Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Circ Res ; 108(6): 695-703, 2011 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-21293004

RESUMEN

RATIONALE: There is strong evidence that oxidative modification of low-density lipoprotein (oxLDL) plays a critical role in atherogenesis and that oxLDL may profoundly influence the mechanical stability of atherosclerotic plaques. OBJECTIVE: To block oxLDL, we designed, expressed, and tested Fc-CD68, a soluble oxLDL binding protein consisting of human Fc and the extracellular domain of the human oxLDL-binding receptor CD68. METHODS AND RESULTS: Fc-CD68 bound with high specific affinity to oxLDL and strongly bound and colocalized with oxLDL in plaques. To study the effects of repeated administrations of Fc-CD68 on the progression of atherosclerosis and plaque vulnerability, 12- and 16-week old cholesterol-fed ApoE(-/-) mice received either Fc-CD68 (n = 6) or Fc control protein (n = 6 to 8) thrice weekly for 4 weeks. Macroscopic and histological analysis of Sudan red lipid staining showed strong and significant reduction of plaque extension in the aorta and in the aortic root, respectively. Histological analysis of pentachrome- and Sirius-stained sections of the brachiocephalic arteries of 20 week-old ApoE(-/-) mice revealed that Fc-CD68 significantly reduced the occurrence of spontaneous ruptures of established plaques by ≈20%, compared with Fc and drastically increased the collagen content of plaques. Furthermore, in immunostained sections of the brachiocephalic artery and the aortic root, Fc-CD68 reduced the infiltration of plaques with T lymphocytes, and macrophages by ≈50% and 30%, respectively. CONCLUSIONS: The oxLDL binding protein Fc-CD68 attenuates atherosclerosis and strengthens the stability of atherosclerotic plaques.


Asunto(s)
Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Aterosclerosis/patología , Tronco Braquiocefálico/efectos de los fármacos , Tronco Braquiocefálico/patología , Fragmentos Fc de Inmunoglobulinas/genética , Placa Aterosclerótica/patología , Proteínas Recombinantes de Fusión/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/patología , Apolipoproteínas E/deficiencia , Aterosclerosis/metabolismo , Unión Competitiva , Proteínas Portadoras/metabolismo , Proteínas Portadoras/farmacología , Colágeno/metabolismo , Progresión de la Enfermedad , Humanos , Inflamación/metabolismo , Inflamación/patología , Metabolismo de los Lípidos , Lipoproteínas LDL/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Placa Aterosclerótica/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/patología
2.
PLoS One ; 18(5): e0284444, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37141296

RESUMEN

Traditional histological stains, such as hematoxylin-eosin (HE), special stains, and immunofluorescence (IF), have defined myriads of cellular phenotypes and tissue structures in a separate stained section. However, the precise connection of information conveyed by the various stains in the same section, which may be important for diagnosis, is absent. Here, we present a new staining modality-Flow chamber stain, which complies with the current staining workflow but possesses newly additional features non-seen in conventional stains, allowing for (1) quickly switching staining modes between destain and restain for multiplex staining in one single section from routinely histological preparation, (2) real-time inspecting and digitally capturing each specific stained phenotype, and (3) efficiently synthesizing graphs containing the tissue multiple-stained components at site-specific regions. Comparisons of its stains with those by the conventional staining fashions using the microscopic images of mouse tissues (lung, heart, liver, kidney, esophagus, and brain), involving stains of HE, Periodic acid-Schiff, Sirius red, and IF for Human IgG, and mouse CD45, hemoglobin, and CD31, showed no major discordance. Repetitive experiments testing on targeted areas of stained sections confirmed the method is reliable with accuracy and high reproducibility. Using the technique, the targets of IF were easily localized and seen structurally in HE- or special-stained sections, and the unknown or suspected components or structures in HE-stained sections were further determined in histological special stains or IF. By the technique, staining processing was videoed and made a backup for off-site pathologists, which facilitates tele-consultation or -education in current digital pathology. Mistakes, which might occur during the staining process, can be immediately found and amended accordingly. With the technique, a single section can provide much more information than the traditional stained counterpart. The staining mode bears great potential to become a common supplementary tool for traditional histopathology.


Asunto(s)
Colorantes , Esófago , Humanos , Animales , Ratones , Reproducibilidad de los Resultados , Coloración y Etiquetado , Esófago/patología , Hematoxilina , Eosina Amarillenta-(YS) , Fenotipo
3.
Circulation ; 123(17): 1891-9, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21502572

RESUMEN

BACKGROUND: Blocking of glycoprotein VI-dependent pathways by interfering in vascular collagen sites is commonly seen as an attractive target for an antiplatelet therapy of acute atherosclerotic diseases such as myocardial infarction or stroke. Revacept (soluble dimeric glycoprotein VI-Fc fusion protein) has been shown to reduce platelet adhesion by blocking vascular collagen in plaques or erosion and to be safe in preclinical studies. A dose-escalating clinical phase I study was performed to assess the safety, tolerability, pharmacokinetics, and pharmacodynamics of Revacept in humans. METHODS AND RESULTS: In a first-in-humans study, 30 healthy men received a single intravenous administration of 10, 20, 40, 80, or 160 mg Revacept. The serum concentration-time courses of each dosage of Revacept showed a narrow variation and a concentration and time dependence. Revacept did not significantly affect the bleeding time. Collagen-induced platelet aggregation was dose-dependently inhibited up to 48 hours at lower doses and for 7 days after higher dose levels. In contrast, ADP- or thrombin receptor activating peptide-dependent platelet aggregation remained unaltered. There were no relevant drug-related adverse events or drug-related changes in laboratory parameters (biochemistry, hematology, and coagulation parameters). There were no drug-related changes in blood pressure, pulse rate, or ECG parameters (including 24-hour Holter monitoring). No anti-Revacept antibodies were detected. CONCLUSION: This phase I study demonstrated that Revacept is a safe and well-tolerated new antiplatelet compound with a clear dose-dependent pharmacokinetic profile with specific, dose-related inhibition of platelet aggregation despite completely unaltered general hemostasis. CLINICAL TRIAL REGISTRATION: URL: www.clinicaltrials.gov. Unique identifier: NCT 01042964. URL: eudract.ema.europa.eu. Identifier: 2005-004656-12.


Asunto(s)
Glicoproteínas/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Inhibidores de Agregación Plaquetaria/administración & dosificación , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/administración & dosificación , Adulto , Animales , Células CHO , Colágeno/administración & dosificación , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Hemostasis/efectos de los fármacos , Humanos , Fragmentos Fc de Inmunoglobulinas/efectos adversos , Inyecciones Intravenosas , Masculino , Inhibidores de Agregación Plaquetaria/efectos adversos , Inhibidores de Agregación Plaquetaria/farmacocinética , Recuento de Plaquetas , Glicoproteínas de Membrana Plaquetaria/efectos adversos , Glicoproteínas de Membrana Plaquetaria/farmacocinética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/farmacocinética , Adulto Joven
4.
Anal Chem ; 84(8): 3523-30, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22397688

RESUMEN

The direct quantification of both the binding affinity and absolute concentration of disease-related biomarkers in biological fluids is particularly beneficial for differential diagnosis and therapy monitoring. Here, we extend microscale thermophoresis to target immunological questions. Optically generated thermal gradients were used to deplete fluorescently marked antigens in 2- and 10-fold-diluted human serum. We devised and validated an autocompetitive strategy to independently fit the concentration and dissociation constant of autoimmune antibodies against the cardiac ß1-adrenergic receptor related to dilated cardiomyopathy. As an artificial antigen, the peptide COR1 was designed to mimic the second extracellular receptor loop. Thermophoresis resolved antibody concentrations from 2 to 200 nM and measured the dissociation constant as 75 nM. The approach quantifies antibody binding in its native serum environment within microliter volumes and without any surface attachments. The simplicity of the mix and probe protocol minimizes systematic errors, making thermophoresis a promising detection method for personalized medicine.


Asunto(s)
Autoanticuerpos/sangre , Biomarcadores/sangre , Electroforesis , Temperatura , Cardiomiopatía Dilatada/diagnóstico , Humanos , Microquímica
5.
Thyroid ; 31(6): 950-963, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33208049

RESUMEN

Background: Antigen-specific lymphocytes are increasingly investigated in autoimmune diseases and immune therapies. We sought to identify thyrotropin receptor (TSHR)-specific lymphocytes in mouse models of Graves' disease, including Graves' patient-specific immunotype human leukocyte antigen (HLA)-DR3, and in frozen and thawed Graves' patient blood samples. Methods and Results: Splenic lymphocytes of adenovirus (Ad)-TSHR-immunized BALB/c mice were stimulated with TSHR-specific peptides C, D, or J. Furthermore, CD154-expressing cells were enriched, expanded in vitro, and analyzed for binding of peptide-major histocompatibility complex (MHC) II multimers ("tetramers," immunotype H2-IAd). Only peptides C and J were able to elicit increased expression/secretion of CD154 and interferon-γ, and tetramers which were loaded with peptide C resulted in antigen-specific signals in splenic lymphocytes from Ad-TSHR-immunized mice. Accordingly, TSHR-specific HLA-DR3-MHC class II tetramers loaded with peptide p10 specifically bound to human HLA-DR3-(allele B1*03:01)-transgenic Bl/6 mouse splenic T lymphocytes. In addition, we fine-tuned a protocol to reliably measure thawed human peripheral blood mononuclear cells (PBMCs), which resulted in reliable recovery after freezing and thawing with regard to vitality and B and T cell subpopulation markers including regulatory T cells (CD3, CD4, CD25, FoxP3, CD25high, CD127low). TSHR-specific HLA-DR3-MHC class II tetramers loaded with peptide p10 identified antigen-specific T cells in HLA-DR3-positive Graves' patients' thawed PBMCs. Moreover, stimulation-dependent release of interleukin (IL)-1beta, IL-6, tumor necrosis factor-alpha from thawed PBMCs occurred at the expected levels. Conclusions: Novel MHC II tetramers identified TSHR-specific T lymphocytes in Ad-TSHR-immunized hyperthyroid BALB/c or HLA-DR3-transgenic mice and in thawed human PBMCs from patients with Graves' disease. These assays may contribute to measure both disease severity and effects of novel immune therapies in future animal studies and clinical investigations of Graves' disease.


Asunto(s)
Enfermedad de Graves/inmunología , Antígeno HLA-DR3/genética , Hipertiroidismo/inmunología , Receptores de Tirotropina/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/inmunología , Ligando de CD40/inmunología , Modelos Animales de Enfermedad , Epítopos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Inmunización , Inmunofenotipificación , Interferón gamma/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Ratones , Ratones Transgénicos , Péptidos , Factor de Necrosis Tumoral alfa/inmunología
6.
Int J Cancer ; 127(8): 1958-74, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20473895

RESUMEN

Proteolytic enzymes expressed on the surface of tumor cells, and thus easily accessible to external interventions, represent useful targets for anticancer and antimetastatic therapies. In our study, we thoroughly evaluated matriptase, a trypsin-like transmembrane serine protease, as potential target for novel inhibitor-based tumor therapies. We applied time-domain near infrared fluorescence (NIRF) imaging to characterize expression and activity of matriptase in vivo in an orthotopic AsPC-1 pancreatic tumor model in nude mice. We show strong and tumor-specific binding of intravenously injected Cy5.5 labeled antimatriptase antibody (MT-Ab*Cy5.5) only to primary AsPC-1 tumors and their metastases over time within living mice, taking into account fluorescence intensities and fluorescence lifetimes of the applied probes. Specific binding of MT-Ab*Cy5.5 to tumor sites was confirmed by ex vivo NIRF imaging of tumor tissue, NIRF microscopy and by coregistration of the in vivo acquired NIRF intensity maps to anatomical structures visualized by flat-panel volume computed tomography (fpVCT) in living mice. Moreover, using an activatable synthetic substrate S*DY-681 we could clearly demonstrate that matriptase is proteolytically active in vitro as well as in vivo in tumor-bearing mice, and that application of synthetic active-site inhibitors having high affinity and selectivity toward matriptase can efficiently inhibit its proteolytic activity for at least 24 hr. We thus successfully applied NIRF imaging in combination with fpVCT to characterize matriptase as a promising molecular target for inhibitor-based cancer therapies.


Asunto(s)
Carbocianinas/metabolismo , Neoplasias Mamarias Experimentales/terapia , Neoplasias Pancreáticas/terapia , Serina Endopeptidasas/metabolismo , Animales , Anticuerpos Bloqueadores , Masculino , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Pronóstico , Espectroscopía Infrarroja Corta , Imagen de Cuerpo Entero , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Endocrinol ; 246(2): 135-147, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32573180

RESUMEN

Graves' disease is an autoimmune disorder, which is characterized by stimulatory antibodies targeting the human thyrotropin receptor (TSHR), resulting in hyperthyroidism and multiple organ damage. We systematically investigated monomeric and dimeric fusion proteins of the A subunit of TSHR for efficacy to bind to the monoclonal patient antibody M22, to interact with Graves' patient serum samples, and to impact on anti-TSHR antibody titers, hyperthyroidism, tachycardia and other in vivo read-outs in a long-term mouse model of Graves' disease induced by immunization with a recombinant adenovirus encoding TSHR A. Binding assays and functional measurements of TSHR-dependent cAMP formation showed binding of monomeric TSHR-His and dimeric TSHR-Fc to the anti-TSHR antibody M22 at low-effective concentrations (EC50 of 5.7 nmol/L and 8.6 nmol/L) and inhibition of the effects of this antibody at high efficiencies (IC50 values of 16-20 nmol/L). Both proteins also block the effects of polyclonal anti-TSHR antibodies occurring in Graves' patient sera with somewhat lower average efficiencies (mean IC50 values of 29 nmol/L and 68 nmol/L). However, in vivo characterization of epicutaneous patch administrations of TSHR-Fc at doses of 0.3 and 0.6 mg/kg body weight in a murine Graves' disease model did not result in any improvement of disease parameters. In conclusion, high affinity binding of TSHR-Fc to pathological anti-TSHR antibodies was not matched by efficacy to improve Graves' disease parameter in a long-term mouse model.


Asunto(s)
Enfermedad de Graves/metabolismo , Receptores de Tirotropina/metabolismo , Animales , Autoinmunidad/genética , Autoinmunidad/fisiología , AMP Cíclico/genética , AMP Cíclico/metabolismo , Electrocardiografía , Ensayo de Inmunoadsorción Enzimática , Femenino , Enfermedad de Graves/genética , Frecuencia Cardíaca/genética , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Ratones , Péptidos/genética , Péptidos/metabolismo , Receptores de Tirotropina/genética , Temperatura
8.
Thromb Haemost ; 120(6): 977-993, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32492725

RESUMEN

Glycoprotein VI (GPVI), a platelet collagen receptor, is crucial in mediating atherothrombosis. Besides collagen, injured plaques expose tissue factor (TF) that triggers fibrin formation. Previous studies reported that GPVI also is a platelet receptor for fibrinogen and fibrin. We studied the effect of anti-GPVI antibodies and inhibitors of GPVI signaling kinases (Syk and Btk) on platelet adhesion and aggregate formation onto immobilized fibrinogen and different types of fibrin under arterial flow conditions. Fibrin was prepared from isolated fibrinogen ("pure fibrin"), recombinant fibrinogen ("recombinant fibrin"), or generated more physiologically from endogenous fibrinogen in plasma ("plasma fibrin") or by exposing TF-coated surfaces to flowing blood ("blood fibrin"). Inhibition of GPVI and Syk did not inhibit platelet adhesion and aggregate formation onto fibrinogen. In contrast anti-GPVI antibodies, inhibitors of Syk and Btk and the anti-GPIb antibody 6B4 inhibited platelet aggregate formation onto pure and recombinant fibrin. However, inhibition of GPVI and GPVI signaling did not significantly reduce platelet coverage of plasma fibrin and blood fibrin. Plasma fibrin contained many proteins incorporated during clot formation. Advanced optical imaging revealed plasma fibrin as a spongiform cushion with thicker, knotty, and long fibers and little activation of adhering platelets. Albumin intercalated in plasma fibrin fibers left only little space for platelet attachment. Pure fibrin was different showing a dense mesh of thin fibers with strongly activated platelets. We conclude that fibrin formed in plasma and blood contains plasma proteins shielding GPVI-activating epitopes. Our findings do not support a role of GPVI for platelet activation by physiologic fibrin.


Asunto(s)
Plaquetas/metabolismo , Fibrina/metabolismo , Glicoproteínas de Membrana Plaquetaria/fisiología , Receptores de Péptidos/metabolismo , Agammaglobulinemia Tirosina Quinasa/sangre , Agammaglobulinemia Tirosina Quinasa/fisiología , Activación Enzimática , Fibrinógeno/metabolismo , Hemorreología , Humanos , Microscopía Confocal/métodos , Plasma , Adhesividad Plaquetaria , Agregación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Glicoproteínas de Membrana Plaquetaria/inmunología , Unión Proteica , Proteínas Recombinantes/metabolismo , Quinasa Syk/antagonistas & inhibidores , Quinasa Syk/sangre , Quinasa Syk/fisiología , Tromboplastina/metabolismo
9.
Int J Oncol ; 35(2): 347-57, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19578749

RESUMEN

Matriptase, also known as MT-SP1, is a type II transmembrane serine protease strongly implicated in both the development and progression of a variety of epithelial cancers. Evidence comes from studies of its expression in human cancers and from mouse models of spontaneous cancer. Matriptase is considered to be a major activator of two key stimulators of invasive growth, namely hepatocyte growth factor/scatter factor and urokinase-type plasminogen activator. The aim of this study was to examine the role of matriptase in pancreatic ductal adenocarcinoma by expression analysis and functional assays in vitro. Immunohistochemical analysis of matriptase performed on microtissue arrays and large samples of 55 pancreatic ductal adenocarcinomas and on 31 samples of normal pancreatic ducts revealed that although matriptase expression differed greatly in both malignant and normal ductal pancreatic tissue, matriptase scores were significantly (p=0.02) elevated in pancreatic ductal adenocarcinoma compared to normal pancreatic ducts. To evaluate the role of matriptase during development of pancreatic cancer, we studied the effects of newly designed matriptase inhibitors on the processing of the zymogen of urokinase-type plasminogen activator in the human adenocarcinoma cell lines AsPC-1 and BxPC-3. In both cell lines, at 1 microM, all matriptase inhibitors completely prevented zymogen activation. At lower inhibitor concentrations, the degree of inhibition of zymogen processing correlated with the affinities of the inhibitors towards matriptase indicating that this is a specific result of matriptase inhibition. Furthermore, matriptase inhibitors reduced the phosphorylation of the HGF receptor/cMet and the overall cellular invasiveness of the human pancreatic adenocarcinoma cell line AsPC-1. Our findings demonstrate for the first time that matriptase may be involved in the progression of pancreatic ductal adenocarcinoma and that matriptase inhibition may contribute to preventing the progression of this devastating disease.


Asunto(s)
Adenocarcinoma/enzimología , Carcinoma Ductal Pancreático/enzimología , Neoplasias Pancreáticas/enzimología , Serina Endopeptidasas/fisiología , Inhibidores de Serina Proteinasa/farmacología , Adenocarcinoma/patología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Humanos , Inmunohistoquímica , Conductos Pancreáticos/enzimología , Neoplasias Pancreáticas/patología , Fosforilación , Proteínas Proto-Oncogénicas c-met/metabolismo , Serina Endopeptidasas/análisis
10.
PLoS One ; 13(8): e0201160, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30125285

RESUMEN

RATIONALE: Despite advances in pharmacotherapy, heart failure still incurs significant morbidity and mortality. Stimulating antibodies directed against the secondextracellular loop of the human ß1-adrenergic receptor (anti-ß1EC2) cause myocyte damage and heart failure in rats. This receptor domain is 100% homologous between rats and humans. OBJECTIVE: ß1EC2-mimicking cyclopeptides (25-meric) markedly improved the development and/or course of anti-ß1EC2-mediated cardiomyopathy. Further developments should be investigated. METHODS AND RESULTS: The shortened 18-meric cyclic peptide COR-1, in which one of the two disulphide bonds was removed to enable reproducible GMP production, can also be used to treat cardiomyopathic rats. Echocardiography, catheterization and histopathology of the rat hearts revealed that monthly intravenous administrations of COR-1 almost fully reversed the cardiomyopathic phenotype within 6 months at doses of 1 to 4 mg/kg body weight. Administration of COR-1 resulted in markedly reduced anti-ß1EC2-expressing memory B lymphocytes in the spleen despite continued antigenic boosts, but did not significantly decrease overall peripheral anti-ß1EC2 titers. COR-1 did not induce any anti-ß1EC2 or other immune response in naïve rats (corresponding to findings in healthy human volunteers). It did not cause any toxic side effects in GLP studies in dogs, rats or mice, and the "no observed adverse effect level" (NOAEL) exceeded the therapeutic doses by 100-fold. CONCLUSION: The second generation immunomodulating epitope-mimicking cyclopeptide COR-1 (also termed JNJ-5442840) offers promise to treat immune-mediated cardiac diseases.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , Receptores Adrenérgicos beta 1/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Especificidad de Anticuerpos , Modelos Animales de Enfermedad , Femenino , Cobayas , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Imitación Molecular/inmunología , Miocardio/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/uso terapéutico , Péptidos Cíclicos/química , Péptidos Cíclicos/toxicidad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Lew , Ratas Wistar , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/genética
11.
Thromb Haemost ; 118(2): 351-361, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29378359

RESUMEN

Platelet glycoprotein VI (GPVI) acts as a decisive collagen receptor in atherothrombosis. Besides collagen, injured atherosclerotic plaques expose tissue factor (TF) that triggers fibrin formation. Two recent studies reported that platelet GPVI also functions as fibrin receptor, which would importantly widen the mode of action of GPVI-targeted antithrombotic drugs. We studied the binding of two GPVI fusion proteins to fibrin under static and arterial flow conditions. Fibrin was prepared from purified fibrinogen or generated more physiologically from endogenous fibrinogen by coagulating plasma with thrombin. Fibrin formation was also triggered by exposing TF-coated surfaces or human atherosclerotic plaque slices to arterially flowing blood. By binding studies and advanced optical imaging, we found that recombinant dimeric GPVI-Fc fusion proteins with Fc from either IgG1 (GPVI-Fc1) or IgG2 (GPVI-Fc2) bound to collagen fibres, but neither to fibrin prepared from purified fibrinogen obtained from three suppliers, nor to physiological fibrin formed by thrombin in plasma or triggered by exposing TF or atherosclerotic plaque slices to arterially flowing blood. Our findings do not support a role of dimeric platelet GPVI as receptor for fibrin. This is important for the understanding of plaque-triggered platelet thrombus formation and is clinically relevant for future GPVI-targeting therapies with recombinant GPVI-Fc and anti-GPVI antibodies.


Asunto(s)
Colágeno/metabolismo , Fibrina/metabolismo , Fibrinógeno/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombina/metabolismo , Aterosclerosis/metabolismo , Coagulación Sanguínea , Plaquetas/metabolismo , Humanos , Microscopía Fluorescente , Placa Aterosclerótica/metabolismo , Activación Plaquetaria , Adhesividad Plaquetaria , Agregación Plaquetaria , Unión Proteica , Multimerización de Proteína , Proteínas Recombinantes
12.
Thromb Haemost ; 117(8): 1651-1659, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28569920

RESUMEN

The efficiency of current dual antiplatelet therapy might be further improved by its combination with a glycoprotein (GP) VI-targeting strategy without increasing bleeding. GPVI-Fc, a recombinant dimeric fusion protein binding to plaque collagen and concealing binding sites for platelet GPVI, acts as a lesion-focused antiplatelet drug, and does not increase bleeding in vivo. We investigated, whether GPVI-Fc added in vitro on top of acetylsalicylic acid (ASA), the P2Y12 antagonist ticagrelor, and the fibrinogen receptor antagonist abciximab alone or in combination would increase inhibition of platelet activation by atherosclerotic plaque. Under static conditions, GPVI-Fc inhibited plaque-induced platelet aggregation by 53 %, and increased platelet inhibition by ASA (51 %) and ticagrelor (64 %) to 66 % and 80 %, respectively. Under arterial flow, GPVI-Fc inhibited plaque-induced platelet aggregation by 57 %, and significantly increased platelet inhibition by ASA (28 %) and ticagrelor (47 %) to about 81 % each. The triple combination of GPVI-Fc, ASA and ticagrelor achieved almost complete inhibition of plaque-induced platelet aggregation (93 %). GPVI-Fc alone or in combination with ASA or ticagrelor did not increase closure time measured by the platelet function analyzer (PFA)-200. GPVI-Fc added on top of abciximab, a clinically used anti-fibrinogen receptor antibody which blocks platelet aggregation, strongly inhibited total (81 %) and stable (89 %) platelet adhesion. We conclude that GPVI-Fc added on top of single or dual antiplatelet therapy with ASA and/or a P2Y12 antagonist is likely to improve anti-atherothrombotic protection without increasing bleeding risk. In contrast, the strong inhibition of platelet adhesion by GPVI-Fc in combination with GPIIb/IIIa inhibitors could be harmful.


Asunto(s)
Adenosina/análogos & derivados , Anticuerpos Monoclonales/farmacología , Aspirina/farmacología , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Glicoproteínas/farmacología , Fragmentos Fab de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Placa Aterosclerótica , Inhibidores de Agregación Plaquetaria/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Trombosis/prevención & control , Abciximab , Adenosina/farmacología , Adenosina/toxicidad , Anticuerpos Monoclonales/toxicidad , Aspirina/toxicidad , Plaquetas/metabolismo , Quimioterapia Combinada , Glicoproteínas/toxicidad , Hemorragia/inducido químicamente , Humanos , Fragmentos Fab de Inmunoglobulinas/toxicidad , Fragmentos Fc de Inmunoglobulinas/toxicidad , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/toxicidad , Antagonistas del Receptor Purinérgico P2Y/toxicidad , Trombosis/sangre , Trombosis/patología , Ticagrelor , Factores de Tiempo
13.
J Med Chem ; 49(14): 4116-26, 2006 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-16821772

RESUMEN

Matriptase is an epithelium-derived type II transmembrane serine protease and has been implicated in the activation of substrates such as pro-HGF/SF and pro-uPA, which are likely involved in tumor progression and metastasis. Through screening, we have identified bis-basic secondary amides of sulfonylated 3-amidinophenylalanine as matriptase inhibitors. X-ray analyses of analogues 8 and 31 in complex with matriptase revealed that these inhibitors occupy, in addition to part of the previously described S4-binding site, the cleft formed by the molecular surface and the unique 60 loop of matriptase. Therefore, optimization of the inhibitors included the incorporation of appropriate sulfonyl substituents that could improve binding of these inhibitors into both characteristic matriptase subsites. The most potent derivatives inhibit matriptase highly selective with K(i) values below 5 nM. Molecular modeling revealed that their improved affinity results from interaction with the S4 site of matriptase. Analogues 8 and 59 were studied in an orthotopic xenograft mouse model of prostate cancer. Compared to control, both inhibitors reduced tumor growth, as well as tumor dissemination.


Asunto(s)
Amidas/síntesis química , Amidinas/síntesis química , Fenilalanina/análogos & derivados , Fenilalanina/síntesis química , Serina Endopeptidasas/metabolismo , Inhibidores de Serina Proteinasa/síntesis química , Sulfonas/síntesis química , Amidas/farmacología , Amidinas/farmacología , Animales , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Cinética , Masculino , Ratones , Ratones Desnudos , Modelos Moleculares , Metástasis de la Neoplasia , Fenilalanina/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Serina Endopeptidasas/química , Inhibidores de Serina Proteinasa/farmacología , Relación Estructura-Actividad , Sulfonas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
JACC Basic Transl Sci ; 1(3): 131-142., 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27766315

RESUMEN

To enhance the antithrombotic properties of recombinant glycoprotein VI fragment crystallizable (GPVI-Fc), the authors incubated GPVI-Fc with anti-human Fc antibodies to cross-link the Fc tails of GPVI-Fc. Cross-linking potentiated the inhibition of human plaque- and collagen-induced platelet aggregation by GPVI-Fc under static and flow conditions without increasing bleeding time in vitro. Cross-linking with anti-human-Fc Fab2 was even superior to anti-human-Fc immunoglobulin G (IgG). Advanced optical imaging revealed a continuous sheath-like coverage of collagen fibers by cross-linked GPVI-Fc complexes. Cross-linking of GPVI into oligomeric complexes provides a new, highly effective, and probably safe antithrombotic treatment as it suppresses platelet GPVI-plaque interaction selectively at the site of acute atherothrombosis.

15.
Int J Oncol ; 27(4): 1061-70, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16142324

RESUMEN

Matriptase, also known as membrane-type-serine-protease 1 (MT-SP 1), is a type II transmembrane serine protease involved in the activation of the precursor form of hepatocyte growth factor/scatter factor (pro-HGF/SF). Since HGF/SF is a well-known extracellular signal, which plays a key role in the control of invasive growth, we investigated the effects of matriptase inhibition in cell lines derived from colon (DLD-1) or prostate (PC-3) carcinomas. Biochemical analysis showed that matriptase was very efficient in the proteolytic conversion of the inactive HGF/SF precursor into HGF/SF. Inhibition of endogenous matriptase synthesis in DLD-1 or PC-3 cells by specific small interfering RNAs impaired the conversion of pro-HGF/SF into HGF/SF at the cell surface and inhibited cell scattering upon pro-HGF/SF stimulation. The same effect was observed after treatment of these cells with matriptase inhibitors of the 3-amidinophenylalanine-type, CJ-697 or CJ-730. Inhibition of matriptase significantly reduced invasion of the extracellular matrix as well. Interestingly, this reduction was observed even in the presence of pre-activated HGF/SF. It is concluded that matriptase plays a dual-role in the events unleashing the invasive phenotype, one 'upstream' from the HGF/SF signalling cascade and one 'downstream', most likely at the level of the plasminogen activation system. These data provide a proof of concept for the targeting of matriptase in the search for anti-invasive drugs.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Serina Endopeptidasas/metabolismo , Western Blotting , Dominio Catalítico , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Colágeno/química , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Activación Enzimática , Femenino , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora , Cinética , Laminina/química , Masculino , Modelos Químicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Péptido Hidrolasas/química , Fenilalanina/metabolismo , Precursores de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteoglicanos/química , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA