely result in hepatic fibrinogen storage disease (HFSD). Herein, we report on the first Turkish family carrying the mutation p.Arg375Trp (fibrinogen Aguadilla) in the γ-chain of the fibrinogen (FGG) gene. METHODS: Clinical, laboratory and histopathological findings of the patient were documented. Molecular study of fibrinogen gene was performed in the patient and her family members. RESULTS: The proband was 5 years old girl presenting with advanced liver fibrosis of unknown origin. The child had very low plasma levels of fibrinogen and hypobetalipoproteinemia. Immunomorphologic and electron microscopic studies showed selective and exclusive accumulation of fibrinogen within the endoplasmic reticulum in liver biopsy of the patient. Patient, mother, two sisters and one brother carried p.Arg375Trp mutation (fibrinogen Aguadilla) in FGG gene. The patient was treated with ursodeoxycholic acid and carbamazepine. After 3 months, carbamazepine was suspended upon family decision and unresponsiveness of carbamazepine. CONCLUSIONS: HFSD is characterized by hypofibrinogenemia and accumulation of abnormal fibrinogen within hepatocytes. In addition, hypofibrinogenemia is associated with hypobetalipoproteinemia in Aguadilla mutation.


Asunto(s)
Afibrinogenemia , Carbamazepina/administración & dosificación , Fibrinógeno , Hipobetalipoproteinemias , Cirrosis Hepática , Ácido Ursodesoxicólico/administración & dosificación , Afibrinogenemia/diagnóstico , Afibrinogenemia/etiología , Afibrinogenemia/metabolismo , Preescolar , Colagogos y Coleréticos/administración & dosificación , Inductores del Citocromo P-450 CYP3A/administración & dosificación , Femenino , Fibrinógeno/análisis , Fibrinógeno/genética , Humanos , Hipobetalipoproteinemias/complicaciones , Hipobetalipoproteinemias/diagnóstico , Hipobetalipoproteinemias/genética , Hipobetalipoproteinemias/fisiopatología , Hígado/patología , Cirrosis Hepática/diagnóstico , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/etiología , Cirrosis Hepática/fisiopatología , Mutación Missense , Resultado del Tratamiento
18.
J Hepatol ; 59(3): 626-30, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23707368

RESUMEN

Fibrinogen storage disease (FSD) is a rare autosomal-dominant hereditary disorder characterized by hypofibrinogenemia and accumulation of fibrinogen aggregates within the hepatocellular endoplasmatic reticulum (ER). Some FSD patients present with elevated amino-transferases and fibrosis/cirrhosis similar to alpha-1-antitrypsin deficiency (ATD), also an ER storage disease. Pharmacological stimulation of autophagy has been shown to mediate clearance of protein aggregates and halt progression of liver fibrosis in in vivo models of ATD. Our aim was to evaluate the presence of autophagy and a possible response to autophagy-enhancing therapy in patients with FSD. Hepatic fibrosis was assessed by transient elastography in 2 newly identified FSD families with fibrinogen Aguadilla and Brescia mutations, encompassing 8 affected members. Available liver biopsies were assessed for autophagy. Two patients, who had had elevated alanine amino-transaminase levels (2-5 above upper limit of normal), were treated with the autophagy enhancer carbamazepine (CBZ). Transient elastography did not show evidence of significant fibrosis in any affected family members. Quantitative electron microscopy of one patient showed a 5.15-fold increase of late stage autophagocytic vacuoles compared to control livers. CBZ at low anticonvulsive treatment levels led to rapid normalization of alanine-aminotransferase and decrease of caspase-cleaved and uncleaved cytokeratin-18 fragments (M30 and M65). These effects reversed after discontinuation of treatment. Response to CBZ may be mediated by pharmacologically enhanced autophagy resulting in reduction of aggregate-related toxicity in FSD. These results suggest clinical applicability of pharmacological stimulation of autophagy in FSD, but potentially also in other related disorders.


Asunto(s)
Afibrinogenemia/tratamiento farmacológico , Carbamazepina/uso terapéutico , Fibrinógeno/metabolismo , Afibrinogenemia/metabolismo , Afibrinogenemia/patología , Autofagia/efectos de los fármacos , Niño , Preescolar , Análisis Mutacional de ADN , Diagnóstico por Imagen de Elasticidad , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Femenino , Fibrinógeno/genética , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Linaje , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico , Deficiencia de alfa 1-Antitripsina/metabolismo , Deficiencia de alfa 1-Antitripsina/patología
19.
Am J Pathol ; 178(3): 1117-25, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21356363

RESUMEN

Chronic cholestatic liver injury induced by cholestasis in rodents is associated with hepatic fibrin deposition, and we found evidence of fibrin deposition in livers of patients with cholestasis. Key components of the fibrinolytic pathway modulate cholestatic liver injury by regulating activation of hepatocyte growth factor. However, the exact role of hepatic fibrin deposition in chronic cholestasis is not known. We tested the hypothesis that fibrinogen (Fbg) deficiency worsens liver injury induced by cholestasis. Fbg-deficient mice (Fbgα(-/-) mice) and heterozygous control mice (Fbgα(+/-) mice) were fed either the control diet or a diet containing 0.025% α-naphthylisothiocyanate (ANIT), which selectively injures bile duct epithelial cells in the liver, for 2 weeks. Hepatic fibrin and collagen deposits were evident in livers of heterozygous control mice fed the ANIT diet. Complete Fbg deficiency was associated with elevated serum bile acids, periportal necrosis, and increased serum alanine aminotransferase activity in mice fed the ANIT diet. Fbg deficiency was associated with enhanced hepatic expression of the transcription factor early growth response-1 (Egr-1) and enhanced induction of genes encoding the Egr-1-regulated proinflammatory chemokines monocyte chemotactic protein-1, KC growth-regulated protein, and macrophage inflammatory protein-2. Interestingly, peribiliary collagen deposition was not evident near necrotic areas in Fbg-deficient mice. The results suggest that in this model of chronic cholestasis, fibrin constrains the release of bile constituents from injured intrahepatic bile ducts, thereby limiting the progression of hepatic inflammation and hepatocellular injury.


Asunto(s)
Afibrinogenemia/complicaciones , Afibrinogenemia/metabolismo , Colestasis/complicaciones , Colestasis/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Fibrinógeno/metabolismo , Hígado/patología , 1-Naftilisotiocianato/administración & dosificación , Afibrinogenemia/patología , Anciano , Animales , Conductos Biliares/patología , Colestasis/inducido químicamente , Colestasis/patología , Enfermedad Crónica , Colágeno/metabolismo , Dieta , Modelos Animales de Enfermedad , Conducta Alimentaria , Femenino , Fibrina/deficiencia , Fibrina/metabolismo , Regulación de la Expresión Génica , Humanos , Hiperplasia , Inflamación/complicaciones , Inflamación/genética , Inflamación/patología , Hígado/metabolismo , Cirrosis Hepática/complicaciones , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones , Persona de Mediana Edad , Neutrófilos/metabolismo , Xenobióticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA