Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Immunol ; 355: 104151, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32615414

RESUMO

B cells with regulatory properties (Bregs) were identified in human and in mice among different B-cell subsets. Their regulatory properties rely mainly on the production of anti-inflammatory cytokines, in particular IL10, IL-35 and TGFß, and were extensively studied in mouse models of autoimmune and inflammatory diseases. However, the exact nature of the stimulatory signals conferring regulatory properties to B cells is still not clear. We serendipitously observed that fluorescein isothiocyanate (FITC) binds to a significant proportion of naïve mouse B cells. Binding of FITC to the B-cell surface implicated at least in part the B-cell receptor. It triggered IL-10 production and allowed the endocytosis of FITC-coupled antigens followed by their presentation to CD4+ T cells. In particular, B cells incubated with FITC-OVA polarized OTII T cells towards a Tr1/Th2 phenotype in vitro. Further, the adoptive transfer of B cells incubated with FITC-labeled myelin oligodendrocyte glycoprotein peptide protected mice from experimental autoimmune encephalomyelitis, a T-cell-dependent autoimmune model. Together, the data show that FITC-stimulated B cells polarize immune responses towards Tr1/Th2 and acquire immuno-modulatory properties.


Assuntos
Linfócitos B Reguladores/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Animais , Subpopulações de Linfócitos B/imunologia , Linfócitos B/imunologia , Linfócitos B Reguladores/imunologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular/imunologia , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Feminino , Fluoresceína/metabolismo , Fluoresceína/farmacologia , Fluoresceína-5-Isotiocianato/química , Fluoresceína-5-Isotiocianato/metabolismo , Fluoresceína-5-Isotiocianato/farmacologia , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucinas/imunologia , Interleucinas/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Glicoproteína Mielina-Oligodendrócito/imunologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/metabolismo
2.
Haematologica ; 105(4): 1129-1137, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31289204

RESUMO

The treatment or prevention of bleeding in patients with hemophilia A relies on replacement therapy with different factor VIII (FVIII)-containing products or on the use of by-passing agents, i.e., activated prothrombin complex concentrates or recombinant activated factor VII. Emerging approaches include the use of bispecific anti-factor IXa/factor X antibodies, anti-tissue factor pathway inhibitor antibodies, interfering RNA to antithrombin, and activated protein C-specific serpins or gene therapy. The latter strategies are, however, hampered by the short clinical experience and potential adverse effects including the absence of tight temporal and spatial control of coagulation and the risk of uncontrolled insertional mutagenesis. Systemic delivery of mRNA allows endogenous production of the corresponding encoded protein. Thus, injection of erythropoietin-encoding mRNA in a lipid nanoparticle formulation resulted in increased erythropoiesis in mice and macaques. Here, we demonstrate that a single injection of in vitro transcribed B domain-deleted FVIII-encoding mRNA to FVIII-deficient mice enables endogenous production of pro-coagulant FVIII. Circulating FVIII:C levels above 5% of normal levels were maintained for up to 72 h, with an estimated half-life of FVIII production of 17.9 h, and corrected the bleeding phenotype in a tail clipping assay. The endogenously produced FVIII did however exhibit low specific activity and induced a potent neutralizing IgG response upon repeated administration of the mRNA. Our results suggest that the administration of mRNA is a plausible strategy for the endogenous production of proteins characterized by poor translational efficacy. The use of alternative mRNA delivery systems and improved FVIII-encoding mRNA should foster the production of functional molecules and reduce their immunogenicity.


Assuntos
Anticorpos Biespecíficos , Hemofilia A , Animais , Fator VIII/genética , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Hemorragia/terapia , Humanos , Camundongos , RNA Mensageiro/genética
3.
Haematologica ; 104(5): 1046-1054, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30545924

RESUMO

Hemophilia A is a rare hemorrhagic disorder caused by the lack of functional pro-coagulant factor VIII. Factor VIII replacement therapy in patients with severe hemophilia A results in the development of inhibitory anti-factor VIII IgG in up to 30% of cases. To date, immune tolerance induction, with daily injection of large amounts of factor VIII, is the only strategy to eradicate factor VIII inhibitors. This strategy is, however, efficient in only 60-80% of patients. We investigated whether blocking B-cell receptor signaling upon inhibition of Bruton tyrosine kinase prevents anti-factor VIII immune responses in a mouse model of severe hemophilia A. Factor VIII-naïve and factor VIII-sensitized factor VIII-deficient mice were fed with the selective inhibitor of Bruton tyrosine kinase, (R)-5-amino-1-(1-cyanopiperidin-3-yl)-3-(4-[2,4-difluorophenoxyl] phenyl)-1H pyrazole-4-carboxamide (PF-06250112), to inhibit B-cell receptor signaling prior to challenge with exogenous factor VIII. The consequences on the anti-factor VIII immune response were studied. Inhibition of Bruton tyrosine kinase during the primary anti-factor VIII immune response in factor VIII-naïve mice did not prevent the development of inhibitory anti-factor VIII IgG. In contrast, the anti-factor VIII memory B-cell response was consistently reduced upon treatment of factor VIII-sensitized mice with the Bruton tyrosine kinase inhibitor. The Bruton tyrosine kinase inhibitor reduced the differentiation of memory B cells ex vivo and in vivo following adoptive transfer to factor VIII-naïve animals. Taken together, our data identify inhibition of Bruton tyrosine kinase using PF-06250112 as a strategy to limit the reactivation of factor VIII-specific memory B cells upon re-challenge with therapeutic factor VIII.


Assuntos
Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Linfócitos B/imunologia , Modelos Animais de Doenças , Fator VIII/fisiologia , Hemofilia A/imunologia , Memória Imunológica/imunologia , Piperidinas/farmacologia , Pirazóis/farmacologia , Animais , Formação de Anticorpos , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Fator VIII/administração & dosagem , Fator VIII/antagonistas & inibidores , Hemofilia A/tratamento farmacológico , Hemofilia A/metabolismo , Tolerância Imunológica/efeitos dos fármacos , Tolerância Imunológica/imunologia , Imunoglobulina G/efeitos dos fármacos , Imunoglobulina G/imunologia , Memória Imunológica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
Trends Biochem Sci ; 39(5): 221-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24685657

RESUMO

Antigen-binding specificity of immunoglobulins is important for their function in immune defense. However, immune repertoires contain a considerable fraction of immunoglobulins with promiscuous binding behavior, the physicochemical basis of which is not well understood. Evolution of immunoglobulin specificity occurs through iterative processes of mutation and selection, referred to as affinity maturation. Recent studies reveal that some somatic mutations could compromise the thermodynamic stability of the variable regions of immunoglobulins. By integrating this observation with the wealth of data on the evolution of novel enzyme activities, we propose that antibody specificity is linked to the thermodynamic stability of the antigen-binding regions, which provides a quantitative distinction between highly specific and promiscuous antibodies.


Assuntos
Imunoglobulinas/química , Imunoglobulinas/imunologia , Animais , Especificidade de Anticorpos , Humanos , Região Variável de Imunoglobulina/química , Região Variável de Imunoglobulina/imunologia , Estabilidade Proteica , Termodinâmica
5.
Cell Immunol ; 325: 64-68, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29395036

RESUMO

The development of antibodies against therapeutic factor VIII (FVIII) represents the major complication of replacement therapy in patients with severe hemophilia A. Amongst the environmental risk factors that influence the anti-FVIII immune response, the presence of active bleeding or hemarthrosis has been evoked. Endothelium damage is typically associated with the release of oxidative compounds. Here, we addressed whether oxidation contributes to FVIII immunogenicity. The control with N-acetyl cysteine of the oxidative status in FVIII-deficient mice, a model of severe hemophilia A, reduced the immune response to exogenous FVIII. Ex vivo exposure of therapeutic FVIII to HOCl induced a mild oxidation of the molecule as evidenced by the loss of free amines and resulted in increased FVIII immunogenicity in vivo when compared to native FVIII. The increased immunogenicity of oxidized FVIII was not reverted by treatment of mice with N-acetyl cysteine, and did not implicate an increased maturation of professional antigen-presenting cells. Our data document that oxidation influences the immunogenicity of therapeutic FVIII.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Hemofilia A/metabolismo , Acetilcisteína/farmacologia , Animais , Anticorpos/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Modelos Animais de Doenças , Fator VIII/metabolismo , Fator VIII/farmacologia , Hemofilia A/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Estresse Oxidativo/imunologia
6.
Semin Thromb Hemost ; 44(6): 517-530, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29864775

RESUMO

The immunogenicity of therapeutic factor VIII (FVIII) in patients with hemophilia A has been puzzling scientific and clinical communities for more than 3 decades. Indeed, the development of inhibitory antibodies to FVIII remains a major clinical challenge and is associated with enormous societal costs. Thus, the reasons for which a presumably innocuous, short-lived, intravenously administered glycoprotein triggers such a deleterious, long-lasting neutralizing immune response is an enigma. This review does not pretend to bring an answer to this challenging question. It will however summarize the latest findings regarding the molecular interactions at play in the recognition of FVIII by the immune cells, the validity of the proposed risk factors for FVIII alloimmunization, and the different solutions that allow induction of FVIII-specific tolerance in preclinical models of hemophilia A.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Humanos
7.
Haematologica ; 103(2): 351-360, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29146705

RESUMO

Development of neutralizing antibodies against therapeutic Factor VIII (FVIII) is the most serious complication of the treatment of hemophilia A. There is growing evidence to show the multifactorial origin of the anti-FVIII immune response, combining both genetic and environmental factors. While a role for the complement system on innate as well as adaptive immunity has been documented, the implication of complement activation on the onset of the anti-FVIII immune response is unknown. Here, using in vitro assays for FVIII endocytosis by human monocyte-derived dendritic cells and presentation to T cells, as well as in vivo complement depletion in FVIII-deficient mice, we show a novel role for complement C3 in enhancing the immune response against therapeutic FVIII. In vitro, complement C3 and its cleavage product C3b enhanced FVIII endocytosis by dendritic cells and presentation to a FVIII-specific CD4+ T-cell hybridoma. The C1 domain of FVIII had previously been shown to play an important role in FVIII endocytosis, and alanine substitutions of the K2092, F2093 and R2090 C1 residues drastically reduce FVIII uptake in vitro Interestingly, complement activation rescued the endocytosis of the FVIII C1 domain triple mutant. In a mouse model of severe hemophilia A, transient complement C3 depletion by humanized cobra venom factor, which does not generate anaphylatoxin C5a, significantly reduced the primary anti-FVIII immune response, but did not affect anti-FVIII recall immune responses. Taken together, our results suggest an important adjuvant role for the complement cascade in the initiation of the immune response to therapeutic FVIII.


Assuntos
Anticorpos Neutralizantes/imunologia , Complemento C3/farmacologia , Fator VIII/imunologia , Animais , Apresentação de Antígeno/imunologia , Ativação do Complemento , Células Dendríticas/fisiologia , Endocitose/efeitos dos fármacos , Humanos , Imunidade/efeitos dos fármacos , Camundongos
8.
Haematologica ; 102(2): 271-281, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27758819

RESUMO

The development of inhibitory antibodies to therapeutic factor VIII is the major complication of replacement therapy in patients with hemophilia A. The first step in the initiation of the anti-factor VIII immune response is factor VIII interaction with receptor(s) on antigen-presenting cells, followed by endocytosis and presentation to naïve CD4+ T cells. Recent studies indicate a role for the C1 domain in factor VIII uptake. We investigated whether charged residues in the C2 domain participate in immunogenic factor VIII uptake. Co-incubation of factor VIII with BO2C11, a monoclonal C2-specific immunoglobulin G, reduced factor VIII endocytosis by dendritic cells and presentation to CD4+ T cells, and diminished factor VIII immunogenicity in factor VIII-deficient mice. The mutation of basic residues within the BO2C11 epitope of C2 replicated reduced in vitro immunogenic uptake, but failed to prevent factor VIII immunogenicity in mice. BO2C11 prevents factor VIII binding to von Willebrand factor, thus potentially biasing factor VIII immunogenicity by perturbing its half-life. Interestingly, a factor VIIIY1680C mutant, that does not bind von Willebrand factor, demonstrated unaltered endocytosis by dendritic cells as well as immunogenicity in factor VIII-deficient mice. Co-incubation of factor VIIIY1680C with BO2C11, however, resulted in decreased factor VIII immunogenicity in vivo In addition, a previously described triple C1 mutant showed decreased uptake in vitro, and reduced immunogenicity in vivo, but only in the absence of endogenous von Willebrand factor. Taken together, the results indicate that residues in the C1 and/or C2 domains of factor VIII are implicated in immunogenic factor VIII uptake, at least in vitro Conversely, in vivo, the binding to endogenous von Willebrand factor masks the reducing effect of mutations in the C domains on factor VIII immunogenicity.


Assuntos
Domínios C2 , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Endocitose/imunologia , Fator VIII/imunologia , Fator VIII/metabolismo , Domínios Proteicos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Fator VIII/química , Fator VIII/genética , Técnicas de Inativação de Genes , Hemofilia A/genética , Hemofilia A/imunologia , Hemofilia A/metabolismo , Humanos , Ativação Linfocitária/imunologia , Camundongos , Mutação , Ligação Proteica , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Fator de von Willebrand/metabolismo
9.
J Biol Chem ; 290(8): 5203-5213, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25564611

RESUMO

The healthy immune repertoire contains a fraction of antibodies that bind to various biologically relevant cofactors, including heme. Interaction of heme with some antibodies results in induction of new antigen binding specificities and acquisition of binding polyreactivity. In vivo, extracellular heme is released as a result of hemolysis or tissue damage; hence the post-translational acquisition of novel antigen specificities might play an important role in the diversification of the immunoglobulin repertoire and host defense. Here, we demonstrate that seronegative immune repertoires contain antibodies that gain reactivity to HIV-1 gp120 upon exposure to heme. Furthermore, a panel of human recombinant antibodies was cloned from different B cell subpopulations, and the prevalence of antibodies with cofactor-induced specificity for gp120 was determined. Our data reveal that upon exposure to heme, ∼24% of antibodies acquired binding specificity for divergent strains of HIV-1 gp120. Sequence analyses reveal that heme-sensitive antibodies do not differ in their repertoire of variable region genes and in most of the molecular features of their antigen-binding sites from antibodies that do not change their antigen binding specificity. However, antibodies with cofactor-induced gp120 specificity possess significantly lower numbers of somatic mutations in their variable region genes. This study contributes to the understanding of the significance of cofactor-binding antibodies in immunoglobulin repertoires and of the influence that the tissue microenvironment might have in shaping adaptive immune responses.


Assuntos
Linfócitos B/imunologia , Anticorpos Anti-HIV , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/imunologia , Região Variável de Imunoglobulina , Imunidade Adaptativa/genética , Anticorpos Anti-HIV/genética , Anticorpos Anti-HIV/imunologia , Humanos , Região Variável de Imunoglobulina/genética , Região Variável de Imunoglobulina/imunologia
10.
Blood ; 123(1): 121-5, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24014239

RESUMO

Vascular endothelial cells (ECs) link hemostasis, thrombosis, and complement. ECs synthesize both the clotting initiator von Willebrand factor (VWF) and the complement regulator factor H (FH). VWF is stored in EC Weibel-Palade bodies (WPBs), but the intracellular location of FH is not well defined. We found that FH colocalizes with VWF in WPBs of human umbilical vein ECs. Moreover, FH bound to VWF with an apparent nanomolar affinity and the complex was present in normal plasma. The binding of VWF to FH enhanced FH cofactor activity toward factor I-mediated downregulation of complement activation. Besides, this interaction inhibited ADAMTS13-mediated proteolysis of VWF and promoted platelet aggregation. Here, we describe a novel interaction between complement and hemostasis. The simultaneous secretion of VWF and FH by activated ECs may promote adhesion of platelets to endothelial injury sites to assure wound healing, simultaneously dampening the proinflammatory effect of complement to limit bystander tissue damage.


Assuntos
Fator H do Complemento/química , Trombose , Fator de von Willebrand/química , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Ativação do Complemento , Fator H do Complemento/metabolismo , Endotélio Vascular/citologia , Ensaio de Imunoadsorção Enzimática , Hemostasia , Heterozigoto , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Imunoprecipitação , Inflamação , Ligação Proteica , Mapeamento de Interação de Proteínas , Ressonância de Plasmônio de Superfície , Corpos de Weibel-Palade/metabolismo , Fator de von Willebrand/metabolismo
11.
Biochemistry ; 54(11): 2061-72, 2015 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-25742488

RESUMO

A fraction of antibodies from healthy immune repertoires binds to heme and acquires the ability to recognize multiple antigens. The mechanism and functional consequences of heme-mediated antigen binding promiscuity (polyreactivity) are not understood. Here, we used SPE7, a mouse monoclonal IgE specific for dinitrophenyl that has been thoroughly characterized at the molecular level, as a model antibody to elucidate the mechanism and functional consequences of heme-mediated polyreactivity. We first demonstrate that exposure of SPE7 to heme results in a substantial increase in its antigen binding polyreactivity. Comparison of the binding kinetics and thermodynamics of interaction of native and heme-bound SPE7 indicates that the binding of heme to SPE7 confers binding affinities in the low nanomolar range toward several antigens but has no influence on the mechanism of recognition of dinitrophenyl. In vitro cellular assays further demonstrate that heme-bound SPE7 does not promote the degranulation of basophils in the presence of new target antigens, while degranulation is observed in the presence of dinitrophenyl. Molecular docking and fluorescence spectroscopy revealed binding of heme to the variable region of SPE7 at a distance from the conventional binding site for dinitrophenyl, explaining the extension of binding polyreactivity without abrogation of the interaction with its cognate antigen. In addition, our data suggest that heme, when bound to IgE, is solvent-exposed and may serve as an interfacial cofactor mediating binding to diverse proteins. This study contributes to the understanding of the mechanisms of heme-induced antibody polyreactivity. It also paves the way toward the delineation of the functional impact of polyreactivity and cross-reactivity of IgE.


Assuntos
Anticorpos Monoclonais/metabolismo , Especificidade de Anticorpos , Heme/metabolismo , Imunoglobulina E/metabolismo , Modelos Moleculares , Animais , Anticorpos Monoclonais/química , Afinidade de Anticorpos , Basófilos/efeitos dos fármacos , Basófilos/imunologia , Basófilos/metabolismo , Sítios de Ligação de Anticorpos , Degranulação Celular/efeitos dos fármacos , Linhagem Celular , Reações Cruzadas , Dinitrobenzenos/química , Dinitrobenzenos/metabolismo , Dinitrobenzenos/farmacologia , Heme/química , Hemina/química , Hemina/metabolismo , Imunoglobulina E/química , Indicadores e Reagentes/química , Indicadores e Reagentes/metabolismo , Indicadores e Reagentes/farmacologia , Cinética , Camundongos , Simulação de Acoplamento Molecular , Ratos , Espectrometria de Fluorescência
12.
J Biol Chem ; 289(25): 17767-79, 2014 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-24802758

RESUMO

Polyreactive antibodies play an important role for neutralization of human immunodeficiency virus (HIV). In addition to intrinsic polyreactive antibodies, the immune system of healthy individuals contains antibodies with cryptic polyreactivity. These antibodies acquire promiscuous antigen binding potential post-translationally, after exposure to various redox-active substances such as reactive oxygen species, iron ions, and heme. Here, we characterized the interaction of a prototypic human antibody that acquires binding potential to glycoprotein (gp) 120 after exposure to heme. The kinetic and thermodynamic analyses of interaction of the polyreactive antibody with distinct clades of gp120 demonstrated that the antigen-binding promiscuity of the antibody compensates for the molecular heterogeneity of the target antigen. Thus, the polyreactive antibody recognized divergent gp120 clades with similar values of the binding kinetics and quantitatively identical changes in the activation thermodynamic parameters. Moreover, this antibody utilized the same type of noncovalent forces for formation of complexes with gp120. In contrast, HIV-1-neutralizing antibodies isolated from HIV-1-infected individuals, F425 B4a1 and b12, demonstrated different binding behavior upon interaction with distinct variants of gp120. This study contributes to a better understanding of the physiological role and binding mechanism of antibodies with cryptic polyreactivity. Moreover, this study might be of relevance for understanding the basic aspects of HIV-1 interaction with human antibodies.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Neutralizantes/química , Anticorpos Anti-HIV/química , Proteína gp120 do Envelope de HIV/química , HIV-1/química , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Sítios de Ligação de Anticorpos , Anticorpos Anti-HIV/imunologia , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Cinética , Termodinâmica
13.
J Immunol ; 191(3): 993-9, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23873158

RESUMO

An Ab molecule or a BCR that is able to bind multiple structurally unrelated Ags is defined as polyreactive. Polyreactive Abs and BCRs constitute an important part of immune repertoires under physiological conditions and may play essential roles in immune defense and in the maintenance of immune homeostasis. In this review, we integrate and discuss different findings that reveal the indispensable role of Ag-binding polyreactivity in the immune system. First, we describe the functional and molecular characteristics of polyreactive Abs. The following part of the review concentrates on the biological roles attributed to polyreactive Abs and to polyreactive BCRs. Finally, we discuss recent studies that link Ig polyreactivity with distinct pathological conditions.


Assuntos
Anticorpos/imunologia , Especificidade de Anticorpos , Reações Antígeno-Anticorpo , Imunoglobulinas/imunologia , Anticorpos/metabolismo , Afinidade de Anticorpos , Linfócitos B/imunologia , Epitopos/imunologia , Humanos , Região Variável de Imunoglobulina/imunologia , Imunoglobulinas/metabolismo , Estrutura Secundária de Proteína , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo
15.
Blood ; 117(7): 2257-64, 2011 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-21131590

RESUMO

Acquired hemophilia is a rare bleeding disorder characterized by the spontaneous occurrence of inhibitory antibodies against endogenous factor VIII (FVIII). IgG from some patients with acquired hemophilia hydrolyze FVIII. Because of the complex etiology of the disease, no clinical parameter, including the presence of FVIII-hydrolyzing IgG, has been associated with patient's survival or death. Here, we demonstrate the presence of anti-FIX antibodies in acquired hemophilia patients. IgG from some patients were found to hydrolyze FIX. In most cases, IgG-mediated FIX-hydrolysis resulted in FIX activation. IgG-mediated hydrolysis of FIX thus led to the significant generation of activated FIX in 25 of 65 patients. Based on the estimated kinetic parameters, patients' IgG activated up to 0.3nM FIX in 24 hours, an amount that restored thrombin generation in vitro provided the presence of more than or equal to 3% residual FVIII activity in plasma. This work identifies proteolytic IgG as novel molecules able to activate FIX under pathologic conditions. IgG-mediated FIX activation is a prevalent phenomenon among acquired hemophilia patients. The presence of FIX-activating IgG may partly compensate for the antibody-mediated inhibition of endogenous FVIII in restoring thrombin generation. This clinical trial was registered at www.clinicaltrials.gov as #NCT00213473.


Assuntos
Autoanticorpos/sangue , Fator IX/imunologia , Fator IX/metabolismo , Imunoglobulina G/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Fator VIII/imunologia , Fator VIII/metabolismo , Feminino , Hemofilia A/sangue , Hemofilia A/imunologia , Humanos , Hidrólise , Técnicas In Vitro , Cinética , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo
16.
Haematologica ; 98(10): 1650-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23716558

RESUMO

Induction of heme oxygenase-1, a stress-inducible enzyme with anti-inflammatory activity, reduces the immunogenicity of therapeutic factor VIII in experimental hemophilia A. In humans, heme oxygenase-1 expression is modulated by polymorphisms in the promoter of the heme oxygenase-1-encoding gene (HMOX1). We investigated the relationship between polymorphisms in the HMOX1 promoter and factor VIII inhibitor development in severe hemophilia A. We performed a case-control study on 99 inhibitor-positive patients and 263 patients who did not develop inhibitors within the first 150 cumulative days of exposure to therapeutic factor VIII. Direct sequencing and DNA fragment analysis were used to study (GT)n polymorphism and single nucleotide polymorphisms located at -1135 and -413 in the promoter of HMOX1. We assessed associations between the individual allele frequencies or genotypes, and inhibitor development. Our results demonstrate that inhibitor-positive patients had a higher frequency of alleles with large (GT)n repeats (L: n≥30), which are associated with lesser heme oxygenase-1 expression (odds ratio 2.31; 95% confidence interval 1.46-3.66; P<0.001]. Six genotypes (L/L, L/M, L/S, M/M, M/S and S/S) of (GT)n repeats were identified (S: n<21; M: 21≤n<30). The genotype group including L alleles (L/L, L/M and L/S) was statistically more frequent among inhibitor-positive than inhibitor-negative patients, as compared to the other genotypes (33.3% versus 17.1%) (odds ratio 2.21, 95% confidence interval 1.30-3.76; P<0.01). To our knowledge, this is the first association identified between HMOX1 promoter polymorphism and development of anti-drug antibodies. Our study paves the way towards modulation of the endogenous anti-inflammatory machinery of hemophilia patients to reduce the risk of inhibitor development.


Assuntos
Fator VIII/uso terapêutico , Heme Oxigenase-1/genética , Hemofilia A/genética , Repetições de Microssatélites/genética , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Anticorpos/sangue , Estudos de Casos e Controles , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Humanos , Índice de Gravidade de Doença
17.
Biochemistry ; 51(20): 4108-16, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22559004

RESUMO

Factor VIII (FVIII) is a glycoprotein that plays an important role in the intrinsic pathway of coagulation. In circulation, FVIII is protected upon binding to von Willebrand factor (VWF), a chaperone molecule that regulates its half-life, distribution, and activity. Despite the biological significance of this interaction, its molecular mechanisms are not fully characterized. We determined the equilibrium and activation thermodynamics of the interaction between FVIII and VWF. The equilibrium affinity determined by surface plasmon resonance was temperature-dependent with a value of 0.8 nM at 35 °C. The FVIII-VWF interaction was characterized by very fast association (8.56 × 10(6) M(-1) s(-1)) and fast dissociation (6.89 × 10(-3) s(-1)) rates. Both the equilibrium association and association rate constants, but not the dissociation rate constant, were dependent on temperature. Binding of FVIII to VWF was characterized by favorable changes in the equilibrium and activation entropy (TΔS° = 89.4 kJ/mol, and -TΔS(++) = -8.9 kJ/mol) and unfavorable changes in the equilibrium and activation enthalpy (ΔH° = 39.1 kJ/mol, and ΔH(++) = 44.1 kJ/mol), yielding a negative change in the equilibrium Gibbs energy. Binding of FVIII to VWF in solid-phase assays demonstrated a high sensitivity to acidic pH and a sensitivity to ionic strength. Our data indicate that the interaction between FVIII and VWF is mediated mainly by electrostatic forces, and that it is not accompanied by entropic constraints, suggesting the absence of conformational adaptation but the presence of rigid "pre-optimized" binding surfaces.


Assuntos
Fator VIII/química , Termodinâmica , Fator de von Willebrand/química , Humanos , Concentração Osmolar , Ligação Proteica , Eletricidade Estática , Ressonância de Plasmônio de Superfície
18.
J Biol Chem ; 286(18): 16459-69, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21454703

RESUMO

C1q is the recognition subunit of the first component of the classical complement pathway. It participates in clearance of immune complexes and apoptotic cells as well as in defense against pathogens. Inappropriate activation of the complement contributes to cellular and tissue damage in different pathologies, urging the need for the development of therapeutic agents that are able to inhibit the complement system. In this study, we report heme as an inhibitor of C1q. Exposure of C1q to heme significantly reduced the activation of the classical complement pathway, mediated by C-reactive protein (CRP) and IgG. Interaction analyses revealed that heme reduces the binding of C1q to CRP and IgG. Furthermore, we demonstrated that the inhibition of C1q interactions results from a direct binding of heme to C1q. Formation of complex of heme with C1q caused changes in the mechanism of recognition of IgG and CRP. Taken together, our data suggest that heme is a natural negative regulator of the classical complement pathway at the level of C1q. Heme may play a role at sites of excessive tissue damage and hemolysis where large amounts of free heme are released.


Assuntos
Complemento C1q/metabolismo , Via Clássica do Complemento/fisiologia , Heme/metabolismo , Proteína C-Reativa/química , Proteína C-Reativa/metabolismo , Complemento C1q/antagonistas & inibidores , Complemento C1q/química , Heme/química , Hemólise/fisiologia , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Ligação Proteica
19.
Br J Haematol ; 156(1): 3-12, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21988190

RESUMO

Acquired haemophilia A (AHA) is a rare bleeding disorder characterized by the sudden generation of autoantibodies against factor VIII (FVIII) in individuals with no previous history of abnormal haemostasis. Understanding the pathogenesis of this disease has been hampered by the rarity of the patients and the difficulty in obtaining biological material from untreated patients. Still, progress has been made recently in understanding the pathogenesis of AHA. In particular, the importance of CD4(+) T cells in AHA development has been documented and the epitopes targeted by T cells on FVIII have been delineated. Accordingly, a polymorphism in the cytotoxic T-lymphocyte-associated protein 4 gene (CTLA4), known to participate in the regulation of CD4(+) T-cell responses, and a preferential usage of certain human leukocyte antigen class II haplotypes, have been associated with the disease. Recent findings have documented the presence of immunoglobulin G (IgG) with proteolytic activity against FVIII and factor IX (FIX) in patients with AHA. While FVIII-hydrolysing IgG has been shown to inactivate FVIII, FIX-hydrolysing IgG from AHA patients activate FIX in vitro. Here, we describe the latest findings on the immuno-pathogenesis of AHA, with a special focus on the potential role played by antibodies endowed with proteolytic properties.


Assuntos
Autoanticorpos/metabolismo , Hemofilia A/imunologia , Imunoglobulina G/metabolismo , Animais , Autoanticorpos/imunologia , Autoimunidade , Epitopos/imunologia , Fator IX/imunologia , Fator IX/metabolismo , Fator VIII/genética , Fator VIII/imunologia , Fator VIII/metabolismo , Antígenos HLA/genética , Antígenos HLA/imunologia , Hemofilia A/genética , Humanos , Imunoglobulina G/imunologia , Proteólise , Fatores de Risco , Linfócitos T/imunologia
20.
Blood ; 115(13): 2682-5, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19890094

RESUMO

Replacement therapy with exogenous factor VIII (FVIII) to treat hemorrhages induces anti-FVIII inhibitory immunoglobulin G in up to 30% of patients with hemophilia A. Chronic inflammation associated with recurrent bleedings is a proposed risk factor for FVIII inhibitor development. Heme oxygenase-1 (HO-1) is a stress-inducible enzyme with potent anti-inflammatory activity. Here, we demonstrate that induction of HO-1 before FVIII administration drastically reduces the onset of the anti-FVIII humoral immune response. The protective effect was specific for HO-1 because it was reproduced on administration of the end products of HO-1 activity, carbon monoxide, and bilirubin, and prevented by the pharmacologic inhibition of HO-1 using tin mesoporphyrin IX. HO-1 induction was associated with decreased major histocompatibility complex class II expression by splenic antigen-presenting cells and reduced T-cell proliferation. Triggering the endogenous anti-inflammatory machinery before FVIII administration may represent a novel therapeutic option for preventing the development of FVIII inhibitors in hemophilia A patients.


Assuntos
Fator VIII/uso terapêutico , Heme Oxigenase-1/fisiologia , Hemina/administração & dosagem , Hemofilia A/imunologia , Imunoglobulina G/biossíntese , Isoanticorpos/biossíntese , Proteínas de Membrana/fisiologia , Animais , Células Apresentadoras de Antígenos/imunologia , Esquema de Medicação , Fator VIII/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Hemina/farmacologia , Hemina/uso terapêutico , Hemofilia A/tratamento farmacológico , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Imunoglobulina G/imunologia , Inflamação , Isoanticorpos/imunologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Metaloporfirinas/farmacologia , Camundongos , Camundongos Knockout , Baço/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA