Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 280(39): 33620-6, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16009714

ABSTRACT

Epstein-Barr virus is a human herpesvirus that causes infectious mononucleosis and lymphoproliferative malignancies. LMP1 (latent membrane protein-1), which is encoded by this virus and which is essential for transformation of B lymphocytes, acts as a constitutively active mimic of the tumor necrosis factor receptor (TNFR) CD40. LMP1 is an integral membrane protein containing six transmembrane segments and a cytoplasmic domain at the C terminus that binds to intracellular TNFR-associated factors (TRAFs). TRAFs are intracellular co-inducers of downstream signaling from CD40 and other TNFRs, and TRAF3 is required for activation of B lymphocytes by LMP1. Cytoplasmic C-terminal activation region 1 of LMP1 bears a motif (PQQAT) that conforms to the TRAF recognition motif PVQET in CD40. In this study, we report the crystal structure of this portion of LMP1 C-terminal activation region-1 (204PQQATDD210) bound in complex with TRAF3. The PQQAT motif is bound in the same binding crevice on TRAF3 where CD40 is bound, providing a molecular mechanism for LMP1 to act as a CD40 decoy for TRAF3. The LMP1 motif is presented in the TRAF3 crevice as a close structural mimic of the PVQET motif in CD40, and the intermolecular contacts are similar. However, the viral protein makes a unique contact: a hydrogen bond network formed between Asp210 in LMP1 and Tyr395 and Arg393 in TRAF3. This intermolecular contact is not made in the CD40-TRAF3 complex. The additional hydrogen bonds may stabilize the complex and strengthen the binding to permit LMP1 to compete with CD40 for binding to the TRAF3 crevice, influencing downstream signaling to B lymphocytes and contributing to dysregulated signaling by LMP1.


Subject(s)
B-Lymphocytes/metabolism , CD40 Antigens/chemistry , Herpesvirus 4, Human/chemistry , TNF Receptor-Associated Factor 3/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Alanine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , B-Lymphocytes/virology , Binding Sites , Blotting, Western , CD40 Antigens/metabolism , Cell Transformation, Viral , Crystallography, X-Ray , Humans , Lymphocyte Activation , Mice , Microscopy, Fluorescence , Models, Molecular , Precipitin Tests , Transfection , Viral Matrix Proteins/genetics
2.
J Immunol ; 173(12): 7394-400, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15585864

ABSTRACT

B cell-activating factor belonging to the TNF family receptor (BAFF-R), a member of the TNFR superfamily, plays a role in autoimmunity after ligation with BAFF ligand (also called TALL-1, BLyS, THANK, or zTNF4). BAFF/BAFF-R interactions are critical for B cell regulation, and signaling from this ligand-receptor complex results in NF-kappaB activation. Most TNFRs transmit signals intracellularly by recruitment of adaptor proteins called TNFR-associated factors (TRAFs). However, BAFF-R binds only one TRAF adaptor, TRAF3, and this interaction negatively regulates activation of NF-kappaB. In this study, we report the crystal structure of a 24-residue fragment of the cytoplasmic portion of BAFF-R bound in complex with TRAF3. The recognition motif (162)PVPAT(166) in BAFF-R is accommodated in the same binding crevice on TRAF3 that binds two related TNFRs, CD40 and LTbetaR, but is presented in a completely different structural framework. This region of BAFF-R assumes an open conformation with two extended strands opposed at right angles that each make contacts with TRAF3. The recognition motif is located in the N-terminal arm and intermolecular contacts mediate TRAF recognition. In the C-terminal arm, key stabilizing contacts are made, including critical hydrogen bonds with Gln(379) in TRAF3 that define the molecular basis for selective binding of BAFF-R solely to this member of the TRAF family. A dynamic conformational adjustment of Tyr(377) in TRAF3 occurs forming a new intermolecular contact with BAFF-R that stabilizes the complex. The structure of the complex provides a molecular explanation for binding affinities and selective protein interactions in TNFR-TRAF interactions.


Subject(s)
Intracellular Fluid/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/immunology , TNF Receptor-Associated Factor 3/chemistry , TNF Receptor-Associated Factor 3/metabolism , Amino Acid Motifs , Amino Acid Sequence , B-Cell Activation Factor Receptor , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Crystallography, X-Ray , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , DNA Mutational Analysis , Humans , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/genetics , Protein Binding/immunology , Protein Conformation , Receptors, Tumor Necrosis Factor/genetics , Signal Transduction/genetics , TNF Receptor-Associated Factor 3/genetics , Thermodynamics
3.
J Biol Chem ; 278(50): 50523-9, 2003 Dec 12.
Article in English | MEDLINE | ID: mdl-14517219

ABSTRACT

Lymphotoxin-beta receptor (LTbetaR) and CD40 are members of the tumor necrosis factor family of signaling receptors that regulate cell survival or death through activation of NF-kappaB. These receptors transmit signals through downstream adaptor proteins called tumor necrosis factor receptor-associated factors (TRAFs). In this study, the crystal structure of a region of the cytoplasmic domain of LTbetaR bound to TRAF3 has revealed an unexpected new recognition motif, 388IPEEGD393, for TRAF3 binding. Although this motif is distinct in sequence and structure from the PVQET motif in CD40 and PIQCT in the regulator TRAF-associated NF-kappaB activator (TANK), recognition is mediated in the same binding crevice on the surface of TRAF3. The results reveal structurally adaptive "hot spots" in the TRAF3-binding crevice that promote molecular interactions driving specific signaling after contact with LTbetaR, CD40, or the downstream regulator TANK.


Subject(s)
Adaptor Proteins, Signal Transducing , CD40 Antigens/biosynthesis , Receptors, Tumor Necrosis Factor/chemistry , Signal Transduction , Amino Acid Motifs , Amino Acid Sequence , CD40 Antigens/chemistry , Cell Line , Cell Survival , Crystallography, X-Ray , DNA, Complementary/metabolism , Electrons , Glutathione Transferase/metabolism , Humans , Lymphotoxin beta Receptor , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Fusion Proteins/metabolism
4.
J Mol Recognit ; 16(3): 121-4, 2003.
Article in English | MEDLINE | ID: mdl-12833566

ABSTRACT

Caspases are cysteine proteases that play a critical role in the initiation and regulation of apoptosis. These enzymes act in a cascade to promote cell death through proteolytic cleavage of intracellular proteins. Since activation of apoptosis is implicated in human diseases such as cancer and neurodegenerative disorders, caspases are targets for drugs designed to modulate their action. Active caspases are heterodimeric enzymes with two symmetrically arranged active sites at opposite ends of the molecule. A number of crystal structures of caspases with peptides or proteins bound at the active sites have defined the mechanism of action of these enzymes, but molecular information about the active sites before substrate engagement has been lacking. As part of a study of peptidyl inhibitors of caspase-3, we crystallized a complex where the inhibitor did not bind in the active site. Here we present the crystal structure of the unoccupied substrate-binding site of caspase-3. No large conformational differences were apparent when this site was compared with that in enzyme-inhibitor complexes. Instead, the 1.9 A structure reveals critical side chain movements in a hydrophobic pocket in the active site. Notably, the side chain of tyrosine204 is rotated by approximately 90 degrees so that the phenol group occupies the S2 subsite in the active site. Thus, binding of substrate or inhibitors is impeded unless rotation of this side chain opens the area. The positions of these side chains may have important implications for the directed design of inhibitors of caspase-3 or caspase-7.


Subject(s)
Caspases/chemistry , Caspases/metabolism , Apoptosis , Binding Sites , Caspase 3 , Cell Line , Crystallization , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein
5.
J Biol Chem ; 278(3): 1837-40, 2003 Jan 17.
Article in English | MEDLINE | ID: mdl-12421809

ABSTRACT

Seven in absentia homologue (Siah) family proteins bind ubiquitin-conjugating enzymes and target proteins for proteasome-mediated degradation. Recently we identified a novel Siah-interacting protein (SIP) that is a Sgt1-related molecule that provides a physical link between Siah family proteins and the Skp1-Cullin-F-box ubiquitin ligase component Skp1. In the present study, a structure-based approach was used to identify interacting residues in Siah that are required for association with SIP. In Siah1 a large concave surface is formed across the dimer interface. Analysis of the electrostatic surface potential of the Siah1 dimer reveals that the beta-sheet concavity is predominately electronegative, suggesting that the protein-protein interactions between Siah1 and SIP are mediated by ionic contacts. The structural prediction was confirmed by site-directed mutagenesis of these electronegative residues, resulting in loss of binding of Siah1 to SIP in vitro and in cells. The results also provide a structural basis for understanding the mechanism by which Siah family proteins interact with partner proteins such as SIP.


Subject(s)
Calcium-Binding Proteins , Carrier Proteins/metabolism , Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Cell Line , Dimerization , Humans , Models, Molecular , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Protein Conformation , Ubiquitin-Protein Ligases
6.
Acta Crystallogr D Biol Crystallogr ; 58(Pt 8): 1340-2, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12136149

ABSTRACT

Tumor necrosis factor receptors (TNFR) signal events in immune responses, Ig class switching, activation of NF-kappaB or regulation of apoptosis. TNFR-associated factors (TRAFs) are adaptor proteins that connect TNFRs to downstream signaling pathways, including the NF-kappaB and c-JUN N-terminal kinase (JNK) pathways. Members of the TRAF family exist as trimers and share a conserved TRAF domain that mediates binding to the cytoplasmic domains of TNFRs. The TRAF domain from TRAF3 has been crystallized. In addition, an N-terminally truncated form of the domain has been crystallized in space group P321 with a shortened c axis and markedly improved diffraction (2.5 A resolution).


Subject(s)
Proteins/chemistry , Crystallization , Crystallography, X-Ray , Humans , Molecular Structure , Protein Structure, Tertiary , Recombinant Proteins/chemistry , TNF Receptor-Associated Factor 3
7.
Structure ; 10(3): 403-11, 2002 Mar.
Article in English | MEDLINE | ID: mdl-12005438

ABSTRACT

TRAFs (tumor necrosis factor receptor [TNFR]-associated factors) bind to the cytoplasmic portion of liganded TNFRs and stimulate activation of NF-kappaB or JNK pathways. A modulator of TRAF signaling, TANK, serves as either an enhancer or an inhibitor of TRAF-mediated signaling pathways. The crystal structure of a region of TANK bound to TRAF3 has been determined and compared to a similar CD40/TRAF3 complex. TANK and CD40 bind to the same crevice on TRAF3. The recognition motif PxQxT is presented in a boomerang-like structure in TANK that is markedly different from the hairpin loop that forms in CD40 upon binding to TRAF3. Critical TANK contact residues were confirmed by mutagenesis to be required for binding to TRAF3 or TRAF2. Binding affinity, measured by isothermal titration calorimetry and competition assays, demonstrated that TANK competes with CD40 for the TRAF binding site.


Subject(s)
Adaptor Proteins, Signal Transducing , CD40 Ligand/metabolism , Protein Structure, Quaternary , Proteins/chemistry , Proteins/metabolism , Binding Sites , CD40 Ligand/chemistry , Calorimetry , Cell Line , Crystallography, X-Ray , Humans , Models, Molecular , Point Mutation , Protein Binding , Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , TNF Receptor-Associated Factor 3
SELECTION OF CITATIONS
SEARCH DETAIL
...