Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
FASEB J ; 31(2): 761-770, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27836986

RESUMEN

Endothelial thrombomodulin (TM) regulates coagulation and inflammation via several mechanisms, including production of activated protein C (APC). Recombinant APC and soluble fragments of TM (sTM) have been tested in settings associated with insufficiency of the endogenous TM/APC pathway, such as sepsis. We previously designed a fusion protein of TM [single-chain variable fragment antibody (scFv)/TM] targeted to red blood cells (RBCs) to improve pharmacokinetics and antithrombotic effects without increasing bleeding. Here, scFv/TM was studied in mouse models of systemic inflammation and ischemia-reperfusion injury. Injected concomitantly with or before endotoxin, scFv/TM provided more potent protection against liver injury and release of pathological mediators than sTM, showing similar efficacy at up to 50-fold lower doses. scFv/TM provided protection when injected after endotoxin, whereas sTM did not, and augmented APC production by thrombin ∼50-fold more than sTM. However, scFv/TM injected after endotoxin did not reduce thrombin/antithrombin complexes; nor did antibodies that block APC anticoagulant activity suppress the prophylactic anti-inflammatory effect of scFv/TM. Therefore, similar to endogenous TM, RBC-anchored scFv/TM activates several protective pathways. Finally, scFv/TM was more effective at reducing cerebral infarct volume and alleviated neurological deficits than sTM after cerebral ischemia/reperfusion injury. These results indicate that RBC-targeted scFv/TM exerts multifaceted cytoprotective effects and may find utility in systemic and focal inflammatory and ischemic disorders.-Carnemolla, R., Villa, C. H., Greineder, C. F., Zaitseva, S., Patel, K. R., Kowalska, M. A., Atochin, D. N., Cines, D. B., Siegel, D. L., Esmon, C. T., Muzykantov, V. R. Targeting thrombomodulin to circulating red blood cells augments its protective effects in models of endotoxemia and ischemia-reperfusion injury.


Asunto(s)
Endotoxemia/prevención & control , Eritrocitos/metabolismo , Daño por Reperfusión/prevención & control , Trombomodulina/administración & dosificación , Trombomodulina/uso terapéutico , Animales , Inflamación/tratamiento farmacológico , Masculino , Proteínas de la Fusión de la Membrana , Ratones , Ratones Endogámicos C57BL , Trombomodulina/química
2.
FASEB J ; 29(8): 3483-92, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25953848

RESUMEN

Anchoring pharmacologic agents to the vascular lumen has the potential to modulate critical processes at the blood-tissue interface, avoiding many of the off-target effects of systemically circulating agents. We report a novel strategy for endothelial dual targeting of therapeutics, which both enhances drug delivery and enables targeted agents to partner enzymatically to generate enhanced biologic effect. Based on the recent discovery that paired antibodies directed to adjacent epitopes of platelet endothelial cell adhesion molecule (PECAM)-1 stimulate each other's binding, we fused single-chain fragments (scFv) of paired anti-mouse PECAM-1 antibodies to recombinant murine thrombomodulin (TM) and endothelial protein C receptor (EPCR), endothelial membrane proteins that partner in activation of protein C (PC). scFv/TM and scFv/EPCR bound to mouse endothelial PECAM-1 with high affinity (EC50 1.5 and 3.8 nM, respectively), and codelivery induced a 5-fold increase in PC activation not seen when TM and EPCR are anchored to distinct cell adhesion molecules. In a mouse model of acute lung injury, dual targeting reduces both the expression of lung inflammatory markers and trans-endothelial protein leak by as much as 40%, as compared to either agent alone. These findings provide proof of principle for endothelial dual targeting, an approach with numerous potential biomedical applications.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Preparaciones Farmacéuticas/administración & dosificación , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos/métodos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Epítopos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteína C/metabolismo , Receptores de Superficie Celular/metabolismo , Trombomodulina/metabolismo
3.
Blood ; 122(9): 1565-75, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23798715

RESUMEN

Despite continued achievements in antithrombotic pharmacotherapy, difficulties remain in managing patients at high risk for both thrombosis and hemorrhage. Utility of antithrombotic agents (ATAs) in these settings is restricted by inadequate pharmacokinetics and narrow therapeutic indices. Use of advanced drug delivery systems (ADDSs) may help to circumvent these problems. Various nanocarriers, affinity ligands, and polymer coatings provide ADDSs that have the potential to help optimize ATA pharmacokinetics, target drug delivery to sites of thrombosis, and sense pathologic changes in the vascular microenvironment, such as altered hemodynamic forces, expression of inflammatory markers, and structural differences between mature hemostatic and growing pathological clots. Delivery of ATAs using biomimetic synthetic carriers, host blood cells, and recombinant fusion proteins that are activated preferentially at sites of thrombus development has shown promising outcomes in preclinical models. Further development and translation of ADDSs that spare hemostatic fibrin clots hold promise for extending the utility of ATAs in the management of acute thrombotic disorders through rapid, transient, and targeted thromboprophylaxis. If the potential benefit of this technology is to be realized, a systematic and concerted effort is required to develop clinical trials and translate the use of ADDSs to the clinical arena.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Fibrinolíticos/administración & dosificación , Trombosis/tratamiento farmacológico , Animales , Disponibilidad Biológica , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Fibrinolíticos/farmacocinética , Semivida , Humanos
4.
Blood ; 119(20): 4779-85, 2012 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-22493296

RESUMEN

Thrombin generates fibrin and activates platelets and endothelium, causing thrombosis and inflammation. Endothelial thrombomodulin (TM) changes thrombin's substrate specificity toward cleavage of plasma protein C into activated protein C (APC), which opposes its thrombotic and inflammatory activities. Endogenous TM activity is suppressed in pathologic conditions, and antithrombotic interventions involving soluble TM are limited by rapid blood clearance. To overcome this problem, we fused TM with a single chain fragment (scFv) of an antibody targeted to red blood cells. scFv/TM catalyzes thrombin-mediated generation of activated protein C and binds to circulating RBCs without apparent damage, thereby prolonging its circulation time and bioavailability orders of magnitude compared with soluble TM. In animal models, a single dose of scFv/TM, but not soluble TM, prevents platelet activation and vascular occlusion by clots. Thus, scFv/TM serves as a prodrug and provides thromboprophylaxis at low doses (0.15 mg/kg) via multifaceted mechanisms inhibiting platelets and coagulation.


Asunto(s)
Quimioprevención/métodos , Sistemas de Liberación de Medicamentos/métodos , Eritrocitos/efectos de los fármacos , Trombomodulina/administración & dosificación , Trombosis/prevención & control , Animales , Células Cultivadas , Drosophila , Eritrocitos/metabolismo , Eritrocitos/fisiología , Humanos , Ratones , Modelos Biológicos , Terapia Molecular Dirigida/métodos , Unión Proteica , Proteína C/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Anticuerpos de Cadena Única/administración & dosificación , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/metabolismo , Anticuerpos de Cadena Única/uso terapéutico , Trombomodulina/química , Trombomodulina/metabolismo , Trombomodulina/uso terapéutico
5.
J Pharmacol Exp Ther ; 347(2): 339-45, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23965383

RESUMEN

Thrombomodulin (TM) is a glycoprotein normally present in the membrane of endothelial cells that binds thrombin and changes its substrate specificity to produce activated protein C (APC) that has antithrombotic and anti-inflammatory features. To compensate for loss of endogenous TM in pathology, we have fused recombinant TM with single chain variable fragment (scFv) of an antibody to mouse platelet endothelial cell adhesion molecule-1 (PECAM). This fusion, anti-PECAM scFv/TM, anchors on the endothelium, stimulates APC production, and provides therapeutic benefits superior to sTM in animal models of acute thrombosis and inflammation. However, in conditions of oxidative stress typical of vascular inflammation, TM is inactivated via oxidation of the methionine 388 (M388) residue. Capitalizing on the reports that M388L mutation renders TM resistant to oxidative inactivation, in this study we designed a mutant anti-PECAM scFv/TM M388L. This mutant has the same APC-producing capacity and binding to target cells, yet, in contrast to wild-type fusion, it retains APC-producing activity in an oxidizing environment in vitro and in vivo. Therefore, oxidant resistant mutant anti-PECAM scFv/TM M388L is a preferable targeted biotherapeutic to compensate for loss of antithrombotic and anti-inflammatory TM functions in the context of vascular oxidative stress.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/farmacología , Proteína C/biosíntesis , Proteínas Recombinantes de Fusión/farmacología , Trombomodulina/genética , Animales , Línea Celular , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Tiempo de Tromboplastina Parcial , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Anticuerpos de Cadena Única/genética , Especificidad por Sustrato , Trombina/metabolismo
6.
J Lipid Res ; 53(5): 951-963, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22402133

RESUMEN

Plasma HDL levels are inversely associated with atherosclerosis. Inbred mouse strains differ in plasma HDL levels and susceptibility to atherosclerosis. Atherosclerosis-susceptible C57BL/6J mice possess plasma HDL levels 2-fold lower than atherosclerosis-resistant FVB/NJ mice. Polymorphisms have been previously identified between the two mouse strains in the major HDL apolipoproteins, ApoA-I and ApoA-II, which may affect their function on HDL. To begin to understand the HDL differences, we here report on a detailed comparison of the lipid-associated functions of the two mouse ApoA-I proteins. We demonstrate that these polymorphisms significantly alter the protein self-association properties, the ability of the proteins to clear lipid micelles from solution, and their binding affinity for mature mouse HDL. The changes in lipid binding do not appear to alter the ability of the protein to promote cholesterol efflux from cells or the formation of nascent HDL from primary hepatocytes. These apolipoprotein polymorphisms do not change the rate at which HDL protein or cholesterol are catabolized in vivo. Although the presence of the polymorphisms in ApoA-I alters important factors in HDL formation, the basis for the differences in the HDL plasma levels observed in the various mouse strains is more complex and requires additional investigation.


Asunto(s)
Apolipoproteína A-I/genética , HDL-Colesterol/metabolismo , Polimorfismo Genético , Animales , Apolipoproteína A-I/metabolismo , Transporte Biológico/genética , Línea Celular , HDL-Colesterol/biosíntesis , HDL-Colesterol/sangre , Activación Enzimática/genética , Femenino , Humanos , Hígado/metabolismo , Ratones , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo
7.
IUBMB Life ; 63(8): 632-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21766410

RESUMEN

In this review we discuss the limited efficacy for current pharmacological agents used in prophylaxis and treatment of thrombosis and highlight targeted delivery of anti-thrombotic agents to fibrin, platelets, red blood cells and endothelium.


Asunto(s)
Antitrombinas/administración & dosificación , Sistemas de Liberación de Medicamentos , Trombosis/tratamiento farmacológico , Antitrombinas/uso terapéutico , Humanos
8.
Semin Thromb Hemost ; 36(3): 332-42, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20490983

RESUMEN

The endothelium is one of the key targets for pharmacological interventions in oxidative stress and thrombosis, two conditions that are notoriously difficult to treat due to limited efficacy and precision of action of current drugs. Design of molecular and nano-devices that deliver potent antioxidant and antithrombotic therapeutic enzymes to the endothelium holds promise to improve the potency, localization, timing, specificity, safety, and mechanistic precision of these interventions. In particular, cell adhesion molecules expressed on the surface of resting and pathologically altered endothelial cells can be used for drug delivery to the endothelial surface (preferable for thrombolytics) and into intracellular compartments (preferable for antioxidants). Drug delivery platforms including protein conjugates, recombinant fusion constructs, and stealth polymer carriers designed to target these drugs to endothelium are reviewed in this article.


Asunto(s)
Antioxidantes/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Endotelio Vascular/efectos de los fármacos , Fibrinolíticos/administración & dosificación , Humanos
9.
J Clin Invest ; 113(5): 764-73, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14991075

RESUMEN

Accelerated atherosclerosis is a major cause of morbidity and death in insulin-resistant states such as obesity and the metabolic syndrome, but the underlying mechanisms are poorly understood. We show that macrophages from obese (ob/ob) mice have increased binding and uptake of oxidized LDL, in part due to a post-transcriptional increase in CD36 protein. Macrophages from ob/ob mice are also insulin resistant, as shown by reduced expression and signaling of insulin receptors. Three lines of evidence indicate that the increase in CD36 is caused by defective insulin signaling: (a) Treatment of wild-type macrophages with LY294002, an inhibitor of insulin signaling via PI3K, results in an increase in CD36; (b) insulin receptor knockout macrophages show a post-transcriptional increase in CD36 protein; and (c) administration of thiazolidinediones to intact ob/ob mice and ob/ob, LDL receptor-deficient mice results in a reversal of macrophage insulin receptor defects and decreases CD36 protein. The last finding contrasts with the increase in CD36 that results from treatment of macrophages with these drugs ex vivo. The results suggest that defective macrophage insulin signaling predisposes to foam cell formation and atherosclerosis in insulin-resistant states and that this is reversed in vivo by treatment with PPAR-gamma activators.


Asunto(s)
Antígenos CD36/biosíntesis , Insulina/metabolismo , Macrófagos/inmunología , Transducción de Señal , Animales , Northern Blotting , Western Blotting , Cromonas/farmacología , Electroforesis en Gel de Poliacrilamida , Inhibidores Enzimáticos/farmacología , Células Espumosas/metabolismo , Glutatión/metabolismo , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Morfolinas/farmacología , Oxígeno/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Unión Proteica , Receptores Citoplasmáticos y Nucleares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rosiglitazona , Tiazolidinedionas/farmacología , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética , Tirosina/metabolismo
10.
PLoS One ; 8(11): e80110, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244621

RESUMEN

The use of targeted therapeutics to replenish pathologically deficient proteins on the luminal endothelial membrane has the potential to revolutionize emergency and cardiovascular medicine. Untargeted recombinant proteins, like activated protein C (APC) and thrombomodulin (TM), have demonstrated beneficial effects in acute vascular disorders, but have failed to have a major impact on clinical care. We recently reported that TM fused with an scFv antibody fragment to platelet endothelial cell adhesion molecule-1 (PECAM-1) exerts therapeutic effects superior to untargeted TM. PECAM-1 is localized to cell-cell junctions, however, whereas the endothelial protein C receptor (EPCR), the key co-factor of TM/APC, is exposed in the apical membrane. Here we tested whether anchoring TM to the intercellular adhesion molecule (ICAM-1) favors scFv/TM collaboration with EPCR. Indeed: i) endothelial targeting scFv/TM to ICAM-1 provides ~15-fold greater activation of protein C than its PECAM-targeted counterpart; ii) blocking EPCR reduces protein C activation by scFv/TM anchored to endothelial ICAM-1, but not PECAM-1; and iii) anti-ICAM scFv/TM fusion provides more profound anti-inflammatory effects than anti-PECAM scFv/TM in a mouse model of acute lung injury. These findings, obtained using new translational constructs, emphasize the importance of targeting protein therapeutics to the proper surface determinant, in order to optimize their microenvironment and beneficial effects.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Antígenos CD/inmunología , Inmunoconjugados/inmunología , Molécula 1 de Adhesión Intercelular/inmunología , Receptores de Superficie Celular/inmunología , Anticuerpos de Cadena Única/inmunología , Trombomodulina/metabolismo , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Animales , Línea Celular , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Receptor de Proteína C Endotelial , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Endotelio Vascular/patología , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacología , Uniones Intercelulares/efectos de los fármacos , Uniones Intercelulares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Proteína C/agonistas , Proteína C/metabolismo , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/farmacología , Trombomodulina/química , Trombomodulina/inmunología
11.
J Immunol Methods ; 384(1-2): 21-4, 2012 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-22750541

RESUMEN

Thrombomodulin-bound thrombin cleaves protein C (PC) zymogen in blood plasma producing activated protein C (APC), which exerts anti-coagulant, anti-inflammatory, anti-apoptotic and CNS-protective effects. Recombinant APC and thrombomodulin (TM) are both in clinical studies for management of acute conditions including sepsis. Methods that permit accurate measurement of APC in plasma are needed for clinical monitoring and mechanistic studies in animal models. However, the two existing methods require either long incubation periods with substrate, resulting in high background or they also recognize protein C inhibitor (PCI) complexed with APC (APC:PCI), which convolutes analysis of the amount of APC generated. Here we describe a robust quantitative in vivo assay that measures APC generation at both low levels of human protein C seen in chronic inflammatory disease and at physiological levels that shows a >99% fit with in vitro data.


Asunto(s)
Precursores Enzimáticos/metabolismo , Proteína C/metabolismo , Trombina/metabolismo , Trombomodulina/metabolismo , Animales , Bovinos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Precursores Enzimáticos/sangre , Ensayo de Inmunoadsorción Enzimática , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Trombina/farmacología
12.
J Biol Chem ; 283(23): 15779-88, 2008 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-18385132

RESUMEN

Humans have two major high density lipoprotein (HDL) sub-fractions, HDL(2) and HDL(3), whereas mice have a monodisperse HDL profile. Epidemiological evidence has suggested that HDL(2) is more atheroprotective; however, currently there is no direct experimental evidence to support this postulate. The amino acid sequence of apoA-I is a primary determinant of HDL subclass formation. The majority of the alpha-helical repeats in human apoA-I are proline-punctuated. A notable exception is the boundary between helices 7 and 8, which is located in the transitional segment between the stable N-terminal domain and the C-terminal hydrophobic domain. In this study we ask whether the substitution of a proline-containing sequence (PCS) separating other helices in human apoA-I for the non-proline-containing sequence (NPCS) between helices 7 and 8 (residues 184-190) influences HDL subclass association. The human apoA-I mutant with PCS2 replacing NPCS preferentially bound to HDL(2). In contrast, the mutant where PCS3 replaced NPCS preferentially associated with HDL(3). Thus, the specific amino acid sequence between helices 7 and 8 influences HDL subclass association. The wild-type and mutant proteins exhibited similar physicochemical properties except that the two mutants displayed greater lipid-associated stability versus wild-type human apoA-I. These results focus new attention on the influence of the boundary between helices 7 and 8 on the properties of apoA-I. The expression of these mutants in mice may result in the preferential generation of HDL(2) or HDL(3) and allow us to examine experimentally the anti-atherogenicity of the HDL subclasses.


Asunto(s)
Apolipoproteína A-I/química , Lipoproteínas HDL2/química , Lipoproteínas HDL3/química , Secuencia de Aminoácidos/genética , Animales , Apolipoproteína A-I/genética , Apolipoproteína A-I/metabolismo , Humanos , Lipoproteínas HDL2/genética , Lipoproteínas HDL2/metabolismo , Lipoproteínas HDL3/genética , Lipoproteínas HDL3/metabolismo , Ratones , Mutación , Unión Proteica/genética , Estructura Secundaria de Proteína/genética , Estructura Terciaria de Proteína/genética , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA