Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Haemophilia ; 25(6): 1073-1082, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31469483

RESUMO

INTRODUCTION: Deficiencies of plasminogen and plasminogen activator inhibitor type 1 (PAI-1) are rare disorders of fibrinolysis. Current laboratory assays for analysis of activity of plasminogen and PAI-1 do not provide an accurate correlation with clinical phenotype. METHODS: The Nijmegen Hemostasis Assay (NHA) was used to simultaneously measure thrombin and plasmin generation in 5 patients with plasminogen deficiency (PLGD) and 10 patients with complete PAI-1 deficiency. Parameters analysed included: lag time ratio, thrombin peak time ratio, thrombin peak height, thrombin potential (AUC), fibrin lysis time, plasmin peak height and plasmin potential. Parameters were expressed as a percentage compared to a reference value of 53 healthy normal controls. RESULTS: Patients with PLGD demonstrated a short lag time and thrombin peak time, with normal thrombin peak height but an increased AUC. Plasmin generation was able to be detected in only one (23% plasminogen activity) of the five PLGD patients. All ten PAI-1 deficient patients demonstrated a short lag and thrombin peak time, low thrombin peak height with normal AUC. Plasmin generation revealed an increased plasmin peak and plasmin potential; interestingly, there was a large variation between individual patients despite all patients having the same homozygous defect. CONCLUSION: Patients with either PLGD or PAI-1 deficiency show distinct abnormalities in plasmin and thrombin generation in the NHA. The differences observed in the propagation phase of thrombin generation may be explained by plasmin generation. These results suggest that disorders of fibrinolysis also influence coagulation and a global assay measuring both activities may better correlate with clinical outcome.


Assuntos
Transtornos de Proteínas de Coagulação/metabolismo , Fibrinolisina/biossíntese , Transtornos Hemorrágicos/metabolismo , Inibidor 1 de Ativador de Plasminogênio/deficiência , Trombina/biossíntese , Adulto , Criança , Transtornos de Proteínas de Coagulação/genética , Feminino , Genótipo , Transtornos Hemorrágicos/genética , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo
2.
J Thromb Haemost ; 17(11): 1798-1807, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31271700

RESUMO

BACKGROUND: Congenital disorders of glycosylation are rare inherited diseases affecting many different proteins. The lack of glycosylation notably affects the hemostatic system and leads to deficiencies of both procoagulant and anticoagulant factors. OBJECTIVE: To assess the hemostatic balance in patients with multiple coagulation disorders by using a thrombin generation assay. METHOD: We performed conventional coagulation assays and a thrombin generation assay on samples from patients with congenital disorder of glycosylation. The thrombin generation assay was performed before and after activation of the protein C system by the addition of soluble thrombomodulin. RESULTS: A total of 35 patients were included: 71% and 57% had low antithrombin and factor XI levels, respectively. Protein C and protein S levels were abnormally low in 29% and 26% of the patients, respectively, whereas only 11% displayed low factor IX levels. Under baseline conditions, the thrombin generation assay revealed a significantly higher endogenous thrombin potential and thrombin peak in patients, relative to controls. After spiking with thrombomodulin, we observed impaired involvement of the protein C system. Hence, 54% of patients displayed a hypercoagulant phenotype in vitro. All the patients with a history of stroke-like episodes or thrombosis displayed this hypercoagulant phenotype. CONCLUSION: A thrombin generation assay revealed a hypercoagulant in vitro phenotype under baseline condition; this was accentuated by impaired involvement of the protein C system. This procoagulant phenotype may thus reflect the risk of severe vascular complications. Further research will have to determine whether the thrombin generation assay is predictive of vascular events.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/sangue , Transtornos de Proteínas de Coagulação/sangue , Defeitos Congênitos da Glicosilação/sangue , Trombina/metabolismo , Adolescente , Coagulação Sanguínea/genética , Transtornos Herdados da Coagulação Sanguínea/diagnóstico , Transtornos Herdados da Coagulação Sanguínea/genética , Criança , Pré-Escolar , Transtornos de Proteínas de Coagulação/diagnóstico , Transtornos de Proteínas de Coagulação/genética , Defeitos Congênitos da Glicosilação/diagnóstico , Defeitos Congênitos da Glicosilação/genética , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Paris , Fenótipo , Estudos Retrospectivos , Espanha
3.
Int J Mol Sci ; 20(12)2019 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-31234407

RESUMO

Molecular strategies tailored to promote/correct the expression and/or processing of defective coagulation factors would represent innovative therapeutic approaches beyond standard substitutive therapy. Here, we focus on the molecular mechanisms and determinants underlying innovative approaches acting at DNA, mRNA and protein levels in inherited coagulation factor deficiencies, and in particular on: (i) gene editing approaches, which have permitted intervention at the DNA level through the specific recognition, cleavage, repair/correction or activation of target sequences, even in mutated gene contexts; (ii) the rescue of altered pre-mRNA processing through the engineering of key spliceosome components able to promote correct exon recognition and, in turn, the synthesis and secretion of functional factors, as well as the effects on the splicing of missense changes affecting exonic splicing elements; this section includes antisense oligonucleotide- or siRNA-mediated approaches to down-regulate target genes; (iii) the rescue of protein synthesis/function through the induction of ribosome readthrough targeting nonsense variants or the correction of folding defects caused by amino acid substitutions. Overall, these approaches have shown the ability to rescue the expression and/or function of potentially therapeutic levels of coagulation factors in different disease models, thus supporting further studies in the future aimed at evaluating the clinical translatability of these new strategies.


Assuntos
Transtornos de Proteínas de Coagulação/genética , Transtornos de Proteínas de Coagulação/terapia , Terapia Genética/métodos , Animais , Fatores de Coagulação Sanguínea/genética , Sistemas CRISPR-Cas , DNA/genética , Edição de Genes/métodos , Humanos , RNA Mensageiro/genética
5.
Orphanet J Rare Dis ; 12(1): 66, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28388959

RESUMO

BACKGROUND: Autosomal recessive bleeding disorders (ARBDs) include deficiencies of clotting factors I, II, V, VII, X, XI, XIII, vitamin K dependent clotting factors, combined factor V & VIII, Von Willebrand Disease (vWD) type 3, Glanzmann's thrombasthenia (GT) and Bernard-Soulier syndrome. Patients with primary bleeding disorders from all the major provincial capitals of Pakistan were screened for ARBDs. Prothrombin (PT), activated partial thromboplastin time (APTT), bleeding time (BT) and fibrinogen levels were measured. Cases with isolated prolonged APTT were tested for factors VIII and IX using factor assays This was followed by FXI:C level assessment in cases with normal FVIII and FIX levels. vWD was screened in patients with low FVIII levels. Factors II, V and X were tested in patients with simultaneous prolongation of PT and APTT. Peripheral blood film examination and platelet aggregation studies were performed to assess platelet disorders. Urea clot solubility testing was done to detect Factor XIII levels where platelet function tests were normal. Descriptive analysis was done using SPSS version 16. RESULTS: Of the 429 suspected bleeding disorder patients, 148 (35%) were diagnosed with hemophilia A and 211 (49.1%) patients had ARBDs. 70 patients (16.3%) remained undiagnosed. Out of 211 patients with ARBD; 95 (33.8%) had vWD type 3. Fibrinogen deficiency was found in 34 patients (12%), GT in 27 (9.6%), factor XIII deficiency in 13 (4.6%), factor VII deficiency in 12 (4.3%), factor V deficiency in 9 (3.2%). Eight patients (2.8%) had vitamin K-dependent clotting factor deficiency, Bernard-Soulier syndrome was diagnosed in seven patients (2.5%), factor X deficiency in 2 (0.7%), factor II deficiency in 2 (0.7%), factor XI deficiency and combined factor V and VIII deficiency in 1 (0.4%) patient each. CONCLUSION: vWD type 3 was the most common ARBD found in our sample of patients in Pakistan, followed by fibrinogen deficiency and GT in respective order.


Assuntos
Transtornos de Proteínas de Coagulação/epidemiologia , Transtornos de Proteínas de Coagulação/genética , Genes Recessivos , Adolescente , Criança , Consanguinidade , Estudos Transversais , Feminino , Humanos , Masculino , Paquistão/epidemiologia
6.
J Thromb Thrombolysis ; 44(1): 71-75, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28251495

RESUMO

Recently several variants of clotting factors have shown a peculiar behavior so that they appear as new defects. The factors involved are FII, FV and FIX. Prothrombin deficiency is usually associated with bleeding. Recently a few prothrombin abnormalities involving Arg396 mutations, have been demonstrated to show antithrombin resistance with the consequent appearance of a thrombophilic state and venous thromboses in young age. The same is true for an abnormal FIX (FIX Padua). The thrombotic manifestations in the latter condition are also venous. The abnormal FIX (FIX Padua) is characterized by a great increase in FIX activity whereas FIX antigen is only slightly increased. The condition is due to an Arg338Lys mutation. The increased intrinsic clotting activity of this abnormal FIX is being investigated as a useful therapeutic approach in homophile B patients. Another new clotting disorder is represented by two abnormal FV (FV East Texas and FV Amsterdam). These are characterized by a deletion of part of the B domain of FV resulting in a "short" FV. The condition is characterized by a mild bleeding tendency due to high levels of Tissue Factor pathway inhibitor. The "short" factor V is in fact resistant to the action of Tissue Factor pathway inhibitor which is sharply increased in these patients. These new clotting entities have again demonstrated that the study of patients who show a tendency to venous thrombosis or a mild bleeding condition that cannot be explained on the basis of our current concepts of blood coagulation, may represent "new" coagulation disorders. All persons interested in thrombotic or hemorrhagic disorders should be informed about these new clinical and laboratory conditions.


Assuntos
Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/metabolismo , Transtornos de Proteínas de Coagulação , Transtornos de Proteínas de Coagulação/sangue , Transtornos de Proteínas de Coagulação/classificação , Transtornos de Proteínas de Coagulação/genética , Transtornos de Proteínas de Coagulação/terapia , Humanos
7.
Blood Cells Mol Dis ; 67: 63-68, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28222949

RESUMO

Rare diseases are defined as life-threatening or chronically debilitating diseases with a prevalence of less than one per 2000 according to the European Union or one per 1250 according to the USA. Congenital rare bleeding disorders RBD are reported in most populations, with incidence varying from 1 in 5000 (Hemophilia A), 1:30,000 (Hemophilia B) to much rarer (1:500,000 for FVII deficiency, 1-3 million for Prothrombin or FXIII deficiency). Acquired Hemophilia A is also a rare bleeding disorder with estimated frequency of 1 in million. Most RBDs are inherited as autosomal recessive (AR); however, heterozygous carriers with varying degrees of corresponding factor deficiency may render an unpredictable propensity for bleeding. In patients with bleeding symptoms, laboratory assessment and especially molecular techniques currently enable accurate diagnosis and may provide tools for prenatal and family counseling. Currently hemostasis control is mainly based upon replacement of the missing coagulation factors (unless presence of inhibitors renders it impossible), however future gene therapy and disruptive, non-replacement alternatives may be promising for patients with RBD.


Assuntos
Transtornos de Proteínas de Coagulação/diagnóstico , Hemorragia/diagnóstico , Doenças Raras/diagnóstico , Animais , Coagulação Sanguínea , Fatores de Coagulação Sanguínea/uso terapêutico , Transtornos de Proteínas de Coagulação/sangue , Transtornos de Proteínas de Coagulação/genética , Transtornos de Proteínas de Coagulação/terapia , Terapia Genética/métodos , Hemorragia/sangue , Hemorragia/genética , Hemorragia/terapia , Humanos , Doenças Raras/sangue , Doenças Raras/genética , Doenças Raras/terapia
8.
Ann Dermatol Venereol ; 143(4): 279-83, 2016 Apr.
Artigo em Francês | MEDLINE | ID: mdl-26944767

RESUMO

BACKGROUND: Pseudoxanthoma elasticum (PXE)-like syndrome is characterized by the association of PXE and cutis laxa (CL) features with a deficiency of vitamin K-dependent clotting factors. It was first described in 1971 and was identified as a distinct genetic entity in 2007 with analysis of the GGCX (γ-glutamyl carboxylase) gene, which is involved in congenital deficiency in vitamin K-dependent clotting factors. Here we report a new case of this extremely rare syndrome. PATIENTS AND METHODS: A 23-year-old female patient was seen for the emergence of loose and redundant skin following extensive weight loss. She also presented a deficiency of vitamin K-dependent clotting factors. Physical examination revealed excessive, leathery skin folds in the axillary and neck regions. A skin biopsy revealed polymorphous and fragmented elastic fibers in the reticular dermis. These were mineralized, as was demonstrated by Von Kossa staining. The clinical features of CL associated with the histopathological features of PXE and vitamin K-dependent clotting factor deficiency led us to a diagnosis of PXE-like syndrome. A molecular study of the GGCX gene showed compound heterozygosity. DISCUSSION: The GGCX gene is usually responsible for PXE-like syndrome. GGCX encodes a γ-glutamyl carboxylase necessary for activation of gla-proteins. Gla-proteins are involved both in coagulation factors in the liver and in the prevention of ectopic mineralization of soft tissues. Uncarboxylated forms of gla-proteins in fibroblast would thus enable mineralization and fragmentation of elastic fibers.


Assuntos
Carbono-Carbono Ligases/deficiência , Transtornos de Proteínas de Coagulação/diagnóstico , Cútis Laxa/diagnóstico , Pseudoxantoma Elástico/diagnóstico , Biópsia , Carbono-Carbono Ligases/genética , Transtornos de Proteínas de Coagulação/genética , Transtornos de Proteínas de Coagulação/patologia , Cútis Laxa/genética , Cútis Laxa/patologia , Feminino , Heterozigoto , Humanos , Mutação de Sentido Incorreto , Processamento de Proteína Pós-Traducional , Pseudoxantoma Elástico/genética , Pseudoxantoma Elástico/patologia , Pele/patologia , Redução de Peso , Adulto Jovem
10.
Scand J Clin Lab Invest ; 73(7): 585-90, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24000886

RESUMO

BACKGROUND: Dysfibrinogenemia is a rare group of qualitative fibrinogen disorders caused by structural abnormalities in the fibrinogen molecule. The laboratory diagnosis of dysfibrinogenemia is controversial. Fibrinogen Paris V, clinically termed Dusart Syndrome, is a dysfibrinogenemia caused by a single base substitution in the gene coding for the Aα-chain of the fibrinogen molecule. OBJECTIVES: To diagnose the first Scandinavian family with Fibrinogen Paris V affecting several family members; the proband, a seven-year-old boy with cerebral vein thrombosis. METHODS: The diagnosis was established following the ISTH guideline for laboratory testing supplemented with fibrin structure analysis and fibrinogen gene analysis. RESULTS: Prolonged thrombin time and reduced ratio between the functional and the protein concentration of fibrinogen were observed in four family members who also were characterized by significantly reduced fibrin polymerization (p < 0.001), reduced fibrin fibre diameter (p < 0.001), reduced fibrin mass-length ratio (p < 0.001) and significantly reduced t-PA-induced fibrinolysis of the fibrin clots (p < 0.001) when compared to controls. Fibrinogen gene analysis demonstrated that five of the family members carried the Fibrinogen Paris V mutation. All laboratory tests were normal in the family members carrying the wild type of the fibrinogen gene. Four of the affected patients had suffered from thrombotic episodes. A noticeable feature in the present family was the presence of both venous and arterial thrombosis. CONCLUSIONS: Excellent concordance was observed between the screening and confirmatory tests, fibrin structure analysis and fibrinogen gene analysis. Fibrin structure analysis should be considered in the laboratory algorithm for diagnosis of dysfibrinogenemia.


Assuntos
Transtornos de Proteínas de Coagulação/congênito , Trombose/sangue , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Transtornos de Proteínas de Coagulação/sangue , Transtornos de Proteínas de Coagulação/genética , Análise Mutacional de DNA , Feminino , Fibrina/química , Fibrinogênios Anormais/genética , Fibrinogênios Anormais/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Multimerização Proteica , Países Escandinavos e Nórdicos , Síndrome , Tempo de Trombina , Trombose/genética , Adulto Jovem , gama-Glutamiltransferase/sangue
11.
Iran J Kidney Dis ; 7(5): 340-5, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24072143

RESUMO

Hemolytic-uremic syndrome (HUS) is a rare life-threatening disorder characterized by microangiopathic hemolytic anemia, thrombocytopenia, and impaired renal function. A thrombotic microangiopathy underlies the clinical features of HUS. In the majority of cases, HUS follows an infection with toxin-producing bacteria such as verotoxin-producing Escherichia coli. In some cases, HUS is not preceded by a clinically apparent infection, and therefore, is named atypical HUS. The prognosis of atypical HUS is poor. While mortality approaches 25% during the acute phase, end-stage renal disease develops in nearly half of patients within a year. Evidence is accumulating that complement activation through the alternative pathway is at the heart of the pathophysiology leading to atypical HUS. Genetic abnormalities involving complement regulatory proteins and complement components form the molecular basis for complement activation. Since microvascular thrombosis is a quintessential feature of atypical HUS, complements and the coagulation system must work in tandem to give rise to the pathologic alterations observed in this condition. Here, a brief discussion of clinical and morphologic features of atypical HUS is followed by a concise presentation of the complement and coagulation systems. The interplay between complements and the coagulation system is graphically highlighted. Last but not least, conventional and emerging therapies for atypical HUS are outlined.


Assuntos
Transtornos de Proteínas de Coagulação/fisiopatologia , Proteínas do Sistema Complemento/metabolismo , Síndrome Hemolítico-Urêmica/patologia , Síndrome Hemolítico-Urêmica/fisiopatologia , Síndrome Hemolítico-Urêmica Atípica , Transtornos de Proteínas de Coagulação/genética , Transtornos de Proteínas de Coagulação/metabolismo , Proteínas do Sistema Complemento/genética , Síndrome Hemolítico-Urêmica/complicações , Síndrome Hemolítico-Urêmica/terapia , Humanos , Falência Renal Crônica/etiologia , Prognóstico
12.
Int J Lab Hematol ; 35(3): 290-6, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23590657

RESUMO

The investigation of inherited bleeding disorders with routine tests of hemostasis will yield clear diagnostic information in the majority of subjects with an unequivocal history of bleeding and especially in those where the phenotypic severity is severe and where an obvious family history of bleeding is present. Nevertheless, a significant minority of subjects with obvious bleeding symptoms will remain without a definite diagnosis after extensive hemostatic testing. With these facts in mind, the role of molecular testing for inherited disorders of hemostasis now includes the following: confirmation of a phenotypic diagnosis through targeted genetic analysis, the distinction of bleeding phenocopies by molecular analysis, and provision of genetic testing as the investigation of choice in situations such as prenatal diagnosis and detection of the carrier state for inherited bleeding traits. In addition, molecular testing can sometimes be used to provide supplementary knowledge that can be used to enhance clinical care. Finally, the utility of genome-wide approaches to identify novel genetic associations may provide new information to explain the cause of bleeding in the population of bleeders without established diagnoses.


Assuntos
Transtornos de Proteínas de Coagulação/genética , Testes Genéticos/métodos , Hemostasia/genética , Técnicas de Diagnóstico Molecular/métodos , Fatores de Coagulação Sanguínea/genética , Transtornos de Proteínas de Coagulação/sangue , Transtornos de Proteínas de Coagulação/diagnóstico , Predisposição Genética para Doença/genética , Testes Genéticos/tendências , Estudo de Associação Genômica Ampla/métodos , Humanos , Técnicas de Diagnóstico Molecular/tendências , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
Ann Fr Anesth Reanim ; 32(3): 198-205, 2013 Mar.
Artigo em Francês | MEDLINE | ID: mdl-23422342

RESUMO

Rare inherited bleeding disorders include fibrinogen disorders, and deficiencies of factors II (prothrombin), V, VII, X, XI, XIII, and combined V+VIII, and combined vitamin K-dependent factors, with general population prevalence rates between 1/500,000 and 1/2,000,000. These inherited disorders, transmitted as autosomal recessive traits, are characterized by a heterogeneous clinical presentation (asymptomatic, mild, moderate or severe bleeding tendency); this variability is more important for deficiencies with factor levels ranging from 5 to 50%. Individual bleeding risk assessment before an invasive procedure or during peri-partum period remains difficult, although an essential step to decide whether a substitution with clotting factor is necessary or not. Because there is a poor correlation between factor activity levels and the severity of bleeding symptoms, factor correction before an invasive procedure should not be based on factor level only, but physicians must also take into account the patient phenotype as well as the haemorrhagic risk of the procedure.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/genética , Transtornos de Proteínas de Coagulação/genética , Hemorragia/etiologia , Transtornos Herdados da Coagulação Sanguínea/tratamento farmacológico , Transtornos Herdados da Coagulação Sanguínea/epidemiologia , Fatores de Coagulação Sanguínea/efeitos adversos , Fatores de Coagulação Sanguínea/análise , Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/uso terapêutico , Transtornos de Proteínas de Coagulação/tratamento farmacológico , Transtornos de Proteínas de Coagulação/epidemiologia , Gerenciamento Clínico , Emergências , Feminino , Genes Recessivos , Hemorragia/prevenção & controle , Humanos , Masculino , Hemorragia Pós-Operatória/etiologia , Hemorragia Pós-Operatória/prevenção & controle , Hemorragia Pós-Parto/etiologia , Hemorragia Pós-Parto/prevenção & controle , Gravidez , Prevalência , Medição de Risco , Avaliação de Sintomas , Trombose/induzido quimicamente
14.
Ann Neurol ; 72(4): 550-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23109149

RESUMO

OBJECTIVE: Congenital disorders of glycosylation (CDG) are a group of metabolic diseases due to defects in protein and lipid glycosylation. We searched for the primary defect in 3 children from 2 families with a severe neurological phenotype, including profound developmental delay, intractable epilepsy, progressive microcephaly, severe hypotonia with elevated blood creatine kinase levels, and early fatal outcome. There was clinical evidence of a muscular dystrophy-dystroglycanopathy syndrome, supported by deficient O-mannosylation by muscle immunohistochemistry. METHODS: Biochemical and molecular methods were combined to pinpoint the defect in the glycosylation pathway in the endoplasmic reticulum. RESULTS: Metabolic investigations revealed CDG-I, pointing to a defect in protein N-glycosylation in the endoplasmic reticulum. Analysis of lipid-linked oligosaccharides in fibroblasts showed accumulation of Dol-PP-GlcNAc(2) -Man(5) . DNA analysis revealed mutations in DPM2, 1 of the subunits of the dolichol-phosphate-mannose (DPM) synthase; the patient in the first family is compound heterozygous for 2 mutations (c.68A>G, predicting a missense mutation p.Y23C and c.4-1G>C, a splice mutation), whereas the patients in the second family are homozygous for the same missense mutation (c.68A>G, p.Y23C). INTERPRETATION: We describe a new CDG, due to a deficiency of DPM2. Hence, mutations have now been described in the genes for the 3 subunits of DPM: DPM1, DPM2, and DPM3, whereby DPM2-CDG links the congenital disorders of glycosylation to the congenital muscular dystrophies.


Assuntos
Defeitos Congênitos da Glicosilação/genética , Epilepsia/genética , Manosiltransferases/genética , Distrofias Musculares/genética , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Transtornos de Proteínas de Coagulação/genética , Defeitos Congênitos da Glicosilação/complicações , Análise Mutacional de DNA , Resistência a Medicamentos , Distroglicanas/metabolismo , Eletromiografia , Retículo Endoplasmático , Epilepsia/etiologia , Feminino , Fibroblastos/metabolismo , Glicosilação , Humanos , Lactente , Focalização Isoelétrica , Hepatopatias/complicações , Hepatopatias/genética , Masculino , Manose/metabolismo , Microcefalia/genética , Microcefalia/patologia , Pessoa de Meia-Idade , Dados de Sequência Molecular , Distrofias Musculares/complicações , Mutação/genética , Mutação/fisiologia , Mutação de Sentido Incorreto/genética , Mutação de Sentido Incorreto/fisiologia , Gravidez , Transtornos da Visão/genética , Transtornos da Visão/patologia , Adulto Jovem
15.
Curr Opin Hematol ; 19(5): 380-4, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22872158

RESUMO

PURPOSE OF REVIEW: By definition, rare factor deficiencies have a prevalence of less than 200,000 in the US population, or an incidence of less than one in 2000 in Europe. The very small numbers of patients with rare disorders present challenges in diagnosis, evaluation of bleeding risk and treatment. Use of new assays, full genome sequencing, and global clotting assays will significantly improve diagnosis of patients with rare bleeding disorders. RECENT FINDINGS: In addition to new assays available for monitoring patients, new therapy, both recombinant and plasma derived, is now available. Registries and clinical trials have demonstrated decreased bleeding and improved outcomes when patients are treated with these agents. Expanding international registries have been initiated to correlate genotype and bleeding phenotype in conjunction with global assays. SUMMARY: Ongoing research continues to expand our understanding of the pathophysiology of rare factor deficiencies. This work complements medical practice to incorporate early diagnosis and new treatment options for patients, resulting in safer and less sensitizing regimens and much improved clinical outcomes.


Assuntos
Fatores de Coagulação Sanguínea/uso terapêutico , Transtornos de Proteínas de Coagulação/terapia , Doenças Raras/terapia , Transtornos de Proteínas de Coagulação/diagnóstico , Transtornos de Proteínas de Coagulação/genética , Humanos , Doenças Raras/diagnóstico , Doenças Raras/genética
16.
Ann Med ; 44(5): 405-18, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21585250

RESUMO

Hemostasis is traditionally defined as a physiological response to blood vessel injury and bleeding, which entails a co-ordinated process involving the blood vessel, platelets, and blood clotting proteins (i.e. coagulation factors). Hemostasis can be divided into primary and secondary components. The former rapidly initiates after endothelial damage and is characterized by vascular contraction, platelet adhesion, and formation of a soft aggregate plug. The latter is initiated following the release of tissue factor and involves a complex sequence of events known as the blood coagulation cascade, encompassing serial steps where each coagulation factor activates another in a chain reaction that culminates in the conversion of fibrinogen to fibrin. Patients carrying abnormalities of the coagulation cascade (i.e. deficiencies of coagulation factors) have an increased bleeding tendency, where the clinical severity is mostly dependent upon the type and the plasma level of the factor affected. These disorders also impose a heavy medical and economic burden on individual patients and society in general. The aim of this article is to provide a general overview on the pathophysiology, clinics, diagnostics, and therapy of inherited disorders of coagulation factors.


Assuntos
Transtornos da Coagulação Sanguínea/genética , Coagulação Sanguínea/fisiologia , Transtornos de Proteínas de Coagulação/genética , Hemofilia A/genética , Hemofilia B/genética , Humanos , Doenças de von Willebrand/genética
18.
Thromb Haemost ; 103(6): 1188-92, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20431847

RESUMO

Antithrombin (AT), in the presence of heparin, is able to inhibit the catalytic activity of factor VIIa bound to tissue factor (TF) on cell surfaces. The clinical meaning of FVIIa-AT complexes plasma levels is unknown. It was the objective of this study to evaluate FVIIa-AT complexes in subjects with thrombosis. Factor VIIa-AT complexes plasma levels in 154 patients consecutively referred to our Department with arterial or venous thrombosis and in a group of 154 healthy subjects, were measured. Moreover, FVIIa-AT complexes were determined in: i) n = 53 subjects belonging to 10 families with inherited factor VII deficiency; ii) n = 58 subjects belonging to seven families with AT deficiency; iii) n = 49 patients undergoing oral anticoagulant therapy (OAT). Factor VIIa-AT levels were determined by a specific ELISA kit (R&D, Diagnostica Stago, Gennevilliers, France). Factor VIIa-AT complexes mean plasma levels were lower in patients with either acute arterial (136 +/- 40 pM) or venous (142 +/- 53 pM) thrombosis than subjects with previous thrombosis (arterial 164 +/- 33 pM and venous 172 +/- 61 pM, respectively) and than healthy controls (156 +/- 63 pM). Differences between acute and previous thrombosis, were statistically significant (p < 0.05). Subjects with inherited and acquired (under OAT) factor VII deficiency had statistically significant lower FVIIa-AT complexes plasma levels (80 +/- 23 pM and 55 +/- 22 pM, respectively) than controls (150 +/- 51 pM, p < 0.0001 and 156 +/- 63 pM, p < 0.00001, respectively). Factor VIIa-AT complexes are positively correlated with plasma factor VII/VIIa levels. Further investigations are needed to verify the possible role of higher FVIIa-AT complex plasma levels in predicting hypercoagulable states and thrombosis.


Assuntos
Antitrombinas/metabolismo , Transtornos de Proteínas de Coagulação/metabolismo , Fator VIIa/metabolismo , Complexos Multiproteicos/metabolismo , Trombose Venosa/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antitrombinas/genética , Transtornos de Proteínas de Coagulação/sangue , Transtornos de Proteínas de Coagulação/genética , Fator V/metabolismo , Fator VIIa/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ligação Proteica , Trombose Venosa/sangue , Trombose Venosa/genética
19.
Proc Natl Acad Sci U S A ; 107(8): 3716-21, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-20133705

RESUMO

A severe coagulopathy is a life-threatening complication of acute promyelocytic leukemia (APL) and is ascribable mainly to the excessive levels of tissue factor (TF) in APL cells regulated in response to the promyelocytic leukemia/retinoic acid receptor alpha (PML/RARalpha) fusion protein. The underlying molecular mechanisms for this regulation remain ill-defined. With U937-PR9 cell lines stably expressing luciferase reporter gene under the control of different mutants of the TF promoter, both luciferase and ChIP data allowed the localization of the PML/RARalpha-responsive sequence in a previously undefined region of the TF promoter at position -230 to -242 devoid of known mammalian transcription factor binding sites. Within this sequence a GAGC motif (-235 to -238) was shown to be crucial because deletion or mutation of these nucleotides impaired both PML/RARalpha interaction and promoter transactivation. However, EMSA results showed that PML/RARalpha did not bind to DNA probes encompassing the -230 to -242 sequences, precluding a direct DNA association. Mutational experiments further suggest that the activator protein 1 (AP-1) sites of the TF promoter are dispensable for PML/RARalpha regulation. This study shows that PML/RARalpha transactivates the TF promoter through an indirect interaction with an element composed of a GAGC motif and the flanking nucleotides, independent of AP-1 binding.


Assuntos
Transtornos de Proteínas de Coagulação/genética , Regulação Leucêmica da Expressão Gênica , Leucemia Promielocítica Aguda/complicações , Proteínas de Fusão Oncogênica/farmacologia , Tromboplastina/genética , Ativação Transcricional , Sequência de Bases , Linhagem Celular Tumoral , Transtornos de Proteínas de Coagulação/etiologia , DNA/metabolismo , Humanos , Regiões Promotoras Genéticas , Fator de Transcrição AP-1/metabolismo
20.
Acta Neurochir (Wien) ; 152(2): 257-62, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19826759

RESUMO

BACKGROUND: We investigated whether genes influencing coagulation are associated with the occurrence of aneurysmal subarachnoid hemorrhage (SAH) and with secondary cerebral ischemia and rebleeding in patients with aneurysmal SAH. METHOD: Genotyping for factor V Leiden (G1691A), prothrombin G20210A, methylenetetetrahydrofolate reductase (MTHFR) C677T, factor XIII subunit A Val34Leu, Tyr204Phe and Pro564Leu, and factor XIII subunit B His95Arg was performed in 208 patients with aneurysmal SAH and in 925 controls. Secondary cerebral ischemia occurred in 49 (24%) patients and rebleeding in 28 (14%) during their clinical course of 3 months after the aneurysmal SAH. The risk of aneurysmal SAH was assessed as odds ratio (OR) with 95% confidence interval (95% CI). The risk of secondary cerebral ischemia and rebleeding was assessed as hazard ratio (HR) with 95% CI using Cox regression. FINDINGS: Carriers of the subunit B His95Arg factor XIII polymorphism had an increased risk of aneurysmal SAH with 23% of the patients homozygous or heterozygous for the variant allele compared to 17% of control subjects (OR 1.5, 95% CI 1.0-2.2). For the remaining genetic variants no effect on the risk of aneurysmal SAH could be demonstrated. A clear relation with the risk of secondary cerebral ischemia and of rebleeding could not be established for any of the genetic variants. CONCLUSIONS: We found that aneurysmal SAH patients are more often carriers of the subunit B His95Arg factor XIII polymorphism compared to controls. This suggests that carriers of the subunit B His95Arg factor XIII polymorphism have an increased risk of aneurysmal SAH. Larger studies should confirm our results. As aneurysmal SAH patients who died soon after admission could not be included in the present study, our results only apply to a population of patients who survived the initial hours after the hemorrhage. For the other studied genetic factors involved in coagulation, no association with the occurrence of aneurysmal SAH or with the occurrence of secondary cerebral ischemia or rebleeding after aneurysmal SAH could be demonstrated.


Assuntos
Isquemia Encefálica/genética , Transtornos de Proteínas de Coagulação/genética , Expressão Gênica/genética , Predisposição Genética para Doença/genética , Hemorragia Subaracnóidea/complicações , Hemorragia Subaracnóidea/genética , Adulto , Idoso , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Proteínas Sanguíneas/genética , Isquemia Encefálica/fisiopatologia , Transtornos de Proteínas de Coagulação/metabolismo , Transtornos de Proteínas de Coagulação/fisiopatologia , Análise Mutacional de DNA , Fator XIII/genética , Feminino , Frequência do Gene/genética , Marcadores Genéticos/genética , Testes Genéticos , Variação Genética/genética , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo Genético/genética , Fatores de Risco , Prevenção Secundária , Hemorragia Subaracnóidea/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA