Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
MAbs ; 10(6): 901-912, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29958069

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Asparagina/química , Imunoglobulina G/química , Modelos Químicos , Amidas/química , Sítios de Ligação de Anticorpos , Concentração de Íons de Hidrogênio , Conformação Molecular , Estrutura Molecular
2.
MAbs ; 10(3): 406-415, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29436927

RESUMO

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.


Assuntos
Anticorpos Monoclonais/análise , Cadeias Pesadas de Imunoglobulinas/análise , Animais , Anticorpos Monoclonais/química , Células CHO , Cricetulus , Medição da Troca de Deutério , Glicosilação , Humanos , Cadeias Pesadas de Imunoglobulinas/química , Relação Estrutura-Atividade
3.
Proc Natl Acad Sci U S A ; 114(21): E4223-E4232, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28484017

RESUMO

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.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Antineoplásicos Imunológicos/farmacologia , Sítios de Ligação de Anticorpos/imunologia , Antígeno CTLA-4/antagonistas & inibidores , Ipilimumab/farmacologia , Melanoma/tratamento farmacológico , Fatores Biológicos/farmacologia , Antígeno CTLA-4/imunologia , Linhagem Celular , Cristalografia por Raios X , Células HEK293 , Humanos , Imunoterapia/métodos , Ligação Proteica , Estrutura Terciária de Proteína
4.
SLAS Discov ; 22(3): 324-331, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27932698

RESUMO

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.


Assuntos
Ensaios de Triagem em Larga Escala , Fatores Imunológicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Receptores de Antígenos de Linfócitos T/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Proteína-Tirosina Quinase ZAP-70/genética , Sistema Livre de Células/química , Polarização de Fluorescência/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , Expressão Gênica , Humanos , Fatores Imunológicos/química , Peptídeos/antagonistas & inibidores , Peptídeos/genética , Peptídeos/imunologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/química , Receptores de Antígenos de Linfócitos T/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transdução de Sinais , Bibliotecas de Moléculas Pequenas/química , Proteína-Tirosina Quinase ZAP-70/antagonistas & inibidores , Proteína-Tirosina Quinase ZAP-70/imunologia
5.
Elife ; 52016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27700984

RESUMO

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.


Assuntos
Proteína Tirosina Quinase p56(lck) Linfócito-Específica/química , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Eletricidade Estática , Proteína-Tirosina Quinase ZAP-70/química , Proteína-Tirosina Quinase ZAP-70/metabolismo , Células HEK293 , Humanos , Especificidade por Substrato
6.
Anal Chem ; 88(19): 9495-9502, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27575380

RESUMO

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.


Assuntos
Anticorpos Monoclonais/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulina G/metabolismo , Metionina/química , Receptores Fc/metabolismo , Anticorpos Monoclonais/química , Deutério , Antígenos de Histocompatibilidade Classe I/química , Humanos , Imunoglobulina G/química , Espectrometria de Massas/métodos , Oxirredução , Ligação Proteica , Conformação Proteica , Processamento de Proteína Pós-Traducional , Receptores Fc/química
7.
Proc Natl Acad Sci U S A ; 113(29): 8218-23, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27370798

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Membrana/metabolismo , Fosfotirosina/metabolismo , Proteínas Son Of Sevenless/metabolismo , Proteína Adaptadora GRB2/metabolismo , Cinética , Membranas Artificiais , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Proteínas ras
8.
Biochem J ; 465(1): 149-61, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25287889

RESUMO

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.


Assuntos
Cisteína/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfopeptídeos/metabolismo , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Proteína-Tirosina Quinase ZAP-70/química , Proteína-Tirosina Quinase ZAP-70/metabolismo , Domínios de Homologia de src , Motivos de Aminoácidos , Sítios de Ligação , Humanos , Peróxido de Hidrogênio/farmacologia , Modelos Moleculares , Oxirredução/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Compostos de Sulfidrila/metabolismo , Quinase Syk , Proteína-Tirosina Quinase ZAP-70/antagonistas & inibidores
9.
Mol Cell Biol ; 33(11): 2188-201, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23530057

RESUMO

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.


Assuntos
Proteína-Tirosina Quinase ZAP-70/química , Proteína-Tirosina Quinase ZAP-70/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cristalografia por Raios X , Transferência Ressonante de Energia de Fluorescência , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/genética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Fosforilação , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Quinase Syk , Tirosina/química , Tirosina/metabolismo , Proteína-Tirosina Quinase ZAP-70/genética , Domínios de Homologia de src , Quinases da Família src/química , Quinases da Família src/metabolismo
10.
Structure ; 19(2): 162-71, 2011 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-21300286

RESUMO

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.


Assuntos
Conformação Molecular , Membro 6b de Receptores do Fator de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Clonagem Molecular , Cristalização , Cristalografia por Raios X , Drosophila melanogaster , Proteína Ligante Fas/imunologia , Proteína Ligante Fas/metabolismo , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Estrutura Terciária de Proteína , Membro 6b de Receptores do Fator de Necrose Tumoral/genética , Membro 6b de Receptores do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
11.
Biochemistry ; 48(32): 7636-45, 2009 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-19522538

RESUMO

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.


Assuntos
Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Dissulfetos/química , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Dobramento de Proteína , Multimerização Proteica , Membro 6b de Receptores do Fator de Necrose Tumoral/química , Membro 6b de Receptores do Fator de Necrose Tumoral/metabolismo , Alinhamento de Sequência , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética
12.
Immunol Rev ; 229(1): 356-86, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19426233

RESUMO

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.


Assuntos
Ativação Linfocitária/genética , Receptores de Superfície Celular/química , Receptores de Superfície Celular/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Cristalografia por Raios X , Genômica , Humanos , Dados de Sequência Molecular , Conformação Proteica , Receptores de Superfície Celular/genética , Alinhamento de Sequência
13.
Proc Natl Acad Sci U S A ; 105(30): 10483-8, 2008 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-18641123

RESUMO

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.


Assuntos
Antígenos de Superfície/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Cristalografia por Raios X/métodos , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Escherichia coli/metabolismo , Ligantes , Ativação Linfocitária , Camundongos , Dados de Sequência Molecular , Mutação , Proteína 2 Ligante de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1 , Conformação Proteica , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos , Linfócitos T/citologia
14.
Proc Natl Acad Sci U S A ; 104(25): 10583-8, 2007 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-17563375

RESUMO

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.


Assuntos
Antígenos CD/química , Antígenos CD/metabolismo , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos , Antígenos CD/genética , Sequência Conservada , Cristalografia por Raios X , Dimerização , Humanos , Cinética , Modelos Químicos , Modelos Moleculares , Dados de Sequência Molecular , Mutação Puntual , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Família de Moléculas de Sinalização da Ativação Linfocitária , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária , Ultracentrifugação , Difração de Raios X
15.
Immunity ; 25(4): 559-70, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17045824

RESUMO

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.


Assuntos
Antígenos CD/química , Receptores de Superfície Celular/química , Sequência de Aminoácidos , Antígenos CD/imunologia , Antígenos CD2/química , Antígenos CD58/química , Cristalografia por Raios X , Dimerização , Humanos , Ativação Linfocitária , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Receptores de Superfície Celular/imunologia , Transdução de Sinais , Família de Moléculas de Sinalização da Ativação Linfocitária , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA