Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Korean Med Sci ; 36(31): e223, 2021 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-34402235

RESUMO

Vaccination with an adenoviral vector vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can result in the rare development of thrombosis with thrombocytopenia mediated by platelet-activating antibodies against platelet factor 4 (PF4). This is a life-threating condition that may be accompanied by bleeding due to thrombocytopenia with thrombosis of the cerebral venous sinus or splanchnic vein. Herein, we describe the first fatal case of thrombosis with thrombocytopenia syndrome in Korea, presenting with intracranial hemorrhage caused by cerebral venous sinus thrombosis. A 33-year-old Korean man received the first dose of the ChAdOx1 nCoV-19 vaccination. He developed severe headache with vomiting 9 days after the vaccination. Twelve days after vaccination, he was admitted to the hospital with neurological symptoms and was diagnosed with cerebral venous sinus thrombosis, which was accompanied by intracranial hemorrhage. Thrombocytopenia and D-dimer elevation were observed, and the result of the PF4 enzyme-linked immunosorbent assay antibody test was reported to be strongly positive. Despite intensive treatment, including intravenous immunoglobulin injection and endovascular mechanical thrombectomy, the patient died 19 days after vaccination. Physicians need to be aware of thrombosis with thrombocytopenia syndrome (TTS) in adenoviral vector-vaccinated patients. Endovascular mechanical thrombectomy might be a useful therapeutic option for the treatment of TTS with cerebral venous sinus thrombosis.


Assuntos
Vacinas contra COVID-19/efeitos adversos , Hemorragia Cerebral/mortalidade , Hemorragia Cerebral/patologia , Trombocitopenia/patologia , Trombose/patologia , Adenoviridae/imunologia , Adulto , COVID-19/imunologia , COVID-19/prevenção & controle , ChAdOx1 nCoV-19 , Humanos , Masculino , Fator Plaquetário 4/antagonistas & inibidores , Fator Plaquetário 4/imunologia , República da Coreia , SARS-CoV-2/imunologia , Trombose/mortalidade , Vacinação/efeitos adversos
2.
MAbs ; 13(1): 1887628, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33596779

RESUMO

ABT-736 is a humanized monoclonal antibody generated to target a specific conformation of the amyloid-beta (Aß) protein oligomer. Development of ABT-736 for Alzheimer's disease was discontinued due to severe adverse effects (AEs) observed in cynomolgus monkey toxicity studies. The acute nature of AEs observed only at the highest doses suggested potential binding of ABT-736 to an abundant plasma protein. Follow-up investigations indicated polyspecificity of ABT-736, including unintended high-affinity binding to monkey and human plasma protein platelet factor 4 (PF-4), known to be involved in heparin-induced thrombocytopenia (HIT) in humans. The chronic AEs observed at the lower doses after repeat administration in monkeys were consistent with HIT pathology. Screening for a backup antibody revealed that ABT-736 possessed additional unintended binding characteristics to other, unknown factors. A subsequently implemented screening funnel focused on nonspecific binding led to the identification of h4D10, a high-affinity Aß oligomer binding antibody that did not bind PF-4 or other unintended targets and had no AEs in vivo. This strengthened the hypothesis that ABT-736 toxicity was not Aß target-related, but instead was the consequence of polyspecificity including PF-4 binding, which likely mediated the acute and chronic AEs and the HIT-like pathology. In conclusion, thorough screening of antibody candidates for nonspecific interactions with unrelated molecules at early stages of discovery can eliminate candidates with polyspecificity and reduce potential for toxicity caused by off-target binding.


Assuntos
Vacinas contra Alzheimer/imunologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Anticorpos Monoclonais Humanizados/toxicidade , Plaquetas/efeitos dos fármacos , Imunidade Heteróloga , Fator Plaquetário 4/antagonistas & inibidores , Púrpura Trombocitopênica Idiopática/induzido quimicamente , Vacinas contra Alzheimer/farmacocinética , Vacinas contra Alzheimer/toxicidade , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacocinética , Especificidade de Anticorpos , Plaquetas/imunologia , Plaquetas/metabolismo , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos Endogâmicos BALB C , Nível de Efeito Adverso não Observado , Ativação Plaquetária/efeitos dos fármacos , Fator Plaquetário 4/imunologia , Púrpura Trombocitopênica Idiopática/sangue , Púrpura Trombocitopênica Idiopática/imunologia , Medição de Risco , Fatores de Tempo , Testes de Toxicidade Aguda , Testes de Toxicidade Crônica
3.
J Med Chem ; 64(4): 2242-2253, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33586962

RESUMO

Bovine intestinal heparins are structurally distinct from porcine intestinal heparins and exhibit lower specific anticoagulant activity (units/mg). The reduced content of N-sulfo, 3-O-sulfo glucosamine, the central and critical residue in heparin's antithrombin III binding site, is responsible for bovine intestinal heparin's reduced activity. Previous studies demonstrate that treatment of bovine intestinal heparin with 3-O-sulfotransferase in the presence of 3'-phosphoadenosine-5'-phosphosulfate afforded remodeled bovine heparin with an enhanced activity reaching the United States Pharmacopeia's requirements. Starting from this remodeled bovine intestinal heparin, we report the preparation of a bovine intestinal low molecular weight heparin having the same structural properties and anti-factor IIa and anti-factor Xa activities of Enoxaparin. Moreover, this bovine intestinal heparin-derived "Enoxaparin" showed comparable platelet factor-4 binding affinity, suggesting that it should exhibit similarly low levels of heparin induced thrombocytopeneia, HIT.


Assuntos
Anticoagulantes/farmacologia , Enoxaparina/farmacologia , Animais , Anticoagulantes/síntese química , Anticoagulantes/metabolismo , Antitrombina III/antagonistas & inibidores , Antitrombina III/metabolismo , Sequência de Carboidratos , Bovinos , Enoxaparina/síntese química , Enoxaparina/metabolismo , Peso Molecular , Fator Plaquetário 4/antagonistas & inibidores , Fator Plaquetário 4/metabolismo , Sulfotransferases/química , Suínos
4.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165604, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31740404

RESUMO

Retinal neovascularization (RNV) is a common pathology of blinding proliferative retinopathies. The current treatments to RNV, however, are hindered by limited efficacy, side effects, and drug resistance. A naturally-occurring cytokine in retina that is amicable to immune system and possesses robust anti-neovascular function would facilitate to overcome the hurdles. In this study, retinas from a mouse model of oxygen-induced retinopathy (OIR) underwent a protein array to screen the naturally-occurring cytokines that may antagonize RNV. Among the 62 angiogenesis-associated cytokines, platelet factor 4 (Pf4) stood out with the most prominent upregulation and statistical significance. Moreover, an intravitreal injection of mouse Pf4 demonstrated dramatic anti-vaso-obliteration and anti-neovascularization effects dose dependently in the OIR model; whereas human PF4 inhibited the proliferation, migration, and tubulogenesis of monkey retinal vascular endothelial cells treated with VEGF and TNF-α. These previously undescribed angiostatic effects of PF4 in OIR retinas and retinal vascular endothelial cells support translation of this naturally-occurring chemokine into a therapeutic modality to RNV supplementary to the anti-VEGFs. Mechanistically, a phosphorylation array and western blots indicated that downregulation of proline-rich Akt substrate of 40 kDa (Pras40) and its phosphorylation were necessary for Pf4's anti-neovascular effects in the OIR retinas. Indeed, overexpression of the wildtype Pras40 and the mutant version with deficient phosphorylation abolished and mimicked the Pf4's angiostatic effects in the OIR retinas, respectively. The similar effects were also observed in vitro. This study, for the first time, links PF4's anti-RNV function to an intracellular signaling molecule PRAS40 and its phosphorylation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Oxigênio/metabolismo , Fosfoproteínas/antagonistas & inibidores , Fator Plaquetário 4/antagonistas & inibidores , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Retinopatia da Prematuridade/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fosforilação/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Int J Lab Hematol ; 41 Suppl 1: 15-25, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31069988

RESUMO

Heparin-induced thrombocytopenia (HIT) is a clinical-pathological disorder; thus, laboratory testing for the pathogenic platelet-activating antiplatelet factor 4 (PF4)/heparin antibodies is central for diagnosis. The "iceberg" model summarizes the inter-relationship between platelet activation assays and PF4-dependent immunoassays, with platelet-activating antibodies comprising a subset of anti-PF4/heparin antibodies. The platelet serotonin-release assay (SRA), performed by reference laboratories, has high sensitivity and specificity for HIT (~95% each), and is especially suited for detecting highly pathogenic HIT sera containing both heparin-dependent and heparin-independent platelet-activating antibodies; this latter subgroup of antibodies explains "autoimmune HIT" disorders (delayed-onset, persisting, spontaneous, heparin "flush," fondaparinux-associated). Recently, SRA-negative HIT has become recognized, in which serum from some HIT patients contains subthreshold levels of platelet-activating antibodies (by SRA) that become detectable using a PF4-enhanced platelet activation assay. Unusual immunologic features of HIT include early antibody detectability (at onset of platelet count fall) and antibody transience (seroreversion). Widely available PF4-dependent enzyme immunoassays (EIAs) have high sensitivity but poor specificity for HIT, although specificity is enhanced with IgG-specific EIAs and strong positive results; unfortunately, EIA results are usually not available in real time. Automated rapid immunoassays, such as the chemiluminescence immunoassay (CLIA) and latex immunoturbidimetric assay (LIA), facilitate real-time laboratory diagnosis. Recently available likelihood ratio (LR) data for positive (LR+) and negative (LR-) test results allow clinicians to adjust their pretest probabilities for HIT, using Bayesian analysis, into real-time posttest probabilities that are dramatically increased (test positive) or decreased (test negative). Moreover, (semi-)quantitative CLIA- and LIA-positive results (weak, moderate, strong positive) can further refine the posttest probability of HIT.


Assuntos
Anticoagulantes/efeitos adversos , Heparina/efeitos adversos , Púrpura Trombocitopênica Idiopática/induzido quimicamente , Púrpura Trombocitopênica Idiopática/diagnóstico , Anticoagulantes/uso terapêutico , Autoanticorpos/imunologia , Técnicas de Laboratório Clínico , Heparina/uso terapêutico , Humanos , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/imunologia , Fator Plaquetário 4/antagonistas & inibidores , Fator Plaquetário 4/imunologia , Púrpura Trombocitopênica Idiopática/imunologia
6.
Nat Commun ; 8: 14945, 2017 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-28530237

RESUMO

Antibodies recognizing complexes of the chemokine platelet factor 4 (PF4/CXCL4) and polyanions (P) opsonize PF4-coated bacteria hereby mediating bacterial host defense. A subset of these antibodies may activate platelets after binding to PF4/heparin complexes, causing the prothrombotic adverse drug reaction heparin-induced thrombocytopenia (HIT). In autoimmune-HIT, anti-PF4/P-antibodies activate platelets in the absence of heparin. Here we show that antibodies with binding forces of approximately 60-100 pN activate platelets in the presence of polyanions, while a subset of antibodies from autoimmune-HIT patients with binding forces ≥100 pN binds to PF4 alone in the absence of polyanions. These antibodies with high binding forces cluster PF4-molecules forming antigenic complexes which allow binding of polyanion-dependent anti-PF4/P-antibodies. The resulting immunocomplexes induce massive platelet activation in the absence of heparin. Antibody-mediated changes in endogenous proteins that trigger binding of otherwise non-pathogenic (or cofactor-dependent) antibodies may also be relevant in other antibody-mediated autoimmune disorders.


Assuntos
Anticorpos/farmacologia , Autoimunidade/efeitos dos fármacos , Fator Plaquetário 4/antagonistas & inibidores , Polímeros/farmacologia , Anticorpos/isolamento & purificação , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Heparina/metabolismo , Humanos , Agregação Plaquetária/efeitos dos fármacos , Polieletrólitos , Ligação Proteica/efeitos dos fármacos
7.
Blood ; 129(21): 2864-2872, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28416511

RESUMO

Heparin-induced thrombocytopenia (HIT) is an immune complication of heparin therapy caused by antibodies to complexes of platelet factor 4 (PF4) and heparin. Pathogenic antibodies to PF4/heparin bind and activate cellular FcγRIIA on platelets and monocytes to propagate a hypercoagulable state culminating in life-threatening thrombosis. It is now recognized that anti-PF4/heparin antibodies develop commonly after heparin exposure, but only a subset of sensitized patients progress to life-threatening complications of thrombocytopenia and thrombosis. Recent scientific developments have clarified mechanisms underlying PF4/heparin immunogenicity, disease susceptibility, and clinical manifestations of disease. Insights from clinical and laboratory findings have also been recently harnessed for disease prevention. This review will summarize our current understanding of HIT by reviewing pathogenesis, essential clinical and laboratory features, and management.


Assuntos
Autoanticorpos , Heparina/efeitos adversos , Fator Plaquetário 4 , Receptores de IgG , Trombocitopenia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Humanos , Fator Plaquetário 4/antagonistas & inibidores , Fator Plaquetário 4/sangue , Fator Plaquetário 4/imunologia , Receptores de IgG/sangue , Receptores de IgG/imunologia , Trombocitopenia/sangue , Trombocitopenia/induzido quimicamente , Trombocitopenia/imunologia , Trombocitopenia/prevenção & controle , Trombose/sangue , Trombose/etiologia , Trombose/imunologia , Trombose/prevenção & controle
10.
Cancer Biol Ther ; 16(12): 1775-83, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26479470

RESUMO

The recurrence of colorectal cancer after chemotherapy is the leading cause of its high mortality. We propose that elucidating the mechanisms of tumor regrowth after chemotherapy in tumor-bearing mice may provide new insights into tumor relapse in cancer patients. We firstly report the identification of a chemokine, CXCL4, that plays an important role in the molecular mechanism of cancer regrowth after chemotherapy. A syngenic transplantation tumor model was established with murine colon cancer CT26 cells and treated with 5-FU. Genome-wide gene expression analysis determined that CXCL4 was transiently upregulated in the tumor model. Systemic overexpression of CXCL4 accelerated cancer growth in vivo, but not in vitro. Conversely, the anti-CXCL4 monoclonal antibody (CXCL4-mab) retarded tumor-regrowth after 5-FU treatment in immune-competent mice, but not nude mice. The CXCL4-mab treatment increased the local expression levels of IFN-γ and Gran-b genes in the tumor-bed, and elevated the function of CTLs against CT26 cells. Thus, the colon cancer cells in responding to the cytotoxic stress of 5-FU produce a high level of CXCL4, which suppresses antitumor immunity to confer the residual cancer cells an advantage for regrowth after chemotherapy. Our findings provide a novel target for developing therapeutics aiming to increase antitumor immunity after chemotherapy.


Assuntos
Imunomodulação , Neoplasias/imunologia , Neoplasias/metabolismo , Fator Plaquetário 4/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Fluoruracila/farmacologia , Perfilação da Expressão Gênica , Humanos , Imunomodulação/efeitos dos fármacos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Fator Plaquetário 4/antagonistas & inibidores , Fator Plaquetário 4/genética , Transcriptoma , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Clin Appl Thromb Hemost ; 21(1): 66-71, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23698726

RESUMO

The underlying inflammatory or infectious condition in disseminated intravascular coagulation (DIC) may stimulate the formation of antiheparin/platelet factor 4 (PF4) antibody, and the resulting antibody may affect the clinical course of DIC. We investigated the prognosis of antiheparin/PF4 antibodies in patients with suspected DIC. We measured heparin/PF4 immunoglobulin G (IgG) and total antibody levels using an automated chemiluminescence system in 118 patients with DIC. Of the 118 patients, 13 (11.0%) patients were positive for total antiheparin/PF4, and 6 (5.1%) patients were positive for antiheparin/PF4 IgG. These 13 patients were negative for platelet-activating antibody and had low-heparin-induced thrombocytopenia probability scores. Patients with antiheparin/PF4 IgG were older and had lower antithrombin levels than patients without antiheparin/PF4 IgG. Patients with antiheparin/PF4 IgG had a higher risk of mortality than those without antiheparin/PF4 IgG. The presence of antiheparin/PF4 IgG in old age or low antithrombin level patients with DIC with old age or low antithrombin level suggests a poor prognosis.


Assuntos
Coagulação Intravascular Disseminada/imunologia , Imunoglobulina G/sangue , Fator Plaquetário 4/antagonistas & inibidores , Fator Plaquetário 4/imunologia , Idoso , Idoso de 80 Anos ou mais , Coagulação Intravascular Disseminada/sangue , Coagulação Intravascular Disseminada/mortalidade , Feminino , Heparina/efeitos adversos , Heparina/imunologia , Humanos , Estimativa de Kaplan-Meier , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Prognóstico , Fatores de Risco , Trombocitopenia/sangue , Trombocitopenia/etiologia , Trombocitopenia/imunologia
13.
Arterioscler Thromb Vasc Biol ; 33(4): 718-26, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23288157

RESUMO

OBJECTIVE: Macrophages are critical contributors to abdominal aortic aneurysm (AAA) disease. We examined the ability of MKEY, a peptide inhibitor of CXCL4-CCL5 interaction, to influence AAA progression in murine models. APPROACH AND RESULTS: AAAs were created in 10-week-old male C57BL/6J mice by transient infrarenal aortic porcine pancreatic elastase infusion. Mice were treated with MKEY via intravenous injection either (1) before porcine pancreatic elastase infusion or (2) after aneurysm initiation. Immunostaining demonstrated CCL5 and CCR5 expression on aneurysmal aortae and mural monocytes/macrophages, respectively. MKEY treatment partially inhibited migration of adaptively transferred leukocytes into aneurysmal aortae in recipient mice. Although all vehicle-pretreated mice developed AAAs, aneurysms formed in only 60% (3/5) and 14% (1/7) of mice pretreated with MKEY at 10 and 20 mg/kg, respectively. MKEY pretreatment reduced aortic diameter enlargement, preserved medial elastin fibers and smooth muscle cells, and attenuated mural macrophage infiltration, angiogenesis, and aortic metalloproteinase 2 and 9 expression after porcine pancreatic elastase infusion. MKEY initiated after porcine pancreatic elastase infusion also stabilized or reduced enlargement of existing AAAs. Finally, MKEY treatment was effective in limiting AAA formation after angiotensin II infusion in apolipoprotein E-deficient mice. CONCLUSIONS: MKEY suppresses AAA formation and progression in 2 complementary experimental models. Peptide inhibition of CXCL4-CCL5 interactions may represent a viable translational strategy to limit progression of human AAA disease.


Assuntos
Aorta Abdominal/efeitos dos fármacos , Aneurisma da Aorta Abdominal/prevenção & controle , Quimiocina CCL5/antagonistas & inibidores , Oligopeptídeos/farmacologia , Fator Plaquetário 4/antagonistas & inibidores , Angiotensina II , Animais , Aorta Abdominal/imunologia , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/imunologia , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Injeções Intravenosas , Leucócitos/efeitos dos fármacos , Leucócitos/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/imunologia , Oligopeptídeos/administração & dosagem , Elastase Pancreática , Fator Plaquetário 4/metabolismo , Receptores CCR5/metabolismo , Fatores de Tempo
14.
Blood ; 119(25): 5955-62, 2012 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-22452981

RESUMO

Patients with heparin-induced thrombocytopenia (HIT) remain at risk for recurrent thromboembolic complications despite improvements in management. HIT is caused by antibodies that preferentially recognize ultralarge complexes (ULCs) of heparin and platelet factor 4 (PF4) tetramers. We demonstrated previously that a variant PF4(K50E) forms dimers but does not tetramerize or form ULCs. Here, we identified small molecules predicted to bind PF4 near the dimer-dimer interface and that interfere with PF4 tetramerization. Screening a library of small molecules in silico for binding at this site, we identified 4 compounds that inhibited tetramerization at micromolar concentrations, designated PF4 antagonists (PF4As). PF4As also inhibited formation of pathogenic ULCs, and 3 of these PF4As promoted the breakdown of preformed ULCs. To characterize the ability of PF4As to inhibit cellular activation, we developed a robust and reproducible assay that measures cellular activation by HIT antibodies via FcγRIIA using DT40 cells. PF4As inhibit FcγRIIA-dependent activation of DT40 cells by HIT antibodies as well as platelet activation, as measured by serotonin release. PF4As provide new tools to probe the pathophysiology of HIT. They also may provide insight into the development of novel, disease-specific therapeutics for the treatment of thromboembolic complications in HIT.


Assuntos
Anticoagulantes/isolamento & purificação , Anticoagulantes/uso terapêutico , Descoberta de Drogas/métodos , Fator Plaquetário 4/antagonistas & inibidores , Trombocitopenia/tratamento farmacológico , Animais , Anticoagulantes/química , Células Cultivadas , Galinhas , Biologia Computacional , Drosophila , Avaliação Pré-Clínica de Medicamentos , Heparina/efeitos adversos , Heparina/uso terapêutico , Humanos , Modelos Biológicos , Modelos Moleculares , Racionalização , Projetos de Pesquisa , Trombocitopenia/induzido quimicamente
15.
Int J Radiat Oncol Biol Phys ; 80(5): 1533-40, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21740995

RESUMO

PURPOSE: Factors affecting the severity of radiation-induced thrombocytopenia (RIT) are not well described. We address whether platelet factor 4 (PF4; a negative paracrine for megakaryopoiesis) affects platelet recovery postradiation. METHODS AND MATERIALS: Using conditioned media from irradiated bone marrow (BM) cells from transgenic mice overexpressing human (h) PF4 (hPF4+), megakaryocyte colony formation was assessed in the presence of this conditioned media and PF4 blocking agents. In a model of radiation-induced thrombocytopenia, irradiated mice with varying PF4 expression levels were treated with anti-hPF4 and/or thrombopoietin (TPO), and platelet count recovery and survival were examined. RESULTS: Conditioned media from irradiated BM from hPF4+ mice inhibited megakaryocyte colony formation, suggesting that PF4 is a negative paracrine released in RIT. Blocking with an anti-hPF4 antibody restored colony formation of BM grown in the presence of hPF4+ irradiated media, as did antibodies that block the megakaryocyte receptor for PF4, low-density lipoprotein receptor-related protein 1 (LRP1). Irradiated PF4 knockout mice had higher nadir platelet counts than irradiated hPF4+/knockout litter mates (651 vs. 328 × 106/mcL, p = 0.02) and recovered earlier (15 days vs. 22 days, respectively, p <0.02). When irradiated hPF4+ mice were treated with anti-hPF4 antibody and/or TPO, they showed less severe thrombocytopenia than untreated mice, with improved survival and time to platelet recovery, but no additive effect was seen. CONCLUSIONS: Our studies show that in RIT, damaged megakaryocytes release PF4 locally, inhibiting platelet recovery. Blocking PF4 enhances recovery while released PF4 from megakaryocytes limits TPO efficacy, potentially because of increased release of PF4 stimulated by TPO. The clinical value of blocking this negative paracrine pathway post-RIT remains to be determined.


Assuntos
Plaquetas/efeitos da radiação , Megacariócitos/efeitos da radiação , Fator Plaquetário 4/fisiologia , Trombocitopenia/etiologia , Animais , Plaquetas/citologia , Medula Óssea/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Ensaio de Unidades Formadoras de Colônias/métodos , Meios de Cultivo Condicionados/efeitos da radiação , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Contagem de Plaquetas , Fator Plaquetário 4/antagonistas & inibidores , Receptores de LDL/antagonistas & inibidores , Trombocitopenia/sangue , Trombocitopenia/tratamento farmacológico , Trombopoese/efeitos dos fármacos , Trombopoese/fisiologia , Trombopoese/efeitos da radiação , Trombopoetina/farmacologia , Proteínas Supressoras de Tumor/antagonistas & inibidores
16.
Thromb Haemost ; 106(2): 353-62, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21655676

RESUMO

Atherosclerosis is an inflammatory and thrombotic disease. Both platelets and lymphocytes play important roles in atherogenesis. However, information on their interaction is limited. We therefore studied how platelets regulate CD4+ T cell activation and differentiation. Human CD4+ T cells and autologous platelets were co-cultured. Platelets concentration-dependently enhanced anti-CD3/CD28-induced IFNγ production by CD4+ T cells, but attenuated their proliferation. Abrogation of heterotypic cell-cell contact partially reversed the enhancement, and supernatant from activated platelets partially mimicked the enhancement, suggesting that platelets exert their effects via both soluble mediators and direct cell-cell contact. Platelets enhanced the production of IL-10 and cytokines characteristic for type 1 T helper (TH1) (IFNγ/TNFα) and TH17 (IL-17) cells, but influenced TH2 cytokines (IL-4/IL-5) little. The cytokine responses were accompanied by enhanced TH1/TH17/TReg differentiation. Using neutralising antibodies and recombinant PF4, RANTES, and TGFß, we found that platelet-derived PF4 and RANTES enhanced both pro- and anti-inflammatory cytokine production, whilst recombinant TGFß enhanced IL-10 but not TNFα production. In conclusion, platelets enhance the differentiation and cytokine production of anti-CD3/CD28-stimulated CD4+ T cells via both multiple chemokines and direct cell-cell contact. Our study provides new insights into the cross-talk between thrombosis and adaptive immunity, and indicates that platelets can enhance T-effector cell development.


Assuntos
Plaquetas/imunologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Quimiocinas/biossíntese , Imunidade Adaptativa , Adulto , Aterosclerose/sangue , Aterosclerose/etiologia , Aterosclerose/imunologia , Comunicação Celular , Diferenciação Celular/imunologia , Quimiocina CCL5/antagonistas & inibidores , Técnicas de Cocultura , Citocinas/biossíntese , Feminino , Humanos , Interleucina-10/biossíntese , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Ativação Plaquetária , Fator Plaquetário 4/antagonistas & inibidores , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th1/imunologia , Células Th17/citologia , Células Th17/imunologia , Células Th2/citologia , Células Th2/imunologia , Fator de Crescimento Transformador beta/antagonistas & inibidores
17.
J Biol Chem ; 286(1): 502-10, 2011 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-21041299

RESUMO

Thrombomodulin (TM) is a cofactor for thrombin-mediated activation of protein C and thrombin-activatable fibrinolysis inhibitor (TAFI) and thereby helps coordinate coagulation, anticoagulation, fibrinolysis, and inflammation. Platelet factor 4 (PF4), a platelet α-granule protein and a soluble cofactor for TM-dependent protein C activation, stimulates protein C activation in vitro and in vivo. In contrast to stimulation of protein C activation, PF4 is shown here to inhibit activation of TAFI by thrombin-TM. Consequences of inhibition of TAFI activation by PF4 included loss of TM-dependent prolongation of clot lysis times in hemophilia A plasma and loss of TM-stimulated conversion of bradykinin (BK) to des-Arg(9)-BK by TAFIa in normal plasma. Thus, PF4 modulates the substrate specificity of the thrombin-TM complex by selectively enhancing protein C activation while inhibiting TAFI activation, thereby preventing the generation of the antifibrinolytic and anti-inflammatory activities of TAFIa. To block the inhibitory effects of PF4 on TAFI activation, heparin derivatives were tested for their ability to retain high affinity binding to PF4 despite having greatly diminished anticoagulant activity. N-acetylated heparin (NAc-Hep) lacked detectable anticoagulant activity in activated partial thromboplastin time clotting assays but retained high affinity binding to PF4 and effectively reversed PF4 binding to immobilized TM. NAc-Hep permitted BK conversion to des-Arg(9)-BK by TAFIa in the presence of PF4. In a clot lysis assay on TM-expressing cells using hemophilia A plasma, NAc-Hep prevented PF4-mediated inhibition of TAFI activation and the antifibrinolytic functions of TAFIa. Accordingly, NAc-Hep or similar heparin derivatives might provide therapeutic benefits by diminishing bleeding complications in hemophilia A via restoration of TAFIa-mediated protection of clots against premature lysis.


Assuntos
Carboxipeptidase B2/metabolismo , Fator Plaquetário 4/farmacologia , Trombina/antagonistas & inibidores , Trombina/farmacologia , Trombomodulina/metabolismo , Animais , Coagulação Sanguínea/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Hemofilia A/sangue , Heparina/metabolismo , Heparina/farmacologia , Humanos , Fator Plaquetário 4/antagonistas & inibidores , Proteína C/metabolismo , Coelhos , Especificidade por Substrato , Trombina/metabolismo
18.
Biochim Biophys Acta ; 1804(9): 1895-901, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20601223

RESUMO

Propofol, an anesthetic drug, has been shown to exhibit antioxidant and anti-inflammatory properties in vitro and in vivo. Hypercoagulopathy is a common clinical feature of sepsis, but the effects of propofol on the coagulation system in septic conditions are unclear. Using the gel-based comparative proteomic approach, together with Western blot analysis, ELISA, antithrombin III activity assay, and blood coagulation test, the effect of propofol on serum proteomic profiles in endotoxemic rats was examined. We identified that serum platelet factor-4 (PF4), an endogenous pro-coagulant, was up-regulated in LPS-challenged rats (p<0.001). Endotoxemia also resulted in hypercoagulopathy as evidenced by the shortening of thromboplastin time and thrombin time. Administration of propofol attenuated LPS-stimulated PF4 release and partially reversed the effect of LPS on thromboplastin time (p=0.0012) and thrombin time (p=0.0072). We demonstrated that propofol reduces serum levels of PF4 and partially corrects the hypercoagulopathy associated with endotoxemia in rats.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Endotoxemia/tratamento farmacológico , Endotoxemia/metabolismo , Hipnóticos e Sedativos/farmacologia , Fator Plaquetário 4/sangue , Propofol/farmacologia , Animais , Antitrombina III/metabolismo , Testes de Coagulação Sanguínea , Proteínas Sanguíneas/metabolismo , Western Blotting , Eletroforese em Gel Bidimensional , Endotoxemia/patologia , Ensaio de Imunoadsorção Enzimática , Lipopolissacarídeos/farmacologia , Masculino , Fator Plaquetário 4/antagonistas & inibidores , Fator Plaquetário 4/efeitos dos fármacos , Proteoma/análise , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Tempo de Trombina
20.
Presse Med ; 35(1 Pt 1): 58-60, 2006 Jan.
Artigo em Francês | MEDLINE | ID: mdl-16462666

RESUMO

INTRODUCTION: Thrombocytosis induced by heparin is rarely reported in the literature. CASE: We report here four cases of thrombocytosis, three in patients under treatment for a stable myeloproliferative disorder. Thrombocytosis always regressed when heparin treatment was discontinued, and no thrombotic events occurred. DISCUSSION: Following several reports suggesting an association between heparin and thrombocytosis, the French adverse drug reporting (pharmacovigilance) network identified 51 cases. Thrombocytosis associated with heparin is probably explained by the latter's potentiation of megakaryocytopoiesis, in particular, by inhibition of platelet factor 4 (PF4). Thrombocytosis is a further reason to monitor platelet counts during heparin treatment.


Assuntos
Anticoagulantes/efeitos adversos , Fibrinolíticos/efeitos adversos , Heparina/efeitos adversos , Trombocitose/induzido quimicamente , Adulto , Sistemas de Notificação de Reações Adversas a Medicamentos , Idoso , Enoxaparina/efeitos adversos , Feminino , Seguimentos , França , Heparina de Baixo Peso Molecular/efeitos adversos , Humanos , Masculino , Transtornos Mieloproliferativos/tratamento farmacológico , Contagem de Plaquetas , Fator Plaquetário 4/antagonistas & inibidores , Trombopoese/efeitos dos fármacos , Fatores de Tempo , Tinzaparina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...