Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Scand J Immunol ; 88(1): e12678, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29855061

RESUMO

Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder characterized by increased peripheral immune platelet destruction and megakaryocyte defects in the bone marrow. Although ITP was originally thought to be primarily due to antibody-mediated autoimmunity, it is now clear that T cells also play a significant role in the disease. However, the exact interplay between platelet destruction, megakaryocyte dysfunction and the elements of both humoral and cell-mediated immunity in ITP remains incompletely defined. While most studies have focused on immune platelet destruction in the spleen, an additional possibility is that the antiplatelet antibodies can also destroy bone marrow megakaryocytes. To address this, we negated the effects of T cells by utilizing an in vivo passive ITP model where BALB/c mice were administered various anti-αIIb, anti-ß3 or anti-GPIb antibodies or antisera and platelet counts and bone marrow megakaryocytes were enumerated. Our results show that after 24 hours, all the different antiplatelet antibodies/sera induced variable degrees of thrombocytopenia in recipient mice. Compared with naïve control mice, however, histological examination of the bone marrow revealed that only 2 antibody preparations (mouse-anti-mouse ß3 sera and an anti- αIIb monoclonal antibody (MWReg30) could affect bone marrow megakaryocyte counts. Our study shows that while most antiplatelet antibodies induce acute thrombocytopenia, the majority of them do not affect the number of megakaryocytes in the bone marrow. This suggests that other mechanisms may be responsible for megakaryocyte abnormalities seen during immune thrombocytopenia.


Assuntos
Autoanticorpos/imunologia , Plaquetas/imunologia , Megacariócitos/patologia , Púrpura Trombocitopênica Idiopática/imunologia , Púrpura Trombocitopênica Idiopática/patologia , Animais , Células da Medula Óssea/patologia , Camundongos , Camundongos Endogâmicos BALB C
2.
Vox Sang ; 98(3 Pt 2): 385-94, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19821958

RESUMO

The increase in platelets in patients with immune thrombocytopenia (ITP) by intravenous administration of human immunoglobulin concentrates (IVIG) reflects a therapeutic immunomodulatory intervention targeted at the disturbed immune response in many inflammatory and autoimmune disorders. These immunoglobulin concentrates contain large numbers of antibodies as well as trace levels of various other immunologically active molecules. Clinical and laboratory studies have documented various mechanisms of action of IVIG. The complex network of immunological reactions resulting from the infusion of IVIG includes changes in several cytokines, interactions with dendritic cells, T- and B- lymphocyte effects, macrophage effects, mediated by distinct Fc-gamma receptors. In addition, effects on complement components and apoptosis have also been observed. Synergism between the different elements of the immune response characterizes the beneficial effects of IVIG in inflammatory and autoimmune disorders. They have immunopathogeneses and clinical manifestations which are difficult to define and therefore IVIG treatment indications remain heterogeneous. Dose finding studies are missing for most of the indications of the drug. In future research, defining the appropriate subgroups of patients should be undertaken. This may be accomplished by prospective registries collecting data on large numbers of patients with long-term follow-up. Controlled clinical and laboratory studies may follow based on new, validated patient selection criteria and focused on mechanisms of action, leading to more evidence-based indications.


Assuntos
Doenças Autoimunes/terapia , Imunoglobulinas Intravenosas/farmacologia , Fatores Imunológicos/farmacologia , Doenças Autoimunes/imunologia , Citocinas/fisiologia , Células Dendríticas/imunologia , Previsões , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Inflamação/imunologia , Inflamação/terapia , Subpopulações de Linfócitos/imunologia , Macrófagos/imunologia , Fagocitose/efeitos dos fármacos , Receptores de IgG/fisiologia
3.
J Thromb Haemost ; 7 Suppl 1: 245-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19630809

RESUMO

Intravenous immunoglobulin (IVIg) is used to treat an ever-increasing number of autoimmune diseases. While the exact mechanism of action of IVIg has remained elusive, many theories have been suggested, including mononuclear phagocytic system blockade, autoantibody neutralization by anti-idiotype antibodies, accelerated pathogenic autoantibody clearance by saturation of the neonatal Fc receptor, cytokine modulation and complement neutralization. More recently, a key role for dendritic cells (DC) in the amelioration of autoimmunity by IVIg has been suggested. Here we will focus on the role that DC may play in IVIg function using data from both mouse and human studies.


Assuntos
Células Dendríticas/imunologia , Imunoglobulinas Intravenosas/farmacologia , Animais , Autoimunidade , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Camundongos
4.
J Thromb Haemost ; 1(2): 337-46, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12871509

RESUMO

The interaction between von Willebrand factor (VWF) and glycoprotein (GP) Ib results in platelet agglutination and activation of many signaling intermediates. To determine if VWF-dependent platelet activation requires the participation of pivotal transmembrane signaling pathways, we analyzed VWF-dependent platelet activation profiles following inhibition of several transmembrane signaling intermediates. This was accomplished using porcine VWF, which has been shown to interact with human GPIb independently of shear stress or ristocetin. Platelet alpha (CD62) and lysozomal granule release (CD63), microparticle formation, and platelet agglutination/aggregation were evaluated. The ability of signaling inhibitors to prevent VWF-dependent platelet activation was compared to their ability to inhibit thrombin-dependent activation. The results demonstrate that VWF-dependent platelet activation can occur independently of the activities of protein kinase C (PKC), wortmannin-sensitive phosphatidylinositide 3-kinase, and phospholipase C, as well as independently of elevations in the concentration of intracellular calcium. In sharp contrast, these transmembrane signaling intermediates are required for thrombin-dependent platelet activation. In addition, thrombin-dependent but not VWF-dependent platelet activation was associated with elevations in the concentration of intracellular calcium under the conditions used. The family of signaling intermediates which appeared to be pivotal for both thrombin- and VWF-dependent platelet activation were the protein tyrosine phosphatases and the serine/threonine phosphatases. It is concluded that thrombin-dependent platelet activation relies on the activation of several transmembrane signaling pathways, whereas VWF-dependent platelet activation is dependent upon the activity of protein phosphatases. Inhibition of these phosphatases in vivo may provide a novel therapeutic approach for treating VWF-dependent platelet disorders such as thrombotic thrombocytopenic purpura or arterial thrombosis.


Assuntos
Ativação Plaquetária/fisiologia , Fator de von Willebrand/fisiologia , Difosfato de Adenosina/farmacologia , Animais , Antígenos CD/sangue , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Cálcio/sangue , Membrana Celular/fisiologia , Humanos , Técnicas In Vitro , Selectina-P/sangue , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/sangue , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/fisiologia , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas , Transdução de Sinais , Especificidade da Espécie , Sus scrofa , Tetraspanina 30 , Trombina/farmacologia , Trombina/fisiologia , Fator de von Willebrand/farmacologia
6.
Am J Hematol ; 69(3): 192-9, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11891806

RESUMO

Activation of platelets and coagulation in vivo was studied in nine patients with hemophilia A and inhibitors to human Factor VIII, prior to and following treatment with porcine Factor VIII (PFVIII; HYATE:C). In addition, six hemophiliac patients were similarly studied after treatment with recombinant Factor VIII (rFVIII). Platelet activation was also examined in vitro using porcine von Willebrand factor (PvWF)-enriched and PvWF-depleted fractions obtained by fractionation of PFVIII. Coagulation was assessed by measuring the concentrations of plasma prothrombin fragment 1+2 concentrations (prothrombinase generation) and Factor Xa-ATIII. Patients treated with PFVIII had significantly increased numbers of circulating platelets expressing CD62 and CD63 (markers of platelet activation) and annexin V (marker of platelet procoagulant activity) compared to patients treated with rFVIII; the former patients also demonstrated an increase in plasma coagulability after therapy. In in vitro experiments it was observed that the platelet-activating and procoagulant capacity of PFVIII resided in the PvWF-enriched fraction, and the same was true for the plasma hypercoagulability following exposure of platelets to PFVIII. These results support the hypothesis that PFVIII-induced platelet activation provides a mechanism for enhancing hemostasis, separate from, and additional to, that due to increased circulating Factor VIII, and it is due to residual PvWF in the PFVIII preparation.


Assuntos
Fator VIII/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Trombofilia/induzido quimicamente , Adolescente , Adulto , Animais , Plaquetas/imunologia , Plaquetas/metabolismo , Estudos de Casos e Controles , Criança , Contaminação de Medicamentos , Fator VIII/administração & dosagem , Fator VIII/normas , Hemofilia A/sangue , Hemofilia A/complicações , Hemofilia A/tratamento farmacológico , Hemostasia/efeitos dos fármacos , Humanos , Imunofenotipagem , Cinética , Pessoa de Meia-Idade , Suínos , Trombofilia/sangue , Fator de von Willebrand/análise , Fator de von Willebrand/farmacologia
7.
Br J Haematol ; 115(3): 679-86, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11736954

RESUMO

Although the mechanism of action of intravenous immunoglobulin (IVIg) in treating antibody-dependent thrombocytopenia remains unclear, most studies have suggested that IVIg blocks the function of Fc receptors in the reticuloendothelial system (RES) and/or the protective effect may be due to the presence of variable region-reactive (anti-idiotype) antibodies within IVIg. We evaluated the effect of IVIg on platelet counts in a murine model of passively induced immune thrombocytopenia (PIT). Although IVIg was unable to neutralize the binding of two platelet-specific monoclonal antibodies to their target antigens either in vivo or in vitro, it was able to prevent PIT as well as ameliorate pre-established PIT mediated by these antibodies. IVIg adsorbed against the antibody used to induce thrombocytopenia or endogenous murine immunoglobulin also protected against PIT, indicating that antibodies with anti-idiotype activity present in IVIg are not necessary for its effective treatment of PIT. IVIg significantly blocked the ability of the RES to clear antibody-sensitized red blood cells. F(ab')2 fragments of IVIg, which are unable to block the RES but retain the idiotypic regions, were ineffective at protecting mice from PIT. Our data suggest that IVIg exerts its rapid effect by inhibiting RES function and that anti-idiotype interactions are not required.


Assuntos
Imunoglobulinas Intravenosas/uso terapêutico , Sistema Fagocitário Mononuclear/imunologia , Trombocitopenia/imunologia , Trombocitopenia/terapia , Adsorção , Animais , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Monoclonais/imunologia , Feminino , Fragmentos Fab das Imunoglobulinas/imunologia , Camundongos , Camundongos SCID , Modelos Animais , Contagem de Plaquetas , Estatísticas não Paramétricas , Trombocitopenia/prevenção & controle
8.
Br J Haematol ; 110(2): 481-7, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10971412

RESUMO

Presensitization of donor platelets with allo-specific immunoglobulin (Ig)G results in a diminished immune response against subsequent transfusions of platelets. To understand better the mechanism of how alloantibody presensitization results in a decreased alloimmune response, we have used murine monoclonal antibodies directed to polymorphic and non-polymorphic regions of human leucocyte antigen (HLA) as well as platelet-specific molecules. Here, we demonstrated that presensitization with anti-human HLA class I antibodies, as well as beta2-microglobulin-specific antibody, protected against alloantibody production to five subsequent untreated platelet challenges. Use of complement fixing, non-fixing or F(ab')2 fragments of HLA-specific antibody also resulted in complete inhibition of alloantibody production. This protection was not seen when the platelets were presensitized with monoclonal antibodies to CD42a (GPIX), CD32 (low-affinity IgG/Fcgamma receptor) or murine IgG and was thus independent of B-cell FcgammaRII-mediated immune suppression. The inhibition observed was independent of HLA alloantigenic specificity as antibodies directed at the beta2-microglobulin portion of HLA class I were as effective as antibodies against any of the HLA-alpha regions (either polymorphic or non-polymorphic) of class I. This work demonstrates that monoclonal antibody-mediated suppression of the human HLA alloimmune response to platelet transfusion is antigen specific and is independent of FcgammaRII-mediated immune regulation, complement fixing or HLA alloantigenic specificity.


Assuntos
Anticorpos Monoclonais/fisiologia , Autoimunidade/fisiologia , Antígenos HLA/imunologia , Tolerância Imunológica/fisiologia , Transfusão de Plaquetas , Receptores de IgG/imunologia , Animais , Feminino , Humanos , Camundongos , Camundongos SCID
9.
Br J Haematol ; 109(4): 851-6, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10929041

RESUMO

The interaction of platelets with subendothelial von Willebrand factor (VWF), especially under high shear stress, is considered to be the first activation step which primes platelets for subsequent haemostatic events. The signalling cascade which results from the interaction of VWF and its receptor GPIbIX has only been partially defined. Mitogen-activated protein kinases (MAPKs) are a family of downstream transmembrane signalling serine-threonine kinases and have been demonstrated to be present and functional in platelets; these include the extracellular signal-related kinases (ERKs), c-Jun amino-terminal kinases (JNKs) and p38 MAPK. Previously, we showed that p38 MAPK was not required in VWF-induced human platelet activation. It is not known whether VWF-dependent platelet activation involves the activation of the JNK and ERK family of signalling molecules. This report demonstrates that porcine von Willebrand factor (pVWF) induced a sustained and stable JNK activation measurable by 1 min after activation. Thrombin also induced JNK activation assessed at 1 min after activation. In contrast to thrombin, pVWF did not induce ERK2 activation at any time point tested. To ensure that ERK activation was unnecessary for pVWF-dependent platelet activation, we functionally inhibited ERK-dependent signalling with PD98059, a potent and selective inhibitor of the MAP kinase kinase (MEK-1), which is the upstream kinase of ERK1 and ERK2. Although PD98059 inhibited ERK2 activation in platelets, it had no effect on pVWF- or thrombin-induced platelet alpha or lysozomal granule release, modulation of membrane glycoprotein CD41, microparticle formation, platelet shape change or platelet agglutination. It is concluded that pVWF and thrombin induced JNK activation, but whereas thrombin induced ERK2 activation VWF did not; functional ERK2 activity was also not required for pVWF- or thrombin-dependent platelet activation.


Assuntos
Plaquetas/enzimologia , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Trombina/farmacologia , Fator de von Willebrand/farmacologia , Animais , Antígenos CD/metabolismo , Plaquetas/efeitos dos fármacos , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Humanos , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Glicoproteínas da Membrana de Plaquetas/metabolismo , Estimulação Química , Suínos , Tetraspanina 30
10.
Transfus Sci ; 22(1-2): 19-27, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10771375

RESUMO

Idiopathic thrombocytopenic purpura (ITP) is characterized by the development of a specific anti-platelet autoantibody immune response mediating the development of thrombocytopenia. Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of a wide variety of autoantibodies. In 15-20% of SLE cases, patients develop thrombocytopenia which appears to be autoimmune in nature (SLE-TP). To better understand the pathogenesis of the thrombocytopenia associated with SLE, we investigated the overlapping platelet and cellular immune features between SLE and ITP. Thirty-one patients with SLE, eight with SLE-TP, and 17 with ITP, were studied and compared to 60 healthy controls. We evaluated platelet-associated IgG, platelet microparticles, reticulated platelets, platelet HLA-DR expression, in vivo cytokine levels, lymphocyte proliferation, and the T lymphocyte anti-platelet immune response in these patients. Patients with SLE-TP and those with ITP had increased platelet-associated IgG, an increased percentage of platelet microparticles, a higher percentage of reticulated platelets and larger platelets, suggesting antibody-mediated platelet destruction and increased platelet production. More than 50% of patients with ITP had increased HLA-DR on their platelet surface whereas subjects with SLE-TP did not. Analysis of serum cytokines demonstrated increased levels of IL-10, IL-15 and TNF-alpha in patients with SLE, but in those with ITP, only increased levels of IL-15 were seen, no increases in any of these cytokines were observed in patients with in SLE-TP. The ability of lymphocytes to proliferate in response to phorbol myristate acetate (PMA) stimulation was increased in SLE-TP, but was normal in both SLE and ITP. Lymphocytes from subjects with ITP displayed an increased ability to proliferate on exposure to platelets, in contrast, those with SLE-TP did not. While the number of subjects evaluated with SLE-TP was small, these data reveal a number of differences in the immunopathogenesis between SLE-TP and ITP.


Assuntos
Plaquetas/imunologia , Lúpus Eritematoso Sistêmico/sangue , Púrpura Trombocitopênica Idiopática/sangue , Adulto , Células Apresentadoras de Antígenos/citologia , Plaquetas/citologia , Divisão Celular/efeitos dos fármacos , Citocinas/sangue , Técnicas Citológicas , Humanos , Imunidade , Lúpus Eritematoso Sistêmico/imunologia , Ativação Linfocitária , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/imunologia , Mitógenos/farmacologia , Contagem de Plaquetas , Testes de Função Plaquetária , Púrpura Trombocitopênica Idiopática/imunologia
11.
Br J Haematol ; 107(3): 532-8, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10583254

RESUMO

We have investigated the role of p38 mitogen-activated protein kinase (MAPK) in von Willebrand factor (VWF)-dependent platelet activation. The interaction of platelets with subendothelial VWF, especially under high shear stress, is considered to be the first activation step which primes platelets for subsequent haemostatic events. As a model of VWF-dependent platelet activation, porcine VWF was employed. Porcine VWF induced p38 MAPK activation by 1 min post-addition; assessed by phosphorylation of a recombinant p38 MAPK fusion protein substrate termed glutathione S-transferase-MAPK activated protein kinase-2. To determine if p38 MAPK was necessary for porcine VWF-induced platelet activation, we functionally inhibited p38 MAPK activity with SB203580 before exposure of the platelets to porcine VWF. Inhibition of p38 MAPK had no effect on VWF-induced platelet alpha or lysozomal granule release, expression of activated GPIIb IIIa, modulation of membrane glycoprotein CD41, expression of phosphatidylserine as assessed by annexin V binding, microparticle formation, or platelet agglutination. It was concluded that SB203580-inhibitable p38 MAPK activity induced by porcine VWF is not necessary for platelet activation.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/fisiologia , Ativação Plaquetária/fisiologia , Fator de von Willebrand/fisiologia , Humanos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno
12.
Transfusion ; 39(8): 818-23, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10504115

RESUMO

BACKGROUND: Alloimmunization requires a process known as co-stimulation. An important co-stimulatory pathway for most immune responses is mediated by the interaction of CD40 on antigen-presenting cells with CD154 (CD40L) on host T cells. Blockade of this co-stimulatory pathway simultaneous with exposure to challenge with HLA-incompatible cells is hypothesized to inhibit alloimmunization. STUDY DESIGN AND METHODS: Severe combined immune-deficient (SCID) mice were reconstituted with human peripheral blood lymphocytes (Hu-PBL-SCID mice) from a subject primed to HLA antigens and challenged with HLA-incompatible lymphocytes. Mice were challenged in the presence or absence of an 18-kDa soluble recombinant active form of human CD154 (18-kDa CD154). Human IgG production, alloimmunization, and in vitro T-cell responsiveness were assessed. RESULTS: There was no significant effect of 18-kDa CD154 on human IgG levels in these mice, but it inhibited the development of HLA-specific alloantibody in this model to five subsequent untreated white cell challenges. In vitro T-cell proliferation in a mixed lymphocyte culture was also prevented by 18-kDa CD154. CONCLUSION: The recombinant protein 18-kDa CD154 inhibited the ability of the Hu-PBL-SCID mice to mount a secondary immune response to allostimulation. This implies that transfusion-induced alloimmunization utilizes CD40-CD154 co-stimulation and that blockade of this pathway can inhibit T-cell function and interfere with the development of alloimmunization.


Assuntos
Isoanticorpos/imunologia , Isoantígenos/imunologia , Transfusão de Linfócitos , Glicoproteínas de Membrana/imunologia , Animais , Formação de Anticorpos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Ligante de CD40 , Feminino , Doença Enxerto-Hospedeiro/imunologia , Humanos , Imunização , Imunoglobulina G/metabolismo , Imunoglobulina M/metabolismo , Memória Imunológica , Ativação Linfocitária , Camundongos , Camundongos SCID , Proteínas Recombinantes/imunologia , Imunologia de Transplantes
13.
Transfus Med ; 9(2): 147-54, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10354385

RESUMO

Accurate assessment of in vivo or in vitro platelet activation requires optimal preanalytical conditions to prevent artefactual in vitro activation of the platelets. The choice of anticoagulant is one of the critical preanalytical conditions as anticoagulants exert different effects on the activation of platelets ex vivo. We tested the effectiveness of Diatube-H (also known as CTAD; sodium citrate, theophylline, adenosine and dipyridamole) and citrate vacutainer tubes in preventing artefactual activation of platelets and preserving functional reserve. Platelet surface expression of the CD62P (reflecting alpha granule release), CD63 (reflecting lysosomal release) and modulation of normal platelet membrane glycoproteins CD41a and CD42b, were measured in whole blood and in isolated platelets immediately after collection and at 6, 24 and 48 h after venipuncture. Samples taken into Diatube-H showed less spontaneous platelet activation than did those taken into citrate. To measure in vitro platelet functional reserve, thrombin was added as agonist to blood stored for varying periods up to 48 h. Although Diatube-H suppressed in vitro platelet activation for up to 4 h, in samples kept for 6-24 h before thrombin addition, the inhibitory effect was lost and platelets responded fully to agonist activation. Hence, Diatube-H preserved platelets and allowed for measurement of in vivo platelet activation as well as thrombin-induced in vitro platelet activation after 6-24 h, in both whole blood and isolated platelets.


Assuntos
Anticoagulantes , Citometria de Fluxo , Ativação Plaquetária , Adenosina/farmacologia , Adulto , Antígenos CD/análise , Plaquetas/imunologia , Plaquetas/fisiologia , Preservação de Sangue , Citratos/farmacologia , Dipiridamol/farmacologia , Feminino , Humanos , Masculino , Selectina-P/sangue , Ativação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/análise , Complexo Glicoproteico GPIb-IX de Plaquetas/análise , Glicoproteínas da Membrana de Plaquetas/análise , Citrato de Sódio , Tetraspanina 30 , Teofilina/farmacologia , Trombina/farmacologia
14.
Br J Haematol ; 104(4): 919-24, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10192460

RESUMO

Severe combined immune deficient (SCID) mice were engrafted with human (Hu) peripheral blood lymphocytes (PBL) from a previously alloimmunized donor and transfused with HLA-mismatched platelets. We have previously shown this to be a useful model for platelet transfusion. These engrafted mice (Hu-PBL-SCID mice) produced high levels of alloantibody in response to standard platelet preparations. However, when the first platelet challenge was presensitized with anti-HLA antibody and then transfused there was a marked reduction in the amount of alloantibody produced to five subsequent untreated platelet transfusions. Platelets pretreated with platelet-specific anti-HPA-1a (PL(A1)) sera did not induce a decrease in the anti-HLA alloantibody response. This demonstrated that platelet-induced HLA alloimmunization can be blocked by anti-HLA antibody-sensitized cells.


Assuntos
Autoimunidade/imunologia , Plaquetas/imunologia , Isoanticorpos/imunologia , Transfusão de Plaquetas , Animais , Formação de Anticorpos , Linfócitos B/citologia , Divisão Celular , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoglobulina G/análise , Isoanticorpos/análise , Camundongos , Camundongos SCID , Linfócitos T/citologia
15.
Blood ; 93(2): 713-20, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9885234

RESUMO

In a murine model of platelet alloimmunization, we examined the definitive role that mononuclear cells (MC) have in modulating platelet immunity by using platelets from severe combined immunodeficient (SCID) mice. CB.17 (H-2(d)) SCID or BALB/c (H-2(d)) mouse platelets were transfused weekly into fully allogeneic CBA (H-2(k)) mice and antidonor antibodies measured by flow cytometry. MC levels in BALB/c platelets were 1.1 +/- 0.6/microL and SCID mouse platelets could be prepared to have significantly lower (<0. 05/microL) MC numbers. Transfusions with 10(8) BALB/c platelets (containing approximately 100 MC/transfusion) stimulated IgG antidonor antibodies in 100% of the recipients by the fifth transfusion, whereas 10(8) SCID mouse platelets (containing approximately 5 MC/transfusion) stimulated higher-titered IgG alloantibodies by the second transfusion. When titrations of BALB/c peripheral blood MC were added to the SCID mouse platelets, levels approaching 1 MC/microL reduced SCID platelet immunity to levels similar to BALB/c platelets. Characterization of the alloantibodies showed that the low levels of MC significantly influenced the isotype of the antidonor IgG; the presence of 1 MC/microL was associated with induction of noncomplement fixing IgG1 antidonor antibodies, whereas platelet transfusions, devoid of MC (<0. 05/microL), were responsible for complement-fixing IgG2a production. When magnetically sorted defined subpopulations of MC were added to the SCID platelets, major histocompatability complex (MHC) class II positive populations, particularly B cells, were found to be primarily responsible for the reduced SCID mouse platelet immunity. The presence of low numbers of MC within the platelets was also associated with an age-dependent reduction in platelet immunogenicity; this relationship however, was not observed with SCID mouse platelets devoid of MC. The results suggest that a residual number of MHC class II positive B cells within allogeneic platelets are required for maximally reducing alloimmunization.


Assuntos
Linfócitos B/imunologia , Plaquetas/imunologia , Antígenos de Histocompatibilidade Classe II/análise , Transfusão de Plaquetas , Animais , Feminino , Imunoglobulina G/sangue , Isoanticorpos/sangue , Contagem de Leucócitos , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos SCID
16.
Acta Paediatr Suppl ; 424: 21-5, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9736214

RESUMO

Adult chronic idiopathic thrombocytopenic purpura (ITP) is an autoimmune disease characterized by production of autoreactive antibodies to platelet antigens. It is now becoming clear that autoantibody production, in general, is regulated by T helper (Th) cells. Several recent studies have examined potential defects in T cell function in this disease and have demonstrated that patients with ITP possess abnormal lymphocyte activation and Th1/Th2-mediated cytokine production. Although the underlying cause(s) of aberrant T cell function in this disease are not known, studies from other models of autoimmune disease indicate that defects in T cell transmembrane signalling can be causally linked to abnormal T cell activation and cytokine production. This review will present some of the major T cell signalling pathways and discuss how altered T cell signalling may be linked to autoimmunity with an emphasis on ITP. Recent preliminary findings of a potential defect in the signal transduction apparatus in lymphocytes from three patients with ITP will also be presented.


Assuntos
Proteínas Tirosina Quinases/metabolismo , Púrpura Trombocitopênica Idiopática/imunologia , Transdução de Sinais , Linfócitos T/fisiologia , Autoimunidade/imunologia , Humanos , Ativação Linfocitária , Fosforilação , Linfócitos T/imunologia
17.
Cytometry ; 32(3): 206-13, 1998 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9667510

RESUMO

Expression of the T-cell receptor (TCR) zeta chain in normal individuals was studied by two-color flow cytometric analysis using digitonin-permeabilized human peripheral blood lymphocytes. Optimal detection of the TCR zeta chain involved fixation of cells in 0.25% paraformaldehyde for 2 min and permeabilization with 500 microg/ml of digitonin at 4 degrees C. Permeabilized lymphocytes and monocytes displayed decreased forward light scatter properties. Neutrophils did not survive this permeabilization/fixation/washing procedure. Permeabilization did not affect the ability of antibodies to CD3, CD4, CD8, CD16, and CD19 to detect CD antigens on lymphocytes, and the percentage of lymphocytes reacting with these antibodies was comparable between untreated and permeabilized cells. Staining of the TCR zeta chain was accomplished by incubating permeabilized cells with unconjugated antibody to the zeta chain, followed by an FITC-conjugated F(ab')2 goat anti-IgG. Following TCR zeta chain staining, cells were blocked with 25 microg/ml of mouse IgG for 20 min to saturate the goat anti-mouse antibody and then incubated with a phycoerythrin-conjugated mouse antibody to a variety of lymphocyte surface antigens. The TCR zeta chain was observed in lymphocytes displaying CD3, CD4, CD8, CD16, or TCRgammadelta markers. This permeabilization/fixation/washing procedure was also validated for detection of another intracellular T-cell protein known as the cytolytic granule-associated protein, recognized by the TIA-1 antibody. Thus the technique can be applied to detection of at least two different intracellular T-cell markers.


Assuntos
Citometria de Fluxo/métodos , Proteínas de Membrana/análise , Receptores de Antígenos de Linfócitos T/análise , Subpopulações de Linfócitos T/química , Fixação de Tecidos/métodos , Anticorpos Monoclonais/química , Antígenos CD/imunologia , Permeabilidade da Membrana Celular , Digitonina , Fixadores , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Luz , Espalhamento de Radiação , Subpopulações de Linfócitos T/metabolismo
18.
Am J Hematol ; 57(3): 200-5, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9495369

RESUMO

We studied the effects of porcine factor VIII (P-FVIII; Hyate:C) and other coagulation products employed in the management of patients with hemophilia A, on platelet activation in vitro. Exposure of normal resting platelets to P-FVIII resulted in platelet activation, as manifested by increased expression of the platelet surface activation markers CD62, CD63, and activated-GPIIbIIIa, and by activation-induced modulation of expression of normal platelet membrane glycoproteins CD41, CD42, and CD36. In contrast, platelet activation was not observed after exposure of the platelets to human FVIII, FEIBA, recombinant FVIIa, or cryosupernatant plasma. As with thrombin, exposure of platelets to P-FVIII resulted in the generation of platelet microparticles, an effect not seen not with the other products. In contrast to the characteristic reduction in expression in the number of CD42 molecules detected on thrombin-activated platelets, P-FVIII-stimulated platelets showed a small increase in CD42 expression. In contrast to thrombin, P-FVIII did not cause platelet dense granule release. The results indicate that therapeutic P-FVIII activates platelets, likely in ways that are different from the platelet activation seen with thrombin. The observed platelet activation and microparticle generation may provide a "hypercoagulable" mechanism for hemostasis with P-FVIII therapy separate from, and additional to, that due to increased circulating FVIII levels.


Assuntos
Plaquetas/fisiologia , Fator VIII/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Animais , Antígenos CD , Moléculas de Adesão Celular/metabolismo , Selectina E/metabolismo , Citometria de Fluxo , Humanos , Técnicas In Vitro , Microcorpos/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas , Suínos , Tetraspanina 30 , Fator de von Willebrand/farmacologia
19.
Transfus Sci ; 19(3): 245-51, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10351136

RESUMO

Chronic autoimmune thrombocytopenic purpura (AITP) is an organ specific autoimmune bleeding disease in which autoantibodies are directed against the individual's own platelets, resulting in increased Fc-mediated platelet destruction by macrophages in the reticuloendothelial system. Although AITP is primarily mediated by IgG auto-antibodies, their production is regulated by the influence of T lymphocytes and antigen presenting cells (APC). This review argues that enhanced T helper cell/antigen presenting cell interactions in patients with AITP may be responsible for IgG anti-platelet auto-antibody production. Understanding these cellular immune responses in AITP may lead to the development of more immune specific therapies for the management of this disease.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Doenças Autoimunes/imunologia , Subpopulações de Linfócitos/imunologia , Púrpura Trombocitopênica Idiopática/imunologia , Antígenos de Plaquetas Humanas/imunologia , Autoanticorpos/imunologia , Autoantígenos/imunologia , Doenças Autoimunes/patologia , Linfócitos B/imunologia , Plaquetas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Humanos , Imunidade Celular , Imunoglobulina G/imunologia , Cooperação Linfocítica , Ativação de Macrófagos , Fator Estimulador de Colônias de Macrófagos/fisiologia , Macrófagos/fisiologia , Modelos Imunológicos , Sistema Fagocitário Mononuclear/patologia , Púrpura Trombocitopênica Idiopática/patologia , Subpopulações de Linfócitos T/imunologia
20.
Transfus Sci ; 19(3): 289-94, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10351141

RESUMO

Infusion of large amounts of intravenous immunoglobulin (IVIG) or anti-D can reverse the low platelet count in patients with ITP within hours of the initiation of treatment. In some cases, the effects of IVIG appear to far outlast several half-lives of the product. Several mechanisms have been proposed to explain these rapid and long term effects and these will be discussed in this review.


Assuntos
Doenças Autoimunes/terapia , Imunoglobulinas Intravenosas/uso terapêutico , Púrpura Trombocitopênica Idiopática/terapia , Anticorpos Anti-Idiotípicos/imunologia , Autoanticorpos/imunologia , Doenças Autoimunes/imunologia , Plaquetas/imunologia , Ensaios Clínicos como Assunto , Meia-Vida , Humanos , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Isoanticorpos/uso terapêutico , Ativação de Macrófagos , Sistema Fagocitário Mononuclear/fisiopatologia , Púrpura Trombocitopênica Idiopática/imunologia , Imunoglobulina rho(D)
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...