Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Nat Commun ; 12(1): 5218, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34471125

RESUMEN

CD47 is the only 5-transmembrane (5-TM) spanning receptor of the immune system. Its extracellular domain (ECD) is a cell surface marker of self that binds SIRPα and inhibits macrophage phagocytosis, and cancer immuno-therapy approaches in clinical trials are focused on blocking CD47/SIRPα interaction. We present the crystal structure of full length CD47 bound to the function-blocking antibody B6H12. CD47 ECD is tethered to the TM domain via a six-residue peptide linker (114RVVSWF119) that forms an extended loop (SWF loop), with the fundamental role of inserting the side chains of W118 and F119 into the core of CD47 extracellular loop region (ECLR). Using hydrogen-deuterium exchange and molecular dynamics simulations we show that CD47's ECLR architecture, comprised of two extracellular loops and the SWF loop, creates a molecular environment stabilizing the ECD for presentation on the cell surface. These findings provide insights into CD47 immune recognition, signaling and therapeutic intervention.


Asunto(s)
Biomarcadores , Antígeno CD47/química , Antígeno CD47/metabolismo , Proteínas Portadoras/metabolismo , Receptores Inmunológicos/metabolismo , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Antígenos de Diferenciación/inmunología , Sitios de Unión , Antígeno CD47/efectos de los fármacos , Antígeno CD47/genética , Humanos , Macrófagos/metabolismo , Modelos Moleculares , Fagocitosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
2.
J Mol Biol ; 433(4): 166766, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33359099

RESUMEN

Interleukin-1ß (IL-1ß) is a potent pleiotropic cytokine playing a central role in protecting cells from microbial pathogen infection or endogenous stress. After it binds to IL-1RI and recruits IL-1 receptor accessory protein (IL-1RAcP), signaling culminates in activation of NF-κB. Many pathophysiological diseases have been attributed to the derailment of IL-1ß regulation. Several blocking reagents have been developed based on two mechanisms: blocking the binding of IL-1ß to IL-1RI or inhibiting the recruitment of IL-1RAcP to the IL-1ß initial complex. In order to simultaneously fulfill these two actions, a human anti-IL-1ß neutralizing antibody IgG26 was screened from human genetic phage-display library and furthered structure-optimized to final version, IgG26AW. IgG26AW has a sub-nanomolar binding affinity for human IL-1ß. We validated IgG26AW-neutralizing antibodies specific for IL-1ß in vivo to prevent human IL-1ß-driving IL-6 elevation in C56BL/6 mice. Mice underwent treatments with IgG26AW in A549 and MDA-MB-231 xenograft mouse cancer models have also been observed with tumor shrank and inhibition of tumor metastasis. The region where IgG26 binds to IL-1ß also overlaps with the position where IL-1RI and IL-1RAcP bind, as revealed by the 26-Fab/IL-1ß complex structure. Meanwhile, SPR experiments showed that IL-1ß bound by IgG26AW prevented the further binding of IL-1RI and IL-1RAcP, which confirmed our inference from the result of protein structure. Therefore, the inhibitory mechanism of IgG26AW is to block the assembly of the IL-1ß/IL-1RI/IL-1RAcP ternary complex which further inhibits downstream signaling. Based on its high affinity, high neutralizing potency, and novel binding epitope simultaneously occupying both IL-1RI and IL-1RAcP residues that bind to IL-1ß, IgG26AW may be a new candidate for treatments of inflammation-related diseases or for complementary treatments of cancers in which the role of IL-1ß is critical to pathogenesis.


Asunto(s)
Anticuerpos Bloqueadores/química , Anticuerpos Monoclonales/química , Proteína Accesoria del Receptor de Interleucina-1/química , Interleucina-1beta/química , Modelos Moleculares , Conformación Proteica , Receptores Tipo I de Interleucina-1/química , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Sitios de Unión , Línea Celular Tumoral , Mapeo Epitopo/métodos , Epítopos/inmunología , Humanos , Inmunoglobulina G/química , Proteína Accesoria del Receptor de Interleucina-1/metabolismo , Interleucina-1beta/metabolismo , Ratones , Modelos Biológicos , FN-kappa B/metabolismo , Biblioteca de Péptidos , Unión Proteica/efectos de los fármacos , Receptores Tipo I de Interleucina-1/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Proc Natl Acad Sci U S A ; 117(46): 28971-28979, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33127753

RESUMEN

Blocking the action of FSH genetically or pharmacologically in mice reduces body fat, lowers serum cholesterol, and increases bone mass, making an anti-FSH agent a potential therapeutic for three global epidemics: obesity, osteoporosis, and hypercholesterolemia. Here, we report the generation, structure, and function of a first-in-class, fully humanized, epitope-specific FSH blocking antibody with a KD of 7 nM. Protein thermal shift, molecular dynamics, and fine mapping of the FSH-FSH receptor interface confirm stable binding of the Fab domain to two of five receptor-interacting residues of the FSHß subunit, which is sufficient to block its interaction with the FSH receptor. In doing so, the humanized antibody profoundly inhibited FSH action in cell-based assays, a prelude to further preclinical and clinical testing.


Asunto(s)
Tejido Adiposo/metabolismo , Anticuerpos Bloqueadores/inmunología , Huesos/metabolismo , Epítopos , Hormona Folículo Estimulante/inmunología , Animales , Anticuerpos Bloqueadores/química , Anticuerpos Monoclonales , Densidad Ósea , Femenino , Hormona Folículo Estimulante/química , Hormona Folículo Estimulante de Subunidad beta/inmunología , Humanos , Hipercolesterolemia , Ratones , Ratones Endogámicos C57BL , Simulación de Dinámica Molecular , Obesidad , Osteoporosis , Receptores de HFE/metabolismo
4.
J Comput Biol ; 26(9): 962-974, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30570348

RESUMEN

Norovirus is the causing agent of acute gastroenteritis disease globally. Efforts in developing therapeutics against virus infection mostly fail due to emergence of drug resistance that is a consequence of presence of high mutation rates in virus genome during virus' life cycle. In this study, we computationally analyzed the affinity of a drug target, wild type VP1 envelope protein and its three variants to a therapeutic antibody FAB5I2. We have found that mutations break important hydrogen bonds and cause high fluctuations in residues that form VP1-FAB5I2 complex interface. In addition to changes in dynamics, we also revealed that the affinity of FAB5I2 to VP1 protein drops significantly upon mutations in terms of relative binding free energy.


Asunto(s)
Anticuerpos Bloqueadores/química , Anticuerpos Monoclonales/química , Antígenos Virales/química , Simulación de Dinámica Molecular , Mutación , Norovirus/química , Proteínas del Envoltorio Viral/química , Anticuerpos Bloqueadores/genética , Antígenos Virales/genética , Humanos , Norovirus/genética , Proteínas del Envoltorio Viral/genética
5.
Sci Rep ; 7(1): 9000, 2017 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-28827556

RESUMEN

The growth and motility factor Hepatocyte Growth Factor/Scatter Factor (HGF/SF) and its receptor, the product of the MET proto-oncogene, promote invasion and metastasis of tumor cells and have been considered potential targets for cancer therapy. We generated a new Met-blocking antibody which binds outside the ligand-binding site, and determined the crystal structure of the Fab in complex with its target, which identifies the binding site as the Met Ig1 domain. The antibody, 107_A07, inhibited HGF/SF-induced cell migration and proliferation in vitro and inhibited growth of tumor xenografts in vivo. In biochemical assays, 107_A07 competes with both HGF/SF and its truncated splice variant NK1 for MET binding, despite the location of the antibody epitope on a domain (Ig1) not reported to bind NK1 or HGF/SF. Overlay of the Fab-MET crystal structure with the InternalinB-MET crystal structure shows that the 107_A07 Fab comes into close proximity with the HGF/SF-binding SEMA domain when MET is in the "compact", InternalinB-bound conformation, but not when MET is in the "open" conformation. These findings provide further support for the importance of the "compact" conformation of the MET extracellular domain, and the relevance of this conformation to HGF/SF binding and signaling.


Asunto(s)
Anticuerpos Bloqueadores/aislamiento & purificación , Anticuerpos Bloqueadores/metabolismo , Antineoplásicos Inmunológicos/aislamiento & purificación , Antineoplásicos Inmunológicos/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/química , Antineoplásicos Inmunológicos/administración & dosificación , Antineoplásicos Inmunológicos/química , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Glioblastoma/tratamiento farmacológico , Xenoinjertos , Humanos , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/aislamiento & purificación , Fragmentos Fab de Inmunoglobulinas/metabolismo , Ratones Desnudos , Trasplante de Neoplasias , Unión Proteica , Conformación Proteica , Proto-Oncogenes Mas , Resultado del Tratamiento
6.
PLoS One ; 12(8): e0182555, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28771632

RESUMEN

The emergence of multi-drug resistant Neisseria gonorrhoeae has generated an urgent need for novel therapies or a vaccine to prevent gonococcal disease. In this study we investigate the potential of targeting the surface exposed nitrite reductase, AniA, to block activity by producing functional blocking antibodies. AniA activity is essential for anaerobic growth and biofilm formation of N. gonorrhoeae and functional blocking antibodies may prevent colonisation and disease. Seven peptides covering regions adjacent to the active site were designed based on the AniA structure. Six of the seven peptide conjugates generated immune responses. Peptide 7, GALGQLKVEGAEN, was able to elicit antibodies capable of blocking AniA activity. Antiserum raised against the peptide 7 conjugate detected AniA in 20 N. gonorrhoeae clinical isolates. Recombinant AniA protein antigens were also assessed in this study and generated high-titre, functional blocking antibody responses. Peptide 7 conjugates or truncated recombinant AniA antigens have potential for inclusion in a vaccine against N. gonorrhoeae.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Antígenos Bacterianos/química , Proteínas de la Membrana Bacteriana Externa/química , Neisseria gonorrhoeae/enzimología , Péptidos/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/química , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacunas Bacterianas/inmunología , Dominio Catalítico , Femenino , Inmunización , Ratones , Neisseria gonorrhoeae/inmunología , Péptidos/agonistas , Péptidos/síntesis química , Conejos , Proteínas Recombinantes/inmunología
7.
Nature ; 545(7652): 112-115, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28445455

RESUMEN

Protease-activated receptors (PARs) are a family of G-protein-coupled receptors (GPCRs) that are irreversibly activated by proteolytic cleavage of the N terminus, which unmasks a tethered peptide ligand that binds and activates the transmembrane receptor domain, eliciting a cellular cascade in response to inflammatory signals and other stimuli. PARs are implicated in a wide range of diseases, such as cancer and inflammation. PARs have been the subject of major pharmaceutical research efforts but the discovery of small-molecule antagonists that effectively bind them has proved challenging. The only marketed drug targeting a PAR is vorapaxar, a selective antagonist of PAR1 used to prevent thrombosis. The structure of PAR1 in complex with vorapaxar has been reported previously. Despite sequence homology across the PAR isoforms, discovery of PAR2 antagonists has been less successful, although GB88 has been described as a weak antagonist. Here we report crystal structures of PAR2 in complex with two distinct antagonists and a blocking antibody. The antagonist AZ8838 binds in a fully occluded pocket near the extracellular surface. Functional and binding studies reveal that AZ8838 exhibits slow binding kinetics, which is an attractive feature for a PAR2 antagonist competing against a tethered ligand. Antagonist AZ3451 binds to a remote allosteric site outside the helical bundle. We propose that antagonist binding prevents structural rearrangements required for receptor activation and signalling. We also show that a blocking antibody antigen-binding fragment binds to the extracellular surface of PAR2, preventing access of the tethered ligand to the peptide-binding site. These structures provide a basis for the development of selective PAR2 antagonists for a range of therapeutic uses.


Asunto(s)
Receptor PAR-2/química , Receptor PAR-2/metabolismo , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico/efectos de los fármacos , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Bencimidazoles/química , Bencimidazoles/farmacología , Benzodioxoles/química , Benzodioxoles/farmacología , Alcoholes Bencílicos/química , Alcoholes Bencílicos/farmacología , Cristalografía por Rayos X , Humanos , Imidazoles/química , Imidazoles/farmacología , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/farmacología , Cinética , Ligandos , Modelos Moleculares , Receptor PAR-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
8.
Oncotarget ; 8(2): 2781-2799, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27835863

RESUMEN

The invasion-promoting MT1-MMP is a cell surface-associated collagenase with a plethora of critical cellular functions. There is a consensus that MT1-MMP is a key protease in aberrant pericellular proteolysis in migrating cancer cells and, accordingly, a promising drug target. Because of high homology in the MMP family and a limited success in the design of selective small-molecule inhibitors, it became evident that the inhibitor specificity is required for selective and successful MT1-MMP therapies. Using the human Fab antibody library (over 1.25×109 individual variants) that exhibited the extended, 23-27 residue long, VH CDR-H3 segments, we isolated a panel of the inhibitory antibody fragments, from which the 3A2 Fab outperformed others as a specific and potent, low nanomolar range, inhibitor of MT1-MMP. Here, we report the in-depth characterization of the 3A2 antibody. Our multiple in vitro and cell-based tests and assays, and extensive structural modeling of the antibody/protease interactions suggest that the antibody epitope involves the residues proximal to the protease catalytic site and that, in contrast with tissue inhibitor-2 of MMPs (TIMP-2), the 3A2 Fab inactivates the protease functionality by binding to the catalytic domain outside the active site cavity. In agreement with the studies in metastasis by others, our animal studies in acute pulmonary melanoma metastasis support a key role of MT1-MMP in metastatic process. Conversely, the selective anti-MT1-MMP monotherapy significantly alleviated melanoma metastatic burden. It is likely that further affinity maturation of the 3A2 Fab will result in the lead inhibitor and a proof-of-concept for MT1-MMP targeting in metastatic cancers.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Antineoplásicos Inmunológicos/farmacología , Metaloproteinasa 14 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Neoplasias/metabolismo , Neoplasias/patología , Animales , Anticuerpos Bloqueadores/química , Antineoplásicos Inmunológicos/química , Unión Competitiva , Dominio Catalítico , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Colágeno/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Femenino , Xenoinjertos , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/farmacología , Inhibidores de la Metaloproteinasa de la Matriz/química , Ratones , Modelos Moleculares , Conformación Molecular , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias/tratamiento farmacológico , Unión Proteica , Proteolisis , Proteínas Recombinantes/metabolismo
9.
MAbs ; 6(6): 1598-607, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25484067

RESUMEN

In inflammatory disease conditions, the regulation of the cytokine system is impaired, leading to tissue damages. Here, we used protein engineering to develop biologicals suitable for blocking a combination of inflammation driving cytokines by a single construct. From a set of interleukin (IL)-6-binding affibody molecules selected by phage display, five variants with a capability of blocking the interaction between complexes of soluble IL-6 receptor α (sIL-6Rα) and IL-6 and the co-receptor gp130 were identified. In cell assays designed to analyze any blocking capacity of the classical or the alternative (trans) signaling IL-6 pathways, one variant, ZIL-6_13 with an affinity (KD) for IL-6 of ∼500 pM, showed the best performance. To construct fusion proteins ("AffiMabs") with dual cytokine specificities, ZIL-6_13 was fused to either the N- or C-terminus of both the heavy and light chains of the anti-tumor necrosis factor (TNF) monoclonal antibody adalimumab (Humira®). One AffiMab construct with ZIL-6_13 positioned at the N-terminus of the heavy chain, denoted ZIL-6_13-HCAda, was determined to be the most optimal, and it was subsequently evaluated in an acute Serum Amyloid A (SAA) model in mice. Administration of the AffiMab or adalimumab prior to challenge with a mix of IL-6 and TNF reduced the levels of serum SAA in a dose-dependent manner. Interestingly, the highest dose (70 mg/kg body weight) of adalimumab only resulted in a 50% reduction of SAA-levels, whereas the corresponding dose of the ZIL-6_13-HCAda AffiMab with combined IL-6/TNF specificity, resulted in SAA levels below the detection limit.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Anticuerpos Monoclonales Humanizados/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteína Amiloide A Sérica/inmunología , Adalimumab , Animales , Antiinflamatorios/química , Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/farmacología , Afinidad de Anticuerpos/inmunología , Línea Celular Tumoral , Células Cultivadas , Receptor gp130 de Citocinas/inmunología , Receptor gp130 de Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Inflamación/sangre , Inflamación/inmunología , Inflamación/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-6/antagonistas & inhibidores , Interleucina-6/inmunología , Interleucina-6/metabolismo , Subunidad alfa del Receptor de Interleucina-6/inmunología , Subunidad alfa del Receptor de Interleucina-6/metabolismo , Ratones Endogámicos BALB C , Unión Proteica/inmunología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacología , Proteína Amiloide A Sérica/antagonistas & inhibidores , Proteína Amiloide A Sérica/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 10): 1935-45, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24100313

RESUMEN

Glycoprotein D (gD) of herpes simplex virus (HSV) binds to a host cell surface receptor, which is required to trigger membrane fusion for virion entry into the host cell. gD has become a validated anti-HSV target for therapeutic antibody development. The highly inhibitory human monoclonal antibody E317 (mAb E317) was previously raised against HSV gD for viral neutralization. To understand the structural basis of antibody neutralization, crystals of the gD ectodomain bound to the E317 Fab domain were obtained. The structure of the complex reveals that E317 interacts with gD mainly through the heavy chain, which covers a large area for epitope recognition on gD, with a flexible N-terminal and C-terminal conformation. The epitope core structure maps to the external surface of gD, corresponding to the binding sites of two receptors, herpesvirus entry mediator (HVEM) and nectin-1, which mediate HSV infection. E317 directly recognizes the gD-nectin-1 interface and occludes the HVEM contact site of gD to block its binding to either receptor. The binding of E317 to gD also prohibits the formation of the N-terminal hairpin of gD for HVEM recognition. The major E317-binding site on gD overlaps with either the nectin-1-binding residues or the neutralizing antigenic sites identified thus far (Tyr38, Asp215, Arg222 and Phe223). The epitopes of gD for E317 binding are highly conserved between two types of human herpesvirus (HSV-1 and HSV-2). This study enables the virus-neutralizing epitopes to be correlated with the receptor-binding regions. The results further strengthen the previously demonstrated therapeutic and diagnostic potential of the E317 antibody.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Herpesvirus Humano 1/química , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 2/química , Herpesvirus Humano 2/inmunología , Pruebas de Neutralización/métodos , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/metabolismo , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Secuencia Conservada , Cristalografía por Rayos X , Mapeo Epitopo/métodos , Herpesvirus Humano 1/metabolismo , Herpesvirus Humano 2/metabolismo , Humanos , Nectinas , Unión Proteica/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo
11.
J Mol Biol ; 425(22): 4366-78, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23867277

RESUMEN

Quiescin sulfhydryl oxidase 1 (QSOX1) is a catalyst of disulfide bond formation that undergoes regulated secretion from fibroblasts and is over-produced in adenocarcinomas and other cancers. We have recently shown that QSOX1 is required for incorporation of particular laminin isoforms into the extracellular matrix (ECM) of cultured fibroblasts and, as a consequence, for tumor cell adhesion to and penetration of the ECM. The known role of laminins in integrin-mediated cell survival and motility suggests that controlling QSOX1 activity may provide a novel means of combating metastatic disease. With this motivation, we developed a monoclonal antibody that inhibits the activity of human QSOX1. Here, we present the biochemical and structural characterization of this antibody and demonstrate that it is a tight-binding inhibitor that blocks one of the redox-active sites in the enzyme, but not the site at which de novo disulfides are generated catalytically. Sulfhydryl oxidase activity is thus prevented without direct binding of the sulfhydryl oxidase domain, confirming the model for the interdomain QSOX1 electron transfer mechanism originally surmised based on mutagenesis and protein dissection. In addition, we developed a single-chain variant of the antibody and show that it is a potent QSOX1 inhibitor. The QSOX1 inhibitory antibody will be a valuable tool in studying the role of ECM composition and architecture in cell migration, and the recombinant version may be further developed for potential therapeutic applications based on manipulation of the tumor microenvironment.


Asunto(s)
Anticuerpos Bloqueadores/química , Anticuerpos Monoclonales/química , Disulfuros/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Tolueno/análogos & derivados , Secuencia de Aminoácidos , Anticuerpos Bloqueadores/metabolismo , Anticuerpos Bloqueadores/farmacología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Sitios de Unión , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/metabolismo , Tolueno/química
12.
Am J Reprod Immunol ; 70(4): 299-308, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23650919

RESUMEN

PROBLEM: Aim of our study was to investigate whether TIFI, a syntetic peptide able to compete with anti-phospholipid antibodies (aPL) in the binding to endothelium, may restore aPL-inhibited endometrial angiogenesis. METHODS: The protective role of TIFI was evaluated on: i) aPL-inhibited of human endometrial endothelial cells (HEEC) angiogenesis in vitro; ii) aPL-inhibited vascular endothelial growth factor (VEGF) and metalloproteases (MMPs) expression; iii) aPL-inhibited Nuclear Factor-κB (NF-κB) and Extracellular signal-Regulated Kinase (ERK) activation and (iv) angiogenesis in vivo. RESULTS: TIFI restores in a dose-dependent manner: i) aPL-mediated inhibition of HEEC angiogenesis in vitro and in vivo (P < 0.05), ii) VEGF (P < 0.001) and MMP-2 (P < 0.05) expression and iii) NF-κB DNA binding and ERK-1/2 activation (P < 0.05) inhibited by aPL. CONCLUSION: Our results show for the first time the protective effects of TIFI, as represented by its ability to interfere with aPL mediated anti-angiogenic activity.


Asunto(s)
Anticuerpos Antifosfolípidos/metabolismo , Anticuerpos Bloqueadores/farmacología , Endometrio/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Péptidos/farmacología , Anticuerpos Bloqueadores/química , Sitios de Unión de Anticuerpos/efectos de los fármacos , Unión Competitiva/efectos de los fármacos , Células Cultivadas , Endometrio/irrigación sanguínea , Endometrio/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , FN-kappa B/metabolismo , Neovascularización Patológica/inmunología , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/química , Fosfolípidos/metabolismo , Unión Proteica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , beta 2 Glicoproteína I/metabolismo
13.
Immunobiology ; 216(6): 698-706, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21183243

RESUMEN

The continuous regions on botulinum neurotoxin A (BoNT/A) light (L) chain recognized by anti-toxin antibodies (Abs) from mouse, horse and chicken have been mapped. We synthesized a panel of thirty-two 19-residue peptides that overlapped consecutively by 5 residues and encompassed the entire L chain (residues 1-453). Mouse Abs recognized 5 major antigenic regions on the L chain, horse Abs recognized 9 while chicken Abs recognized 8 major antigenic regions. Overall, however, the three host species recognized, to some extent, similar, but not identical, peptides and the levels of Abs directed against a given region varied with the immunized host. Differences in the MHC of the host caused variation in levels of Ab recognition and some epitopes showed right or left frame-shifts among the species. Selected region(s) were also uniquely recognized by one species (e.g., peptide L1 by horse Abs). Mapping of the L chain antigenic regions and the previous localization of the regions on the H chain with the same antisera, has permitted description of the complete antigenic structure of BoNT/A. The locations in the 3-dimensional structure of the antigenic regions of the entire toxin are shown for mouse Abs. In the 3-D structure, the antigenic regions are on the surface of the toxin and when antibodies are bound the enzymatic activity of the light chain is obstructed.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Anticuerpos Bloqueadores/metabolismo , Toxinas Botulínicas Tipo A/inmunología , Toxinas Botulínicas Tipo A/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/química , Especificidad de Anticuerpos/inmunología , Antígenos/química , Antígenos/inmunología , Toxinas Botulínicas Tipo A/química , Ratones , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Péptidos/síntesis química , Péptidos/inmunología , Unión Proteica/inmunología , Conformación Proteica , Especificidad de la Especie
14.
Pharmazie ; 64(4): 238-41, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19435141

RESUMEN

Phage antibody library is a promising tool for rapidly creating in vitro single-chain Fv (scFv) antibodies to various antigens. The scFv can also act like a subcellularly-expressed antibody, known as intrabody, and can either be used as a novel research tool or used efficiently for targeted molecular therapy. However, there are only a few existing reports about the successful expression of scFvs as functional antibodies in the cell, mainly because poor quality scFv phage antibody libraries were used to isolate the intrabody clones. The aim of this study was to isolate intrabody-forming scFv clones from the nonimmune scFv phage antibody library we have generated. Using this library, we isolated a scFv clone against the apoptosis-related intracellular protein Bid in two weeks. To evaluate the intrabody-forming quality of this anti-Bid scFv clone, we expressed it in cultured mammalian cells after fusing it with the fluorescent protein Venus. The expression of the soluble form of anti-Bid scFv-Venus fusion protein was confirmed by fluorescence microscopy analysis. These results show that our scFv phage library is not only optimized for antibody production but can also be used to efficiently generate intrabodies.


Asunto(s)
Anticuerpos/química , Biblioteca de Péptidos , Secuencia de Aminoácidos , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Especificidad de Anticuerpos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/inmunología , Línea Celular Tumoral , Células Cultivadas , Clonación Molecular , Ensayo de Inmunoadsorción Enzimática , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacología
15.
Drug Metab Dispos ; 37(5): 925-31, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19196844

RESUMEN

Cytochrome P450 (P450) is the superfamily of enzymes responsible for biotransformation of endobiotics and xenobiotics. However, their large isoform multiplicity, inducibility, diverse structure, widespread distribution, polymorphic expression, and broad overlapping substrate specificity make it difficult to measure the precise role of each individual P450 to the metabolism of drugs (or carcinogens) and hamper the understanding of the relationship between the genetic/environmental factors that regulate P450 phenotype and the responses of the individual P450s to drugs. The antibodies against P450s have been useful tools for the quantitative determination of expression level and contribution of the epitope-specific P450 to the metabolism of a drug or carcinogen substrate in tissues containing multiple P450 isoforms and for implications in pharmacogenetics and human risk assessment. In particular, the inhibitory antibodies are uniquely suited for reaction phenotyping that helps to predict human pharmacokinetics for clinical drug-drug interaction potential in drug discovery and development.


Asunto(s)
Sistema Enzimático del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/inmunología , Animales , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/química , Isoenzimas/metabolismo , Fenotipo , Medición de Riesgo
16.
Biol Pharm Bull ; 31(7): 1444-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18591790

RESUMEN

We previously prepared 2-hydroxypropyl-beta-cyclodextrin (HPbetaCD) solutions containing disulfiram (DSF) and hydroxypropylmethylcellulose (HPMC, DSF solutions), and found the instillation of this DSF solutions delayed lens opacification in ICR/f rats, a recessive-type hereditary cataractous strain. In this study, we determined the corneal penetration mechanism of DSF solutions using human cornea epithelial cell monolayers based on the immortalized human cornea epithelial cell line (HCE-T) developed by Tropainen et al. [Invest. Ophthalmol. Vis. Sci., 42, 2942-2948 (2001)]. The transepithelial electrical resistance (TER) values of HCE-T cells increases from approximately 275 to 388 Omega.cm(2) by exposure to an air-liquid interface for 2 weeks. The penetration of DSF into the basolateral chamber was prevented by the increase in TER values. The DSF in solution was converted to diethyldithiocarbamate (DDC) during the penetration experiment using HCE-T cell monolayers, and a close relationship between the penetration coefficient of DDC and aldehyde dehydrogenase (ALDH) 3A1 mRNA expression (y=41.202x+18.587, R=0.9413) was observed. In addition, an anti-ALDH3A1 antibody significantly inhibited the DSF-DDC conversion. These results suggest that DSF in DSF solutions is converted to DDC via catalysis by an ALDH3A1 in the cornea, and then transited from the apical side to the basolateral side.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Córnea/metabolismo , Disulfiram/farmacocinética , Células Epiteliales/metabolismo , Aldehído Deshidrogenasa/antagonistas & inhibidores , Anticuerpos Bloqueadores/química , Anticuerpos Bloqueadores/farmacología , Células Cultivadas , Córnea/citología , Ditiocarba/metabolismo , Células Epiteliales/enzimología , Humanos , ARN/biosíntesis , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Absorción Cutánea
17.
Toxicol In Vitro ; 22(3): 779-87, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18207356

RESUMEN

P450s are key enzymes responsible for biotransformation of numerous endogenous and exogenous compounds and are located in almost every tissue. This superfamily is the largest group of enzymes (>6000) that share a high degree of similarity in protein sequence. The human genome contains 57 CYP genes and 58 pseudogenes. A major gap exists in our knowledge about differences in CYP expression on a protein level. DNA and mRNA information are not sufficient because transcription and particularly translation events are not necessarily correlated with levels of expressed proteins. The data reported in this study complete the framework of an integrated proteomic method for the simultaneous qualitative and quantitative analysis of CYP isozyme composition using MALDI-TOF-MS and immunochemistry that has been developed in our laboratory over the last several years (Alterman et al., 2005a,b) and is based on the fact that each P450 isozyme possesses unique tryptic peptide(s) (UTP) that could be used for differential analysis of human CYP expression. Here we demonstrate that three different immunochemical techniques (ELISA, Western blot, and peptide affinity enrichment on magnetic beads with attached antibodies) have potential to be incorporated in an integrated proteomic method combining mass spectrometry and immunochemistry. Fundamentally, this approach is based on the measurement of the same chemical entity (isozyme-specific UTP) in the tryptic digest by two orthogonal analytical techniques, mass spectrometry and immunochemistry. The application of this approach is illustrated with two human CYP isozymes--CYP1A2 and CYP2E1.


Asunto(s)
Anticuerpos Bloqueadores/química , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Péptidos/química , Péptidos/inmunología , Proteómica/métodos , Tripsina/química , Animales , Western Blotting , Cromatografía de Afinidad , Citocromo P-450 CYP1A2/química , Citocromo P-450 CYP1A2/inmunología , Citocromo P-450 CYP2E1/química , Citocromo P-450 CYP2E1/inmunología , Sistema Enzimático del Citocromo P-450/inmunología , Ensayo de Inmunoadsorción Enzimática , Inmunoquímica , Indicadores y Reactivos , Isoenzimas/genética , Isoenzimas/inmunología , Isoenzimas/metabolismo , Espectrometría de Masas , Conejos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
18.
J Mol Biol ; 366(3): 815-29, 2007 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-17196977

RESUMEN

Non-immune (naïve) antibody phage libraries have become an important source of human antibodies. The synthetic phage antibody library described here utilizes a single human framework with a template containing human consensus complementarity-determining regions (CDRs). Diversity of the libraries was introduced at select CDR positions using tailored degenerate and trinucleotide codons that mimic natural human antibodies. Neuropilin-1 (NRP1), a cell-surface receptor for both vascular endothelial growth factor (VEGF) and class 3 semaphorins, is expressed on endothelial cells and neurons. NRP1 is required for vascular development and is expressed widely in the developing vasculature. To investigate the possibility of function blocking antibodies to NRP1 as potential therapeutics, and study the consequence of targeting NRP1 in murine tumor models, panels of antibodies that cross-react with human and murine NRP1 were generated from a designed antibody phage library. Antibody (YW64.3) binds to the CUB domains (a1a2) of NRP1 and completely blocks Sema3A induced neuron collapse; antibody (YW107.4.87) binds to the coagulation factor V/VIII domains (b1b2) of NRP1 and blocks VEGF binding and VEGF induced cell migration. YW107.4.87 inhibits tumor growth in animal xenograft models. These antibodies have provided valuable tools to study the roles of NRP1 in vascular and tumor biology.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Neuropilina-1/inmunología , Biblioteca de Péptidos , Secuencia de Aminoácidos , Animales , Anticuerpos Bloqueadores/química , Afinidad de Anticuerpos/efectos de los fármacos , Células CHO , Movimiento Celular/efectos de los fármacos , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/inmunología , Cricetinae , Cricetulus , Conos de Crecimiento/efectos de los fármacos , Humanos , Inmunoglobulina G/inmunología , Cinética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/genética , Neoplasias/patología , Estructura Terciaria de Proteína/efectos de los fármacos , Semaforina-3A/farmacología
19.
Pharmazie ; 61(10): 889-90, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17069432

RESUMEN

In this study, we converted the immunoglobulin-type anti-human tumor necrosis factor-alpha (TNF-alpha) monoclonal antibody (Mab) to a scFv-type antibody in order to assess its basic properties. The immunoglobulin VH and VL genes were isolated from the hybridoma that produced an anti-TNF-alpha neutralizing Mab, and they were then linked together to create scFvs of the VL-VH or VH-VL-form. The binding affinity to TNF-a was retained in both scFvs. Interestingly, the VL-VH-type scFv effectively inhibited the TNF-alpha-mediated cytotoxicity, while this neutralization activity was dramatically decreased in the VH-VL-type scFv. These results suggest that the VL-VH-type scFv is a suitable template to create improved versions of the anti-TNF-alpha antibody using a phage display system, and they also show that the structural format must be taken into account in manufacturing scFvs.


Asunto(s)
Anticuerpos Monoclonales/química , Región Variable de Inmunoglobulina/química , Biblioteca de Péptidos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Anticuerpos Bloqueadores/química , Anticuerpos Monoclonales/metabolismo , Humanos , Hibridomas , Factor de Necrosis Tumoral alfa/metabolismo
20.
Cytometry B Clin Cytom ; 70(4): 251-8, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16906584

RESUMEN

BACKGROUND: In this study we developed a method to measure the amount of ZAP-70 [zeta accessory protein] in B-CLL cells without relying on the ZAP-70 expression of patient B or T cells to normalize fluorescence intensity. METHODS: B-CLL cells were fixed with formaldehyde before surface staining with gating antibodies CD19PC5 and CD5FITC. The cells were permeabilized with saponin, and the ZAP-70 antigen was blocked in one tube with unlabeled antibody to ZAP-70 [clone 1E7.2]. Zap-70-PE was then added to this tube. ZAP-70-PE was added to a second tube without unlabeled antibody to ZAP-70. The mean fluorescence intensity of the ZAP-70 in the tube without unlabeled antibody divided by the mean fluorescence intensity of the ZAP-70 in the tube with unlabeled antibody equals the RATIO of total fluorescence to non-specific ZAP-70 fluorescence in the B-CLL cells. In a second method of analysis, a region is created in the histogram showing ZAP-70 fluorescence intensity in the tube with unlabeled antibody to ZAP-70. This region is set to 0.9% positive cells. This same region is then used to measure the % positive [%POS] ZAP-70 cells in the tube without unlabeled antibody to ZAP-70. The brighter the ZAP-70 fluorescence above the non-specific background, the higher the %POS. RESULTS: Due to the varying amount of non-specific staining between patient B-CLL cells and other cells, the blocking antibody method yielded a more quantitative and reproducible measure of ZAP-70 in B-CLL cells than other methods, which use the ratio of B-CLL fluorescence to normal B or T-cell fluorescence. Using this improved method, ZAP-70 was determined to be negative if the RATIO was less than 2:1 and positive if the RATIO was greater than 2:1. ZAP-70 was determined to be negative if the %POS was less than 5% and positive if the %POS was greater than 5%, a cut-off value lower than previous values published, due to exclusion of non-specific staining. Both cut-offs were based upon patient specimen distribution profiling. CONCLUSIONS: Use of a blocking antibody resulted in a robust, reproducible clinical B-CLL assay that is not influenced by the need to measure the amount of ZAP-70 in other cells. ZAP-70 results segre gate patients into indolent and aggressive groups suggested by published clinical outcomes.


Asunto(s)
Anticuerpos Bloqueadores/química , Anticuerpos Monoclonales/química , Citometría de Flujo/métodos , Leucemia Linfocítica Crónica de Células B/diagnóstico , Proteína Tirosina Quinasa ZAP-70/análisis , ADP-Ribosil Ciclasa 1/análisis , ADP-Ribosil Ciclasa 1/inmunología , Reacciones Antígeno-Anticuerpo , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/inmunología , Humanos , Inmunofenotipificación , Leucemia Linfocítica Crónica de Células B/inmunología , Reproducibilidad de los Resultados , Coloración y Etiquetado , Fijación del Tejido/métodos , Proteína Tirosina Quinasa ZAP-70/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA