Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 43(6): 1031-1040, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37078286

RESUMEN

BACKGROUND: Current clinical imaging of thromboembolic diseases often relies on indirect detection of thrombi, which may delay diagnosis and ultimately the institution of beneficial, potentially lifesaving treatment. Therefore, the development of targeting tools that facilitate the rapid, specific, and direct imaging of thrombi using molecular imaging is highly sought after. One potential molecular target is FXIIa (factor XIIa), which initiates the intrinsic coagulation pathway but also activates the kallikrein-kinin system, thereby initiating coagulation and inflammatory/immune responses. As FXII (factor XII) is dispensable for normal hemostasis, its activated form (FXIIa) represents an ideal molecular target for diagnostic and therapeutic approaches, the latter combining diagnosis/identification of thrombi and effective antithrombotic therapy. METHODS: We conjugated an FXIIa-specific antibody, 3F7, to a near-infrared (NIR) fluorophore and demonstrated binding to FeCl3-induced carotid thrombosis with 3-dimensional fluorescence emission computed tomography/computed tomography and 2-dimensional fluorescence imaging. We further demonstrated ex vivo imaging of thromboplastin-induced pulmonary embolism and detection of FXIIa in human thrombi produced in vitro. RESULTS: We demonstrated imaging of carotid thrombosis by fluorescence emission computed tomography/computed tomography and measured a significant fold increase in signal between healthy and control vessels from mice injected with 3F7-NIR compared with mice injected with nontargeted probe (P=0.002) ex vivo. In a model of pulmonary embolism, we measured increased NIR signal in lungs from mice injected with 3F7-NIR compared with mice injected with nontargeted probe (P=0.0008) and healthy lungs from mice injected with 3F7-NIR (P=0.021). CONCLUSIONS: Overall, we demonstrate that FXIIa targeting is highly suitable for the specific detection of venous and arterial thrombi. This approach will allow direct, specific, and early imaging of thrombosis in preclinical imaging modalities and may facilitate monitoring of antithrombotic treatment in vivo.


Asunto(s)
Trombosis de las Arterias Carótidas , Embolia Pulmonar , Trombosis , Ratones , Humanos , Animales , Coagulación Sanguínea , Trombosis/diagnóstico por imagen , Factor XII/metabolismo , Factor XIIa/metabolismo , Imagen Molecular
2.
J Immunol ; 197(11): 4392-4402, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27807194

RESUMEN

G-CSF is a hemopoietic growth factor that has a role in steady state granulopoiesis, as well as in mature neutrophil activation and function. G-CSF- and G-CSF receptor-deficient mice are profoundly protected in several models of rheumatoid arthritis, and Ab blockade of G-CSF also protects against disease. To further investigate the actions of blocking G-CSF/G-CSF receptor signaling in inflammatory disease, and as a prelude to human studies of the same approach, we developed a neutralizing mAb to the murine G-CSF receptor, which potently antagonizes binding of murine G-CSF and thereby inhibits STAT3 phosphorylation and G-CSF receptor signaling. Anti-G-CSF receptor rapidly halted the progression of established disease in collagen Ab-induced arthritis in mice. Neutrophil accumulation in joints was inhibited, without rendering animals neutropenic, suggesting an effect of G-CSF receptor blockade on neutrophil homing to inflammatory sites. Consistent with this, neutrophils in the blood and arthritic joints of anti-G-CSF receptor-treated mice showed alterations in cell adhesion receptors, with reduced CXCR2 and increased CD62L expression. Furthermore, blocking neutrophil trafficking with anti-G-CSF receptor suppressed local production of proinflammatory cytokines (IL-1ß, IL-6) and chemokines (KC, MCP-1) known to drive tissue damage. Differential gene expression analysis of joint neutrophils showed a switch away from an inflammatory phenotype following anti-G-CSF receptor therapy in collagen Ab-induced arthritis. Importantly, G-CSF receptor blockade did not adversely affect viral clearance during influenza infection in mice. To our knowledge, we describe for the first time the effect of G-CSF receptor blockade in a therapeutic model of inflammatory joint disease and provide support for pursuing this therapeutic approach in treating neutrophil-associated inflammatory diseases.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Artritis Experimental/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/inmunología , Receptores de Factor Estimulante de Colonias de Granulocito/antagonistas & inhibidores , Animales , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Citocinas/genética , Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/inmunología , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Articulaciones/inmunología , Articulaciones/patología , Masculino , Ratones , Ratones Noqueados , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Neutrófilos/patología , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Receptores de Factor Estimulante de Colonias de Granulocito/inmunología
4.
Methods Mol Biol ; 2702: 433-449, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37679634

RESUMEN

We have previously published protocols for high-throughput IgG reformatting and expression, that enable rapid reformatting of phage-displayed antibody Fab fragments into a single dual expression vector for full IgG expression in Expi293F cells (Chen et al. Nucleic Acids Res 42:e26, 2014; Chen et al. Methods in Molecular Biology, vol 1701, 2018). However, when working with phage clones from a naïve library containing highly diverse N-terminal sequences, where the 5' PCR primers bind, the PCR step can become cumbersome. To overcome this limitation, we have investigated and found that the C-terminal 7 amino acid residues of the human antibody VH1 secretion signal can be replaced with those from ompA or pelB bacterial signals to form hybrid signal sequences that can drive strong IgG expression in Expi293F cells. The use of such hybrid signals allows any Fab fragment in the library to be amplified and cloned into the IgG expression vector using only a single 5' PCR primer targeting the bacterial secretion signal of the light or heavy chain, thus dramatically simplifying the IgG reformatting workflow.


Asunto(s)
Bacteriófagos , Humanos , Secreciones Corporales , Técnicas de Visualización de Superficie Celular , Fragmentos Fab de Inmunoglobulinas/genética , Tecnología , Inmunoglobulina G/genética
5.
Hum Antibodies ; 15(3): 71-80, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17065738

RESUMEN

Previously we identified circulating autoantibodies in high titre in the serum of a patient with biopsy-proven discoid lupus erythematosus reactive to serine/threonine phosphoepitopes of the novel mitotic chromosomal autoantigen we named MCA1. MCA1-reactive antibodies have subsequently been demonstrated to bind to the modified histone H3 (H3K9me3S10ph). In this study we utilised Epstein Barr virus (EBV) infection of peripheral blood mononuclear cells collected from this patient to immortalise B-lymphocytes producing MCA1-reactive antibodies. We found MCA1-reactive antibody production by the EBV-immortalised B-lymphocytes unstable in culture. We discuss the possibilities of producing MCA-1-reactive reagents through protein engineering methods (variable or complementarity-determining region grafting) and to modulate antibody affinity through library technologies (such as phage, yeast or ribosome display). Clinical studies have shown that carcinogenesis is most often linked to the development of proliferative abnormalities and proliferative activity has prognostic significance in a variety of human tumours. Here we demonstrate the potential utility of antibodies reactive to H3K9me3S10ph (MCA1), which is only detected on mitotic chromatin, as a marker for cell proliferation in FACS analysis, tissue section staining and in determination of mitotic indices.


Asunto(s)
Autoantígenos/metabolismo , Biomarcadores/metabolismo , Histonas/metabolismo , Serina/metabolismo , Anciano , Animales , Separación Celular , Femenino , Citometría de Flujo , Células HeLa , Histonas/química , Humanos , Lupus Eritematoso Sistémico/metabolismo , Fosforilación , Ratas
6.
MAbs ; 8(3): 436-53, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26651396

RESUMEN

The ß common-signaling cytokines interleukin (IL)-3, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-5 stimulate pro-inflammatory activities of haematopoietic cells via a receptor complex incorporating cytokine-specific α and shared ß common (ßc, CD131) receptor. Evidence from animal models and recent clinical trials demonstrate that these cytokines are critical mediators of the pathogenesis of inflammatory airway disease such as asthma. However, no therapeutic agents, other than steroids, that specifically and effectively target inflammation mediated by all 3 of these cytokines exist. We employed phage display technology to identify and optimize a novel, human monoclonal antibody (CSL311) that binds to a unique epitope that is specific to the cytokine-binding site of the human ßc receptor. The binding epitope of CSL311 on the ßc receptor was defined by X-ray crystallography and site-directed mutagenesis. CSL311 has picomolar binding affinity for the human ßc receptor, and at therapeutic concentrations is a highly potent antagonist of the combined activities of IL-3, GM-CSF and IL-5 on primary eosinophil survival in vitro. Importantly, CSL311 inhibited the survival of inflammatory cells present in induced sputum from human allergic asthmatic subjects undergoing allergen bronchoprovocation. Due to its high potency and ability to simultaneously suppress the activity of all 3 ß common cytokines, CSL311 may provide a new strategy for the treatment of chronic inflammatory diseases where the human ßc receptor is central to pathogenesis. The coordinates for the ßc/CSL311 Fab complex structure have been deposited with the RCSB Protein Data Bank (PDB 5DWU).


Asunto(s)
Anticuerpos Monoclonales de Origen Murino , Subunidad beta Común de los Receptores de Citocinas , Epítopos , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Interleucina-3 , Interleucina-5 , Animales , Anticuerpos Monoclonales de Origen Murino/química , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Complejo Antígeno-Anticuerpo/química , Complejo Antígeno-Anticuerpo/inmunología , Asma/tratamiento farmacológico , Asma/inmunología , Asma/patología , Cristalografía por Rayos X , Subunidad beta Común de los Receptores de Citocinas/química , Subunidad beta Común de los Receptores de Citocinas/inmunología , Eosinófilos/inmunología , Eosinófilos/patología , Epítopos/química , Epítopos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Interleucina-3/antagonistas & inhibidores , Interleucina-3/inmunología , Interleucina-5/antagonistas & inhibidores , Interleucina-5/inmunología , Ratones
7.
Sci Transl Med ; 6(222): 222ra17, 2014 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-24500405

RESUMEN

Currently used anticoagulants prevent thrombosis but increase bleeding. We show an anticoagulation therapy without bleeding risk based on a plasma protease factor XII function-neutralizing antibody. We screened for antibodies against activated factor XII (FXIIa) using phage display and demonstrated that recombinant fully human antibody 3F7 binds into the FXIIa enzymatic pocket. 3F7 interfered with FXIIa-mediated coagulation, abolished thrombus formation under flow, and blocked experimental thrombosis in mice and rabbits. We adapted an extracorporeal membrane oxygenation (ECMO) cardiopulmonary bypass system used for infant therapy to analyze clinical applicability of 3F7 in rabbits. 3F7 provided thromboprotection as efficiently as heparin, and both drugs prevented fibrin deposition and thrombosis within the extracorporeal circuit. Unlike heparin, 3F7 treatment did not impair the hemostatic capacity and did not increase bleeding from wounds. These data establish that targeting of FXIIa is a safe mode of thromboprotection in bypass systems, and provide a clinically relevant anticoagulation strategy that is not complicated by excess bleeding.


Asunto(s)
Anticuerpos/farmacología , Anticuerpos/uso terapéutico , Circulación Extracorporea/efectos adversos , Factor XIIa/antagonistas & inhibidores , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Trombosis/prevención & control , Animales , Derivación Arteriovenosa Quirúrgica , Coagulación Sanguínea/efectos de los fármacos , Dominio Catalítico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Mapeo Epitopo , Factor XIIa/metabolismo , Heparina/farmacología , Heparina/uso terapéutico , Humanos , Ratones , Conejos , Factores de Riesgo , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA