Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(21): E4223-E4232, 2017 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-28484017

RESUMEN

Rational modulation of the immune response with biologics represents one of the most promising and active areas for the realization of new therapeutic strategies. In particular, the use of function blocking monoclonal antibodies targeting checkpoint inhibitors such as CTLA-4 and PD-1 have proven to be highly effective for the systemic activation of the human immune system to treat a wide range of cancers. Ipilimumab is a fully human antibody targeting CTLA-4 that received FDA approval for the treatment of metastatic melanoma in 2011. Ipilimumab is the first-in-class immunotherapeutic for blockade of CTLA-4 and significantly benefits overall survival of patients with metastatic melanoma. Understanding the chemical and physical determinants recognized by these mAbs provides direct insight into the mechanisms of pathway blockade, the organization of the antigen-antibody complexes at the cell surface, and opportunities to further engineer affinity and selectivity. Here, we report the 3.0 Å resolution X-ray crystal structure of the complex formed by ipilimumab with its human CTLA-4 target. This structure reveals that ipilimumab contacts the front ß-sheet of CTLA-4 and intersects with the CTLA-4:Β7 recognition surface, indicating that direct steric overlap between ipilimumab and the B7 ligands is a major mechanistic contributor to ipilimumab function. The crystallographically observed binding interface was confirmed by a comprehensive cell-based binding assay against a library of CTLA-4 mutants and by direct biochemical approaches. This structure also highlights determinants responsible for the selectivity exhibited by ipilimumab toward CTLA-4 relative to the homologous and functionally related CD28.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Antineoplásicos Inmunológicos/farmacología , Sitios de Unión de Anticuerpos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Ipilimumab/farmacología , Melanoma/tratamiento farmacológico , Factores Biológicos/farmacología , Antígeno CTLA-4/inmunología , Línea Celular , Cristalografía por Rayos X , Células HEK293 , Humanos , Inmunoterapia/métodos , Unión Proteica , Estructura Terciaria de Proteína
2.
Proc Natl Acad Sci U S A ; 113(29): 8218-23, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27370798

RESUMEN

The assembly of cell surface receptors with downstream signaling molecules is a commonly occurring theme in multiple signaling systems. However, little is known about how these assemblies modulate reaction kinetics and the ultimate propagation of signals. Here, we reconstitute phosphotyrosine-mediated assembly of extended linker for the activation of T cells (LAT):growth factor receptor-bound protein 2 (Grb2):Son of Sevenless (SOS) networks, derived from the T-cell receptor signaling system, on supported membranes. Single-molecule dwell time distributions reveal two, well-differentiated kinetic species for both Grb2 and SOS on the LAT assemblies. The majority fraction of membrane-recruited Grb2 and SOS both exhibit fast kinetics and single exponential dwell time distributions, with average dwell times of hundreds of milliseconds. The minor fraction exhibits much slower kinetics, extending the dwell times to tens of seconds. Considering this result in the context of the multistep process by which the Ras GEF (guanine nucleotide exchange factor) activity of SOS is activated indicates that kinetic stabilization from the LAT assembly may be important. This kinetic proofreading effect would additionally serve as a stochastic noise filter by reducing the relative probability of spontaneous SOS activation in the absence of receptor triggering. The generality of receptor-mediated assembly suggests that such effects may play a role in multiple receptor proximal signaling processes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de la Membrana/metabolismo , Fosfotirosina/metabolismo , Proteínas Son Of Sevenless/metabolismo , Proteína Adaptadora GRB2/metabolismo , Cinética , Membranas Artificiales , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Proteínas ras
3.
Anal Chem ; 88(19): 9495-9502, 2016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27575380

RESUMEN

Hydrogen/deuterium exchange mass spectrometry (HDX MS) was used in two case studies to evaluate the impact of methionine (Met) oxidation on the biological functions of IgG1 antibodies. In the first case study, linear correlations were observed between the oxidation of the conserved Fc methionine residues and the loss of neonatal Fc receptor (FcRn) binding and complement-dependent cytotoxicity (CDC) activity. Both heavy chain (HC) residues Met257 and Met433 were located near the FcRn binding interface as indicated by HDX MS and structural modeling; however, HC Met257 oxidation was further demonstrated to have a more significant impact on FcRn binding than HC Met433 oxidation. In addition, oxidation of HC Met257 and HC Met433 could disrupt protein conformation at the CH2-CH3 interface and prevent IgG oligomerization, which is needed for C1q binding and subsequent CDC activity. In the second case study, HDX MS demonstrated that oxidation of the two complementary determining region (CDR) methionine residues had little or no impact on antigen binding of the antibody. Together, these results suggested that HDX MS is a powerful tool for evaluating the impact of individual post translational modifications (PTMs) on the biological activities of antibodies, even when the PTM levels are relatively low. The high selectivity and sensitivity of this method makes it a valuable tool for assisting the critical quality attributes (CQAs) assessment of antibodies.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoglobulina G/metabolismo , Metionina/química , Receptores Fc/metabolismo , Anticuerpos Monoclonales/química , Deuterio , Antígenos de Histocompatibilidad Clase I/química , Humanos , Inmunoglobulina G/química , Espectrometría de Masas/métodos , Oxidación-Reducción , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional , Receptores Fc/química
4.
Biochem J ; 465(1): 149-61, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25287889

RESUMEN

Zeta-chain associated protein of 70 kDa (ZAP-70) and spleen tyrosine kinase (Syk) are non-receptor tyrosine kinases that are essential for T-cell and B-cell antigen receptor signalling respectively. They are recruited, via their tandem-SH2 (Src-homology domain 2) domains, to doubly phosphorylated immunoreceptor tyrosine-based activation motifs (ITAMs) on invariant chains of immune antigen receptors. Because of their critical roles in immune signalling, ZAP-70 and Syk are targets for the development of drugs for autoimmune diseases. We show that three thiol-reactive small molecules can prevent the tandem-SH2 domains of ZAP-70 and Syk from binding to phosphorylated ITAMs. We identify a specific cysteine residue in the phosphotyrosine-binding pocket of each protein (Cys39 in ZAP-70, Cys206 in Syk) that is necessary for inhibition by two of these compounds. We also find that ITAM binding to ZAP-70 and Syk is sensitive to the presence of H2O2 and these two cysteine residues are also necessary for inhibition by H2O2. Our findings suggest a mechanism by which the reactive oxygen species generated during responses to antigen could attenuate signalling through these kinases and may also inform the development of ZAP-70 and Syk inhibitors that bind covalently to their SH2 domains.


Asunto(s)
Cisteína/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosfopéptidos/metabolismo , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Proteína Tirosina Quinasa ZAP-70/química , Proteína Tirosina Quinasa ZAP-70/metabolismo , Dominios Homologos src , Secuencias de Aminoácidos , Sitios de Unión , Humanos , Peróxido de Hidrógeno/farmacología , Modelos Moleculares , Oxidación-Reducción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Unión Proteica/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Compuestos de Sulfhidrilo/metabolismo , Quinasa Syk , Proteína Tirosina Quinasa ZAP-70/antagonistas & inhibidores
5.
Immunol Rev ; 229(1): 356-86, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19426233

RESUMEN

SUMMARY: Costimulatory receptors and ligands trigger the signaling pathways that are responsible for modulating the strength, course, and duration of an immune response. High-resolution structures have provided invaluable mechanistic insights by defining the chemical and physical features underlying costimulatory receptor:ligand specificity, affinity, oligomeric state, and valency. Furthermore, these structures revealed general architectural features that are important for the integration of these interactions and their associated signaling pathways into overall cellular physiology. Recent technological advances in structural biology promise unprecedented opportunities for furthering our understanding of the structural features and mechanisms that govern costimulation. In this review, we highlight unique insights that have been revealed by structures of costimulatory molecules from the immunoglobulin and tumor necrosis factor superfamilies and describe a vision for future structural and mechanistic analysis of costimulation. This vision includes simple strategies for the selection of candidate molecules for structure determination and highlights the critical role of structure in the design of mutant costimulatory molecules for the generation of in vivo structure-function correlations in a mammalian model system. This integrated 'atoms-to-animals' paradigm provides a comprehensive approach for defining atomic and molecular mechanisms.


Asunto(s)
Activación de Linfocitos/genética , Receptores de Superficie Celular/química , Receptores de Superficie Celular/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cristalografía por Rayos X , Genómica , Humanos , Datos de Secuencia Molecular , Conformación Proteica , Receptores de Superficie Celular/genética , Alineación de Secuencia
6.
Proc Natl Acad Sci U S A ; 105(30): 10483-8, 2008 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-18641123

RESUMEN

Programmed death-1 (PD-1) is a member of the CD28/B7 superfamily that delivers negative signals upon interaction with its two ligands, PD-L1 or PD-L2. The high-resolution crystal structure of the complex formed by the complete ectodomains of murine PD-1 and PD-L2 revealed a 1:1 receptor:ligand stoichiometry and displayed a binding interface and overall molecular organization distinct from that observed in the CTLA-4/B7 inhibitory complexes. Furthermore, our structure also provides insights into the association between PD-1 and PD-L1 and highlights differences in the interfaces formed by the two PD-1 ligands (PD-Ls) Mutagenesis studies confirmed the details of the proposed PD-1/PD-L binding interfaces and allowed for the design of a mutant PD-1 receptor with enhanced affinity. These studies define spatial and organizational constraints that control the localization and signaling of PD-1/PD-L complexes within the immunological synapse and provide a basis for manipulating the PD-1 pathways for immunotherapy.


Asunto(s)
Antígenos de Superficie/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Cristalografía por Rayos X/métodos , Péptidos/metabolismo , Secuencia de Aminoácidos , Animales , Escherichia coli/metabolismo , Ligandos , Activación de Linfocitos , Ratones , Datos de Secuencia Molecular , Mutación , Proteína 2 Ligando de Muerte Celular Programada 1 , Receptor de Muerte Celular Programada 1 , Conformación Proteica , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido , Linfocitos T/citología
7.
Biochemistry ; 48(32): 7636-45, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19522538

RESUMEN

TNF-like 1A (TL1A) is a newly described member of the TNF superfamily that is directly implicated in the pathogenesis of autoimmune diseases, including inflammatory bowel disease, atherosclerosis, and rheumatoid arthritis. We report the crystal structure of the human TL1A extracellular domain at a resolution of 2.5 A, which reveals a jelly-roll fold typical of the TNF superfamily. This structural information, in combination with complementary mutagenesis and biochemical characterization, provides insights into the binding interface and the specificity of the interactions between TL1A and the DcR3 and DR3 receptors. These studies suggest that the mode of interaction between TL1A and DcR3 differs from other characterized TNF ligand/receptor complexes. In addition, we have generated functional TL1A mutants with altered disulfide bonding capability that exhibit enhanced solution properties, which will facilitate the production of materials for future cell-based and whole animal studies. In summary, these studies provide insights into the structure and function of TL1A and provide the basis for the rational manipulation of its interactions with cognate receptors.


Asunto(s)
Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cristalografía por Rayos X , Disulfuros/química , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Pliegue de Proteína , Multimerización de Proteína , Miembro 6b de Receptores del Factor de Necrosis Tumoral/química , Miembro 6b de Receptores del Factor de Necrosis Tumoral/metabolismo , Alineación de Secuencia , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
8.
MAbs ; 10(6): 901-912, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29958069

RESUMEN

Identification of asparagine (Asn) sites that are prone to deamidation is critical for the development of therapeutic monoclonal antibodies (mAbs). Despite a common chemical degradation pathway, the rates of Asn deamidation can vary dramatically among different sites, and prediction of the sensitive deamidation sites is still challenging. In this study, characterization of Asn deamidation for five IgG1 and five IgG4 mAbs under both normal and stressed conditions revealed dramatic differences in the Asn deamidation rates. A comprehensive analysis of the deamidation sites indicated that the deamidation rate differences could be explained by differences in the local structure conformation, structure flexibility and solvent accessibility. A decision tree was developed to predict the deamidation propensity for all Asn sites in IgG mAbs based on the analysis of these three structural parameters. This decision tree will allow potential Asn deamidation hot spots to be identified early in development.


Asunto(s)
Anticuerpos Monoclonales/química , Asparagina/química , Inmunoglobulina G/química , Modelos Químicos , Amidas/química , Sitios de Unión de Anticuerpos , Concentración de Iones de Hidrógeno , Conformación Molecular , Estructura Molecular
9.
MAbs ; 10(3): 406-415, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29436927

RESUMEN

Glycation has been observed in antibody therapeutics manufactured by the fed-batch fermentation process. It not only increases the heterogeneity of antibodies, but also potentially affects product safety and efficacy. In this study, non-glycated and glycated fractions enriched from a monoclonal antibody (mAb1) as well as glucose-stressed mAb1 were characterized using a variety of biochemical, biophysical and biological assays to determine the effects of glycation on the structure and function of mAb1. Glycation was detected at multiple lysine residues and reduced the antigen binding activity of mAb1. Heavy chain Lys100, which is located in the complementary-determining region of mAb1, had the highest levels of glycation in both stressed and unstressed samples, and glycation of this residue was likely responsible for the loss of antigen binding based on hydrogen/deuterium exchange mass spectrometry analysis. Peptide mapping and intact liquid chromatography-mass spectrometry (LC-MS) can both be used to monitor the glycation levels. Peptide mapping provides site specific glycation results, while intact LC-MS is a quicker and simpler method to quantitate the total glycation levels and is more useful for routine testing. Capillary isoelectric focusing (cIEF) can also be used to monitor glycation because glycation induces an acidic shift in the cIEF profile. As expected, total glycation measured by intact LC-MS correlated very well with the percentage of total acidic peaks or main peak measured by cIEF. In summary, we demonstrated that glycation can affect the function of a representative IgG1 mAb. The analytical characterization, as described here, should be generally applicable for other therapeutic mAbs.


Asunto(s)
Anticuerpos Monoclonales/análisis , Cadenas Pesadas de Inmunoglobulina/análisis , Animales , Anticuerpos Monoclonales/química , Células CHO , Cricetulus , Medición de Intercambio de Deuterio , Glicosilación , Humanos , Cadenas Pesadas de Inmunoglobulina/química , Relación Estructura-Actividad
10.
SLAS Discov ; 22(3): 324-331, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27932698

RESUMEN

ZAP-70 is a critical molecule in the transduction of T cell antigen receptor signaling and the activation of T cells. Upon activation of the T cell antigen receptor, ZAP-70 is recruited to the intracellular ζ-chains of the T cell receptor, where ZAP-70 is activated and colocalized with its substrates. Inhibitors of ZAP-70 could potentially function as treatments for autoimmune diseases or organ transplantation. In this work, we present the design, optimization, and implementation of a screen for inhibitors that would disrupt the interaction between ZAP-70 and the T cell antigen receptor. The screen is based on a fluorescence polarization assay for peptide binding to ZAP-70.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Factores Inmunológicos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Antígenos de Linfocitos T/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Proteína Tirosina Quinasa ZAP-70/genética , Sistema Libre de Células/química , Polarización de Fluorescencia/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Expresión Génica , Humanos , Factores Inmunológicos/química , Péptidos/antagonistas & inhibidores , Péptidos/genética , Péptidos/inmunología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química , Proteína Tirosina Quinasa ZAP-70/antagonistas & inhibidores , Proteína Tirosina Quinasa ZAP-70/inmunología
11.
Elife ; 52016 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-27700984

RESUMEN

The sequence of events that initiates T cell signaling is dictated by the specificities and order of activation of the tyrosine kinases that signal downstream of the T cell receptor. Using a platform that combines exhaustive point-mutagenesis of peptide substrates, bacterial surface-display, cell sorting, and deep sequencing, we have defined the specificities of the first two kinases in this pathway, Lck and ZAP-70, for the T cell receptor ζ chain and the scaffold proteins LAT and SLP-76. We find that ZAP-70 selects its substrates by utilizing an electrostatic mechanism that excludes substrates with positively-charged residues and favors LAT and SLP-76 phosphosites that are surrounded by negatively-charged residues. This mechanism prevents ZAP-70 from phosphorylating its own activation loop, thereby enforcing its strict dependence on Lck for activation. The sequence features in ZAP-70, LAT, and SLP-76 that underlie electrostatic selectivity likely contribute to the specific response of T cells to foreign antigens.


Asunto(s)
Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/química , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Electricidad Estática , Proteína Tirosina Quinasa ZAP-70/química , Proteína Tirosina Quinasa ZAP-70/metabolismo , Células HEK293 , Humanos , Especificidad por Sustrato
12.
Mol Cell Biol ; 33(11): 2188-201, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23530057

RESUMEN

Serial activation of the tyrosine kinases Lck and ZAP-70 initiates signaling downstream of the T cell receptor. We previously reported the structure of an autoinhibited ZAP-70 variant in which two regulatory tyrosine residues (315 and 319) in the SH2-kinase linker were replaced by phenylalanine. We now present a crystal structure of ZAP-70 in which Tyr 315 and Tyr 319 are not mutated, leading to the recognition of a five-residue sequence register error in the SH2-kinase linker of the original crystallographic model. The revised model identifies distinct roles for these two tyrosines. As seen in a recently reported structure of the related tyrosine kinase Syk, Tyr 315 of ZAP-70 is part of a hydrophobic interface between the regulatory apparatus and the kinase domain, and the integrity of this interface would be lost upon engagement of doubly phosphorylated peptides by the SH2 domains. Tyr 319 is not necessarily dislodged by SH2 engagement, which activates ZAP-70 only ∼5-fold in vitro. In contrast, phosphorylation by Lck activates ZAP-70 ∼100-fold. This difference is due to the ability of Tyr 319 to suppress ZAP-70 activity even when the SH2 domains are dislodged from the kinase domain, providing stringent control of ZAP-70 activity downstream of Lck.


Asunto(s)
Proteína Tirosina Quinasa ZAP-70/química , Proteína Tirosina Quinasa ZAP-70/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cristalografía por Rayos X , Transferencia Resonante de Energía de Fluorescencia , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Humanos , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Fosforilación , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/metabolismo , Quinasa Syk , Tirosina/química , Tirosina/metabolismo , Proteína Tirosina Quinasa ZAP-70/genética , Dominios Homologos src , Familia-src Quinasas/química , Familia-src Quinasas/metabolismo
13.
Structure ; 19(2): 162-71, 2011 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-21300286

RESUMEN

Decoy Receptor 3 (DcR3), a secreted member of the Tumor Necrosis Factor (TNF) receptor superfamily, neutralizes three different TNF ligands: FasL, LIGHT, and TL1A. Each of these ligands engages unique signaling receptors which direct distinct and critical immune responses. We report the crystal structures of the unliganded DcR3 ectodomain and its complex with TL1A, as well as complementary mutagenesis and biochemical studies. These analyses demonstrate that DcR3 interacts with invariant backbone and side-chain atoms in the membrane-proximal half of TL1A which supports recognition of its three distinct TNF ligands. Additional features serve as antideterminants that preclude interaction with other members of the TNF superfamily. This mode of interaction is unique among characterized TNF:TNFR family members and provides a mechanistic basis for the broadened specificity required to support the decoy function of DcR3, as well as for the rational manipulation of specificity and affinity of DcR3 and its ligands.


Asunto(s)
Conformación Molecular , Miembro 6b de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Secuencia de Aminoácidos , Animales , Células Cultivadas , Clonación Molecular , Cristalización , Cristalografía por Rayos X , Drosophila melanogaster , Proteína Ligando Fas/inmunología , Proteína Ligando Fas/metabolismo , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína , Miembro 6b de Receptores del Factor de Necrosis Tumoral/genética , Miembro 6b de Receptores del Factor de Necrosis Tumoral/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología
14.
Proc Natl Acad Sci U S A ; 104(25): 10583-8, 2007 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-17563375

RESUMEN

The signaling lymphocyte activation molecule (SLAM) family includes homophilic and heterophilic receptors that modulate both adaptive and innate immune responses. These receptors share a common ectodomain organization: a membrane-proximal immunoglobulin constant domain and a membrane-distal immunoglobulin variable domain that is responsible for ligand recognition. CD84 is a homophilic family member that enhances IFN-gamma secretion in activated T cells. Our solution studies revealed that CD84 strongly self-associates with a K(d) in the submicromolar range. These data, in combination with previous reports, demonstrate that the SLAM family homophilic affinities span at least three orders of magnitude and suggest that differences in the affinities may contribute to the distinct signaling behavior exhibited by the individual family members. The 2.0 A crystal structure of the human CD84 immunoglobulin variable domain revealed an orthogonal homophilic dimer with high similarity to the recently reported homophilic dimer of the SLAM family member NTB-A. Structural and chemical differences in the homophilic interfaces provide a mechanism to prevent the formation of undesired heterodimers among the SLAM family homophilic receptors. These structural data also suggest that, like NTB-A, all SLAM family homophilic dimers adopt a highly kinked organization spanning an end-to-end distance of approximately 140 A. This common molecular dimension provides an opportunity for all two-domain SLAM family receptors to colocalize within the immunological synapse and bridge the T cell and antigen-presenting cell.


Asunto(s)
Antígenos CD/química , Antígenos CD/metabolismo , Receptores de Superficie Celular/metabolismo , Secuencia de Aminoácidos , Antígenos CD/genética , Secuencia Conservada , Cristalografía por Rayos X , Dimerización , Humanos , Cinética , Modelos Químicos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Puntual , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Ultracentrifugación , Difracción de Rayos X
15.
Immunity ; 25(4): 559-70, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17045824

RESUMEN

The signaling lymphocytic activation molecule (SLAM) family includes homophilic and heterophilic receptors that regulate both innate and adaptive immunity. The ectodomains of most SLAM family members are composed of an N-terminal IgV domain and a C-terminal IgC2 domain. NK-T-B-antigen (NTB-A) is a homophilic receptor that stimulates cytotoxicity in natural killer (NK) cells, regulates bactericidal activities in neutrophils, and potentiates T helper 2 (Th2) responses. The 3.0 A crystal structure of the complete NTB-A ectodomain revealed a rod-like monomer that self-associates to form a highly kinked dimer spanning an end-to-end distance of approximately 100 A. The NTB-A homophilic and CD2-CD58 heterophilic dimers show overall structural similarities but differ in detailed organization and physicochemical properties of their respective interfaces. The NTB-A structure suggests a mechanism responsible for binding specificity within the SLAM family and imposes physical constraints relevant to the colocalization of SLAM-family proteins with other signaling molecules in the immunological synapse.


Asunto(s)
Antígenos CD/química , Receptores de Superficie Celular/química , Secuencia de Aminoácidos , Antígenos CD/inmunología , Antígenos CD2/química , Antígenos CD58/química , Cristalografía por Rayos X , Dimerización , Humanos , Activación de Linfocitos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Receptores de Superficie Celular/inmunología , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA