Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Thromb Haemost ; 122(1): 80-91, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33940654

RESUMO

Hemolytic disorders characterized by complement-mediated intravascular hemolysis, such as autoimmune hemolytic anemia and paroxysmal nocturnal hemoglobinuria, are often complicated by life-threatening thromboembolic complications. Severe hemolytic episodes result in the release of red blood cell (RBC)-derived proinflammatory and oxidatively reactive mediators (e.g., extracellular hemoglobin, heme, and iron) into plasma. Here, we studied the role of these hemolytic mediators in coagulation activation by measuring factor Xa (FXa) and thrombin generation in the presence of RBC lysates. Our results show that hemolytic microvesicles (HMVs) formed during hemolysis stimulate thrombin generation through a mechanism involving FVIII and FIX, the so-called intrinsic tenase complex. Iron scavenging during hemolysis using deferoxamine decreased the ability of the HMVs to enhance thrombin generation. Furthermore, the addition of ferric chloride (FeCl3) to plasma propagated thrombin generation in a FVIII- and FIX-dependent manner suggesting that iron positively affects blood coagulation. Phosphatidylserine (PS) blockade using lactadherin and iron chelation using deferoxamine reduced intrinsic tenase activity in a purified system containing HMVs as source of phospholipids confirming that both PS and iron ions contribute to the procoagulant effect of the HMVs. Finally, the effects of FeCl3 and HMVs decreased in the presence of ascorbate and glutathione indicating that oxidative stress plays a role in hypercoagulability. Overall, our results provide evidence for the contribution of iron ions derived from hemolytic RBCs to thrombin generation. These findings add to our understanding of the pathogenesis of thrombosis in hemolytic diseases.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Micropartículas Derivadas de Células/metabolismo , Cisteína Endopeptidases/metabolismo , Ferro/metabolismo , Proteínas de Neoplasias/metabolismo , Coagulação Sanguínea/fisiologia , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/efeitos dos fármacos , Micropartículas Derivadas de Células/fisiologia , Cisteína Endopeptidases/efeitos adversos , Cisteína Endopeptidases/fisiologia , Eritrócitos/química , Eritrócitos/metabolismo , Eritrócitos/fisiologia , Hemólise/fisiologia , Humanos , Ferro/sangue , Proteínas de Neoplasias/efeitos adversos , Proteínas de Neoplasias/fisiologia , Trombose/metabolismo , Trombose/fisiopatologia
2.
mBio ; 12(2)2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33758081

RESUMO

Complement, contact activation, coagulation, and fibrinolysis are serum protein cascades that need strict regulation to maintain human health. Serum glycoprotein, a C1 inhibitor (C1-INH), is a key regulator (inhibitor) of serine proteases of all the above-mentioned pathways. Recently, an autotransporter protein, virulence-associated gene 8 (Vag8), produced by the whooping cough pathogen, Bordetella pertussis, was shown to bind to C1-INH and interfere with its function. Here, we present the structure of the Vag8-C1-INH complex determined using cryo-electron microscopy at a 3.6-Å resolution. The structure shows a unique mechanism of C1-INH inhibition not employed by other pathogens, where Vag8 sequesters the reactive center loop of C1-INH, preventing its interaction with the target proteases.IMPORTANCE The structure of a 10-kDa protein complex is one of the smallest to be determined using cryo-electron microscopy at high resolution. The structure reveals that C1-INH is sequestered in an inactivated state by burial of the reactive center loop in Vag8. By so doing, the bacterium is able to simultaneously perturb the many pathways regulated by C1-INH. Virulence mechanisms such as the one described here assume more importance given the emerging evidence about dysregulation of contact activation, coagulation, and fibrinolysis leading to COVID-19 pneumonia.


Assuntos
Proteínas de Bactérias/metabolismo , Bordetella pertussis/patogenicidade , Proteína Inibidora do Complemento C1/metabolismo , Evasão da Resposta Imune , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Coagulação Sanguínea , Bordetella pertussis/química , Bordetella pertussis/metabolismo , Proteína Inibidora do Complemento C1/química , Proteínas do Sistema Complemento/metabolismo , Microscopia Crioeletrônica , Fibrinólise , Modelos Moleculares , Mutação , Ligação Proteica , Domínios Proteicos , Sistemas de Secreção Tipo V/genética , Sistemas de Secreção Tipo V/metabolismo , Virulência , Fatores de Virulência de Bordetella
3.
Pharmaceuticals (Basel) ; 14(1)2021 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-33440845

RESUMO

Correct glycosylation of proteins is essential for production of therapeutic proteins as glycosylation is important for protein solubility, stability, half-life and immunogenicity. The heavily glycosylated plasma protein C1-inhibitor (C1-INH) is used in treatment of hereditary angioedema attacks. In this study, we used C1-INH as a model protein to propose an approach to develop recombinant glycoproteins with the desired glycosylation. We produced fully functional recombinant C1-INH in Chinese hamster ovary (CHO) cells. In vivo we observed a biphasic clearance, indicating different glycosylation forms. N-glycan analysis with mass spectrometry indeed demonstrated heterogeneous glycosylation for recombinant C1-INH containing terminal galactose and terminal sialic acid. Using a Ricinus Communis Agglutinin I (RCA120) column, we could reduce the relative abundance of terminal galactose and increase the relative abundance of terminal sialic acid. This resulted in a fully active protein with a similar in vivo clearance rate to plasmaderived C1-INH. In summary, we describe the development of a recombinant human glycoprotein using simple screening tools to obtain a product that is similar in function and in vivo clearance rate to its plasma-derived counterpart. The approach used here is of potential use in the development of other therapeutic recombinant human glycoproteins.

4.
Hum Mutat ; 41(1): 38-57, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31517426

RESUMO

C1 inhibitor (C1Inh) deficiency is responsible for hereditary angioedema (C1-INH-HAE) and caused by variants of the SERPING1/C1INH/C1NH gene. C1Inh is the major control of kallikrein-kinin system. C1Inh deficiency leads to its uncontrolled activation, with subsequent generation of the vasoactive peptide bradykinin. This update documents 748 different SERPING1 variants, including published variants and additional 120 unpublished ones. They were identified as heterozygous variants (n = 729), as homozygous variants in 10 probands and as compound heterozygous variants (nine combinations). Six probands with heterozygous variants exhibited gonadal mosaicism. Probands with heterozygous (n = 72) and homozygous (n = 1) variants were identified as de novo cases. Overall, 58 variants were found at positions showing high residue conservation among serpins, and have been referred to as a mousetrap function of C1Inh: reactive center loop, gate, shutter, breach, and hinge. C1Inh phenotype analysis identified dysfunctional serpin variants with failed serpin-protease association and a residual 105-kDa species after incubation with target protease. Regarding this characteristic, in conditions with low antigenic C1Inh, 74 C1-INH-HAE probands presented with an additional so-called intermediate C1-INH-HAE phenotype. The present update addresses a comprehensive SERPING1 variant spectrum that facilitates genotype-phenotype correlations, highlighting residues of strategic importance for serpin function and for identification of C1Inh deficiency as serpinopathy.


Assuntos
Angioedemas Hereditários/diagnóstico , Angioedemas Hereditários/genética , Proteína Inibidora do Complemento C1/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Mutação , Fenótipo , Alelos , Proteína Inibidora do Complemento C1/química , Biologia Computacional , Bases de Dados Genéticas , Estudos de Associação Genética/métodos , Genótipo , Haploinsuficiência , Humanos , Modelos Moleculares , Conformação Proteica , Splicing de RNA , Relação Estrutura-Atividade
5.
Thromb Haemost ; 118(12): 2134-2144, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30453343

RESUMO

BACKGROUND: C1-inhibitor (C1-inh) therapeutics can reduce neutrophil activity in various inflammatory conditions. This 'novel' anti-inflammatory effect of C1-inh is attributed to the tetrasaccharide sialyl LewisX (SLeX) present on its N-glycans. Via SLeX, C1-inh is suggested to interact with selectins on inflamed endothelium and prevent neutrophil rolling. However, C1-inh products contain plasma glycoprotein α1-antichymotrypsin (ACT) as a co-purified protein impurity. OBJECTIVE: This article investigates the contribution of ACT to the effects observed with C1-inh. MATERIALS AND METHODS: We have separated C1-inh and ACT from a therapeutic C1-inh preparation and investigated the influence of these proteins on SLeX-selectin interactions in a specific in vitro model, which makes use of rolling of SLeX-coated beads on immobilized E-selectin. RESULTS: We find that ACT and not C1-inh, shows a clear sialic acid-dependent interference in SLeX-selectin interactions, at concentrations present in C1-inh therapeutics. Furthermore, we do not find any evidence of SLeX on C1-inh using either Western blotting with anti-SLeX antibodies (CSLEX1 and KM93) or by mass spectrometric analysis of N-glycans. C1-inh reacts weakly to antibody HECA-452, which detects a broad range of selectin ligands, but ACT gives a much stronger signal, suggesting the presence of a selectin ligand on ACT. CONCLUSION: The 'novel' anti-inflammatory effects of C1-inh are unlikely due to SLeX on C1-inh and can in fact be due to SLeX-like glycans on ACT, present in C1-inh products. In view of our results, it is important to assess the role of ACT in vivo and revisit past studies performed with commercial C1-inh.


Assuntos
Anti-Inflamatórios/imunologia , Proteína Inibidora do Complemento C1/uso terapêutico , Endotélio Vascular/fisiologia , Neutrófilos/imunologia , Oligossacarídeos/uso terapêutico , Anticorpos Bloqueadores/farmacologia , Ligação Competitiva , Sistema Livre de Células , Humanos , Migração e Rolagem de Leucócitos , Microesferas , Ativação de Neutrófilo , Preparações Farmacêuticas , Selectinas/metabolismo , Antígeno Sialil Lewis X , alfa 1-Antitripsina/metabolismo
6.
PLoS One ; 12(10): e0186652, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29036225

RESUMO

C1 esterase inhibitor (C1-INH) can inhibit multiple pathways (complement, contact-kinin, coagulation, and fibrinolysis) that are all implicated in the pathophysiology of asthma. We explored the effect of human plasma-derived C1-INH on allergic lung inflammation in a house dust mite (HDM) induced asthma mouse model by daily administration of C1-INH (15 U) during the challenge phase. NaCl and HDM exposed mice had comparable plasma C1-INH levels, while bronchoalveolar lavage fluid (BALF) levels were increased in HDM exposed mice coinciding with slightly reduced activation of complement (C5a). C1-INH treatment reduced Th2 response and enhanced HDM-specific IgG1. Influx of eosinophils in BALF or lung, pulmonary damage, mucus production, procoagulant response or plasma leakage in BALF was similar in both groups. In conclusion, C1-INH dampens Th2 responses during HDM induced allergic lung inflammation.


Assuntos
Asma/tratamento farmacológico , Asma/imunologia , Proteína Inibidora do Complemento C1/farmacologia , Pyroglyphidae/imunologia , Animais , Proteína Inibidora do Complemento C1/uso terapêutico , Feminino , Humanos , Imunoglobulina G/imunologia , Camundongos , Células Th2/efeitos dos fármacos , Células Th2/imunologia
7.
J Immunol Methods ; 426: 114-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26318839

RESUMO

BACKGROUND: Neutralizing autoantibodies (NAbs) against plasma serpin C1-inhibitor (C1-inh) are implicated in the rare disorder, acquired angioedema (AAE). There is insufficient understanding of the process of antibody formation and its correlation with disease progression and severity. We have developed an ELISA for detecting neutralizing capacity of anti-C1-inh positive plasma samples that can be used to study changes in NAb repertoire in patient plasma over the course of disease. METHODS: The ELISA is based on the specific interaction of active C1-inh with its target protease C1s. Decrease in the amount of C1s bound to immobilized C1-inh in the presence of test samples is proportional to the neutralizing capacity of the sample. Assay specificity, intra- and inter-assay variation and assay cut-off are determined using anti-C1-inh antibodies. Assay capability is demonstrated using plasma samples from AAE patients. RESULTS: The assay is specific to a neutralizing anti-C1-inh antibody and shows no interference by a non-neutralizing anti-C1-inh antibody or by the plasma matrix. Intra-assay and inter-assay variations are determined as 17 and 18% respectively. Neutralizing capacity of antibody positive AAE patient plasma samples (n=16) with IgG or IgM type antibodies is readily determined. All samples show positive neutralizing capacity. CONCLUSION: We have developed a robust, specific and semi-quantitative assay to detect the neutralizing capacity of plasma samples containing anti-C1-inh antibodies. This assay can be an important tool for the study of clinical implications of anti-C1-inh NAbs.


Assuntos
Angioedema/sangue , Anticorpos Neutralizantes/sangue , Autoanticorpos/sangue , Proteínas Inativadoras do Complemento 1/imunologia , Ensaio de Imunoadsorção Enzimática , Angioedema/imunologia , Proteína Inibidora do Complemento C1 , Humanos
8.
Drug Deliv ; 21(4): 302-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24215253

RESUMO

CONTEXT: C1-esterase inhibitor (C1-inh) therapy is currently administered to patients with C1-inh deficiency through intravenous injections. The possibility of subcutaneous administration is currently being explored since this would alleviate need for hospitalization and increase mobility and well-being of patients. Recently, it was observed in pigs that C1-inh indeed can effectively be applied by subcutaneous injection. For studies on the effectiveness of C1-inh therapy for other indications than acquired and hereditary angioedema, rats are commonly used as model animal. For rats, however, subcutaneous C1-inh administration has never been investigated. OBJECTIVE: To evaluate the efficacy of subcutaneous C1-inh administration in rats. MATERIALS AND METHODS: Three boli of 100 U/kg human plasma-derived C1-inh were administered to Wistar rats on three consecutive days through subcutaneous injection or intravenous injection. Blood samples were collected from the tail veins 3, 4.5 or 6 h after C1-inh administration for measurement of C1-inh plasma levels. Antigen and activity levels of C1-inh of each plasma sample were determined by means of a specific ELISA. RESULTS: For both C1-inh antigen and C1-inh activity, 21- to 119-fold higher plasma levels were measured after intravenous administration compared with subcutaneous administration. Subcutaneous administration also resulted in C1-inh plasma levels that were less stable and with decreased relative activity. CONCLUSION: These combined results indicate that in rats, subcutaneous injections in the present formulation are not effective as alternative administration route for C1-inh.


Assuntos
Angioedema/tratamento farmacológico , Proteína Inibidora do Complemento C1/administração & dosagem , Modelos Animais de Doenças , Angioedema/sangue , Angioedema/enzimologia , Animais , Proteína Inibidora do Complemento C1/metabolismo , Humanos , Inflamação/sangue , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Injeções Subcutâneas , Masculino , Ratos , Ratos Wistar , Resultado do Tratamento
9.
Crit Care ; 10(5): R150, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17069648

RESUMO

INTRODUCTION: The mechanism of coagulation activation during continuous venovenous hemofiltration (CVVH) has not yet been elucidated. Insight into the mechanism(s) of hemostatic activation within the extracorporeal circuit could result in a more rational approach to anticoagulation. The aim of the present study was to investigate whether CVVH using cellulose triacetate filters causes activation of the contact factor pathway or of the tissue factor pathway of coagulation. In contrast to previous studies, CVVH was performed without anticoagulation. METHODS: Ten critically ill patients were studied prior to the start of CVVH and at 5, 15 and 30 minutes and 1, 2, 3 and 6 hours thereafter, for measurement of prothrombin fragment F1+2, soluble tissue factor, activated factor VII, tissue factor pathway inhibitor, kallikrein-C1-inhibitor and activated factor XII-C1-inhibitor complexes, tissue-type plasminogen activator, plasminogen activator inhibitor type I, plasmin-antiplasmin complexes, protein C and antithrombin. RESULTS: During the study period the prothrombin fragment F1+2 levels increased significantly in four patients (defined as group A) and did not change in six patients (defined as group B). Group A also showed a rapid increase in transmembrane pressure, indicating clotting within the filter. At baseline, the activated partial thromboplastin time, the prothrombin time and the kallikrein-C1-inhibitor complex and activated factor XII-C1-inhibitor complex levels were significantly higher in group B, whereas the platelet count was significantly lower in group B. For the other studied markers the differences between group A and group B at baseline were not statistically significant. During CVVH the difference in the time course between group A and group B was not statistically significant for the markers of the tissue factor system (soluble tissue factor, activated factor VII and tissue factor pathway inhibitor), for the markers of the contact system (kallikrein-C1-inhibitor and activated factor XII-C1-inhibitor complexes) and for the markers of the fibrinolytic system (plasmin-antiplasmin complexes, tissue-type plasminogen activator and plasminogen activator inhibitor type I). CONCLUSION: Early thrombin generation was detected in a minority of intensive care patients receiving CVVH without anticoagulation. Systemic concentrations of markers of the tissue factor system and of the contact system did not change during CVVH. To elucidate the mechanism of clot formation during CVVH we suggest that future studies are needed that investigate the activation of coagulation directly at the site of the filter. Early coagulation during CVVH may be related to lower baseline levels of markers of contact activation.


Assuntos
Fatores de Coagulação Sanguínea/metabolismo , Hemofiltração , Adulto , Idoso , Coagulação Sanguínea/fisiologia , Estudos de Coortes , Feminino , Hemofiltração/efeitos adversos , Hemofiltração/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Taxa de Sobrevida
10.
FEMS Yeast Res ; 5(11): 1069-77, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16269393

RESUMO

Serine proteinases from inflammatory cells, including polymorphonuclear neutrophils, are involved in various inflammatory disorders, like pulmonary emphysema and rheumatoid arthritis. Inhibitors of these serine proteinases are potential drug candidates for the treatment of these disorders, since they prevent the unrestricted proteolysis. This study describes a novel specific antistasin-type inhibitor of neutrophil serine proteinases, we called Fahsin. This inhibitor was purified from the Nile leech Limnatis nilotica, sequenced and heterologously expressed using a synthetic gene in the methylotrophic yeast Pichia pastoris, yielding 0.5 g(-l) of the protein in the culture medium. Recombinant Fahsin was purified to homogeneity and characterised by N-terminal amino acid sequencing and mass spectrometry. Inhibition-kinetic analysis showed that recombinant Fahsin is a fast, tight-binding inhibitor of human neutrophil elastase with inhibition constant in the nanomolar range. Furthermore, recombinant Fahsin was, in contrast to various other neutrophil elastase inhibitors, insensitive to chemical oxidation and biological oxidation via myeloperoxidase-generated free oxygen radicals. Thus, Fahsin constitutes a novel member of a still expanding family of naturally occurring inhibitors of serine proteinases with potential therapeutic use for treatment of human diseases.


Assuntos
Hormônios de Invertebrado/biossíntese , Sanguessugas/química , Pichia/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Inibidores de Serino Proteinase/biossíntese , Animais , Hormônios de Invertebrado/genética , Pichia/genética , Inibidores de Proteases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Inibidores de Serino Proteinase/genética , Inibidores de Serino Proteinase/metabolismo
11.
J Biol Chem ; 278(32): 29463-70, 2003 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-12773530

RESUMO

C1-inhibitor (C1-Inh) is a serine protease inhibitor (serpin) with a unique, non-conserved N-terminal domain of unknown function. Genetic deficiency of C1-Inh causes hereditary angioedema. A novel type of mutation (Delta 3) in exon 3 of the C1-Inh gene, resulting in deletion of Asp62-Thr116 in this unique domain, was encountered in a hereditary angioedema pedigree. Because the domain is supposedly not essential for inhibitory activity, the unexpected loss-of-function of this deletion mutant was further investigated. The Delta 3 mutant and three additional mutants starting at Pro76, Gly98, and Ser115, lacking increasing parts of the N-terminal domain, were produced recombinantly. C1-Inh76 and C1-Inh98 retained normal conformation and interaction kinetics with target proteases. In contrast, C1-Inh115 and Delta 3, which both lack the connection between the serpin and the non-serpin domain via two disulfide bridges, were completely non-functional because of a complex-like and multimeric conformation, as demonstrated by several criteria. The Delta 3 mutant also circulated in multimeric form in plasma from affected family members. The C1-Inh mutant reported here is unique in that deletion of an entire amino acid stretch from a domain not shared by other serpins leads to a loss-of-function. The deletion in the unique N-terminal domain results in a "multimerization phenotype" of C1-Inh, because of diminished stability of the central beta-sheet. This phenotype, as well as the location of the disulfide bridges between the serpin and the non-serpin domain of C1-Inh, suggests that the function of the N-terminal region may be similar to one of the effects of heparin in antithrombin III, maintenance of the metastable serpin conformation.


Assuntos
Mutação , Serpinas/química , Sequência de Aminoácidos , Sequência de Bases , Western Blotting , Centrifugação com Gradiente de Concentração , Proteínas Inativadoras do Complemento 1 , Proteína Inibidora do Complemento C1 , Cristalografia por Raios X , Cisteína/química , DNA/metabolismo , Dissulfetos/química , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Éxons , Deleção de Genes , Glicina/química , Heparina/química , Temperatura Alta , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Fenótipo , Plasmídeos/metabolismo , Reação em Cadeia da Polimerase , Prolina/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Serina/química , Sacarose/farmacologia , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...