Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Biol Cell ; 30(17): 2331-2347, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31216232

RESUMO

Syk/Zap70 family kinases are essential for signaling via multichain immune-recognition receptors such as tetrameric (αßγ2) FcεRI. Syk activation is generally attributed to cis binding of its tandem SH2 domains to dual phosphotyrosines within FcεRIγ-ITAMs (immunoreceptor tyrosine-based activation motifs). However, the mechanistic details of Syk docking on γ homodimers are unresolved. Here, we estimate that multivalent interactions for WT Syk improve cis-oriented binding by three orders of magnitude. We applied molecular dynamics (MD), hybrid MD/worm-like chain polymer modeling, and live cell imaging to evaluate relative binding and signaling output for all possible cis and trans Syk-FcεRIγ configurations. Syk binding is likely modulated during signaling by autophosphorylation on Y130 in interdomain A, since a Y130E phosphomimetic form of Syk is predicted to lead to reduced helicity of interdomain A and alter Syk's bias for cis binding. Experiments in reconstituted γ-KO cells, whose γ subunits are linked by disulfide bonds, as well as in cells expressing monomeric ITAM or hemITAM γ-chimeras, support model predictions that short distances between γ ITAM pairs are required for trans docking. We propose that the full range of docking configurations improves signaling efficiency by expanding the combinatorial possibilities for Syk recruitment, particularly under conditions of incomplete ITAM phosphorylation.


Assuntos
Receptores de IgE/metabolismo , Quinase Syk/metabolismo , Quinase Syk/ultraestrutura , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Modelos Biológicos , Modelos Teóricos , Fosforilação , Fosfotirosina/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores de IgE/ultraestrutura , Transdução de Sinais , Tirosina/metabolismo , Proteína-Tirosina Quinase ZAP-70 , Domínios de Homologia de src
2.
PLoS One ; 7(12): e51669, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23284735

RESUMO

We present a model of the early events in mast cell signaling mediated by FcεRI where the plasma membrane is composed of many small ordered lipid domains (rafts), surrounded by a non-order region of lipids consisting of the remaining plasma membrane. The model treats the rafts as transient structures that constantly form and breakup, but that maintain a fixed average number per cell. The rafts have a high propensity for harboring Lyn kinase, aggregated, but not unaggregated receptors, and the linker for the activation of T cells (LAT). Phosphatase activity in the rafts is substantially reduced compared to the nonraft region. We use the model to analyze published experiments on the rat basophilic leukemia (RBL)-2H3 cell line that seem to contradict the notion that rafts offer protection. In these experiments IgE was cross-linked with a multivalent antigen and then excess monovalent hapten was added to break-up cross-links. The dephosphorylation of the unaggregated receptor (nonraft associated) and of LAT (raft associated) were then monitored in time and found to decay at similar rates, leading to the conclusion that rafts offer no protection from dephosphorylation. In the model, because the rafts are transient, a protein that is protected while in a raft will be subject to dephosphorylation when the raft breaks up and the protein finds itself in the nonraft region of the membrane. We show that the model is consistent with the receptor and LAT dephosphorylation experiments while still allowing rafts to enhance signaling by providing substantial protection from phosphatases.


Assuntos
Imunoglobulina E/metabolismo , Leucemia Basofílica Aguda/metabolismo , Mastócitos/metabolismo , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Receptores de IgE/metabolismo , Quinases da Família src/metabolismo , Animais , Cálcio/metabolismo , Membrana Celular/metabolismo , Simulação por Computador , Haptenos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucemia Basofílica Aguda/patologia , Lipoilação , Mastócitos/patologia , Microdomínios da Membrana/patologia , Mutação/genética , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Ratos , Transdução de Sinais , Quinase Syk , Células Tumorais Cultivadas , Tirosina/metabolismo , Quinases da Família src/genética
3.
PLoS Comput Biol ; 7(10): e1002192, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22022247

RESUMO

Numerous signaling proteins use multivalent binding to increase the specificity and affinity of their interactions within the cell. Enhancement arises because the effective binding constant for multivalent binding is larger than the binding constants for each individual interaction. We seek to gain both qualitative and quantitative understanding of the multivalent interactions of an adaptor protein, growth factor receptor bound protein-2 (Grb2), containing two SH3 domains interacting with the nucleotide exchange factor son-of-sevenless 1 (Sos1) containing multiple polyproline motifs separated by flexible unstructured regions. Grb2 mediates the recruitment of Sos1 from the cytosol to the plasma membrane where it activates Ras by inducing the exchange of GDP for GTP. First, using a combination of evolutionary information and binding energy calculations, we predict an additional polyproline motif in Sos1 that binds to the SH3 domains of Grb2. This gives rise to a total of five polyproline motifs in Sos1 that are capable of binding to the two SH3 domains of Grb2. Then, using a hybrid method combining molecular dynamics simulations and polymer models, we estimate the enhancement in local concentration of a polyproline motif on Sos1 near an unbound SH3 domain of Grb2 when its other SH3 domain is bound to a different polyproline motif on Sos1. We show that the local concentration of the Sos1 motifs that a Grb2 SH3 domain experiences is approximately 1000 times greater than the cellular concentration of Sos1. Finally, we calculate the intramolecular equilibrium constants for the crosslinking of Grb2 on Sos1 and use thermodynamic modeling to calculate the stoichiometry. With these equilibrium constants, we are able to predict the distribution of complexes that form at physiological concentrations. We believe this is the first systematic analysis that combines sequence, structure, and thermodynamic analyses to determine the stoichiometry of the complexes that are dominant in the cellular environment.


Assuntos
Proteína Adaptadora GRB2/metabolismo , Proteína SOS1/metabolismo , Sequência de Aminoácidos , Animais , Proteína Adaptadora GRB2/química , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Estrutura Molecular , Probabilidade , Ligação Proteica , Proteína SOS1/química , Homologia de Sequência de Aminoácidos
4.
J Immunol ; 185(6): 3268-76, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20733205

RESUMO

The term serial engagement was introduced to describe the ability of a single peptide, bound to a MHC molecule, to sequentially interact with TCRs within the contact region between a T cell and an APC. In addition to ligands on surfaces, soluble multivalent ligands can serially engage cell surface receptors with sites on the ligand, binding and dissociating from receptors many times before all ligand sites become free and the ligand leaves the surface. To evaluate the role of serial engagement in Syk activation, we use a detailed mathematical model of the initial signaling cascade that is triggered when FcepsilonRI is aggregated on mast cells by multivalent Ags. Although serial engagement is not required for mast cell signaling, it can influence the recruitment of Syk to the receptor and subsequent Syk phosphorylation. Simulating the response of mast cells to ligands that serially engage receptors at different rates shows that increasing the rate of serial engagement by increasing the rate of dissociation of the ligand-receptor bond decreases Syk phosphorylation. Increasing serial engagement by increasing the rate at which receptors are cross-linked (for example by increasing the forward rate constant for cross-linking or increasing the valence of the ligand) increases Syk phosphorylation. When serial engagement enhances Syk phosphorylation, it does so by partially reversing the effects of kinetic proofreading. Serial engagement rapidly returns receptors that have dissociated from aggregates to new aggregates before the receptors have fully returned to their basal state.


Assuntos
Imunoglobulina E/metabolismo , Fragmentos de Imunoglobulinas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mastócitos/enzimologia , Mastócitos/imunologia , Modelos Imunológicos , Proteínas Tirosina Quinases/metabolismo , Receptores de IgE/metabolismo , Regulação para Cima/imunologia , Animais , Sítios de Ligação de Anticorpos/genética , Linhagem Celular Tumoral , Ativação Enzimática/genética , Ativação Enzimática/imunologia , Imunoglobulina E/química , Imunoglobulina E/fisiologia , Fragmentos de Imunoglobulinas/química , Fragmentos de Imunoglobulinas/fisiologia , Leucemia Basofílica Aguda/enzimologia , Leucemia Basofílica Aguda/imunologia , Ligantes , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Mastócitos/metabolismo , Valor Preditivo dos Testes , Transporte Proteico/genética , Transporte Proteico/imunologia , Ratos , Receptores de IgE/química , Receptores de IgE/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Quinase Syk , Regulação para Cima/genética
5.
Cell Immunol ; 255(1-2): 8-16, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-18845299

RESUMO

The accessory protein MD2 has been implicated in LPS-mediated activation of the innate immune system by functioning as a co-receptor with TLR4 for LPS binding at the cell surface. Epithelial cells that play a role in primary immune response, such as in the lung or gut, often express TLR4, but are dependent on circulating soluble MD2 (sMD2) to bind TLR4 to assemble the functional receptor. In this study, we show that sMD2 incubation with HEK293 epithelial cells transfected with TLR4 increases the cell surface levels of TLR4 in the absence of LPS. Dose response studies reveal that a threshold sMD2 concentration (approximately 450 nM) stimulates maximal TLR4 levels on the cell surface, whereas higher concentrations of sMD2 (approximately 1800 nM) reduce these enhanced TLR4 levels. We show evidence that MD2 multimer formation is increased at these higher concentrations of sMD2 and that addition of LPS to sMD2-stimulated cells masks the enhanced TLR4 cell surface levels, most likely due to the LPS-induced downregulation of TLR4 by endocytosis following receptor stimulation. All together, these results support a model in which sMD2 binds to TLR4 and increases TLR4 levels at the cell surface by preventing TLR4 turnover through the endocytic pathway. Thus, sMD2 may prime epithelial cells for enhanced immunoresponsive function prior to LPS exposure.


Assuntos
Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Antígeno 96 de Linfócito/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Linhagem Celular , Células Epiteliais/citologia , Humanos , Lipopolissacarídeos/imunologia , Antígeno 96 de Linfócito/genética , Receptor 4 Toll-Like/genética
6.
Semin Immunol ; 19(4): 216-24, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17398114

RESUMO

A recent workshop discussed the recognition of multiple distinct ligands by individual T cell and B cell receptors and the implications of this discovery for lymphocyte biology. The workshop recommends general use of the term polyspecificity because it emphasizes two fundamental aspects, the inherent specificity of receptor recognition and the ability to recognize multiple ligands. Many different examples of polyspecificity and the structural mechanisms were discussed, and the group concluded that polyspecificity is a general, inherent feature of TCR and antibody recognition. This review summarizes the relevance of polyspecificity for lymphocyte development, activation and disease processes.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos B/imunologia , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Humanos , Ligantes , Camundongos , Peptídeos/imunologia
7.
J Immunol ; 178(6): 3530-5, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17339448

RESUMO

Cells may discriminate among ligands with different dwell times for receptor binding through a mechanism called kinetic proofreading in which the formation of an activated receptor complex requires a progression of events that is aborted if the ligand dissociates before completion. This mechanism explains how, at equivalent levels of receptor occupancy, a rapidly dissociating ligand can be less effective than a more slowly dissociating analog at generating distal cellular responses. Simple mathematical models predict that kinetic proofreading is limited to the initial complex; once the signal passes to second messengers, the dwell time no longer regulates the signal. This suggests that an assay for kinetic proofreading might be used to determine which activation events occur within the initial signaling complex. In signaling through the high affinity IgE receptor FcepsilonRI, the transmembrane adaptor called linker for activation of T cells (LAT) is thought to nucleate a distinct secondary complex. Experiments in which the concentrations of two ligands with different dwell times are adjusted to equalize the level of LAT phosphorylation in rat basophilic leukemia 2H3 cells show that Erk2 phosphorylation, intracellular Ca(2+), and degranulation exhibit kinetic proofreading downstream of LAT phosphorylation. These results suggest that ligand-bound FcepsilonRI and LAT form a complex that is required for effective signal transmission.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sinalização do Cálcio/fisiologia , Proteínas de Membrana/metabolismo , Modelos Biológicos , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Receptores de IgE/metabolismo , Animais , Linhagem Celular Tumoral , Cinética , Ligantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosforilação , Ratos
8.
Biosystems ; 83(2-3): 136-51, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16233948

RESUMO

We consider a model of early events in signaling by the epidermal growth factor (EGF) receptor (EGFR). The model includes EGF, EGFR, the adapter proteins Grb2 and Shc, and the guanine nucleotide exchange factor Sos, which is activated through EGF-induced formation of EGFR-Grb2-Sos and EGFR-Shc-Grb2-Sos assemblies at the plasma membrane. The protein interactions involved in signaling can potentially generate a diversity of protein complexes and phosphoforms; however, this diversity has been largely ignored in models of EGFR signaling. Here, we develop a model that accounts more fully for potential molecular diversity by specifying rules for protein interactions and then using these rules to generate a reaction network that includes all chemical species and reactions implied by the protein interactions. We obtain a model that predicts the dynamics of 356 molecular species, which are connected through 3749 unidirectional reactions. This network model is compared with a previously developed model that includes only 18 chemical species but incorporates the same scope of protein interactions. The predictions of this model are reproduced by the network model, which also yields new predictions. For example, the network model predicts distinct temporal patterns of autophosphorylation for different tyrosine residues of EGFR. A comparison of the two models suggests experiments that could lead to mechanistic insights about competition among adapter proteins for EGFR binding sites and the role of EGFR monomers in signal transduction.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Hepatócitos/metabolismo , Fígado/metabolismo , Modelos Biológicos , Transdução de Sinais/fisiologia , Animais , Técnicas de Química Combinatória , Simulação por Computador , Regulação da Expressão Gênica/fisiologia , Complexos Multienzimáticos/metabolismo , Ratos
9.
Proc Natl Acad Sci U S A ; 102(13): 4824-9, 2005 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-15772168

RESUMO

CD8(+) T cells recognize peptides of eight to nine amino acid residues long in the context of MHC class I molecules on the surface of antigen-presenting cells (APCs). This recognition event is highly sensitive, as evidenced by the fact that T cells can be activated by cognate peptide/MHC complex (pMHC) at extremely low densities (1-50 molecules). High sensitivity is particularly valuable for detection of antigens at low density, such as those derived from tumor cells and intracellular pathogens, which can down-modulate cognate pMHCs from the surface of APCs to evade recognition by the adaptive immune system. T cell activation is only triggered in response to interactions between the T cell receptor (TCR) and the pMHC ligand that reach a specific half-life threshold. However, interactions with excessively long half-lives result in impaired T cell activation. Thus, efficient T cell activation by pMHC on the surface of APCs requires an optimal dwell time of TCR-pMHC interaction. Here, we show that, although this is a requirement at low cognate pMHC density on the APC surface, at high epitope density there is no impairment of T cell activation by extended TCR-pMHC dwell times. This observation was predicted by mathematical simulations for T cell activation by pMHC at different densities and supported by experiments performed on APCs selected for varied expression of cognate pMHC. According to these results, effective T cell activation depends on a complex interplay between inherent TCR-pMHC binding kinetics and the epitope density on the APC.


Assuntos
Células Apresentadoras de Antígenos/metabolismo , Epitopos de Linfócito T/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Ativação Linfocitária/fisiologia , Modelos Imunológicos , Receptores de Antígenos de Linfócitos T/metabolismo , Células Cultivadas , Citometria de Fluxo , Antígenos H-2/metabolismo , Humanos , Hibridomas/imunologia , Cinética , Ligantes , Ligação Proteica
10.
J Immunol ; 170(7): 3769-81, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12646643

RESUMO

Aggregation of Fc epsilon RI on mast cells and basophils leads to autophosphorylation and increased activity of the cytosolic protein tyrosine kinase Syk. We investigated the roles of the Src kinase Lyn, the immunoreceptor tyrosine-based activation motifs (ITAMs) on the beta and gamma subunits of Fc epsilon RI, and Syk itself in the activation of Syk. Our approach was to build a detailed mathematical model of reactions involving Fc epsilon RI, Lyn, Syk, and a bivalent ligand that aggregates Fc(epsilon)RI. We applied the model to experiments in which covalently cross-linked IgE dimers stimulate rat basophilic leukemia cells. The model makes it possible to test the consistency of mechanistic assumptions with data that alone provide limited mechanistic insight. For example, the model helps sort out mechanisms that jointly control dephosphorylation of receptor subunits. In addition, interpreted in the context of the model, experimentally observed differences between the beta- and gamma-chains with respect to levels of phosphorylation and rates of dephosphorylation indicate that most cellular Syk, but only a small fraction of Lyn, is available to interact with receptors. We also show that although the beta ITAM acts to amplify signaling in experimental systems where its role has been investigated, there are conditions under which the beta ITAM will act as an inhibitor.


Assuntos
Modelos Químicos , Modelos Imunológicos , Receptores de IgE/química , Receptores de IgE/fisiologia , Transdução de Sinais/imunologia , Motivos de Aminoácidos/imunologia , Animais , Dimerização , Relação Dose-Resposta Imunológica , Regulação para Baixo/imunologia , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Ligantes , Fosforilação , Ligação Proteica/imunologia , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Subunidades Proteicas/fisiologia , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Ratos , Receptores de IgE/antagonistas & inibidores , Receptores de IgE/metabolismo , Software , Quinase Syk , Células Tumorais Cultivadas , Tirosina/metabolismo , Regulação para Cima/imunologia , Quinases da Família src/química , Quinases da Família src/metabolismo
11.
Biotechnol Bioeng ; 84(7): 783-94, 2003 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-14708119

RESUMO

Many activities of cells are controlled by cell-surface receptors, which in response to ligands, trigger intracellular signaling reactions that elicit cellular responses. A hallmark of these signaling reactions is the reversible nucleation of multicomponent complexes, which typically begin to assemble when ligand-receptor binding allows an enzyme, often a kinase, to create docking sites for signaling molecules through chemical modifications, such as tyrosine phosphorylation. One function of such docking sites is the co-localization of enzymes with their substrates, which can enhance both enzyme activity and specificity. The directed assembly of complexes can also influence the sensitivity of cellular responses to ligand-receptor binding kinetics and determine whether a cellular response is up- or downregulated in response to a ligand stimulus. The full functional implications of ligand-stimulated complex formation are difficult to discern intuitively. Complex formation is governed by conditional interactions among multivalent signaling molecules and influenced by quantitative properties of both the components in a system and the system itself. Even a simple list of the complexes that can potentially form in response to a ligand stimulus is problematic because of the number of ways signaling molecules can be modified and combined. Here, we review the role of multicomponent complexes in signal transduction and advocate the use of mathematical models that incorporate detail at the level of molecular domains to study this important aspect of cellular signaling.


Assuntos
Modelos Biológicos , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia , Receptores ErbB/química , Receptores ErbB/metabolismo , Ligação Proteica , Receptores de IgE/química , Receptores de IgE/metabolismo
12.
Mol Immunol ; 38(16-18): 1213-9, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12217386

RESUMO

We present a detailed mathematical model of the phosphorylation and dephosphorylation events that occur upon ligand-induced receptor aggregation, for a transfectant expressing FcepsilonRI, Lyn, Syk and endogenous phosphatases that dephosphorylate exposed phosphotyrosines on FcepsilonRI and Syk. Through model simulations we show how changing the ligand concentration, and consequently the concentration of receptor aggregates, can change the nature of a cellular response as well as its amplitude. We illustrate the value of the model in analyzing experimental data by using it to show that the intrinsic rate of dephosphorylation of the FcepsilonRI gamma immunoreceptor tyrosine-based activation motif (ITAM) in rat basophilic leukemia (RBL) cells is much faster than the observed rate, provided that all of the cytosolic Syk is available to receptors.


Assuntos
Modelos Teóricos , Receptores de IgE/metabolismo , Transdução de Sinais , Animais , Basófilos/enzimologia , Basófilos/imunologia , Células CHO , Cricetinae , Precursores Enzimáticos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Ligantes , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Ratos , Quinase Syk , Células Tumorais Cultivadas , Quinases da Família src/metabolismo
13.
Nat Immunol ; 3(10): 926-31, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12244312

RESUMO

To assess the roles of serial engagement and kinetic proofreading in T cell receptor (TCR) internalization, we have developed a mathematical model of this process. Our determination of TCR down-regulation for an array of TCR mutants, interpreted in the context of the model, has provided new information about peptide-induced TCR internalization. The amount of TCR down-regulation increases to a maximum value and then declines as a function of the half-life of the bond between the TCR and peptide-major histocompatibility complex (pMHC). The model shows that this behavior, which reflects competition between serial engagement and kinetic proofreading, arises only if it is postulated that activated TCRs remain marked for internalization after dissociation from pMHC. The model also predicts that because of kinetic proofreading, the range of TCR-pMHC-binding half-lives required for T cell activation depends on the concentrations and localization of intracellular signaling molecules. We show here that kinetic proofreading provides an explanation for the different requirements for activation observed in naïve and memory T cells.


Assuntos
Apresentação de Antígeno/imunologia , Endocitose/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Modelos Imunológicos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Animais , Regulação para Baixo , Cinética , Computação Matemática , Camundongos , Peptídeos/imunologia , Transdução de Sinais
14.
Biochemistry ; 41(8): 2543-51, 2002 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-11851401

RESUMO

Interleukin-2 is the primary T cell growth factor secreted by activated T cells. IL-2 is an alpha-helical cytokine that binds to a multisubunit receptor expressed on the surface of a variety of cell types. IL-2Ralpha, IL-2Rbeta, and IL-2Rgammac receptor subunits expressed on the surface of cells may aggregate to form distinct binding sites of differing affinities. IL-2Rgammac was the last receptor subunit to be identified. It has since been shown to be shared by at least five other cytokine receptors. In this study, we have probed the role of IL-2Rgammac in the assembly of IL-2R complexes and in ligand binding. We demonstrate that in the absence of ligand IL-2Rgammac does not possess detectable affinity for IL-2Ralpha, IL-2Rbeta, or the pseudo-high-affinity binding site composed of preformed IL-2Ralpha/beta. We also demonstrate that IL-2Rgammac possesses an IL-2-dependent affinity for IL-2Rbeta and IL-2Ralpha/beta. We performed a detailed biosensor analysis to examine the interaction of soluble IL-2Rgammac with IL-2-bound IL-2Rbeta and IL-2-bound IL-2Ralpha/beta. The kinetic and equilibrium constants for sIL-2Rgammac binding to these two different liganded complexes were similar, indicating that IL-2Ralpha does not play a role in recruitment of IL-2Rgammac. We also determined that the binding of IL-2 to the isolated IL-2Rgammac was very weak (approximate K(D) = 0.7 mM). The experimental methodologies and principles derived from these studies can be extended to at least five other cytokines that share IL-2Rgammac as a receptor subunit.


Assuntos
Receptores de Interleucina-2/metabolismo , Técnicas Biossensoriais , Humanos , Interleucina-2/metabolismo , Cinética , Ligantes , Ligação Proteica , Ensaio Radioligante , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas , Ultracentrifugação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA