Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 139(13): 2050-2065, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-34752599

RESUMO

Although several members of protein disulfide isomerase (PDI) family support thrombosis, other PDI family members with the CXYC motif remain uninvestigated. ERp46 has 3 CGHC redox-active sites and a radically different molecular architecture than other PDIs. Expression of ERp46 on the platelet surface increased with thrombin stimulation. An anti-ERp46 antibody inhibited platelet aggregation, adenosine triphosphate (ATP) release, and αIIbß3 activation. ERp46 protein potentiated αIIbß3 activation, platelet aggregation, and ATP release, whereas inactive ERp46 inhibited these processes. ERp46 knockout mice had prolonged tail-bleeding times and decreased platelet accumulation in thrombosis models that was rescued by infusion of ERp46. ERp46-deficient platelets had decreased αIIbß3 activation, platelet aggregation, ATP release, and P-selectin expression. The defects were reversed by wild-type ERp46 and partially reversed by ERp46 containing any of the 3 active sites. Platelet aggregation stimulated by an αIIbß3-activating peptide was inhibited by the anti-ERp46 antibody and was decreased in ERp46-deficient platelets. ERp46 bound tightly to αIIbß3 by surface plasmon resonance but poorly to platelets lacking αIIbß3 and physically associated with αIIbß3 upon platelet activation. ERp46 mediated clot retraction and platelet spreading. ERp46 more strongly reduced disulfide bonds in the ß3 subunit than other PDIs and in contrast to PDI, generated thiols in ß3 independently of fibrinogen. ERp46 cleaved the Cys473-Cys503 disulfide bond in ß3, implicating a target for ERp46. Finally, ERp46-deficient platelets have decreased thiols in ß3, implying that ERp46 cleaves disulfide bonds in platelets. In conclusion, ERp46 is critical for platelet function and thrombosis and facilitates αIIbß3 activation by targeting disulfide bonds.


Assuntos
Hemostasia , Tiorredoxinas/metabolismo , Trombose , Animais , Retículo Endoplasmático/metabolismo , Camundongos , Camundongos Knockout , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombose/genética , Trombose/metabolismo
2.
Blood ; 138(21): 2106-2116, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34189574

RESUMO

Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder mediated by ultra-large immune complexes (ULICs) containing immunoglobulin G (IgG) antibodies to a multivalent antigen composed of platelet factor 4 and heparin. The limitations of current antithrombotic therapy in HIT supports the need to identify additional pathways that may be targets for therapy. Activation of FcγRIIA by HIT ULICs initiates diverse procoagulant cellular effector functions. HIT ULICs are also known to activate complement, but the contribution of this pathway to the pathogenesis of HIT has not been studied in detail. We observed that HIT ULICs physically interact with C1q in buffer and plasma, activate complement via the classical pathway, promote codeposition of IgG and C3 complement fragments (C3c) on neutrophil and monocyte cell surfaces. Complement activation by ULICs, in turn, facilitates FcγR-independent monocyte tissue factor expression, enhances IgG binding to the cell surface FcγRs, and promotes platelet adhesion to injured endothelium. Inhibition of the proximal, but not terminal, steps in the complement pathway abrogates monocyte tissue factor expression by HIT ULICs. Together, these studies suggest a major role for complement activation in regulating Fc-dependent effector functions of HIT ULICs, identify potential non-anticoagulant targets for therapy, and provide insights into the broader roles of complement in immune complex-mediated thrombotic disorders.


Assuntos
Anticoagulantes/efeitos adversos , Complexo Antígeno-Anticorpo/imunologia , Ativação do Complemento , Heparina/efeitos adversos , Trombocitopenia/induzido quimicamente , Anticoagulantes/imunologia , Complemento C3/imunologia , Heparina/imunologia , Humanos , Imunoglobulina G/imunologia , Fator Plaquetário 4/imunologia , Receptores de IgG/imunologia , Trombocitopenia/complicações , Trombocitopenia/imunologia , Trombose/etiologia , Trombose/imunologia
3.
Blood ; 135(10): 743-754, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-31722003

RESUMO

Sepsis is characterized by multiorgan system dysfunction that occurs because of infection. It is associated with high morbidity and mortality and is in need of improved therapeutic interventions. Neutrophils play a crucial role in sepsis, releasing neutrophil extracellular traps (NETs) composed of DNA complexed with histones and toxic antimicrobial proteins that ensnare pathogens, but also damage host tissues. At presentation, patients often have a significant NET burden contributing to the multiorgan damage. Therefore, interventions that inhibit NET release would likely be ineffective at preventing NET-based injury. Treatments that enhance NET degradation may liberate captured bacteria and toxic NET degradation products (NDPs) and likely be of limited therapeutic benefit as well. We propose that interventions that stabilize NETs and sequester NDPs may be protective in sepsis. We showed that platelet factor 4 (PF4), a platelet-associated chemokine, binds and compacts NETs, increasing their resistance to DNase I. We now show that PF4 increases NET-mediated bacterial capture, reduces the release of NDPs, and improves outcome in murine models of sepsis. A monoclonal antibody KKO which binds to PF4-NET complexes, further enhances DNase resistance. However, the Fc portion of this antibody activates the immune response and increases thrombotic risk, negating any protective effects in sepsis. Therefore, we developed an Fc-modified KKO that does not induce these negative outcomes. Treatment with this antibody augmented the effects of PF4, decreasing NDP release and bacterial dissemination and increasing survival in murine sepsis models, supporting a novel NET-targeting approach to improve outcomes in sepsis.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Imunoglobulina G/uso terapêutico , Sepse/tratamento farmacológico , Animais , Anticorpos Monoclonais/química , Células Cultivadas , Modelos Animais de Doenças , Feminino , Heparina/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Imunoglobulina G/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator Plaquetário 4/genética , Fator Plaquetário 4/imunologia , Sepse/complicações , Sepse/imunologia , Trombocitopenia/induzido quimicamente , Trombocitopenia/complicações , Trombocitopenia/patologia , Trombocitopenia/terapia
4.
Blood ; 135(15): 1270-1280, 2020 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-32077913

RESUMO

Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder mediated by complexes between platelet factor 4 (PF4) and heparin or other polyanions, but the risk of thrombosis extends beyond exposure to heparin implicating other PF4 partners. We recently reported that peri-thrombus endothelium is targeted by HIT antibodies, but the binding site(s) has not been identified. We now show that PF4 binds at multiple discrete sites along the surface of extended strings of von Willebrand factor (VWF) released from the endothelium following photochemical injury in an endothelialized microfluidic system under flow. The HIT-like monoclonal antibody KKO and HIT patient antibodies recognize PF4-VWF complexes, promoting platelet adhesion and enlargement of thrombi within the microfluidic channels. Platelet adhesion to the PF4-VWF-HIT antibody complexes is inhibited by antibodies that block FcγRIIA or the glycoprotein Ib-IX complex on platelets. Disruption of PF4-VWF-HIT antibody complexes by drugs that prevent or block VWF oligomerization attenuate thrombus formation in a murine model of HIT. Together, these studies demonstrate assembly of HIT immune complexes along VWF strings released by injured endothelium that might propagate the risk of thrombosis in HIT. Disruption of PF4-VWF complex formation may provide a new therapeutic approach to HIT.


Assuntos
Anticorpos/imunologia , Anticoagulantes/efeitos adversos , Heparina/efeitos adversos , Fator Plaquetário 4/imunologia , Trombocitopenia/induzido quimicamente , Trombose/etiologia , Fator de von Willebrand/imunologia , Animais , Anticoagulantes/imunologia , Heparina/imunologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Adesividade Plaquetária , Trombocitopenia/complicações , Trombocitopenia/imunologia , Trombocitopenia/patologia , Trombose/imunologia , Trombose/patologia
5.
Blood ; 135(23): 2085-2093, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32187355

RESUMO

Thromboembolism complicates disorders caused by immunoglobulin G (IgG)-containing immune complexes (ICs), but the underlying mechanisms are incompletely understood. Prior evidence indicates that induction of tissue factor (TF) on monocytes, a pivotal step in the initiation, localization, and propagation of coagulation by ICs, is mediated through Fcγ receptor IIa (FcγRIIa); however, the involvement of other receptors has not been investigated in detail. The neonatal Fc receptor (FcRn) that mediates IgG and albumin recycling also participates in cellular responses to IgG-containing ICs. Here we asked whether FcRn is also involved in the induction of TF-dependent factor Xa (FXa) activity by IgG-containing ICs by THP-1 monocytic cells and human monocytes. Induction of FXa activity by ICs containing IgG antibodies to platelet factor 4 (PF4) involved in heparin-induced thrombocytopenia (HIT), ß-2-glycoprotein-1 implicated in antiphospholipid syndrome, or red blood cells coated with anti-(α)-Rh(D) antibodies that mediate hemolysis in vivo was inhibited by a humanized monoclonal antibody (mAb) that blocks IgG binding to human FcRn. IgG-containing ICs that bind to FcγR and FcRn induced FXa activity, whereas IgG-containing ICs with an Fc engineered to be unable to engage FcRn did not. Infusion of an α-FcRn mAb prevented fibrin deposition after microvascular injury in a murine model of HIT in which human FcγRIIa was expressed as a transgene. These data implicate FcRn in TF-dependent FXa activity induced by soluble and cell-associated IgG-containing ICs. Antibodies to FcRn, now in clinical trials in warm autoimmune hemolytic anemia to lower IgG antibodies and IgG containing ICs may also reduce the risk of venous thromboembolism.


Assuntos
Anticorpos Monoclonais Humanizados/imunologia , Heparina/toxicidade , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulina G/metabolismo , Receptores Fc/metabolismo , Trombocitopenia/imunologia , Tromboplastina/metabolismo , Animais , Anticoagulantes/toxicidade , Complexo Antígeno-Anticorpo , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Masculino , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Fator Plaquetário 4/genética , Fator Plaquetário 4/metabolismo , Receptores Fc/genética , Receptores Fc/imunologia , Trombocitopenia/induzido quimicamente , Trombocitopenia/metabolismo , Trombocitopenia/patologia
6.
J Autoimmun ; 107: 102355, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31732191

RESUMO

Systemic lupus erythematosus (SLE) is associated with a high risk of venous and arterial thrombosis, not necessarily associated with prothrombotic antiphospholipid antibodies (Abs). Alternatively, thrombosis may be due to an increased titer of anti-dsDNA Abs that presumably promote thrombosis via direct platelet activation. Here, we investigated effects of purified anti-dsDNA Abs from the blood of SLE patients, alone or in a complex with dsDNA, on isolated normal human platelets. We showed that anti-dsDNA Abs and anti-dsDNA Ab/dsDNA complexes induced strong platelet activation assessed by enhanced P-selectin expression and dramatic morphological and ultrastructural changes. Electron microscopy revealed a significantly higher percentage of platelets that lost their discoid shape, formed multiple filopodia and had a shrunken body when treated with anti-dsDNA Abs or anti-dsDNA Ab/dsDNA complexes compared with control samples. In addition, these platelets activated with anti-dsDNA Ab/dsDNA complexes typically contained a reduced number of secretory α-granules that grouped in the middle and often merged into a solid electron dense area. Many activated platelets released plasma membrane-derived microvesicles and/or fell apart into subcellular cytoplasmic fragments. Confocal microscopy revealed that platelets treated with anti-dsDNA Ab/dsDNA complex had a heterogeneous distribution of septin2 compared with the homogeneous distribution in control platelets. Structural perturbations were concomitant with mitochondrial depolarization and a decreased content of platelet ATP, indicating energetic exhaustion. Most of the biochemical and morphological changes in platelets induced by anti-dsDNA Abs and anti-dsDNA Ab/dsDNA complexes were prevented by pre-treatment with a monoclonal mAb against FcγRIIA. The aggregate of data indicates that anti-dsDNA Abs alone or in a complex with dsDNA strongly affect platelets via the FcγRIIA receptor. The immune activation of platelets with antinuclear Abs may comprise a prothrombotic mechanism underlying a high risk of thrombotic complications in patients with SLE.


Assuntos
Anticorpos Antinucleares/imunologia , Plaquetas/imunologia , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/imunologia , Ativação Plaquetária/imunologia , Trombose/etiologia , Anticorpos Antinucleares/sangue , Autoantígenos/imunologia , Autoimunidade , Plaquetas/metabolismo , DNA/imunologia , Humanos , Lúpus Eritematoso Sistêmico/metabolismo , Trombose/diagnóstico , Trombose/metabolismo
7.
Blood ; 132(7): 727-734, 2018 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-29914979

RESUMO

Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder initiated by antibodies to platelet factor 4 (PF4)/heparin complexes. PF4 released from platelets binds to surface glycosaminoglycans on hematopoietic and vascular cells that are heterogenous in composition and differ in affinity for PF4. PF4 binds to monocytes with higher affinity than to platelets, and depletion of monocytes exacerbates thrombocytopenia in a murine HIT model. Here we show that the expression of PF4 on platelets and development of thrombocytopenia are modulated by the (re)distribution of PF4 among hematopoietic and endothelial cell surfaces. Binding of PF4 to platelets in whole blood in vitro varies inversely with the white cell count, likely because of the greater affinity of monocytes for PF4. In mice, monocyte depletion increased binding of PF4 to platelets by two- to three-fold. Induction of HIT in mice caused a transient >80-fold increase in binding of HIT antibody to monocytes vs 3.5-fold increase to platelets and rapid transient monocytopenia. Normalization of monocyte counts preceded the return in platelet counts. Exposure of blood to endothelial cells also depletes PF4 from platelet surfaces. These studies demonstrate a dynamic interchange of surface-bound PF4 among hematopoetic and vascular cells that may limit thrombocytopenia at the expense of promoting prothrombotic processes in HIT.


Assuntos
Antígenos/biossíntese , Plaquetas/metabolismo , Heparina/efeitos adversos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Monócitos/metabolismo , Fator Plaquetário 4/biossíntese , Trombocitopenia/metabolismo , Animais , Plaquetas/patologia , Regulação da Expressão Gênica , Heparina/uso terapêutico , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Camundongos , Monócitos/patologia , Trombocitopenia/induzido quimicamente , Trombocitopenia/patologia
8.
Blood ; 132(23): 2431-2440, 2018 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-30309891

RESUMO

The mechanisms by which exposure to heparin initiates antibody responses in many, if not most, recipients are poorly understood. We recently demonstrated that antigenic platelet factor 4 (PF4)/heparin complexes activate complement in plasma and bind to B cells. Here, we describe how this process is initiated. We observed wide stable variation in complement activation when PF4/heparin was added to plasma of healthy donors, indicating a responder "phenotype" (high, intermediate, or low). Proteomic analysis of plasma from these healthy donors showed a strong correlation between complement activation and plasma immunoglobulin M (IgM) levels (r = 0.898; P < .005), but not other Ig isotypes. Complement activation response to PF4/heparin in plasma displaying the low donor phenotype was enhanced by adding pooled IgM from healthy donors, but not monoclonal IgM. Depletion of IgM from plasma abrogated C3c generation by PF4/heparin. The complement-activating features of IgM are likely mediated by nonimmune, or natural, IgM, as cord blood and a monoclonal polyreactive IgM generate C3c in the presence of PF4/heparin. IgM facilitates complement and antigen deposition on B cells in vitro and in patients receiving heparin. Anti-C1q antibody prevents IgM-mediated complement activation by PF4/heparin complexes, indicating classical pathway involvement. These studies demonstrate that variability in plasma IgM levels correlates with functional complement responses to PF4/heparin. Polyreactive IgM binds PF4/heparin, triggers activation of the classical complement pathway, and promotes antigen and complement deposition on B cells. These studies provide new insights into the evolution of the heparin-induced thrombocytopenia immune response and may provide a biomarker of risk.


Assuntos
Linfócitos B/imunologia , Via Clássica do Complemento/imunologia , Heparina/imunologia , Imunoglobulina M/imunologia , Ativação Linfocitária , Fator Plaquetário 4/imunologia , Complemento C3c/imunologia , Via Clássica do Complemento/efeitos dos fármacos , Heparina/farmacologia , Humanos , Fator Plaquetário 4/farmacologia , Proteômica
9.
Int J Mol Sci ; 21(7)2020 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-32272655

RESUMO

Heparin-induced thrombocytopenia (HIT) is an adverse drug reaction characterized by thrombocytopenia and a high risk for venous or arterial thrombosis. HIT is caused by antibodies that recognize complexes of platelet factor 4 and heparin. The pathogenic mechanisms of this condition are not fully understood. In this study, we used flow cytometry, fluorimetry, and Western blot analysis to study the direct effects of pathogenic immune complexes containing platelet factor 4 on human platelets isolated by gel-filtration. HIT-like pathogenic immune complexes initially caused pronounced activation of platelets detected by an increased expression of phosphatidylserine and P-selectin. This activation was mediated either directly through the FcγRIIA receptors or indirectly via protease-activated receptor 1 (PAR1) receptors due to thrombin generated on or near the surface of activated platelets. The immune activation was later followed by the biochemical signs of cell death, such as mitochondrial membrane depolarization, up-regulation of Bax, down-regulation of Bcl-XL, and moderate activation of procaspase 3 and increased calpain activity. The results show that platelet activation under the action of HIT-like immune complexes is accompanied by their death through complex apoptotic and calpain-dependent non-apoptotic pathways that may underlie the low platelet count in HIT.


Assuntos
Apoptose/fisiologia , Plaquetas/fisiologia , Heparina/fisiologia , Ativação Plaquetária/fisiologia , Transdução de Sinais/fisiologia , Trombocitopenia/induzido quimicamente , Anticoagulantes/farmacologia , Apoptose/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Humanos , Receptores de IgG/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trombocitopenia/tratamento farmacológico , Trombocitopenia/metabolismo , Trombose/tratamento farmacológico , Trombose/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
10.
Blood ; 140(5): 413-414, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35925641
11.
Blood ; 130(6): 817-828, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28576878

RESUMO

Several CGHC motif-containing disulfide isomerases support thrombosis. We here report that endoplasmic reticulum protein 72 (ERp72), with 3 CGHC redox-active sites (ao, a, and a'), supports thrombosis. We generated a new conditional knockout mouse model and found that Tie2-Cre/ERp72fl/fl mice with blood and endothelial cells lacking ERp72 had prolonged tail bleeding times and decreased platelet accumulation in laser-induced cremaster arteriole injury and FeCl3-induced mesenteric arterial injury. Fibrin deposition was decreased in the laser injury model. Both platelet and fibrin accumulation defects were fully rescued by infusion of recombinant ERp72 containing functional a and a' CGHC motifs (ERp72(oo-ss-ss)). Infusion of ERp72 containing inactivated a and a' CGHC motifs (ERp72(ss-oo-oo)) inhibited platelet accumulation and fibrin deposition in wild-type mice. Infusion of ERp72(oo-ss-ss) into ß3-null mice increased fibrin deposition in the absence of platelets. ERp72-null platelets had defective aggregation, JON/A binding, P-selectin expression, and adenosine triphosphate (ATP) secretion. The aggregation and ATP secretion defects were fully rescued by ERp72(oo-ss-ss) but partially rescued by ERp72(ss-oo-ss) and ERp72(ss-ss-oo). Aggregation and ATP secretion of human platelets was potentiated by ERp72(oo-ss-ss) but inhibited by ERp72(ss-oo-ss) and ERp72(ss-ss-oo). These data suggest that both the a and a' active sites are required for platelet function. ERp72 bound poorly to ß3-null mouse platelets, and the addition of ERp72(oo-ss-ss) to human platelets generated thiols in αIIbß3, suggesting a direct interaction of ERp72 with αIIbß3. Defective aggregation of ERp72-null platelets was recovered by ERp72, but not other thiol isomerases. In summary, ERp72 plays a critical role in platelet function and coagulation through the a and a' CGHC motifs.


Assuntos
Plaquetas/metabolismo , Glicoproteínas de Membrana/metabolismo , Trombose/metabolismo , Animais , Plaquetas/patologia , Humanos , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Ativação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombose/genética , Trombose/patologia
12.
Blood ; 138(4): 285-286, 2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34323943
13.
Blood ; 128(14): 1789-1799, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27412887

RESUMO

Heparin-induced thrombocytopenia is a prothrombotic disorder caused by antibodies to platelet factor 4 (PF4)/heparin complexes. The mechanism that incites such prevalent anti-PF4/heparin antibody production in more than 50% of patients exposed to heparin in some clinical settings is poorly understood. To investigate early events associated with antigen exposure, we first examined the interaction of PF4/heparin complexes with cells circulating in whole blood. In healthy donors, PF4/heparin complexes bind preferentially to B cells (>90% of B cells bind to PF4/heparin in vitro) relative to neutrophils, monocytes, or T cells. Binding of PF4 to B cells is heparin dependent, and PF4/heparin complexes are found on circulating B cells from some, but not all, patients receiving heparin. Given the high proportion of B cells that bind PF4/heparin, we investigated complement as a mechanism for noncognate antigen recognition. Complement is activated by PF4/heparin complexes, co-localizes with antigen on B cells from healthy donors, and is present on antigen-positive B cells in patients receiving heparin. Binding of PF4/heparin complexes to B cells is mediated through the interaction between complement and complement receptor 2 (CR2 [CD21]). To the best of our knowledge, these are the first studies to demonstrate complement activation by PF4/heparin complexes, opsonization of PF4/heparin to B cells via CD21, and the presence of complement activation fragments on circulating B cells in some patients receiving heparin. Given the critical contribution of complement to humoral immunity, our observations provide new mechanistic insights into the immunogenicity of PF4/heparin complexes.


Assuntos
Antígenos/metabolismo , Linfócitos B/metabolismo , Proteínas do Sistema Complemento/metabolismo , Heparina/efeitos adversos , Receptores de Complemento 3d/metabolismo , Trombocitopenia/induzido quimicamente , Trombocitopenia/imunologia , Heparina/uso terapêutico , Humanos , Modelos Biológicos , Fator Plaquetário 4/metabolismo , Protaminas/metabolismo , Ligação Proteica , Trombocitopenia/sangue
14.
Blood ; 127(4): 464-72, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26518435

RESUMO

Heparin-induced thrombocytopenia (HIT) is characterized by a high incidence of thrombosis, unlike other antibody-mediated causes of thrombocytopenia. We have shown that monocytes complexed with surface-bound platelet factor 4 (PF4) activated by HIT antibodies contribute to the prothrombotic state in vivo, but the mechanism by which this occurs and the relationship to the requirement for platelet activation via fragment crystallizable (Fc)γRIIA is uncertain. Using a microfluidic model and human or murine blood, we confirmed that activation of monocytes contributes to the prothrombotic state in HIT and showed that HIT antibodies bind to monocyte FcγRIIA, which activates spleen tyrosine kinase and leads to the generation of tissue factor (TF) and thrombin. The combination of direct platelet activation by HIT immune complexes through FcγRIIA and transactivation by monocyte-derived thrombin markedly increases Annexin V and factor Xa binding to platelets, consistent with the formation of procoagulant coated platelets. These data provide a model of HIT wherein a combination of direct FcγRIIA-mediated platelet activation and monocyte-derived thrombin contributes to thrombosis in HIT and identifies potential new targets for lessening this risk.


Assuntos
Anticoagulantes/efeitos adversos , Plaquetas/imunologia , Heparina/efeitos adversos , Monócitos/imunologia , Trombocitopenia/induzido quimicamente , Trombocitopenia/complicações , Trombose/etiologia , Animais , Anticoagulantes/imunologia , Células Cultivadas , Heparina/imunologia , Humanos , Camundongos , Técnicas Analíticas Microfluídicas , Ativação Plaquetária , Fator Plaquetário 4/imunologia , Receptores de IgG/imunologia , Trombocitopenia/imunologia , Trombose/imunologia
15.
Blood ; 123(10): 1596-603, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24335500

RESUMO

Contraction of blood clots is necessary for hemostasis and wound healing and to restore flow past obstructive thrombi, but little is known about the structure of contracted clots or the role of erythrocytes in contraction. We found that contracted blood clots develop a remarkable structure, with a meshwork of fibrin and platelet aggregates on the exterior of the clot and a close-packed, tessellated array of compressed polyhedral erythrocytes within. The same results were obtained after initiation of clotting with various activators and also with clots from reconstituted human blood and mouse blood. Such close-packed arrays of polyhedral erythrocytes, or polyhedrocytes, were also observed in human arterial thrombi taken from patients. The mechanical nature of this shape change was confirmed by polyhedrocyte formation from the forces of centrifugation of blood without clotting. Platelets (with their cytoskeletal motility proteins) and fibrin(ogen) (as the substrate bridging platelets for contraction) are required to generate the forces necessary to segregate platelets/fibrin from erythrocytes and to compress erythrocytes into a tightly packed array. These results demonstrate how contracted clots form an impermeable barrier important for hemostasis and wound healing and help explain how fibrinolysis is greatly retarded as clots contract.


Assuntos
Coagulação Sanguínea/fisiologia , Animais , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/metabolismo , Vasos Coronários/patologia , Deutério/metabolismo , Eritrócitos/metabolismo , Eritrócitos/ultraestrutura , Fibrina/metabolismo , Humanos , Hidrogênio/metabolismo , Camundongos , Agregação Plaquetária , Trombose/metabolismo
16.
Blood ; 121(10): 1858-67, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23264598

RESUMO

The integrin family is composed of a series of 24 αß heterodimer transmembrane adhesion receptors that mediate cell-cell and cell-extracellular matrix interactions. Adaptor molecules bearing immunoreceptor tyrosine-based activation motifs (ITAMs) have recently been shown to cooperate with specific integrins to increase the efficiency of transmitting ligand-binding-induced signals into cells. In human platelets, Fc receptor γ-chain IIa (FcγRIIa) has been identified as an ITAM-bearing transmembrane receptor responsible for mediating "outside-in" signaling through αIIbß3, the major adhesion receptor on the platelet surface. To explore the importance of FcγRIIa in thrombosis and hemostasis, we subjected FcγRIIa-negative and FcγRIIa-positive murine platelets to a number of well-accepted models of platelet function. Compared with their FcγRIIa-negative counterparts, FcγRIIa-positive platelets exhibited increased tyrosine phosphorylation of Syk and phospholipase Cγ2 and increased spreading upon interaction with immobilized fibrinogen, retracted a fibrin clot faster, and showed markedly enhanced thrombus formation when perfused over a collagen-coated flow chamber under conditions of arterial and venous shear. They also displayed increased thrombus formation and fibrin deposition in in vivo models of vascular injury. Taken together, these data establish FcγRIIa as a physiologically important functional conduit for αIIbß3-mediated outside-in signaling, and suggest that modulating the activity of this novel integrin/ITAM pair might be effective in controlling thrombosis.


Assuntos
Plaquetas/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Receptores de IgG/fisiologia , Receptores Imunológicos/metabolismo , Transdução de Sinais , Trombose/etiologia , Tirosina/metabolismo , Animais , Arteríolas/metabolismo , Arteríolas/patologia , Fibrina/metabolismo , Fibrinogênio/metabolismo , Hemostasia/fisiologia , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Agregação Plaquetária , Contagem de Plaquetas , Trombose/metabolismo , Trombose/patologia , Veias/metabolismo , Veias/patologia
17.
J Biol Chem ; 288(46): 33060-70, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24097975

RESUMO

Heparin-induced thrombocytopenia (HIT) is a thrombotic complication of heparin therapy mediated by antibodies to complexes between platelet factor 4 (PF4) and heparin or cellular glycosaminoglycans. However, only a fraction of patients with anti-PF4-heparin antibodies develop HIT, implying that only a subset of these antibodies is pathogenic. The basis for the pathogenic potential of anti-PF4-heparin antibodies remains unclear. To elucidate the intrinsic PF4-binding properties of HIT-like monoclonal antibody (KKO) versus non-pathogenic antibody (RTO) at the single-molecule level, we utilized optical trap-based force spectroscopy to measure the strength and probability of binding of surface-attached antibodies with oligomeric PF4 to simulate interactions on cells. To mimic the effect of heparin in bringing PF4 complexes into proximity, we chemically cross-linked PF4 tetramers using glutaraldehyde. Analysis of the force histograms revealed that KKO-PF4 interactions had ∼10-fold faster on-rates than RTO-PF4, and apparent equilibrium dissociation constants differed ∼10-fold with similar force-free off-rates (k(off) = 0.0031 and 0.0029 s(-1)). Qualitatively similar results were obtained for KKO and RTO interacting with PF4-heparin complexes. In contrast to WT PF4, KKO and RTO showed lower and similar binding probabilities to cross-linked PF4(K50E), which forms few if any oligomers. Thus, formation of stable PF4 polymers results in much stronger interactions with the pathogenic antibody without a significant effect on the binding of the non-pathogenic antibody. These results suggest a fundamental difference in the antigen-binding mechanisms between model pathogenic and non-pathogenic anti-PF4 antibodies that might underlie their distinct pathophysiological behaviors.


Assuntos
Anticorpos Monoclonais/química , Especificidade de Anticorpos , Anticoagulantes/química , Autoanticorpos/química , Heparina/química , Fator Plaquetário 4/química , Anticorpos Monoclonais/imunologia , Anticoagulantes/efeitos adversos , Anticoagulantes/imunologia , Anticoagulantes/uso terapêutico , Autoanticorpos/imunologia , Sítios de Ligação de Anticorpos , Heparina/efeitos adversos , Heparina/imunologia , Heparina/uso terapêutico , Humanos , Cinética , Fator Plaquetário 4/imunologia , Trombocitopenia/induzido quimicamente , Trombocitopenia/imunologia
18.
Clin Chem ; 60(9): 1174-82, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24958814

RESUMO

BACKGROUND: Existing approaches for measuring hemostasis parameters require multiple platforms, can take hours to provide results, and generally require 1-25 mL of sample. We developed a diagnostic platform that allows comprehensive assessment of hemostatic parameters on a single instrument and provides results within 15 min using 0.04 mL of blood with minimal sample handling. METHODS: T2 magnetic resonance (T2MR) was used to directly measure integrated reactions in whole blood samples by resolving multiple water relaxation times from distinct sample microenvironments. Clotting, clot contraction, and fibrinolysis stimulated by thrombin or tissue plasminogen activator, respectively, were measured. T2MR signals of clotting samples were compared with images produced by scanning electron microscopy and with standard reference methods for the following parameters: hematocrit, prothrombin time, clot strength, and platelet activity. RESULTS: Application of T2MR methodology revealed conditions under which a unique T2MR signature appeared that corresponded with the formation of polyhedral erythrocytes, the dynamics and morphology of which are dependent on thrombin, fibrinogen, hematocrit, and platelet levels. We also showed that the T2MR platform can be used for precise and accurate measurements of hematocrit (%CV, 4.8%, R(2) = 0.95), clotting time (%CV, 3.5%, R(2) = 0.94), clot strength (R(2) = 0.95), and platelet function (93% agreement with light transmission aggregometry). CONCLUSIONS: This proof-of-concept study demonstrates that T2MR has the potential to provide rapid and sensitive identification of patients at risk for thrombosis or bleeding and to identify new biomarkers and therapeutic targets with a single, simple-to-employ analytic approach that may be suitable for routine use in both research and diverse clinical settings.


Assuntos
Doenças Hematológicas/sangue , Hemostasia , Espectroscopia de Ressonância Magnética , Imagem Corporal Total , Coagulação Sanguínea/fisiologia , Hematócrito , Humanos , Sensibilidade e Especificidade , Fatores de Tempo
19.
Blood ; 119(8): 1798-9, 2012 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-22361971

RESUMO

In this issue of Blood, Nishimura and colleagues present a novel in vivo murine model of thrombosis triggered on the undisrupted, activated endothelium in response to reactive oxygen species (ROS)­producing free radicals derived from oxygen (namely O(2)(-)), which can be present in the vasculature as a result of inflammation (see figure). This new in vivo model involving a novel injury, minimally invasive vessel preparation, and advanced data collection may be particularly useful in dissecting the role of inflammation in thrombosis.


Assuntos
Endotélio Vascular/metabolismo , Interleucina-1/metabolismo , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Animais , Humanos , Masculino
20.
Blood ; 120(5): 1137-42, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22577175

RESUMO

Rapid laboratory assessment of heparin-induced thrombocytopenia (HIT) is important for disease recognition and management. The utility of contemporary immunoassays to detect antiplatelet factor 4 (PF4)/heparin antibodies is hindered by detection of antibodies unassociated with disease. To begin to distinguish properties of pathogenic anti-PF4/heparin antibodies, we compared isotype-matched monoclonal antibodies that bind to different epitopes: KKO causes thrombocytopenia in an in vivo model of HIT, whereas RTO does not. KKO binding to PF4 and heparin is specifically inhibited by human HIT antibodies that activate platelets, whereas inhibition of RTO binding is not differentially affected. Heparin increased the avidity of KKO binding to PF4 without affecting RTO, but it did not increase total binding or binding to nontetrameric PF4(K50E). Single-molecule forced unbinding demonstrated KKO was 8-fold more reactive toward PF4 tetramers and formed stronger complexes than RTO, but not to PF4(K50E) dimers. KKO, but not RTO, promoted oligomerization of PF4 but not PF4(K50E). This study reveals differences in the properties of anti-PF4 antibodies that cause thrombocytopenia not revealed by ELISA that correlate with oligomerization of PF4 and sustained high-avidity interactions that may simulate transient antibody-antigen interactions in vivo. These differences suggest the potential importance of epitope specificity in the pathogenesis of HIT.


Assuntos
Anticorpos/metabolismo , Afinidade de Anticorpos/fisiologia , Trombocitopenia/etiologia , Trombocitopenia/imunologia , Animais , Anticorpos Imobilizados/metabolismo , Anticorpos Monoclonais/metabolismo , Afinidade de Anticorpos/genética , Reações Antígeno-Anticorpo/genética , Reações Antígeno-Anticorpo/fisiologia , Linhagem Celular Tumoral , Drosophila , Ensaio de Imunoadsorção Enzimática , Heparina/efeitos adversos , Humanos , Cinética , Camundongos , Camundongos Transgênicos , Fator Plaquetário 4/genética , Fator Plaquetário 4/imunologia , Ligação Proteica/fisiologia , Trombocitopenia/genética , Trombocitopenia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA