Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Nat Med ; 6(4): 443-6, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10742152

RESUMEN

Inhibitory receptors have been proposed to modulate the in vivo cytotoxic response against tumor targets for both spontaneous and antibody-dependent pathways. Using a variety of syngenic and xenograft models, we demonstrate here that the inhibitory FcgammaRIIB molecule is a potent regulator of antibody-dependent cell-mediated cytotoxicity in vivo, modulating the activity of FcgammaRIII on effector cells. Although many mechanisms have been proposed to account for the anti-tumor activities of therapeutic antibodies, including extended half-life, blockade of signaling pathways, activation of apoptosis and effector-cell-mediated cytotoxicity, we show here that engagement of Fcgamma receptors on effector cells is a dominant component of the in vivo activity of antibodies against tumors. Mouse monoclonal antibodies, as well as the humanized, clinically effective therapeutic agents trastuzumab (Herceptin(R)) and rituximab (Rituxan(R)), engaged both activation (FcgammaRIII) and inhibitory (FcgammaRIIB) antibody receptors on myeloid cells, thus modulating their cytotoxic potential. Mice deficient in FcgammaRIIB showed much more antibody-dependent cell-mediated cytotoxicity; in contrast, mice deficient in activating Fc receptors as well as antibodies engineered to disrupt Fc binding to those receptors were unable to arrest tumor growth in vivo. These results demonstrate that Fc-receptor-dependent mechanisms contribute substantially to the action of cytotoxic antibodies against tumors and indicate that an optimal antibody against tumors would bind preferentially to activation Fc receptors and minimally to the inhibitory partner FcgammaRIIB.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD/inmunología , Linfoma de Células B/inmunología , Melanoma Experimental/inmunología , Receptor ErbB-2/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Anticuerpos Monoclonales de Origen Murino , Antígenos CD/genética , Antineoplásicos/inmunología , Antineoplásicos/farmacología , Femenino , Humanos , Inmunización Pasiva , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Receptores de IgG/genética , Rituximab , Trastuzumab
2.
J Cell Biol ; 125(6): 1395-406, 1994 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7515889

RESUMEN

This study describes the identification of seven amino acid residues of the vascular cell adhesion molecule (VCAM-1) that influence binding to the alpha 4 beta 1 receptor. Using recombinant murine VCAM-1-IgG, which is bound by both mouse (WEHI 231) and human (Ramos) lymphoid cells, two approaches demonstrated the crucial role of the first two NH2-terminal Ig-like domains in binding: (a) blocking monoclonal anti-mouse VCAM-1 antibodies bound to only truncation variants that included the first two domains; (b) site-direct mutagenesis of the first NH2-terminal domain showed that alanine substitution of the amino acid residues R36, D40, K46, S54, N65, T72, and E81 partially or completely reduced adherence by human and/or mouse cells. Of these D40, when mutated to A, N, or K (but not E), showed complete abrogation of adherence by mouse and human cells, as well as inability to bind blocking anti-murine VCAM-1 antibody MVCAM.A429, while not inducing gross structural perturbations in VCAM-1. By molecular modeling, the D40 residue was located on a beta turn connecting two beta strands defined as C and D. The residues R36, K46, S54, N65, T72, and E81, which perturb cell adherence and caused small changes to gross structure, are conformationally near or adjacent to D40. Although these residues, identified as crucial for cell adhesion, are all located in domain 1, it is evident that there is a structural requirement for domains 1 and 2 to be intact so that cell adhesive function can occur.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Adhesión Celular/fisiología , Integrinas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígeno muy Tardío/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Secuencia de Bases , Adhesión Celular/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Clonación Molecular , Análisis Mutacional de ADN , Epítopos , Humanos , Inmunoglobulina G/genética , Integrina alfa4beta1 , Linfocitos/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígeno muy Tardío/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Molécula 1 de Adhesión Celular Vascular
3.
J Cell Biol ; 120(5): 1227-35, 1993 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7679675

RESUMEN

The selectins are a family of three calcium-dependent lectins that mediate adhesive interactions between leukocytes and the endothelium during normal and abnormal inflammatory episodes. Previous work has implicated the carbohydrate sialyl Lewis(x) (sLe(x); sialic acid alpha 2-3 galactose beta 1-4 [Fucose alpha 1-3] N-acetyl glucosamine) as a component of the ligand recognized by E- and P-selectin. In the case of P-selectin, other components of the cell surface, including 2'6-linked sialic acid and sulfatide (galactose-4-sulfate ceramide), have also been proposed for adhesion mediated by this selectin. We have recently defined a region of the E-selectin lectin domain that appears to be directly involved with carbohydrate recognition and cell adhesion (Erbe, D. V., B. A. Wolitzky, L. G. Presta, C. R. Norton, R. J. Ramos, D. K. Burns, R. M. Rumberger, B. N. N. Rao, C. Foxall, B. K. Brandley, and L. A. Lasky. 1992. J. Cell Biol. 119:215-227). Here we describe a similar analysis of the P-selectin lectin domain which demonstrates that a homologous region of this glycoprotein's lectin motif is involved with carbohydrate recognition and cell binding. In addition, we present evidence that is inconsistent with a biological role for either 2'6-linked sialic acid or sulfatide in P-selectin-mediated adhesion. These results suggest that a common region of the E- and P-selectin lectin domains appears to mediate carbohydrate recognition and cell adhesion.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Adhesión Celular , Glucolípidos/metabolismo , Glicoproteínas de Membrana Plaquetaria/fisiología , Secuencia de Aminoácidos , Sitios de Unión , Moléculas de Adhesión Celular/química , Selectina E , Citometría de Flujo , Humanos , Técnicas In Vitro , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Selectina-P , Glicoproteínas de Membrana Plaquetaria/química , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
4.
J Cell Biol ; 119(1): 215-27, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1382077

RESUMEN

E-selectin elicits cell adhesion by binding to the cell surface carbohydrate, sialyl Lewis X (sLe(x)). We evaluated the effects of mutations in the E-selectin lectin domain on the binding of a panel of anti-E-selectin mAbs and on the recognition of immobilized sLe(x) glycolipid. Functional residues were then superimposed onto a three-dimensional model of the E-selectin lectin domain. This analysis demonstrated that the epitopes recognized by blocking mAbs map to a patch near the antiparallel beta sheet derived from the NH2 and COOH termini of the lectin domain and two adjacent loops. Mutations that affect sLe(x) binding map to this same region. These results thus define a small region of the E-selectin lectin domain that is critical for carbohydrate recognition.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Adhesión Celular/fisiología , Antígeno Lewis X/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Sitios de Unión/fisiología , Moléculas de Adhesión Celular/química , Análisis Mutacional de ADN , Selectina E , Técnica del Anticuerpo Fluorescente , Humanos , Antígeno Lewis X/química , Glicoproteínas de Membrana/química , Datos de Secuencia Molecular , Conformación Proteica , Conejos , Receptores Inmunológicos/química , Proteínas Recombinantes de Fusión/metabolismo
5.
Science ; 240(4859): 1632-41, 1988 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-2837824

RESUMEN

The alpha helix, first proposed by Pauling and co-workers, is a hallmark of protein structure, and much effort has been directed toward understanding which sequences can form helices. The helix hypothesis, introduced here, provides a tentative answer to this question. The hypothesis states that a necessary condition for helix formation is the presence of residues flanking the helix termini whose side chains can form hydrogen bonds with the initial four-helix greater than N-H groups and final four-helix greater than C-O groups; these eight groups would otherwise lack intrahelical partners. This simple hypothesis implies the existence of a stereochemical code in which certain sequences have the hydrogen-bonding capacity to function as helix boundaries and thereby enable the helix to form autonomously. The three-dimensional structure of a protein is a consequence of the genetic code, but the rules relating sequence to structure are still unknown. The ensuing analysis supports the idea that a stereochemical code for the alpha helix resides in its boundary residues.


Asunto(s)
Conformación Proteica , Secuencia de Aminoácidos , Animales , Carboxipeptidasas , Carboxipeptidasas A , Grupo Citocromo c , Flavodoxina , Humanos , Enlace de Hidrógeno , Modelos Químicos , Datos de Secuencia Molecular , Muramidasa , Mioglobina , Polipéptido Pancreático , Parvalbúminas , Plastocianina , Ribonucleasas , Venenos de Escorpión , Tetrahidrofolato Deshidrogenasa , Triosa-Fosfato Isomerasa , Inhibidores de Tripsina , Difracción de Rayos X
6.
Nat Biotechnol ; 16(7): 677-81, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9661204

RESUMEN

Production of bispecific IgG (BsIgG) by coexpressing two different antibodies is inefficient due to unwanted pairings of the component heavy and light chains. To overcome this problem, heavy chains were remodeled for heterodimerization using engineered disulfide bonds in combination with previously identified "knobs-into-holes" mutations. One of the variants, S354C:T366W/Y349'C:T366'S:L368'A:Y407++ +'V, gave near quantitative (approximately 95%) heterodimerization. Light chain mispairing was circumvented by using an identical light chain for each arm of the BsIgG. Antibodies with identical light chains that bind to different antigens were identified from an scFv phage library with a very restricted light chain repertoire for the majority (50/55) of antigen pairs tested. A BsIgG capable of simultaneously binding to the human receptors HER3 and cMpI was prepared by coexpressing the common light chain and corresponding remodeled heavy chains followed by protein A chromatography. The engineered heavy chains retain their ability to support antibody-dependent cell-mediated cytotoxicity as demonstrated with an anti-HER2 antibody.


Asunto(s)
Especificidad de Anticuerpos/inmunología , Inmunoglobulina G/biosíntesis , Proteínas de Neoplasias , Receptores Inmunológicos/metabolismo , Complejo Antígeno-Anticuerpo , Citotoxicidad Inmunológica , Dimerización , Disulfuros , Receptores ErbB/metabolismo , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Modelos Moleculares , Mutación/genética , Ingeniería de Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Receptor ErbB-3 , Receptores de Citocinas/metabolismo , Receptores de Trombopoyetina
7.
Mol Biol Cell ; 8(3): 501-15, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9188101

RESUMEN

Intercellular adhesion molecule 1 (ICAM-1, CD54) is a member of the Ig superfamily and is a counterreceptor for the beta 2 integrins: lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18), complement receptor 1 (MAC-1, CD11b/CD18), and p150,95 (CD11c/CD18). Binding of ICAM-1 to these receptors mediates leukocyte-adhesive functions in immune and inflammatory responses. In this report, we describe a cell-free assay using purified recombinant extracellular domains of LFA-1 and a dimeric immunoadhesin of ICAM-1. The binding of recombinant secreted LFA-1 to ICAM-1 is divalent cation dependent (Mg2+ and Mn2+ promote binding) and sensitive to inhibition by antibodies that block LFA-1-mediated cell adhesion, indicating that its conformation mimics that of LFA-1 on activated lymphocytes. We describe six novel anti-ICAM-1 monoclonal antibodies, two of which are function blocking. Thirty-five point mutants of the ICAM-1 immunoadhesin were generated and residues important for binding of monoclonal antibodies and purified LFA-1 were identified. Nineteen of these mutants bind recombinant LFA-1 equivalently to wild type. Sixteen mutants show a 66-2500-fold decrease in LFA-1 binding yet, with few exceptions, retain binding to the monoclonal antibodies. These mutants, along with modeling studies, define the LFA-1 binding site on ICAM-1 as residues E34, K39, M64, Y66, N68, and Q73, that are predicted to lie on the CDFG beta-sheet of the Ig fold. The mutant G32A also abrogates binding to LFA-1 while retaining binding to all of the antibodies, possibly indicating a direct interaction of this residue with LFA-1. These data have allowed the generation of a highly refined model of the LFA-1 binding site of ICAM-1.


Asunto(s)
Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Sitios de Unión , Sitios de Unión de Anticuerpos , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/aislamiento & purificación , Antígeno-1 Asociado a Función de Linfocito/genética , Antígeno-1 Asociado a Función de Linfocito/aislamiento & purificación , Ratones , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Cancer Res ; 55(1): 63-70, 1995 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-7805042

RESUMEN

The humanized anti-p185HER2 antibody, humAb4D5-8, has completed Phase II clinical trials for p185HER2-overexpressing breast cancer. Here, this antibody is used as a building block to engineer a disulfide-linked Fv (dsFv) beta-lactamase fusion protein for use in antibody-dependent enzyme-mediated prodrug therapy using cephalosporin-based prodrugs. Three Fv variants were designed with an interchain disulfide bond buried at the VL/VH interface and secreted from Escherichia coli. One variant, dsFv3 (VL L46C VH D101C0, has similar affinity for antigen (Kd = 0.7 nM) as the wild-type Fv and was used to construct a fusion protein in which beta-lactamase, RTEM-1, is joined to the carboxy terminus of VH. The dsFv3-beta-lactamase fusion protein secreted from E. coli efficiently activates a cephalothin doxorubicin prodrug (PRODOX, kcat/km = 1.5 x 10(5) s-1 M-1). PRODOX is approximately 20-fold less toxic than free doxorubicin against breast tumor cell lines SK-BR-3 and MCF7, which express p185HER2 at elevated and normal levels, respectively. Prebinding the dsFv3-beta-lactamase fusion protein specifically enhances the toxicity level of PRODOX to that of doxorubicin against SK-BR-3 but not MCF7 cells. The fusion protein retains both antigen-binding plus kinetic activity in murine serum and is cleared rapidly as judged by pharmacokinetic analysis in nude mice (initial and terminal half-lives of 0.23 and 1.27 h, respectively). Development and characterization of the dsFv3-beta-lactamase fusion protein is an important step toward targeted prodrug therapy of p185HER2-overexpressing tumors.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Doxorrubicina/metabolismo , Inmunoconjugados/farmacología , Profármacos/metabolismo , Receptor ErbB-2/inmunología , beta-Lactamasas , Animales , Secuencia de Bases , Neoplasias de la Mama , Disulfuros , Femenino , Fragmentos Fab de Inmunoglobulinas , Ratones , Ratones Desnudos , Modelos Moleculares , Datos de Secuencia Molecular , Ingeniería de Proteínas , Células Tumorales Cultivadas
9.
Cancer Res ; 57(20): 4593-9, 1997 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-9377574

RESUMEN

Vascular endothelial growth factor (VEGF) is a major mediator of angiogenesis associated with tumors and other pathological conditions, including proliferative diabetic retinopathy and age-related macular degeneration. The murine anti-human VEGF monoclonal antibody (muMAb VEGF) A.4.6.1 has been shown to potently suppress angiogenesis and growth in a variety of human tumor cells lines transplanted in nude mice and also to inhibit neovascularization in a primate model of ischemic retinal disease. In this report, we describe the humanization of muMAb VEGF A.4.6.1. by site-directed mutagenesis of a human framework. Not only the residues involved in the six complementarity-determining regions but also several framework residues were changed from human to murine. Humanized anti-VEGF F(ab) and IgG1 variants bind VEGF with affinity very similar to that of the original murine antibody. Furthermore, recombinant humanized MAb VEGF inhibits VEGF-induced proliferation of endothelial cells in vitro and tumor growth in vivo with potency and efficacy very similar to those of muMAb VEGF A.4.6.1. Therefore, recombinant humanized MAb VEGF is suitable to test the hypothesis that inhibition of VEGF-induced angiogenesis is a valid strategy for the treatment of solid tumors and other disorders in humans.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/uso terapéutico , Factores de Crecimiento Endotelial/inmunología , Endotelio Vascular/fisiología , Fragmentos Fab de Inmunoglobulinas/biosíntesis , Fragmentos Fab de Inmunoglobulinas/uso terapéutico , Linfocinas/inmunología , Neovascularización Patológica/prevención & control , Rabdomiosarcoma/terapia , Corteza Suprarrenal/irrigación sanguínea , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Capilares , Bovinos , Células Cultivadas , Clonación Molecular , Secuencia de Consenso , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/química , Inmunoglobulina G/uso terapéutico , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Ligeras de Inmunoglobulina/química , Región Variable de Inmunoglobulina/química , Ratones , Ratones Desnudos , Modelos Estructurales , Datos de Secuencia Molecular , Conformación Proteica , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/uso terapéutico , Rabdomiosarcoma/irrigación sanguínea , Alineación de Secuencia , Trasplante Heterólogo , Células Tumorales Cultivadas , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
10.
J Mol Biol ; 198(3): 417-24, 1987 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-3430613

RESUMEN

The crystal structures of porcine pancreatic elastase complexed to two similar benzoxazinone inhibitors are reported to 2.09 A and 1.76 A resolution, and refined to conventional R factors of 0.153 and 0.172.


Asunto(s)
Carbamatos , Oxazinas , Páncreas/enzimología , Elastasa Pancreática , Secuencia de Aminoácidos , Animales , Benzoxazinas , Enlace de Hidrógeno , Modelos Moleculares , Porcinos , Difracción de Rayos X
11.
J Mol Biol ; 189(3): 533-9, 1986 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-3640831

RESUMEN

A single crystal of porcine pancreatic elastase was mounted in a thin-walled capillary and allowed to react with acetyl-Ala-Pro-Ala-paranitroanalide. Diffraction data to 1.65 A resolution were measured and the isomorphous structure was solved from the difference Fourier map. The structure contains two surprises. Two molecules of the product: acetyl-Ala-Pro-Ala molecule are bound in the extended binding site. Both molecules are bound backwards with respect to the established mode of peptide binding.


Asunto(s)
Elastasa Pancreática , Secuencia de Aminoácidos , Animales , Sitios de Unión , Enlace de Hidrógeno , Oligopéptidos , Conformación Proteica , Porcinos , Difracción de Rayos X
12.
J Mol Biol ; 226(4): 1143-59, 1992 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-1518048

RESUMEN

A global census of the hydrogen bonds in 42 X-ray-elucidated proteins was taken and the following demographic trends identified: (1) Most hydrogen bonds are local, i.e. between partners that are close in sequence, the primary exception being hydrogen-bonded ion pairs. (2) Most hydrogen bonds are between backbone atoms in the protein, an average of 68%. (3) All proteins studied have extensive hydrogen-bonded secondary structure, an average of 82%. (4) Almost all backbone hydrogen bonds are within single elements of secondary structure. An approximate rule of thirds applies: slightly more than one-third (37%) form i----i--3 hydrogen bonds, almost one-third (32%) form i----i--4 hydrogen bonds, and slightly less than one-third (26%) reside in paired strands of beta-sheet. The remaining 5% are not wholly within an individual helix, turn or sheet. (5) Side-chain to backbone hydrogen bonds are clustered at helix-capping positions. (6) An extensive network of hydrogen bonds is present in helices. (7) To a close approximation, the total number of hydrogen bonds is a simple function of a protein's helix and sheet content. (8) A unique quantity, termed the reduced number of hydrogen bonds, is defined as the maximum number of hydrogen bonds possible when every donor:acceptor pair is constrained to be 1:1. This quantity scales linearly with chain length, with 0.71 reduced hydrogen bond per residue. Implications of these results for pathways of protein folding are discussed.


Asunto(s)
Aminoácidos/química , Conformación Proteica , Proteínas/química , Algoritmos , Enlace de Hidrógeno , Proteínas/clasificación , Tetrahidrofolato Deshidrogenasa/química , Difracción de Rayos X
13.
Curr Opin Biotechnol ; 3(4): 394-8, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1368441

RESUMEN

Current research into antibody engineering stresses the design of constructs that have both scientific and medical significance. Highlights of the past year include several successful humanizations of non-human antibodies, in which a human antibody is created that possesses the same binding specificity as the non-human one, and phage display of antibodies. This review is also published in Current Opinion in Structural Biology 1992, 2:593-596.


Asunto(s)
Anticuerpos , Anticuerpos/inmunología , Especificidad de Anticuerpos , Catálisis , Humanos , Ingeniería de Proteínas
14.
Protein Sci ; 6(4): 781-8, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9098887

RESUMEN

An anti-p185HER2/anti-CD3 humanized bispecific diabody was previously constructed from two cross-over single-chain Fv in which YH and VL domains of the parent antibodies are present on different polypeptides. Here this diabody is used to evaluate domain interface engineering strategies for enhancing the formation of functional heterodimers over inactive homodimers. A disulfide-stabilized diabody was obtained by introducing two cysteine mutations, VL L46C and VH D101C, at the anti-p185HER2.VL/VH interface. The fraction of recovered diabody that was functional following expression in Escherichia coli was improved for the disulfide-stabilized compared to the parent diabody (> 96% versus 72%), whereas the overall yield was > 60-fold lower. Eleven "knob-into-hole" diabodies were designed by molecular modeling of sterically complementary mutations at the two VL/VH interfaces. Replacements at either interface are sufficient to improve the fraction of functional heterodimer, while maintaining overall recoverable yields and affinity for both antigens close to that of the parent diabody. For example, diabody variant v5 containing the mutations VL Y87A:F98M and VH V37F:L45W at the anti-p185HER2 VL/VH interface was recovered as 92% functional heterodimer while maintaining overall recovered yield within twofold of the parent diabody. The binding affinity of v5 for p185HER2 extracellular domain and T cells is eightfold weaker and twofold stronger than for the parent diabody, respectively. Domain interface remodeling based upon either sterically complementary mutations or interchain disulfide bonds can facilitate the production of a functional diabody heterodimer. This study expands the scope of domain interface engineering by demonstrating the enhanced assembly of proteins interacting via two domain interfaces.


Asunto(s)
Anticuerpos Biespecíficos/química , Receptor ErbB-2/inmunología , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/aislamiento & purificación , Cristalografía por Rayos X , Dimerización , Humanos , Mutagénesis , Ingeniería de Proteínas
15.
Immunol Res ; 16(3): 299-311, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9379078

RESUMEN

The integrin alpha 4 beta 7 and mucosal addressin cell adhesion molecule-1 (MAdCAM-1) are molecules involved in the normal recirculation of lymphocytes between the blood and the gastrointestinal tract. These molecules may play a complementary and significant role in animal models of colitis. We have investigated the structural interaction between alpha 4 beta 7 and MAdCAM-1. Site-directed mutagenesis studies of the MAdCAM-1 molecule has led to the identification of the amino acid residue (LDT) in the loop between beta strands C and D of the Ig-superfamily-like folds being involved in the adhesive and cell activation functions of MAdCAM-1 with alpha 4 beta 7.


Asunto(s)
Colitis/inmunología , Inmunoglobulinas , Mucoproteínas , Receptores Mensajeros de Linfocitos , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular , Colitis/genética , Humanos , Inmunoglobulinas/química , Inmunoglobulinas/genética , Inmunoglobulinas/inmunología , Integrinas/química , Integrinas/genética , Integrinas/inmunología , Datos de Secuencia Molecular , Mucoproteínas/química , Mucoproteínas/genética , Mucoproteínas/inmunología , Mutagénesis Sitio-Dirigida , Receptores Mensajeros de Linfocitos/química , Receptores Mensajeros de Linfocitos/genética , Receptores Mensajeros de Linfocitos/inmunología , Relación Estructura-Actividad
16.
Protein Eng ; 7(7): 895-903, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7971951

RESUMEN

The hepatocyte growth factor receptor (HGFr) transduces a wide range of biological signals, including mitogenesis, motogenesis and morphogenesis. We recently localized a region within the N-terminal 175 amino acids of hepatocyte growth factor (HGF), termed HGF/NK1, that is necessary and sufficient for binding to the HGFr. HGF/NK1 contains an as-yet structurally undefined N-terminal region followed by the first of four HGF kringles. We have used a combination of molecular modeling and mutagenesis to dissect the function of this region of HGF. Two mutation-sensitive patches on the proposed surface regions of HGF kringle one (K1) were identified. The first patch consists of residues E159, S161, E195 and R197, all of which are predicted to be close to each other in the tertiary structure of K1. The second patch, lying on the opposite side of the kringle, consists of residues D171 and Q173. Mutational analysis of the N-terminal region of HGF identified residue D117 which also appeared to influence receptor binding. We also investigated the properties of a naturally occurring HGF variant (delta 5-HGF) that arises from an alternatively spliced transcript and therefore lacks five residues within K1. Our data suggest that in wild-type HGF, F162 is crucial in maintaining the hydrophobic core of the kringle. In delta 5-HGF, the loss of this residue is compensated for by a functional substitution of F162 with Y167, which is predicted to occupy the delta 5-HGF K1 core. Comparison of the models of wild-type and delta 5 kringles reveals that the positions of the presumed receptor binding determinants remain unchanged.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Simulación por Computador , Factor de Crecimiento de Hepatocito/metabolismo , Kringles , Modelos Moleculares , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas Receptoras/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Células CHO , Línea Celular , Cricetinae , Cristalografía por Rayos X , Perros , Fibrina/metabolismo , Riñón , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Proteínas Proto-Oncogénicas c-met , Proteínas Tirosina Quinasas Receptoras/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal
17.
J Biol Chem ; 273(6): 3216-22, 1998 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-9452434

RESUMEN

Flt-1 is one of two receptor tyrosine kinases through which the angiogenic factor vascular endothelial growth factor (VEGF) functions. Placenta growth factor (PlGF) is an additional ligand for Flt-1. The second immunoglobulin-like domain in the extracellular domain of Flt-1 has previously been identified as the region containing the critical ligand-binding determinants. We analyzed the contribution of charged residues within the first three domains of Flt-1 to ligand binding by alanine-scanning mutagenesis. Domain 2 residues Arg159, Glu208 and His223-Arg224 (together) affect both VEGF and PlGF binding, while Glu137, Lys171, His223, and Arg224 affect PlGF but not VEGF. Several charged residues, especially Asp187, are important in maintaining the structural integrity of domain 2. In addition, some residues in domain 3 contribute to binding (Asp231) or provide for additional discrimination between ligands (Arg280-Asp283).


Asunto(s)
Factores de Crecimiento Endotelial/química , Inmunoglobulinas/química , Linfocinas/química , Proteínas Proto-Oncogénicas/química , Proteínas Tirosina Quinasas Receptoras/química , Alanina/química , Alanina/metabolismo , Secuencia de Aminoácidos , Factores de Crecimiento Endotelial/genética , Factores de Crecimiento Endotelial/metabolismo , Humanos , Inmunoglobulinas/metabolismo , Linfocinas/genética , Linfocinas/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Homología de Secuencia de Aminoácido , Factor A de Crecimiento Endotelial Vascular , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
18.
Biochemistry ; 33(33): 9856-64, 1994 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-8060992

RESUMEN

In this work we have investigated the contribution to protein stability of residues forming the boundaries of alpha-helices. At the N-terminus of helix 2 of human growth hormone there are two residues, Ser71 and Glu74, which form two reciprocal hydrogen bonds between the side chains and the backbone nitrogens of either residue (the N-capping box). In order to evaluate the stabilizing effect of each hydrogen bond, site-directed mutagenesis was employed. In addition, the effect of side-chain negative charge on helix stabilization, via charge dipole interaction, was assessed. Ultraviolet spectroscopy and near- and far-UV CD spectroscopy as well as guanidine hydrochloride protein denaturation were used as assays to monitor the conformational and free energy of stabilization changes induced by the point mutations. The results of these experiments can be summarized as follows: (a) receptor binding studies showed that the tertiary conformation of each mutant was similar to that of the native hormone, (b) far-UV CD spectroscopic analyses showed that the overall alpha-helical content was unchanged in the mutants, (c) UV absorption and CD spectroscopic analyses indicated small alterations in helical packing in those mutants in which the hydrogen bond between the side chain of Ser71 and backbone NH of Glu74 was disrupted, (d) the hydrogen bond involving the side chain of Ser71 contributes at least 1.0 kcal/mol to protein stabilization and has a 2-fold larger stabilizing effect than that of the hydrogen bond involving the Glu74 side chain, and (e) the putative charge-dipole interaction of Glu74 with the alpha-helix dipole does not contribute to the stabilization of the tertiary conformation of human growth hormone.


Asunto(s)
Hormona del Crecimiento/química , Dicroismo Circular , Cristalización , Estabilidad de Medicamentos , Hormona del Crecimiento/genética , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Plásmidos , Mutación Puntual , Desnaturalización Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Espectrofotometría Ultravioleta , Relación Estructura-Actividad , Termodinámica
19.
Protein Eng ; 9(7): 617-21, 1996 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8844834

RESUMEN

'Knobs-into-holes' was originally proposed by Crick in 1952 as a model for the packing of amino acid side chains between adjacent alpha-helices. 'Knobs-into-holes' is demonstrated here as a novel and effective design strategy for engineering antibody heavy chain homodimers for heterodimerization. In this approach a 'knob' variant was first obtained by replacement of a small amino acid with a larger one in the CH3 domain of a CD4-IgG immunoadhesin: T366Y. The knob was designed to insert into a 'hole' in the CH3 domain of a humanized anti-CD3 antibody created by judicious replacement of a large residue with a smaller one: Y407T. The anti-CD3/CD4-IgG hybrid represents up to 92% of the protein A purified protein pool following co-expression of these two different heavy chains together with the anti-CD3 light chain. In contrast, only up to 57% of the anti-CD3/CD4-IgG hybrid is recovered following co-expression in which heavy chains contained wild-type CH3 domains. Thus knobs-into-holes engineering facilitates the construction of an antibody/immunoadehsin hybrid and likely other Fc-containing bifunctional therapeutics including bispecific immunoadhesins and bispecific antibodies.


Asunto(s)
Anticuerpos/metabolismo , Antígenos CD/inmunología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Cadenas Pesadas de Inmunoglobulina/metabolismo , Anticuerpos/genética , Complejo CD3/inmunología , Antígenos CD4/inmunología , Dimerización , Variación Genética , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Cadenas Pesadas de Inmunoglobulina/genética , Modelos Moleculares , Unión Proteica , Ingeniería de Proteínas/métodos
20.
Biochemistry ; 35(6): 1897-903, 1996 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-8639672

RESUMEN

This report describes the effect on antigen binding of an isomerized aspartate residue located in the complementarity-determining regions (CDRs) of a recombinant monoclonal antibody. The antibody, which binds human IgE, contains two Asp-Gly sequences within its CDRs, but only one site was found to be labile to isomerization. Isolation and characterization of antibody fragments differing in the labile sequence were facilitated by using a technique involving hydrophobic interaction chromatography (HIC) that separates aspartyl, isoaspartyl, and cyclic imide variants to the residue located in CDR-L1. The variants were isolated for structural characterization and for determination of their relative antigen binding affinities. Mutants were constructed with altered residues to obviate the effects of isomerization and were evaluated for their ability to bind to IgE. Inspection of published crystal structures of CDRs of antibodies indicated that hydrogen binding of the Asp side chain of the unreactive residue may be the constraint that prevents isomerization. The strategy outlined here may prove to be of general utility in the biochemical and immunochemical characterization of recombinant antibodies.


Asunto(s)
Anticuerpos Antiidiotipos/química , Inmunoglobulina E/inmunología , Región Variable de Inmunoglobulina/química , Secuencia de Aminoácidos , Anticuerpos Antiidiotipos/genética , Anticuerpos Antiidiotipos/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Afinidad de Anticuerpos , Ácido Aspártico/química , Variación Genética , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/metabolismo , Técnicas In Vitro , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutación Puntual , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Estereoisomerismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda