Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Mol Cell ; 40(4): 548-57, 2010 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21095585

RESUMO

Inactivating mutations in the ubiquitin (Ub) editing protein A20 promote persistent nuclear factor (NF)-κB signaling and are genetically linked to inflammatory diseases and hematologic cancers. A20 tightly regulates NF-κB signaling by acting as an Ub editor, removing K63-linked Ub chains and mediating addition of Ub chains that target substrates for degradation. However, a precise molecular understanding of how A20 modulates this pathway remains elusive. Here, using structural analysis, domain mapping, and functional assays, we show that A20 zinc finger 4 (ZnF4) does not directly interact with E2 enzymes but instead can bind mono-Ub and K63-linked poly-Ub. Mutations to the A20 ZnF4 Ub-binding surface result in decreased A20-mediated ubiquitination and impaired regulation of NF-κB signaling. Collectively, our studies illuminate the mechanistically distinct but biologically interdependent activities of the A20 ZnF and ovarian tumor (OTU) domains that are inherent to the Ub editing process and, ultimately, to regulation of NF-κB signaling.


Assuntos
NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Dedos de Zinco , Sítios de Ligação , Cristalografia por Raios X , Lisina/metabolismo , Modelos Moleculares , Mutação/genética , Proteínas Nucleares/química , Poliubiquitina/metabolismo , Ligação Proteica , Especificidade por Substrato , Enzimas de Conjugação de Ubiquitina/metabolismo
2.
Mol Cell ; 31(4): 544-556, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18722180

RESUMO

In vitro, the anaphase-promoting complex (APC) E3 ligase functions with E2 ubiquitin-conjugating enzymes of the E2-C and Ubc4/5 families to ubiquitinate substrates. However, only the use of the E2-C family, notably UbcH10, is genetically well validated. Here, we biochemically demonstrate preferential use of UbcH10 by the APC, specified by the E2 core domain. Importantly, an additional E2-E3 interaction mediated by the N-terminal extension of UbcH10 regulates APC activity. Mutating the highly conserved N terminus increases substrate ubiquitination and the number of substrate lysines targeted, allows ubiquitination of APC substrates lacking their destruction boxes, increases resistance to the APC inhibitors Emi1 and BubR1 in vitro, and bypasses the spindle checkpoint in vivo. Fusion of the UbcH10 N terminus to UbcH5 restricts ubiquitination activity but does not direct specific interactions with the APC. Thus, UbcH10 combines a specific E2-E3 interface and regulation via its N-terminal extension to limit APC activity for substrate selection and checkpoint control.


Assuntos
Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Sequência de Aminoácidos , Ciclossomo-Complexo Promotor de Anáfase , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Lisina/metabolismo , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato/efeitos dos fármacos , Complexos Ubiquitina-Proteína Ligase/antagonistas & inibidores , Ubiquitinação/efeitos dos fármacos
3.
Bioorg Med Chem Lett ; 25(21): 4728-4732, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26338362

RESUMO

A fragment-based lead discovery approach was used to discover novel ERK2 inhibitors. The crystal structure of N-benzyl-9H-purin-6-amine 1 in complex with ERK2 elucidated its hinge-binding mode. In addition, the simultaneous binding of an imidazole molecule adjacent to 1 suggested a direction for fragment expansion. Structure-based core hopping applied to 1 led to 5H-pyrrolo[3,2-b]pyrazine (3) that afforded direct vectors to probe the pockets of interest while retaining the essential hinge binding elements. Utilizing the new vectors for SAR exploration, the new core 3 was quickly optimized to compound 39 resulting in a greater than 6600-fold improvement in potency.


Assuntos
Descoberta de Drogas , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Pirazinas/farmacologia , Pirróis/farmacologia , Relação Dose-Resposta a Droga , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Modelos Moleculares , Estrutura Molecular , Pirazinas/síntese química , Pirazinas/química , Pirróis/síntese química , Pirróis/química , Relação Estrutura-Atividade
4.
Proc Natl Acad Sci U S A ; 109(14): 5299-304, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22431598

RESUMO

The Ras gene is frequently mutated in cancer, and mutant Ras drives tumorigenesis. Although Ras is a central oncogene, small molecules that bind to Ras in a well-defined manner and exert inhibitory effects have not been uncovered to date. Through an NMR-based fragment screen, we identified a group of small molecules that all bind to a common site on Ras. High-resolution cocrystal structures delineated a unique ligand-binding pocket on the Ras protein that is adjacent to the switch I/II regions and can be expanded upon compound binding. Structure analysis predicts that compound-binding interferes with the Ras/SOS interactions. Indeed, selected compounds inhibit SOS-mediated nucleotide exchange and prevent Ras activation by blocking the formation of intermediates of the exchange reaction. The discovery of a small-molecule binding pocket on Ras with functional significance provides a new direction in the search of therapeutically effective inhibitors of the Ras oncoprotein.


Assuntos
Nucleotídeos/metabolismo , Proteínas Son Of Sevenless/metabolismo , Proteínas ras/metabolismo , Sítios de Ligação , Linhagem Celular , Humanos , Ligantes , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Proteínas ras/química
5.
Bioorg Med Chem Lett ; 23(11): 3186-94, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23628333

RESUMO

A novel 2-thio-6-oxo-1,6-dihydropyrimidine-containing inhibitor of human lactate dehydrogenase (LDH) was identified by high-throughput screening (IC50=8.1 µM). Biochemical, surface plasmon resonance, and saturation transfer difference NMR experiments indicated that the compound specifically associated with human LDHA in a manner that required simultaneous binding of the NADH co-factor. Structural variation of the screening hit resulted in significant improvements in LDHA biochemical inhibition activity (best IC50=0.48 µM). A crystal structure of an optimized compound bound to human LDHA was obtained and explained many of the observed structure-activity relationships.


Assuntos
Inibidores Enzimáticos/química , L-Lactato Desidrogenase/antagonistas & inibidores , Pirimidinas/química , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Humanos , Ligação de Hidrogênio , L-Lactato Desidrogenase/metabolismo , Espectroscopia de Ressonância Magnética , NAD/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Pirimidinas/síntese química , Pirimidinas/metabolismo , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície
6.
Protein Sci ; 16(11): 2454-71, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17962403

RESUMO

High-temperature requirement A (HtrA) and its homologs contain a serine protease domain followed by one or two PDZ domains. Bacterial HtrA proteins and the mitochondrial protein HtrA2/Omi maintain cell function by acting as both molecular chaperones and proteases to manage misfolded proteins. The biological roles of the mammalian family members HtrA1 and HtrA3 are less clear. We report a detailed structural and functional analysis of the PDZ domains of human HtrA1 and HtrA3 using peptide libraries and affinity assays to define specificity, structural studies to view the molecular details of ligand recognition, and alanine scanning mutagenesis to investigate the energetic contributions of individual residues to ligand binding. In common with HtrA2/Omi, we show that the PDZ domains of HtrA1 and HtrA3 recognize hydrophobic polypeptides, and while C-terminal sequences are preferred, internal sequences are also recognized. However, the details of the interactions differ, as different domains rely on interactions with different residues within the ligand to achieve high affinity binding. The results suggest that mammalian HtrA PDZ domains interact with a broad range of hydrophobic binding partners. This promiscuous specificity resembles that of bacterial HtrA family members and suggests a similar function for recognizing misfolded polypeptides with exposed hydrophobic sequences. Our results support a common activation mechanism for the HtrA family, whereby hydrophobic peptides bind to the PDZ domain and induce conformational changes that activate the protease. Such a mechanism is well suited to proteases evolved for the recognition and degradation of misfolded proteins.


Assuntos
Serina Endopeptidases/química , Sequência de Aminoácidos , Chaperoninas/química , Escherichia coli/metabolismo , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Ligantes , Dados de Sequência Molecular , Domínios PDZ , Peptídeos/química , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
7.
Biomol NMR Assign ; 10(2): 345-9, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27386854

RESUMO

The deubiquitinase Ubiquitin Specific Protease 7 (USP7) is part of the regulatory cascade of proteins that modulates the activity of the tumor suppressor protein p53. Deubiquitination of its target Murine Double Minute 2 (MDM2) leads to increased proteosomal degradation of p53. Consequently, USP7 has emerged as an attractive oncology target because its inhibition stabilizes p53, thereby promoting p53-dependent apoptosis in cancer cells. Here we report the backbone resonance assignment for the 40.5 kDa catalytic domain of USP7.


Assuntos
Domínio Catalítico , Ressonância Magnética Nuclear Biomolecular , Ubiquitina Tiolesterase/química , Sequência de Aminoácidos , Humanos , Ubiquitina Tiolesterase/metabolismo , Peptidase 7 Específica de Ubiquitina
8.
J Mol Biol ; 316(3): 769-87, 2002 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-11866530

RESUMO

Vascular endothelial growth factor (VEGF) is a potent endothelial cell-specific mediator of angiogenesis and vasculogenesis. VEGF is involved pathologically in cancer, proliferative retinopathy and rheumatoid arthritis, and as such represents an important therapeutic target. Three classes of disulfide-constrained peptides that antagonize binding of the VEGF dimer to its receptors, KDR and Flt-1, were identified previously using phage display methods. NMR studies of a representative peptide from the most potent class of these peptide antagonists, v107 (GGNECDAIRMWEWECFERL), were undertaken to characterize its interactions with VEGF. v107 has no defined structure free in solution, but binding to VEGF induces folding of the peptide. The solution structure of the VEGF receptor-binding domain-v107 complex was determined using 3940 (1970 per VEGF monomer) internuclear distance and 476 (238 per VEGF monomer) dihedral angle restraints derived from NMR data obtained using samples containing either (13)C/(15)N-labeled protein plus excess unlabeled peptide or (13)C/(15)N-labeled peptide plus excess unlabeled protein. Residual dipolar coupling restraints supplemented the structure determination of the complex and were found to increase significantly both the global precision of VEGF in the complex and the agreement with available crystal structures of VEGF. The calculated ensemble of structures is of high precision and is in excellent agreement with the experimental restraints. v107 has a turn-helix conformation with hydrophobic residues partitioned to one face of the peptide and polar or charged residues at the other face. Contacts between two v107 peptides and the VEGF dimer are mediated by primarily hydrophobic side-chain interactions. The v107-binding site on VEGF overlaps partially with the binding site of KDR and is similar to that for domain 2 of Flt-1. The structure of the VEGF-v107 complex provides new insight into how binding to VEGF can be achieved that may be useful for the design of small molecule antagonists.


Assuntos
Bacteriófagos/química , Fatores de Crescimento Endotelial/antagonistas & inibidores , Fatores de Crescimento Endotelial/química , Linfocinas/antagonistas & inibidores , Linfocinas/química , Peptídeos/química , Peptídeos/farmacologia , Alanina/genética , Alanina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Dimerização , Dissulfetos/metabolismo , Fatores de Crescimento Endotelial/genética , Fatores de Crescimento Endotelial/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Linfocinas/genética , Linfocinas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Ressonância Magnética Nuclear Biomolecular , Peptídeos/síntese química , Peptídeos/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/antagonistas & inibidores , Receptores de Fatores de Crescimento/química , Receptores de Fatores de Crescimento/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular , Soluções , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
9.
PLoS One ; 9(7): e102695, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25029443

RESUMO

Receptor tyrosine kinase-like orphan receptors (ROR) 1 and 2 are atypical members of the receptor tyrosine kinase (RTK) family and have been associated with several human diseases. The vertebrate RORs contain an ATP binding domain that deviates from the consensus amino acid sequence, although the impact of this deviation on catalytic activity is not known and the kinase function of these receptors remains controversial. Recently, ROR2 was shown to signal through a Wnt responsive, ß-catenin independent pathway and suppress a canonical Wnt/ß-catenin signal. In this work we demonstrate that both ROR1 and ROR2 kinase domains are catalytically deficient while CAM-1, the C. elegans homolog of ROR, has an active tyrosine kinase domain, suggesting a divergence in the signaling processes of the ROR family during evolution. In addition, we show that substitution of the non-consensus residues from ROR1 or ROR2 into CAM-1 and MuSK markedly reduce kinase activity, while restoration of the consensus residues in ROR does not restore robust kinase function. We further demonstrate that the membrane-bound extracellular domain alone of either ROR1 or ROR2 is sufficient for suppression of canonical Wnt3a signaling, and that this domain can also enhance Wnt5a suppression of Wnt3a signaling. Based on these data, we conclude that human ROR1 and ROR2 are RTK-like pseudokinases.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Evolução Molecular , Modelos Moleculares , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/genética , Transdução de Sinais/genética , Sequência de Aminoácidos , Animais , Caenorhabditis elegans , Catálise , Células HEK293 , Humanos , Immunoblotting , Luciferases , Dados de Sequência Molecular , Fosforilação , Estrutura Terciária de Proteína , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/química , Alinhamento de Sequência , Especificidade da Espécie
10.
J Mol Biol ; 415(1): 22-8, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22079049

RESUMO

Jumping Translocation Breakpoint (JTB) is an orphan receptor that is conserved from nematodes to humans and whose gene expression in humans is strikingly upregulated in diverse types of cancers. Translocations occur frequently at the hJTB genomic locus, leading to multiple copies of a truncated JTB gene, which potentially encodes a soluble secreted ectodomain. In addition, JTB and its orthologs likely represent a unique and ancient protein family since homologs could not be identified by direct sequence comparison. In the present study, we have determined the NMR solution structure of the N-terminal ectodomain of human JTB, showing that its fold architecture is a new variant of a three-ß-strand antiparallel ß-meander. The JTB structure has a distant relationship to the midkine/pleiotrophin fold, particularly in the conservation of distinctive disulfide bridge patterns. The structure of this newly characterized small cysteine-rich domain suggests potential involvement of JTB in interactions with proteins or extracellular matrix and may help to uncover the elusive biological functions of this protein.


Assuntos
Citocinas/química , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Clonagem Molecular/métodos , Dissulfetos/química , Humanos , Proteínas de Membrana/metabolismo , Midkina , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Ressonância Magnética Nuclear Biomolecular/métodos , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Translocação Genética/genética
11.
J Mol Biol ; 408(3): 420-31, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21396940

RESUMO

Ubiquitination refers to the covalent addition of ubiquitin (Ub) to substrate proteins or other Ub molecules via the sequential action of three enzymes (E1, E2, and E3). Recent advances in mass spectrometry proteomics have made it possible to identify and quantify Ub linkages in biochemical and cellular systems. We used these tools to probe the mechanisms controlling linkage specificity for UbcH5A. UbcH5A is a promiscuous E2 enzyme with an innate preference for forming polyubiquitin chains through lysine 11 (K11), lysine 48 (K48), and lysine 63 (K63) of Ub. We present the crystal structure of a noncovalent complex between Ub and UbcH5A. This structure reveals an interaction between the Ub surface flanking K11 and residues adjacent to the E2 catalytic cysteine and suggests a possible role for this surface in formation of K11 linkages. Structure-guided mutagenesis, in vitro ubiquitination and quantitative mass spectrometry have been used to characterize the ability of residues in the vicinity of the E2 active site to direct synthesis of K11- and K63-linked polyubiquitin. Mutation of critical residues in the interface modulated the linkage specificity of UbcH5A, resulting in generation of more K63-linked chains at the expense of K11-linkage synthesis. This study provides direct evidence that the linkage specificity of E2 enzymes may be altered through active-site mutagenesis.


Assuntos
Enzimas de Conjugação de Ubiquitina/química , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo , Cristalografia por Raios X , Mutagênese Sítio-Dirigida , Proteínas Mutantes/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Enzimas de Conjugação de Ubiquitina/genética
12.
Structure ; 17(10): 1398-410, 2009 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-19836339

RESUMO

Members of the interleukin-1 (IL-1) family of cytokines play major roles in host defense and immune system regulation in infectious and inflammatory diseases. IL-1 cytokines trigger a biological response in effector cells by assembling a heterotrimeric signaling complex with two IL-1 receptor chains, a high-affinity primary receptor and a low-affinity coreceptor. To gain insights into the signaling mechanism of the novel IL-1-like cytokine IL-33, we first solved its solution structure and then performed a detailed biochemical and structural characterization of the interaction between IL-33, its primary receptor ST2, and the coreceptor IL-1RAcP. Using nuclear magnetic resonance data, we obtained a model of the IL-33/ST2 complex in solution that is validated by small-angle X-ray scattering (SAXS) data and is similar to the IL-1beta/IL-1R1 complex. We extended our SAXS analysis to the IL-33/ST2/IL-1RAcP and IL-1beta/IL-1R1/IL-1RAcP complexes and propose a general model of the molecular architecture of IL-1 ternary signaling complexes.


Assuntos
Proteína Acessória do Receptor de Interleucina-1/química , Interleucina-1/metabolismo , Interleucinas/química , Interleucinas/metabolismo , Receptores de Superfície Celular/química , Transdução de Sinais , Sítios de Ligação , Cristalografia por Raios X , Humanos , Interleucina-1/química , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Modelos Moleculares , Conformação Proteica , Receptores de Superfície Celular/metabolismo
13.
Biochemistry ; 45(39): 11974-82, 2006 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-17002296

RESUMO

Signal transduction pathways are frequently found to repress transcription of target genes in the absence of stimulation and, conversely, to upregulate transcription in the presence of a signal. Transcription factors are central in this dual regulatory mechanism and widely use a generalized mechanism to repress transcription through recruitment of a Sin3-histone deacetylase (HDAC) complex to their binding sites on DNA. The protein SAP18 (Sin3-associated polypeptide of 18 kDa) has been shown to play a key role in gene-specific recruitment of the HDAC complex by a number of transcription factors including Gli, GAGA, and Bicoid. The solution structure of SAP18 reveals a ubiquitin-like fold with several large loop insertions relative to other family members. This fold supports the functional role of SAP18 as a protein-protein adapter module and provides insight for how SAP18 may bridge the Sin3-HDAC complex to transcription factors.


Assuntos
Proteínas de Transporte/química , Dobramento de Proteína , Ubiquitina/química , Proteínas de Transporte/metabolismo , Proteínas Correpressoras , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Humanos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Ressonância Magnética Nuclear Biomolecular , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Ubiquitina/metabolismo
14.
J Biol Chem ; 280(8): 7218-27, 2005 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-15542592

RESUMO

TACI is a member of the tumor necrosis factor receptor superfamily and serves as a key regulator of B cell function. TACI binds two ligands, APRIL and BAFF, with high affinity and contains two cysteine-rich domains (CRDs) in its extracellular region; in contrast, BCMA and BR3, the other known high affinity receptors for APRIL and BAFF, respectively, contain only a single or partial CRD. However, another form of TACI exists wherein the N-terminal CRD is removed by alternative splicing. We find that this shorter form is capable of ligand-induced cell signaling and that the second CRD alone (TACI_d2) contains full affinity for both ligands. Furthermore, we report the solution structure and alanine-scanning mutagenesis of TACI_d2 along with co-crystal structures of APRIL.TACI_d2 and APRIL.BCMA complexes that together reveal the mechanism by which TACI engages high affinity ligand binding through a single CRD, and we highlight sources of ligand-receptor specificity within the APRIL/BAFF system.


Assuntos
Cisteína , Proteínas de Membrana/química , Receptores do Fator de Necrose Tumoral/química , Fator de Necrose Tumoral alfa/química , Processamento Alternativo , Animais , Fator Ativador de Células B , Antígeno de Maturação de Linfócitos B , Cristalização , Cristalografia por Raios X , Humanos , Ligantes , Proteínas de Membrana/genética , Camundongos , Mutagênese , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Estrutura Terciária de Proteína , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Soluções , Proteína Transmembrana Ativadora e Interagente do CAML , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral
15.
Biochemistry ; 42(20): 5977-83, 2003 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-12755599

RESUMO

BAFF/BLyS, a member of the tumor necrosis family (TNF) superfamily of ligands, is a crucial survival factor for B cells. BAFF binds three receptors, TACI, BCMA, and BR3, with signaling through BR3 being essential for promoting B cell function. Typical TNF receptor (TNFR) family members bind their cognate ligands through interactions with two cysteine-rich domains (CRDs). However, the extracellular domain (ECD) of BR3 consists of only a partial CRD, with cysteine spacing distinct from other modules described previously. Herein, we report the solution structure of the BR3 ECD. A core region of only 19 residues adopts a stable structure in solution. The BR3 fold is analogous to the first half of a canonical TNFR CRD but is stabilized by an additional noncanonical disulfide bond. BAFF-binding determinants were identified by shotgun alanine-scanning mutagenesis of the BR3 ECD expressed on phage. Several of the key BAFF-binding residues are presented from a beta-turn that we have shown previously to be sufficient for ligand binding when transferred to a structured beta-hairpin scaffold [Kayagaki, N., Yan, M., Seshasayee, D., Wang, H., Lee, W., French, D. M., Grewal, I. S., Cochran, A. G., Gordon, N. C., Yin, J., Starovasnik, M. A, and Dixit, V. M. (2002) Immunity 10, 515-524]. Outside of the turn, mutagenesis identifies additional hydrophobic contacts that enhance the BAFF-BR3 interaction. The crystal structure of the minimal hairpin peptide, bhpBR3, in complex with BAFF reveals intimate packing of the six-residue BR3 turn into a cavity on the ligand surface. Thus, BR3 binds BAFF through a highly focused interaction site, unprecedented in the TNFR family.


Assuntos
Proteínas de Membrana/química , Receptores do Fator de Necrose Tumoral/química , Fator de Necrose Tumoral alfa/química , Sequência de Aminoácidos , Fator Ativador de Células B , Receptor do Fator Ativador de Células B , Sítios de Ligação , Cisteína/química , Humanos , Técnicas In Vitro , Ligantes , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Soluções , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA