Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Blood ; 143(23): 2373-2385, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38452208

RESUMO

ABSTRACT: Gene therapy using adeno-associated virus (AAV) vectors is a promising approach for the treatment of monogenic disorders. Long-term multiyear transgene expression has been demonstrated in animal models and clinical studies. Nevertheless, uncertainties remain concerning the nature of AAV vector persistence and whether there is a potential for genotoxicity. Here, we describe the mechanisms of AAV vector persistence in the liver of a severe hemophilia A dog model (male = 4, hemizygous; and female = 4, homozygous), more than a decade after portal vein delivery. The predominant vector form was nonintegrated episomal structures with levels correlating with long-term transgene expression. Random integration was seen in all samples (median frequency, 9.3e-4 sites per cell), with small numbers of nonrandom common integration sites associated with open chromatin. No full-length integrated vectors were found, supporting predominant episomal vector-mediated long-term transgene expression. Despite integration, this was not associated with oncogene upregulation or histopathological evidence of tumorigenesis. These findings support the long-term safety of this therapeutic modality.


Assuntos
Dependovirus , Fator VIII , Terapia Genética , Vetores Genéticos , Hemofilia A , Fígado , Animais , Cães , Dependovirus/genética , Hemofilia A/genética , Hemofilia A/terapia , Vetores Genéticos/genética , Fígado/metabolismo , Fígado/patologia , Masculino , Terapia Genética/métodos , Feminino , Fator VIII/genética , Técnicas de Transferência de Genes , Integração Viral , Transgenes , Modelos Animais de Doenças
2.
Blood ; 134(11): 880-891, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31350267

RESUMO

Factor VIII (FVIII) pharmacokinetic (PK) properties show high interpatient variability in hemophilia A patients. Although previous studies have determined that age, body mass index, von Willebrand factor antigen (VWF:Ag) levels, and ABO blood group status can influence FVIII PK, they do not account for all observed variability. In this study, we aim to describe the genetic determinants that modify the FVIII PK profile in a population of 43 pediatric hemophilia A patients. We observed that VWF:Ag and VWF propeptide (VWFpp)/VWF:Ag, but not VWFpp, were associated with FVIII half-life. VWFpp/VWF:Ag negatively correlated with FVIII half-life in patients with non-O blood type, but no correlation was observed for type O patients, suggesting that von Willebrand factor (VWF) half-life, as modified by the ABO blood group, is a strong regulator of FVIII PK. The FVIII-binding activity of VWF positively correlated with FVIII half-life, and the rare or low-frequency nonsynonymous VWF variants p.(Arg826Lys) and p.(Arg852Glu) were identified in patients with reduced VWF:FVIIIB but not VWF:Ag. Common variants at the VWF, CLEC4M, and STAB2 loci, which have been previously associated with plasma levels of VWF and FVIII, were associated with the FVIII PK profile. Together, these studies characterize the mechanistic basis by which VWF clearance and ABO glycosylation modify FVIII PK in a pediatric population. Moreover, this study is the first to identify non-VWF and non-ABO variants that modify FVIII PK in pediatric hemophilia A patients.


Assuntos
Coagulação Sanguínea/genética , Fator VIII/farmacocinética , Hemofilia A/genética , Hemofilia A/metabolismo , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo , Adolescente , Testes de Coagulação Sanguínea , Criança , Fator VIII/uso terapêutico , Feminino , Variação Genética , Genótipo , Meia-Vida , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Humanos , Masculino , Taxa de Depuração Metabólica/genética , Ligação Proteica , Proteólise
3.
Arterioscler Thromb Vasc Biol ; 40(12): 2860-2874, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32967458

RESUMO

OBJECTIVE: Obesity is characterized by chronic low-grade inflammation and consequentially a hypercoagulable state, associating with an increased incidence of venous thromboembolism. Increased VWF (von Willebrand factor) plasma concentration and procoagulant function are independent risk factors for venous thromboembolism and are elevated in obese patients. Here, we explore the pathobiological role of VWF in obesity-associated venous thrombosis using murine models. Approach and Results: We first showed that diet-induced obese mice have increased VWF plasma levels and FVIII (factor VIII) activity compared with littermate controls. Elevated VWF levels appeared to be because of both increased synthesis and impaired clearance. Diet-induced obesity-associated venous thrombosis was assessed using the inferior vena cava-stenosis model of deep vein thrombosis. Diet-induced obese mice developed larger venous thrombi that were rich in VWF, erythrocytes, and leukocytes. Administering a polyclonal anti-VWF antibody or an anti-VWF A1 domain nanobody was protective against obesity-mediated thrombogenicity. Delayed administration (3 hours post-inferior vena cava stenosis) similarly reduced thrombus weight in diet-induced obese mice. CONCLUSIONS: This study demonstrates the critical role of VWF in the complex, thrombo-inflammatory state of obesity. It adds to the growing rationale for targeting VWF-specific interactions in thrombotic disease.


Assuntos
Dieta Hiperlipídica , Obesidade/complicações , Veia Cava Inferior/metabolismo , Trombose Venosa/etiologia , Fator de von Willebrand/metabolismo , Proteína ADAMTS13/genética , Proteína ADAMTS13/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Fibrinolíticos/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/metabolismo , Transdução de Sinais , Anticorpos de Domínio Único/farmacologia , Veia Cava Inferior/efeitos dos fármacos , Veia Cava Inferior/patologia , Trombose Venosa/metabolismo , Trombose Venosa/patologia , Trombose Venosa/prevenção & controle , Fator de von Willebrand/antagonistas & inibidores , Fator de von Willebrand/genética
4.
Blood ; 128(6): 753-62, 2016 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-27354721

RESUMO

In recent years, the traditional view of the hemostatic system as being regulated by a coagulation factor cascade coupled with platelet activation has been increasingly challenged by new evidence that activation of the immune system strongly influences blood coagulation and pathological thrombus formation. Leukocytes can be induced to express tissue factor and release proinflammatory and procoagulant molecules such as granular enzymes, cytokines, and damage-associated molecular patterns. These mediators can influence all aspects of thrombus formation, including platelet activation and adhesion, and activation of the intrinsic and extrinsic coagulation pathways. Leukocyte-released procoagulant mediators increase systemic thrombogenicity, and leukocytes are actively recruited to the site of thrombus formation through interactions with platelets and endothelial cell adhesion molecules. Additionally, phagocytic leukocytes are involved in fibrinolysis and thrombus resolution, and can regulate clearance of platelets and coagulation factors. Dysregulated activation of leukocyte innate immune functions thus plays a role in pathological thrombus formation. Modulation of the interactions between leukocytes or leukocyte-derived procoagulant materials and the traditional hemostatic system is an attractive target for the development of novel antithrombotic strategies.


Assuntos
Hemostasia , Leucócitos/patologia , Trombose/sangue , Trombose/patologia , Animais , Fatores de Coagulação Sanguínea/imunologia , Plaquetas/imunologia , Plaquetas/patologia , Células Endoteliais/imunologia , Células Endoteliais/patologia , Humanos , Leucócitos/imunologia , Fagocitose , Tromboplastina/imunologia , Trombose/imunologia
5.
Haematologica ; 103(11): 1925-1936, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30002126

RESUMO

Immune responses to factor VIII remain the greatest complication in the treatment of severe hemophilia A. Recent epidemiological evidence has highlighted that recombinant factor VIII produced in baby hamster kidney cells is more immunogenic than factor VIII produced in Chinese hamster ovary cells. Glycosylation differences have been hypothesized to influence the immunogenicity of these synthetic concentrates. In two hemophilia A mouse models, baby hamster kidney cell-derived factor VIII elicited a stronger immune response compared to Chinese hamster ovary cell-derived factor VIII. Furthermore, factor VIII produced in baby hamster kidney cells exhibited accelerated clearance from circulation independent of von Willebrand factor. Lectin and mass spectrometry analysis of total N-linked glycans revealed differences in high-mannose glycans, sialylation, and the occupancy of glycan sites. Factor VIII desialylation did not influence binding to murine splenocytes or dendritic cells, nor surface co-stimulatory molecule expression. We did, however, observe increased levels of immunoglobulin M specific to baby hamster kidney-derived factor VIII in naïve hemophilia A mice. De-N-glycosylation enhanced immunoglobulin M binding, suggesting that N-glycan occupancy masks epitopes. Elevated levels of immunoglobulin M and immunoglobulin G specific to baby hamster kidney-derived factor VIII were also observed in healthy individuals, and de-N-glycosylation increased immunoglobulin G binding. Collectively, our data suggest that factor VIII produced in baby hamster kidney cells is more immunogenic than that produced in Chinese hamster ovary cells, and that incomplete occupancy of N-linked glycosylation sites leads to the formation of immunoglobulin M- and immunoglobulin G-factor VIII immune complexes that contribute to the enhanced clearance and immunogenicity in these mouse models of hemophilia A.


Assuntos
Fator VIII , Hemofilia A , Animais , Células CHO , Cricetulus , Modelos Animais de Doenças , Fator VIII/imunologia , Fator VIII/farmacologia , Feminino , Glicosilação , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Hemofilia A/imunologia , Hemofilia A/patologia , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia
6.
Circ Res ; 118(9): 1443-52, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-27126652

RESUMO

Molecular genetic details of the human coagulation system were among the first successes of the genetic revolution in the 1980s. This information led to new molecular diagnostic strategies for inherited disorders of hemostasis and the development of recombinant clotting factors for the treatment of the common inherited bleeding disorders. A longer term goal of this knowledge has been the establishment of gene transfer to provide continuing access to missing or defective hemostatic proteins. Because of the relative infrequency of inherited coagulation factor disorders and the availability of safe and effective alternative means of management, the application of gene therapy for these conditions has been slow to realize clinical application. Nevertheless, the tools for effective and safe gene transfer are now much improved, and we have started to see examples of clinical gene therapy successes. Leading the way has been the use of adeno-associated virus-based strategies for factor IX gene transfer in hemophilia B. Several small phase 1/2 clinical studies using this approach have shown prolonged expression of therapeutically beneficial levels of factor IX. Nevertheless, before the application of gene therapy for coagulation disorders becomes widespread, several obstacles need to be overcome. Immunologic responses to the vector and transgenic protein need to be mitigated, and production strategies for clinical grade vectors require enhancements. There is little doubt that with the development of more efficient and facile strategies for genome editing and the application of other nucleic acid-based approaches to influence the coagulation system, the future of genetic therapies for hemostasis is bright.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/terapia , Fatores de Coagulação Sanguínea/genética , Terapia Genética/métodos , Animais , Fatores de Coagulação Sanguínea/metabolismo , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos
7.
Curr Opin Hematol ; 23(5): 471-8, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27428891

RESUMO

PURPOSE OF REVIEW: In the last nine decades, large advances have been made toward the characterization of the pathogenic basis and clinical management of von Willebrand disease (VWD), the most prevalent inherited bleeding disorder. Pathological variations at the von Willebrand factor (VWF) locus present as a range of both quantitative and qualitative abnormalities that make up the complex clinical spectrum of VWD. This review describes the current understanding of the pathobiological basis of VWD. RECENT FINDINGS: The molecular basis of type 2 (qualitative abnormalities) and type 3 VWD (total quantitative deficiency) have been well characterized in recent decades. However, knowledge of type 1 VWD (partial quantitative deficiency) remains incomplete because of the allelic and locus heterogeneity of this trait, and is complicated by genetic variability at the VWF gene, interactions between the VWF gene and the environment, and the involvement of external modifying loci. Recent genome wide association studies and linkage analyses have sought to identify additional genes that modify the type 1 VWD phenotype. SUMMARY: Understanding the pathogenic basis of VWD will facilitate the development of novel treatment regimens for this disorder, and improve the ability to provide complementary molecular diagnostics for type 1 VWD.


Assuntos
Doenças de von Willebrand/etiologia , Animais , Proteínas de Transporte , Células Endoteliais/metabolismo , Fator VIII/metabolismo , Testes Genéticos , Genótipo , Humanos , Padrões de Herança , Mutação , Fenótipo , Ligação Proteica , Doenças de von Willebrand/diagnóstico , Doenças de von Willebrand/terapia , Fator de von Willebrand/química , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
8.
Blood ; 124(3): 313-5, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-25035147

RESUMO

In this issue of Blood, Yee et al1 have demonstrated that expression or infusion of a truncated von Willebrand factor (VWF) fragment containing the factor VIII (FVIII)-binding D'D3 region of VWF is sufficient to stabilize endogenous FVIII levels in VWF-deficient mice. In the absence of the carrier function of VWF, FVIII is susceptible to rapid proteolysis and clearance resulting in markedly reduced plasma levels of FVIII that contribute to a bleeding diathesis.


Assuntos
Fator VIII/metabolismo , Fragmentos de Peptídeos/metabolismo , Fator de von Willebrand/metabolismo , Animais , Humanos
9.
Blood ; 121(26): 5228-37, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23529928

RESUMO

Genetic variation in or near the C-type lectin domain family 4 member M (CLEC4M) has been associated with plasma levels of von Willebrand factor (VWF) in healthy individuals. CLEC4M is a lectin receptor with a polymorphic extracellular neck region possessing a variable number of tandem repeats (VNTR). A total of 491 participants (318 patients with type 1 von Willebrand disease [VWD] and 173 unaffected family members) were genotyped for the CLEC4M VNTR polymorphism. Family-based association analysis on kindreds with type 1 VWD demonstrated an excess transmission of VNTR 6 to unaffected individuals (P = .0096) and an association of this allele with increased VWF:RCo (P = .029). CLEC4M-Fc bound to VWF. Immunofluorescence and enzyme-linked immunosorbent assay demonstrated that HEK 293 cells transfected with CLEC4M bound and internalized VWF. Cells expressing 4 or 9 copies of the CLEC4M neck region VNTR showed reduced interaction with VWF relative to CLEC4M with 7 VNTR (CLEC4M 4%-60% reduction, P < .001; CLEC4M 9%-45% reduction, P = .006). Mice expressing CLEC4M after hydrodynamic liver transfer have a 46% decrease in plasma levels of VWF (P = .0094). CLEC4M binds to and internalizes VWF, and polymorphisms in the CLEC4M gene contribute to variable plasma levels of VWF.


Assuntos
Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Repetições Minissatélites/genética , Polimorfismo Genético/genética , Doenças de von Willebrand/sangue , Fator de von Willebrand/metabolismo , Adolescente , Adulto , Animais , Estudos de Casos e Controles , Criança , Pré-Escolar , DNA/genética , Ensaio de Imunoadsorção Enzimática , Família , Feminino , Citometria de Fluxo , Estudo de Associação Genômica Ampla , Genótipo , Células HEK293 , Humanos , Técnicas Imunoenzimáticas , Lactente , Desequilíbrio de Ligação , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Adulto Jovem , Doenças de von Willebrand/genética , Doenças de von Willebrand/patologia
10.
Arterioscler Thromb Vasc Biol ; 34(9): 1977-84, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25012129

RESUMO

OBJECTIVE: Activation of neutrophils by microbial or inflammatory stimuli results in the release of neutrophil extracellular traps (NETs) that are composed of DNA, histones, and antimicrobial proteins. In purified systems, cell-free DNA (CFDNA) activates the intrinsic pathway of coagulation, whereas histones promote thrombin generation through platelet-dependent mechanisms. However, the overall procoagulant effects of CFDNA/histone complexes as part of intact NETs are unknown. In this study, we examined the procoagulant potential of intact NETs released from activated neutrophils. We also determined the relative contribution of CFDNA and histones to thrombin generation in plasmas from patients with sepsis. APPROACH AND RESULTS: NETs released from phorbyl myristate-activated neutrophils enhance thrombin generation in platelet-poor plasma. This effect was DNA dependent (confirmed by DNase treatment) and occurred via the intrinsic pathway of coagulation (confirmed with coagulation factor XII- and coagulation factor XI-depleted plasma). In platelet-rich plasma treated with corn trypsin inhibitor, addition of phorbyl myristate-activated neutrophils increased thrombin generation and shortened the lag time in a toll-like receptor-2- and toll-like receptor-4-dependent mechanism. Addition of DNase further augmented thrombin generation, suggesting that dismantling of the NET scaffold increases histone-mediated, platelet-dependent thrombin generation. In platelet-poor plasma samples from patients with sepsis, we found a positive correlation between endogenous CFDNA and thrombin generation, and addition of DNase attenuated thrombin generation. CONCLUSIONS: These studies examine the procoagulant activities of CFDNA and histones in the context of NETs. Our studies also implicate a role for the intrinsic pathway of coagulation in sepsis pathogenesis.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas/fisiologia , Matriz Extracelular/fisiologia , Ativação de Neutrófilo/fisiologia , Sepse/sangue , Trombina/biossíntese , Sistema Livre de Células , DNA/sangue , DNA/farmacologia , Histonas/sangue , Histonas/farmacologia , Humanos , Ativação de Neutrófilo/efeitos dos fármacos , Plasma Rico em Plaquetas , Acetato de Tetradecanoilforbol/farmacologia , Trombina/genética , Receptor 2 Toll-Like/fisiologia , Receptor 4 Toll-Like/fisiologia
11.
Pharmgenomics Pers Med ; 16: 239-252, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36998673

RESUMO

The inherited bleeding disorder hemophilia A involves the quantitative deficiency of the coagulation cofactor factor VIII (FVIII). Prophylactic treatment of severe hemophilia A patients with FVIII concentrates aims to reduce the frequency of spontaneous joint bleeding and requires personalized tailoring of dosing regimens to account for the substantial inter-individual variability of FVIII pharmacokinetics. The strong reproducibility of FVIII pharmacokinetic (PK) metrics between repeat analyses in the same individual suggests this trait is genetically regulated. While the influence of plasma von Willebrand factor antigen (VWF:Ag) levels, ABO blood group, and patient age on FVIII PK is well established, estimates suggest these factors account for less than 35% of the overall variability in FVIII PK. More recent studies have identified genetic determinants that modify FVIII clearance or half-life including VWF gene variants that impair VWF-FVIII binding resulting in the accelerated clearance of VWF-free FVIII. Additionally, variants in receptors that regulate the clearance of FVIII or the VWF-FVIII complex have been associated with FVIII PK. The characterization of genetic modifiers of FVIII PK will provide mechanistic insight into a subject of clinical significance and support the development of personalized treatment plans for patients with hemophilia A.

12.
J Thromb Haemost ; 21(8): 2007-2019, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37085036

RESUMO

Quantitative abnormalities in factor VIII (FVIII) and its binding partner, von Willebrand factor (VWF), are associated with an increased risk of bleeding or thrombosis, and pathways that regulate the clearance of VWF-FVIII can strongly influence their plasma levels. In 2010, the Cohorts for Heart and Aging Research in Genome Epidemiology (CHARGE) on genome-wide association study meta-analysis identified variants in the genes for the sinusoidal endothelial receptors C-type lectin domain family 4 member M (CLEC4M), stabilin-2, and scavenger receptor class A member 5 (SCARA5) as being associated with plasma levels of VWF and/or FVIII in normal individuals. The ability of these receptors to bind, internalize, and clear the VWF-FVIII complex from the circulation has now been reported in a series of studies using in vitro and in vivo models. The receptor stabilin-2 has also been shown to modulate the immune response to infused VWF-FVIII concentrates in a murine model. In addition, the influence of genetic variants in CLEC4M, STAB2, and SCARA5 on type 1 von Willebrand disease/low VWF phenotype, FVIII pharmacokinetics, and the risk of venous thromboembolism has been described in a number of patient-based studies. Understanding the role of these receptors in the regulation of VWF-FVIII clearance has led to significant insights into the genomic architecture that modulates plasma VWF and FVIII levels, improving the understanding of pathways that regulate VWF-FVIII clearance and the mechanistic basis of quantitative VWF-FVIII pathologies.


Assuntos
Hemostáticos , Trombose , Doenças de von Willebrand , Animais , Camundongos , Fator de von Willebrand/metabolismo , Estudo de Associação Genômica Ampla , Fator VIII/genética , Hemostasia/genética , Trombose/genética , Trombose/metabolismo , Células Endoteliais/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Doenças de von Willebrand/metabolismo , Receptores Depuradores Classe A/genética
13.
J Thromb Haemost ; 21(3): 586-598, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36696220

RESUMO

BACKGROUND: Although the concept of immunothrombosis has established a link between inflammation and thrombosis, the role of inflammation in the pathogenesis of deep vein thrombosis remains to be fully elucidated. Further, although various constituents of venous thrombi have been identified, their localizations and cellular and molecular interactions are yet to be combined in a single, multiplexed analysis. OBJECTIVES: The objective of this study was to investigate the role of the von Willebrand factor (VWF) in inflammation-associated venous thrombosis. We also performed a proof-of-concept study of imaging mass cytometry to quantitatively and simultaneously analyze the localizations and interactions of 10 venous thrombus constituents. METHODS: We combined the murine inferior vena cava stenosis model of deep vein thrombosis with the lipopolysaccharide model of endotoxemia. We also performed a proof-of-concept study of imaging mass cytometry to assess the feasibility of this approach in analyzing the structural composition of thrombi. RESULTS: We found that lipopolysaccharide-treated mice had significantly higher incidences of venous thrombosis, an effect that was mitigated when VWF was inhibited using inhibitory αVWF antibodies. Our detailed structural analysis also showed that most thrombus components are localized in the white thrombus regardless of endotoxemia. Moreover, although endotoxemia modulated the relative representation and interactions of VWF with other thrombus constituents, the scaffolding network, comprised VWF, fibrin, and neutrophil extracellular traps, remained largely unaffected. CONCLUSIONS: We observe a key role for VWF in the pathogenesis of inflammation-associated venous thrombosis while providing a more comprehensive insight into the molecular interactions that constitute the architecture of venous thrombi.


Assuntos
Endotoxemia , Trombose , Trombose Venosa , Camundongos , Animais , Fator de von Willebrand , Lipopolissacarídeos , Trombose Venosa/etiologia , Trombose/complicações
14.
Crit Care ; 16(4): R151, 2012 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-22889177

RESUMO

INTRODUCTION: Although sepsis is the leading cause of death in noncoronary critically ill patients, identification of patients at high risk of death remains a challenge. In this study, we examined the incremental usefulness of adding multiple biomarkers to clinical scoring systems for predicting intensive care unit (ICU) mortality in patients with severe sepsis. METHODS: This retrospective observational study used stored plasma samples obtained from 80 severe sepsis patients recruited at three tertiary hospital ICUs in Hamilton, Ontario, Canada. Clinical data and plasma samples were obtained at study inclusion for all 80 patients, and then daily for 1 week, and weekly thereafter for a subset of 50 patients. Plasma levels of cell-free DNA (cfDNA), interleukin 6 (IL-6), thrombin, and protein C were measured and compared with clinical characteristics, including the primary outcome of ICU mortality and morbidity measured with the Multiple Organ Dysfunction (MODS) score and Acute Physiology and Chronic Health Evaluation (APACHE) II scores. RESULTS: The level of cfDNA in plasma at study inclusion had better prognostic utility than did MODS or APACHE II scores, or the biomarkers measured. The area under the receiver operating characteristic (ROC) curves for cfDNA to predict ICU mortality is 0.97 (95% CI, 0.93 to 1.00) and to predict hospital mortality is 0.84 (95% CI, 0.75 to 0.94). We found that a cfDNA cutoff value of 2.35 ng/µl had a sensitivity of 87.9% and specificity of 93.5% for predicting ICU mortality. Sequential measurements of cfDNA suggested that ICU mortality may be predicted within 24 hours of study inclusion, and that the predictive power of cfDNA may be enhanced by combining it with protein C levels or MODS scores. DNA-sequence analyses and studies with Toll-like receptor 9 (TLR9) reporter cells suggests that the cfDNA from sepsis patients is host derived. CONCLUSIONS: These studies suggest that cfDNA provides high prognostic accuracy in patients with severe sepsis. The serial data suggest that the combination of cfDNA with protein C and MODS scores may yield even stronger predictive power. Incorporation of cfDNA in sepsis risk-stratification systems may be valuable for clinical decision making or for inclusion into sepsis trials.


Assuntos
DNA/sangue , Sepse/sangue , Sepse/mortalidade , APACHE , Fatores Etários , Idoso , Biomarcadores/sangue , Feminino , Mortalidade Hospitalar , Humanos , Unidades de Terapia Intensiva , Interleucina-6/sangue , Masculino , Pessoa de Meia-Idade , Insuficiência de Múltiplos Órgãos/mortalidade , Valor Preditivo dos Testes , Prognóstico , Proteína C/metabolismo , Curva ROC , Estudos Retrospectivos , Trombina/metabolismo , Fatores de Tempo , Receptor Toll-Like 9/sangue
15.
Blood Adv ; 6(9): 2908-2919, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35020809

RESUMO

von Willebrand factor (VWF) is an extremely cysteine-rich multimeric protein that is essential for maintaining normal hemostasis. The cysteine residues of VWF monomers form intra- and intermolecular disulfide bonds that regulate its structural conformation, multimer distribution, and ultimately its hemostatic activity. In this study, we investigated and characterized the molecular and pathogenic mechanisms through which a novel cysteine variant p.(Cys1084Tyr) causes an unusual, mixed phenotype form of von Willebrand disease (VWD). Phenotypic data including bleeding scores, laboratory values, VWF multimer distribution, and desmopressin response kinetics were investigated in 5 members (2 parents and 3 daughters) of a consanguineous family. VWF synthesis and secretion were also assessed in a heterologous expression system and in a transient transgenic mouse model. Heterozygosity for p.(Cys1084Tyr) was associated with variable expressivity of qualitative VWF defects. Heterozygous individuals had reduced VWF:GPIbM (<0.40 IU/mL) and VWF:CB (<0.35 IU/mL), as well as relative reductions in high-molecular-weight multimers, consistent with type 2A VWD. In addition to these qualitative defects, homozygous individuals also displayed reduced factor VIII (FVIII):C/VWF:Ag, leading to very low FVIII levels (0.03-0.1 IU/mL) and reduced VWF:Ag (<0.40 IU/mL) and VWF:GPIbM (<0.30 IU/mL). Accelerated VWF clearance and impaired VWF secretion contributed to the fully expressed homozygous phenotype with impaired secretion arising because of disordered disulfide connectivity.


Assuntos
Doença de von Willebrand Tipo 2 , Doenças de von Willebrand , Animais , Cisteína/genética , Dissulfetos , Camundongos , Doença de von Willebrand Tipo 2/genética , Doenças de von Willebrand/genética , Fator de von Willebrand/metabolismo
16.
J Thromb Haemost ; 19(3): 654-663, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33219619

RESUMO

BACKGROUND: Factor VIII (FVIII) pharmacokinetics (PK) in adult hemophilia A populations are highly variable and have been previously determined to be influenced by von Willebrand factor:antigen (VWF:Ag), ABO blood group, and age. However, additional genetic determinants of FVIII PK are largely unknown. OBJECTIVES: The contribution of VWF clearance, VWF-FVIII-binding activity, and genetic variants in VWF clearance receptors to FVIII PK in adult patients were assessed. METHODS: FVIII PK assessment was performed in 44 adult subjects (age 18-61 years) with moderate or severe hemophilia A. VWF:Ag, VWF propeptide (VWFpp), VWFpp/VWF:Ag, and VWF:FVIII binding activity were measured. The VWF modifying loci CLEC4M, SCARA5, STAB2, and ABO, and the D'D3 FVIII-binding region of the VWF gene were genotyped. RESULTS: VWF:Ag, VWFpp, and VWF:FVIIIB positively correlated with FVIII half-life and negatively correlated with FVIII clearance. VWFpp/VWF:Ag negatively correlated with FVIII half-life and positively correlated with FVIII clearance. The correlation between VWFpp/VWF:Ag and FVIII half-life was stronger for type non-O patients than for type O patients, suggesting that slower VWF clearance increases FVIII half-life. Patients heterozygous for the CLEC4M rs868875 variant had increased FVIII clearance when compared with individuals homozygous for the reference allele. The CLEC4M variable number of tandem repeat (VNTR) alleles were also associated with the rate of FVIII clearance. When compared with the quartile of patients with the fastest FVIII clearance, the quartile of patients with the slowest FVIII clearance had a decreased frequency of the CLEC4M 5-VNTR. CONCLUSIONS: VWF-FVIII binding activity and genetic determinants of VWF clearance are important contributors to FVIII pharmacokinetics in adult patients.


Assuntos
Hemofilia A , Doenças de von Willebrand , Adolescente , Adulto , Fator VIII/genética , Meia-Vida , Hemofilia A/diagnóstico , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Humanos , Pessoa de Meia-Idade , Receptores Depuradores Classe A , Adulto Jovem , Fator de von Willebrand/genética
17.
J Thromb Haemost ; 19(10): 2440-2453, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34152080

RESUMO

BACKGROUND: Stabilin-2 is an endocytic scavenger receptor that mediates the clearance of glycosaminoglycans, phosphatidylserine-expressing cells, and the von Willebrand factor-factor VIII (FVIII) complex. In a genome-wide screening study, pathogenic loss-of-function variants in the human STAB2 gene associated with an increased incidence of unprovoked venous thromboembolism (VTE). However, the specific mechanism(s) by which stabilin-2 deficiency influences the pathogenesis of VTE is unknown. OBJECTIVES: The aim of this study was to assess the influence of stabilin-2 on deep vein thrombosis (DVT) and to characterize the underlying prothrombotic phenotype of stabilin-2 deficiency in a mouse model. METHODS: DVT was induced using the inferior vena cava (IVC) stenosis model in two independent cohorts (littermates and non-littermates) of wild-type (Stab2+/+ ) and stabilin-2 (Stab2-/- )-deficient mice. Thrombus structure and contents were quantified by immunohistochemistry. Plasma procoagulant activity was assessed and complete blood counts were performed. RESULTS: Incidence of thrombus formation was not altered between Stab2+/+ and Stab2-/- mice. When thrombi were formed, Stab2-/- mice developed significantly larger thrombi than Stab2+/+ controls. Thrombi from Stab2-/- mice contained significantly more leukocytes and citrullinated histone H3 than Stab2+/+ thrombi. Stab2-/- mice had increased FVIII activity. Circulating levels of monocytes and granulocytes were significantly elevated in Stab2-/- mice, and Stab2-/- mice had elevated plasma cell-free DNA 24 hours post-IVC stenosis compared to their Stab2+/+ counterparts. CONCLUSIONS: These data suggest that stabilin-2 deficiency associates with a prothrombotic phenotype involving elevated levels of neutrophil extracellular trap-releasing leukocytes coupled with endogenous procoagulant activity, resulting in larger and qualitatively distinct venous thrombi.


Assuntos
Armadilhas Extracelulares , Trombose , Animais , Moléculas de Adesão Celular Neuronais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Veias , Fator de von Willebrand
18.
Pathophysiol Haemost Thromb ; 37(2-4): 88-97, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21430357

RESUMO

Although thromboembolism is a problematic complication of chemotherapy, the pathogenic mechanisms by which chemotherapeutic agents exert prothrombotic effects in vivo are unclear.The objective of this study was to examine the effects of adjuvant chemotherapy on thrombin generation, the protein C anticoagulant pathway, and microparticle tissue factor (MP TF) activity in 26 breast cancer patients (stages I to III). The patients received cyclophosphamide, 5-fluorouracil, and methotrexate, epirubicin, or doxorubicin. Plasma samples were collected on day 1 (baseline), day 2, and day 8 for the first 2 cycles of chemotherapy. Levels of thrombin-antithrombin (TAT) complexes, MP TF activity, and components of the protein C anticoagulant pathway, including protein C, activated protein C (APC), soluble thrombomodulin (sTM), and soluble endothelial protein C receptor (sEPCR), were measured. Compared to prechemotherapy baseline levels, plasma TAT, protein C, and APC were significantly different following the administration of chemotherapy (p < 0.01 for each). Plasma TAT was higher in cycle 1, day 2, and cycle 2, day 8, compared to baseline. Plasma protein C levels were lower in cycle 2, day 8, whereas plasma APC levels were lower in cycle 2, day 1, and cycle 2, day 8. No significant changes were found in plasma sEPCR, sTM, or MP TF activity. This study suggests that adjuvant chemotherapy in women with breast cancer increases thrombin generation and impairs the endothelium-based protein C anticoagulant pathway.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias da Mama/sangue , Neoplasias da Mama/tratamento farmacológico , Proteína C/metabolismo , Trombina/metabolismo , Adulto , Antígenos CD/sangue , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Antitrombina III , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Receptor de Proteína C Endotelial , Epirubicina/administração & dosagem , Epirubicina/efeitos adversos , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/efeitos adversos , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Peptídeo Hidrolases/sangue , Receptores de Superfície Celular/sangue , Trombomodulina/sangue , Tromboplastina/metabolismo , Trombose/sangue , Trombose/induzido quimicamente , Fatores de Tempo
19.
J Thromb Haemost ; 17(4): 681-694, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30740857

RESUMO

Essentials CLEC4M is an endocytic receptor for factor FVIII. CLEC4M interacts with FVIII in a VWF-dependent and independent manner. CLEC4M binds to mannose-containing glycans on FVIII. CLEC4M internalization of FVIII involves clathrin coated pits. SUMMARY: Background von Willebrand factor (VWF) and factor VIII (FVIII) circulate in the plasma as a non-covalent complex, and the majority of FVIII is likely to be cleared by VWF-dependent pathways. Clearance of VWF-free FVIII is rapid and underlies the pathological basis of some quantitative FVIII deficiencies. The receptor pathways that regulate the clearance of VWF-bound and VWF-free FVIII are incompletely uncharacterized. The human liver-expressed endothelial lectin CLEC4M has been previously characterized as a clearance receptor for VWF, although its influence on FVIII is unknown. Objective The interaction between FVIII and CLEC4M was characterized in the presence or absence of VWF. Methods FVIII interactions with CLEC4M were evaluated by in vitro cell-based and solid phase binding assays. Interactions between FVIII and CLEC4M or liver sinusoidal endothelial cells were evaluated in vivo by immunohistochemistry. Results CLEC4M-expressing HEK 293 cells bound and internalized recombinant and plasma-derived FVIII through VWF-dependent and independent mechanisms. CLEC4M binding to recombinant FVIII was dependent on mannose-exposed N-linked glycans. CLEC4M mediated FVIII internalization via a clathrin-coated pit-dependent mechanism, resulting in transport of FVIII from early and late endosomes for catabolism by lysosomes. In vivo hepatic expression of CLEC4M after hydrodynamic liver transfer was associated with a decrease in plasma levels of endogenous murine FVIII:C in normal mice, whereas infused recombinant human FVIII was associated with sinusoidal endothelial cells in the presence or absence of VWF. Conclusions These findings suggest that CLEC4M is a novel clearance receptor that interacts with mannose-exposed glycans on FVIII in the presence or absence of VWF.


Assuntos
Moléculas de Adesão Celular/metabolismo , Endocitose , Células Endoteliais/metabolismo , Fator VIII/metabolismo , Lectinas Tipo C/metabolismo , Fígado/irrigação sanguínea , Receptores de Superfície Celular/metabolismo , Fator de von Willebrand/metabolismo , Animais , Sítios de Ligação , Moléculas de Adesão Celular/genética , Clatrina/metabolismo , Endossomos/metabolismo , Fator VIII/genética , Células HEK293 , Humanos , Lectinas Tipo C/genética , Lisossomos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligação Proteica , Transporte Proteico , Proteólise , Receptores de Superfície Celular/genética , Fator de von Willebrand/genética
20.
Res Pract Thromb Haemost ; 3(2): 254-260, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31011709

RESUMO

BACKGROUND: The multimeric glycoprotein von Willebrand factor (VWF) mediates platelet adhesion and aggregation at the site of vessel injury. The adhesive activity of VWF is influenced by its multimer length which is regulated by the metalloprotease ADAMTS13. The ability of ADAMTS13 to regulate platelet thrombus growth in a shear-dependent manner has been described, however, the mechanistic basis of this action has not been well characterized. METHODS: We developed an mCherry-tagged murine ADAMTS13 protein and utilized an ex vivo flow chamber system to visualize the localization of ADAMTS13 within the platelet thrombus under different conditions of shear. Using this system, we also assessed the influence of platelet-mediated tensile force on ADAMTS13 localization within the thrombus using gain-of-function GPIb binding and loss-of-function GPIIbIIIa binding mutants in VWF/ADAMTS13 DKO mice. RESULTS: ADAMTS13 was visualized on the growing platelet thrombus under very high shear using ADAMTS13-mcherry. ADAMTS13-mCherry localized particularly at the top portion of the thrombus and reduced thrombus size as it grew to occlusion. At the pathological high shear of 7500 s-1, platelet-mediated tensile force, involving GPIb but not GPIIbIIIa receptors, influenced localization of ADAMTS13 to the thrombus under conditions of shear. CONCLUSIONS: Tensile force applied on VWF produced by shear stress and platelet GPIb binding has a crucial role in ADAMTS13 activity at the site of thrombus formation. These results suggest that ADAMTS13 activity at the site of platelet thrombus formation is regulated by a shear stress and platelet-dependent feedback mechanism to prevent vessel occlusion and pathological thrombosis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA