Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Clin Exp Immunol ; 200(1): 1-11, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31853959

RESUMEN

Carbamylation is a post-translational modification that can be detected on a range of proteins, including immunoglobulin (Ig)G, in several clinical conditions. Carbamylated IgG (ca-IgG) was reported to lose its capacity to trigger complement activation, but the mechanism remains unclear. Because C1q binds with high affinity to hexameric IgG, we analyzed whether carbamylation of IgG affects binding of C1q, hexamerization and complement-dependent cytotoxicity (CDC). Synovial tissues of rheumatoid arthritis (RA) patients were analyzed for the presence of ca-IgG in vivo. Synovial tissues from RA patients were analyzed for the presence of ca-IgG using mass spectrometry (MS). Monomeric or hexameric antibodies were carbamylated in vitro and quality in solution was controlled. The capacity of ca-IgG to activate complement was analyzed in enzyme-linked immunosorbent (ELISAs) and cellular CDC assays. Using MS, we identified ca-IgG to be present in the joints of RA patients. Using in vitro carbamylated antibodies, we observed that ca-IgG lost its capacity to activate complement in both solid-phase and CDC assays. Mixing ca-IgG with non-modified IgG did not result in effective inhibition of complement activation by ca-IgG. Carbamylation of both monomeric IgG and preformed hexameric IgG greatly impaired the capacity to trigger complement activation. Furthermore, upon carbamylation, the preformed hexameric IgG dissociated into monomeric IgG in solution, indicating that carbamylation influences both hexamerization and C1q binding. In conclusion, ca-IgG can be detected in vivo and has a strongly reduced capacity to activate complement which is, in part, mediated through a reduced ability to form hexamers.


Asunto(s)
Artritis Reumatoide/inmunología , Activación de Complemento/inmunología , Complemento C1q/inmunología , Inmunoglobulina G/inmunología , Anciano , Secuencia de Aminoácidos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Artritis Reumatoide/metabolismo , Línea Celular Tumoral , Complemento C1q/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Carbamilación de Proteína/inmunología , Multimerización de Proteína/inmunología , Líquido Sinovial/inmunología , Líquido Sinovial/metabolismo , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo
2.
Scand J Immunol ; 86(4): 196-206, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28708284

RESUMEN

Human γδ T cells are innate-like T cells which are able to kill a broad range of tumour cells and thus may have potential for cancer immunotherapy. The activating receptor natural killer group 2 member D (NKG2D) plays a key role in regulating immune responses driven by γδ T cells. Here, we explored whether recombinant immunoligands consisting of a CD20 single-chain fragment variable (scFv) linked to a NKG2D ligand, either MHC class I chain-related protein A (MICA) or UL16 binding protein 2 (ULBP2), could be employed to engage γδ T cells for tumour cell killing. The two immunoligands, designated MICA:7D8 and ULBP2:7D8, respectively, enhanced cytotoxicity of ex vivo-expanded γδ T cells against CD20-positive lymphoma cells. Both Vδ1 and Vδ2 γδ T cells were triggered by MICA:7D8 or ULBP2:7D8. Killing of CD20-negative tumour cells was not induced by the immunoligands, indicating their antigen specificity. MICA:7D8 and ULBP2:7D8 acted in a dose-dependent manner and induced cytotoxicity at nanomolar concentrations. Importantly, chronic lymphocytic leukaemia (CLL) cells isolated from patients were sensitized by the two immunoligands for γδ T cell cytotoxicity. In a combination approach, the immunoligands were combined with bromohydrin pyrophosphate (BrHPP), an agonist for Vδ2 γδ T cells, which further enhanced the efficacy in target cell killing. Thus, employing tumour-directed recombinant immunoligands which engage NKG2D may represent an attractive strategy to enhance antitumour cytotoxicity of γδ T cells.


Asunto(s)
Antígenos CD20/metabolismo , Citotoxicidad Inmunológica , Inmunoterapia/métodos , Linfoma/terapia , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Anticuerpos de Cadena Única/uso terapéutico , Linfocitos T/fisiología , Antígenos CD20/inmunología , Difosfatos/uso terapéutico , Quimioterapia Combinada , Proteínas Ligadas a GPI/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Inmunización , Péptidos y Proteínas de Señalización Intercelular/genética , Linfoma/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Anticuerpos de Cadena Única/genética , Células Tumorales Cultivadas
3.
Arch Dermatol Res ; 298(9): 449-55, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17091277

RESUMEN

CD4(+) T cells, in activated or malignant form, are involved in a number of diseases including inflammatory skin diseases such as psoriasis, and T cell lymphomas such as the majority of cutaneous T cell lymphomas (CTCL). Targeting CD4 with an antibody that inhibits and/or eliminates disease-driving T cells in situ may therefore be a useful approach in the treatment of inflammatory and malignant skin diseases. Depletion of CD4(+) T cells in intact inflamed human skin tissue by Zanolimumab, a fully human therapeutic monoclonal antibody (IgG1, kappa) against CD4, was studied in a human psoriasis xenograft mouse model. Zanolimumab treatment was shown to induce a significant reduction in the numbers of inflammatory mononuclear cells in upper dermis. This reduction in inflammatory mononuclear cells in situ was primarily due to a significant reduction in the numbers of skin-infiltrating CD4(+), but not CD8(+) CD3(+) T cells. The capacity of Zanolimumab to deplete the CD4(+) T cells in the skin may be of importance in diseases where CD4(+) T cells play a central role. Indeed, in a phase II clinical trial Zanolimumab has shown a dose-dependent clinical response in patients with CTCL and the antibody is currently in a phase III clinical trial for CTCL, a disease for which there is no safe and effective treatment available today.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Piel/efectos de los fármacos , Animales , Anticuerpos Monoclonales Humanizados , Biopsia , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/patología , Modelos Animales de Enfermedad , Humanos , Linfoma Cutáneo de Células T/tratamiento farmacológico , Ratones , Ratones SCID , Psoriasis/tratamiento farmacológico , Piel/citología , Trasplante Heterólogo
4.
Biosens Bioelectron ; 87: 388-395, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27589401

RESUMEN

Biolayer interferometry (BLI) is a well-established optical label-free technique to study biomolecular interactions. Here we describe for the first time a cell-based BLI (cBLI) application that allows label-free real-time monitoring of signal transduction in living cells. Human A431 epidermoid carcinoma cells were captured onto collagen-coated biosensors and serum-starved, followed by exposure to agonistic compounds targeting various receptors, while recording the cBLI signal. Stimulation of the epidermal growth factor receptor (EGFR) with EGF, the ß2-adrenoceptor with dopamine, or the hepatocyte growth factor receptor (HGFR/c-MET) with an agonistic antibody resulted in distinct cBLI signal patterns. We show that the mechanism underlying the observed changes in cBLI signal is mediated by rearrangement of the actin cytoskeleton, a process referred to as dynamic mass redistribution (DMR). A panel of ligand-binding blocking and non-blocking anti-EGFR antibodies was used to demonstrate that this novel BLI application can be efficiently used as a label-free cellular assay for compound screening and characterization.


Asunto(s)
Técnicas Biosensibles/métodos , Interferometría/métodos , Línea Celular Tumoral , Células Inmovilizadas/citología , Células Inmovilizadas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Células Epidérmicas , Epidermis/metabolismo , Receptores ErbB/agonistas , Receptores ErbB/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-met/metabolismo , Receptores Adrenérgicos beta 2/metabolismo
5.
Leukemia ; 27(1): 190-201, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22660187

RESUMEN

Bispecific antibodies (bsab) offer a promising approach for optimizing antibody-based therapies. In the present study, [(CD20)(2)xCD16], a recombinant CD20- and CD16-directed bsab in the tribody format, was designed to optimize recruitment of FcγRIII (CD16)-positive effector cells. [(CD20)(2)xCD16] retained the antigen specificities of the parental monoclonal antibodies and binding to FcγRIIIa was not compromised by the F/V polymorphism at amino-acid position 158. [(CD20)(2)xCD16] mediated potent lysis of lymphoma cell lines and freshly isolated tumor cells from patients, even at low picomolar concentrations (∼10 pM). Irrespective of the CD16a allotype, potency as well as efficacy of lysis obtained with the tribody was significantly higher than lysis triggered by rituximab. Tumor cell killing also occurred when autologous NK cells were used as effector cells. Compared with rituximab, the tribody demonstrated depletion of autologous B cells in ex vivo whole blood assays at 100-fold lower antibody concentration. In mice with a reconstituted humanized hematopoietic system, established by transplantation of human CD34-positive cord blood cells, this novel tribody significantly depleted autologous human B cells. Thus, tribodies such as [(CD20)(2)xCD16], recruiting CD16-positive effector cells, may represent promising candidates for clinical development.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD20/inmunología , Leucemia de Células B/terapia , Linfoma de Células B/terapia , Receptores de IgG/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Animales Recién Nacidos , Especificidad de Anticuerpos , Femenino , Sangre Fetal/citología , Sangre Fetal/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Leucemia de Células B/inmunología , Depleción Linfocítica , Linfoma de Células B/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Receptores de IgG/metabolismo
6.
J Immunol Methods ; 371(1-2): 122-33, 2011 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-21756911

RESUMEN

Antibody-drug conjugates (ADC) represent promising agents for targeted cancer therapy. To allow rational selection of human antibodies with favorable characteristics for ADC development a screening tool was designed obviating the need of preparing individual covalently linked conjugates. Therefore, α-kappa-ETA' was designed as a fusion protein consisting of a human kappa light chain binding antibody fragment and a truncated version of Pseudomonas exotoxin A. α-kappa-ETA' specifically bound to human kappa light chains of human or human-mouse chimeric antibodies and Fab fragments. Antibody-redirected α-kappa-ETA' specifically inhibited proliferation of antigen-expressing cell lines at low toxin and antibody concentrations. Selected antibodies that efficiently delivered α-kappa-ETA' in the novel assay system were used to generate scFv-based covalently linked immunotoxins. These molecules efficiently triggered apoptosis of target cells, indicating that antibodies identified in our assay system can be converted to functional immunoconjugates. Finally, a panel of human epidermal growth factor receptor (EGFR) antibodies was screened--demonstrating favorable characteristics with antibody 2F8. These data suggest that antibodies with potential for Pseudomonas exotoxin A-based ADC development can be identified using the novel α-kappa-ETA' conjugate.


Asunto(s)
ADP Ribosa Transferasas/inmunología , Toxinas Bacterianas/inmunología , Exotoxinas/inmunología , Cadenas kappa de Inmunoglobulina/aislamiento & purificación , Inmunotoxinas/aislamiento & purificación , Factores de Virulencia/inmunología , ADP Ribosa Transferasas/uso terapéutico , Animales , Toxinas Bacterianas/uso terapéutico , Línea Celular , Citotoxicidad Inmunológica , Ensayo de Inmunoadsorción Enzimática , Receptores ErbB/inmunología , Exotoxinas/uso terapéutico , Citometría de Flujo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Cadenas kappa de Inmunoglobulina/química , Cadenas kappa de Inmunoglobulina/uso terapéutico , Inmunotoxinas/química , Inmunotoxinas/uso terapéutico , Ratones , Modelos Moleculares , Neoplasias/inmunología , Neoplasias/terapia , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/uso terapéutico , Factores de Virulencia/uso terapéutico , Exotoxina A de Pseudomonas aeruginosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA