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

País/Región como asunto
Intervalo de año de publicación
1.
J Korean Med Sci ; 36(31): e223, 2021 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-34402235

RESUMEN

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.


Asunto(s)
Vacunas contra la COVID-19/efectos adversos , Hemorragia Cerebral/mortalidad , Hemorragia Cerebral/patología , Trombocitopenia/patología , Trombosis/patología , Adenoviridae/inmunología , Adulto , COVID-19/inmunología , COVID-19/prevención & control , ChAdOx1 nCoV-19 , Humanos , Masculino , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/inmunología , República de Corea , SARS-CoV-2/inmunología , Trombosis/mortalidad , Vacunación/efectos adversos
2.
Blood ; 129(21): 2864-2872, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28416511

RESUMEN

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.


Asunto(s)
Autoanticuerpos , Heparina/efectos adversos , Factor Plaquetario 4 , Receptores de IgG , Trombocitopenia , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Humanos , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/sangre , Factor Plaquetario 4/inmunología , Receptores de IgG/sangre , Receptores de IgG/inmunología , Trombocitopenia/sangre , Trombocitopenia/inducido químicamente , Trombocitopenia/inmunología , Trombocitopenia/prevención & control , Trombosis/sangre , Trombosis/etiología , Trombosis/inmunología , Trombosis/prevención & control
3.
Blood ; 119(25): 5955-62, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-22452981

RESUMEN

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.


Asunto(s)
Anticoagulantes/aislamiento & purificación , Anticoagulantes/uso terapéutico , Descubrimiento de Drogas/métodos , Factor Plaquetario 4/antagonistas & inhibidores , Trombocitopenia/tratamiento farmacológico , Animales , Anticoagulantes/química , Células Cultivadas , Pollos , Biología Computacional , Drosophila , Evaluación Preclínica de Medicamentos , Heparina/efectos adversos , Heparina/uso terapéutico , Humanos , Modelos Biológicos , Modelos Moleculares , Racionalización , Proyectos de Investigación , Trombocitopenia/inducido químicamente
4.
Arterioscler Thromb Vasc Biol ; 33(4): 718-26, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23288157

RESUMEN

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.


Asunto(s)
Aorta Abdominal/efectos de los fármacos , Aneurisma de la Aorta Abdominal/prevención & control , Quimiocina CCL5/antagonistas & inhibidores , Oligopéptidos/farmacología , Factor Plaquetario 4/antagonistas & inhibidores , Angiotensina II , Animales , Aorta Abdominal/inmunología , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/inmunología , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inyecciones Intravenosas , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/inmunología , Oligopéptidos/administración & dosificación , Elastasa Pancreática , Factor Plaquetario 4/metabolismo , Receptores CCR5/metabolismo , Factores de Tiempo
5.
J Biol Chem ; 286(1): 502-10, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21041299

RESUMEN

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.


Asunto(s)
Carboxipeptidasa B2/metabolismo , Factor Plaquetario 4/farmacología , Trombina/antagonistas & inhibidores , Trombina/farmacología , Trombomodulina/metabolismo , Animales , Coagulación Sanguínea/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células HEK293 , Hemofilia A/sangre , Heparina/metabolismo , Heparina/farmacología , Humanos , Factor Plaquetario 4/antagonistas & inhibidores , Proteína C/metabolismo , Conejos , Especificidad por Sustrato , Trombina/metabolismo
6.
Biochim Biophys Acta ; 1804(9): 1895-901, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20601223

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Endotoxemia/tratamiento farmacológico , Endotoxemia/metabolismo , Hipnóticos y Sedantes/farmacología , Factor Plaquetario 4/sangre , Propofol/farmacología , Animales , Antitrombina III/metabolismo , Pruebas de Coagulación Sanguínea , Proteínas Sanguíneas/metabolismo , Western Blotting , Electroforesis en Gel Bidimensional , Endotoxemia/patología , Ensayo de Inmunoadsorción Enzimática , Lipopolisacáridos/farmacología , Masculino , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/efectos de los fármacos , Proteoma/análisis , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tiempo de Trombina
7.
J Med Chem ; 64(4): 2242-2253, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33586962

RESUMEN

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.


Asunto(s)
Anticoagulantes/farmacología , Enoxaparina/farmacología , Animales , Anticoagulantes/síntesis química , Anticoagulantes/metabolismo , Antitrombina III/antagonistas & inhibidores , Antitrombina III/metabolismo , Secuencia de Carbohidratos , Bovinos , Enoxaparina/síntesis química , Enoxaparina/metabolismo , Peso Molecular , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/metabolismo , Sulfotransferasas/química , Porcinos
8.
MAbs ; 13(1): 1887628, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33596779

RESUMEN

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.


Asunto(s)
Vacunas contra el Alzheimer/inmunología , Péptidos beta-Amiloides/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/toxicidad , Plaquetas/efectos de los fármacos , Inmunidad Heteróloga , Factor Plaquetario 4/antagonistas & inhibidores , Púrpura Trombocitopénica Idiopática/inducido químicamente , Vacunas contra el Alzheimer/farmacocinética , Vacunas contra el Alzheimer/toxicidad , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacocinética , Especificidad de Anticuerpos , Plaquetas/inmunología , Plaquetas/metabolismo , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones Endogámicos BALB C , Nivel sin Efectos Adversos Observados , Activación Plaquetaria/efectos de los fármacos , Factor Plaquetario 4/inmunología , Púrpura Trombocitopénica Idiopática/sangre , Púrpura Trombocitopénica Idiopática/inmunología , Medición de Riesgo , Factores de Tiempo , Pruebas de Toxicidad Aguda , Pruebas de Toxicidad Crónica
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(3): 165604, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31740404

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Oxígeno/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Factor Plaquetario 4/antagonistas & inhibidores , Retina/metabolismo , Neovascularización Retiniana/metabolismo , Retinopatía de la Prematuridad/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Fosforilación/fisiología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Int J Lab Hematol ; 41 Suppl 1: 15-25, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31069988

RESUMEN

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.


Asunto(s)
Anticoagulantes/efectos adversos , Heparina/efectos adversos , Púrpura Trombocitopénica Idiopática/inducido químicamente , Púrpura Trombocitopénica Idiopática/diagnóstico , Anticoagulantes/uso terapéutico , Autoanticuerpos/inmunología , Técnicas de Laboratorio Clínico , Heparina/uso terapéutico , Humanos , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/inmunología , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/inmunología , Púrpura Trombocitopénica Idiopática/inmunología
12.
J Clin Invest ; 61(4): 1102-6, 1978 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-207723

RESUMEN

We have studied the platelet release reaction and thrombin generation during the spontaneous clotting of whole blood in vitro. Both thrombin formation and secretion of platelet Factor 4 were detected at least 12 min before clotting (clotting time, 22--26 min). Initially, at low thrombin concentrations (2--5 ng/ml), there is a small increase in plasma platelet Factor 4 (less than 1% of the amount present in serum). This is followed by a gradual increase in both platelet Factor 4 and thrombin concentrations over a 12 to 20-min interval. Finally, 5 min 5 before clotting, there is a rapid increase in both thrombin generation and platelet secretion. Thus, we have shown that the release of platelet Factor 4 is a prolonged reactoin and the extent to which it occurs parallel thrombin generation. It is only when thrombin concentrations are high (45--90) ng/ml)--during the period of clot formation--that the major part of platelet Factor 4 secretion occurs. Release of platelet Factor 4, like fibrin formation, occurs in the last step of in vitro coagulation.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Coagulación Sanguínea , Factor Plaquetario 4/metabolismo , Trombina/metabolismo , Plaquetas/metabolismo , Bucladesina/farmacología , Humanos , Técnicas In Vitro , Cinética , Factor Plaquetario 4/antagonistas & inhibidores , Prostaglandinas E/farmacología
13.
Nat Commun ; 8: 14945, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28530237

RESUMEN

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.


Asunto(s)
Anticuerpos/farmacología , Autoinmunidad/efectos de los fármacos , Factor Plaquetario 4/antagonistas & inhibidores , Polímeros/farmacología , Anticuerpos/aislamiento & purificación , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Plaquetas/ultraestructura , Heparina/metabolismo , Humanos , Agregación Plaquetaria/efectos de los fármacos , Polielectrolitos , Unión Proteica/efectos de los fármacos
14.
Presse Med ; 35(1 Pt 1): 58-60, 2006 Jan.
Artículo en Francés | MEDLINE | ID: mdl-16462666

RESUMEN

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.


Asunto(s)
Anticoagulantes/efectos adversos , Fibrinolíticos/efectos adversos , Heparina/efectos adversos , Trombocitosis/inducido químicamente , Adulto , Sistemas de Registro de Reacción Adversa a Medicamentos , Anciano , Enoxaparina/efectos adversos , Femenino , Estudios de Seguimiento , Francia , Heparina de Bajo-Peso-Molecular/efectos adversos , Humanos , Masculino , Trastornos Mieloproliferativos/tratamiento farmacológico , Recuento de Plaquetas , Factor Plaquetario 4/antagonistas & inhibidores , Trombopoyesis/efectos de los fármacos , Factores de Tiempo , Tinzaparina
16.
Clin Appl Thromb Hemost ; 21(1): 66-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23698726

RESUMEN

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.


Asunto(s)
Coagulación Intravascular Diseminada/inmunología , Inmunoglobulina G/sangre , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/inmunología , Anciano , Anciano de 80 o más Años , Coagulación Intravascular Diseminada/sangre , Coagulación Intravascular Diseminada/mortalidad , Femenino , Heparina/efectos adversos , Heparina/inmunología , Humanos , Estimación de Kaplan-Meier , Modelos Logísticos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Trombocitopenia/sangre , Trombocitopenia/etiología , Trombocitopenia/inmunología
17.
Cancer Biol Ther ; 16(12): 1775-83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26479470

RESUMEN

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.


Asunto(s)
Inmunomodulación , Neoplasias/inmunología , Neoplasias/metabolismo , Factor Plaquetario 4/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Fluorouracilo/farmacología , Perfilación de la Expresión Génica , Humanos , Inmunomodulación/efectos de los fármacos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/genética , Transcriptoma , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Thromb Haemost ; 67(1): 137-43, 1992 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-1377413

RESUMEN

We have produced a panel of monoclonal antibodies (mAbs) against rabbit platelet factor 4 (PF4). Two of these mAbs have been characterized in this study. In particular the antibody called 10B2, which also recognizes the human molecule, is able to block PF4's ability to neutralize heparin in a modified Heparin-Factor Xa chromogenic assay. The inhibition appears to be more than 95% at 1:1 mAb/PF4 molar ratio both for purified rabbit and human PF4. Similar results were obtained using supernatants from stimulated human platelets (90% of inhibition at 1:1 mAb/PF4 molar ratio) or using Fab fragments from 10B2. Studies to determine the antigenic determinant against which 10B2 is directed, show that this is an assembled epitope which involves disulfide bonds of the PF4.


Asunto(s)
Anticuerpos Monoclonales , Antagonistas de Heparina/inmunología , Factor Plaquetario 4/inmunología , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Sitios de Unión , Plaquetas/inmunología , Epítopos/química , Humanos , Fragmentos Fab de Inmunoglobulinas , Técnicas In Vitro , Datos de Secuencia Molecular , Pruebas de Neutralización , Oligopéptidos/química , Oligopéptidos/inmunología , Factor Plaquetario 4/antagonistas & inhibidores , Factor Plaquetario 4/química , Conejos
19.
Neoplasma ; 34(5): 609-14, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3696302

RESUMEN

The effect of lomustin on platelet functions was investigated in vitro using a wide battery of tests. Lomustin was found to act as a specific inhibitor of platelet aggregation, release reaction and clot retraction and to induce acquired thrombocytopathy. Thus, impairment of platelet functions might play a role in hemorrhagic complications accompanying lomustin therapy in some cases.


Asunto(s)
Plaquetas/efectos de los fármacos , Lomustina/farmacología , Pruebas de Función Plaquetaria , Adenosina Difosfato/farmacología , Retracción del Coagulo , Colágeno/farmacología , Epinefrina/farmacología , Humanos , Agregación Plaquetaria/efectos de los fármacos , Factor Plaquetario 3/antagonistas & inhibidores , Factor Plaquetario 4/antagonistas & inhibidores , Antagonistas de la Serotonina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA