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











Publication year range
1.
Molecules ; 23(10)2018 Oct 22.
Article in English | MEDLINE | ID: mdl-30360356

ABSTRACT

µ-Conotoxins are potent and highly specific peptide blockers of voltage-gated sodium channels. In this study, the solution structure of µ-conotoxin GIIIC was determined using 2D NMR spectroscopy and simulated annealing calculations. Despite high sequence similarity, GIIIC adopts a three-dimensional structure that differs from the previously observed conformation of µ-conotoxins GIIIA and GIIIB due to the presence of a bulky, non-polar leucine residue at position 18. The side chain of L18 is oriented towards the core of the molecule and consequently the N-terminus is re-modeled and located closer to L18. The functional characterization of GIIIC defines it as a canonical µ-conotoxin that displays substantial selectivity towards skeletal muscle sodium channels (NaV), albeit with ~2.5-fold lower potency than GIIIA. GIIIC exhibited a lower potency of inhibition of NaV1.4 channels, but the same NaV selectivity profile when compared to GIIIA. These observations suggest that single amino acid differences that significantly affect the structure of the peptide do in fact alter its functional properties. Our work highlights the importance of structural factors, beyond the disulfide pattern and electrostatic interactions, in the understanding of the functional properties of bioactive peptides. The latter thus needs to be considered when designing analogues for further applications.


Subject(s)
Conotoxins/chemistry , Magnetic Resonance Spectroscopy , Amino Acid Sequence , Conotoxins/chemical synthesis , Conotoxins/pharmacology , Disulfides/chemistry , Leucine/chemistry , Models, Molecular , Peptides/chemical synthesis , Peptides/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , Sodium Channel Blockers/chemical synthesis , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Sodium Channels/chemistry , Sodium Channels/metabolism , Structure-Activity Relationship
2.
J Biol Chem ; 290(49): 29217-30, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26468282

ABSTRACT

Inflammasomes mediate inflammatory and cell death responses to pathogens and cellular stress signals via activation of procaspases-1 and -8. During inflammasome assembly, activated receptors of the NLR or PYHIN family recruit the adaptor protein ASC and initiate polymerization of its pyrin domain (PYD) into filaments. We show that ASC filaments in turn nucleate procaspase-8 death effector domain (DED) filaments in vitro and in vivo. Interaction between ASC PYD and procaspase-8 tandem DEDs optimally required both DEDs and represents an unusual heterotypic interaction between domains of the death fold superfamily. Analysis of ASC PYD mutants showed that interaction surfaces that mediate procaspase-8 interaction overlap with those required for ASC self-association and interaction with the PYDs of inflammasome initiators. Our data indicate that multiple types of death fold domain filaments form at inflammasomes and that PYD/DED and homotypic PYD interaction modes are similar. Interestingly, we observed condensation of procaspase-8 filaments containing the catalytic domain, suggesting that procaspase-8 interactions within and/or between filaments may be involved in caspase-8 activation. Procaspase-8 filaments may also be relevant to apoptosis induced by death receptors.


Subject(s)
Caspase 8/metabolism , Cytoskeletal Proteins/metabolism , Inflammasomes/metabolism , Apoptosis , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Catalytic Domain , Cell Death , HEK293 Cells , Humans , Inflammation , Microscopy, Fluorescence , Mutation , Protein Binding , Signal Transduction
3.
J Biol Chem ; 289(34): 23504-19, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-25006247

ABSTRACT

Inflammasomes are macromolecular complexes that mediate inflammatory and cell death responses to pathogens and cellular stress signals. Dysregulated inflammasome activation is associated with autoinflammatory syndromes and several common diseases. During inflammasome assembly, oligomerized cytosolic pattern recognition receptors recruit procaspase-1 and procaspase-8 via the adaptor protein ASC. Inflammasome assembly is mediated by pyrin domains (PYDs) and caspase recruitment domains, which are protein interaction domains of the death fold superfamily. However, the molecular details of their interactions are poorly understood. We have studied the interaction between ASC and pyrin PYDs that mediates ASC recruitment to the pyrin inflammasome, which is implicated in the pathogenesis of familial Mediterranean fever. We demonstrate that both the ASC and pyrin PYDs have multifaceted binding modes, involving three sites on pyrin PYD and two sites on ASC PYD. Molecular docking of pyrin-ASC PYD complexes showed that pyrin PYD can simultaneously interact with up to three ASC PYDs. Furthermore, ASC PYD can self-associate and interact with pyrin, consistent with previous reports that pyrin promotes ASC clustering to form a proinflammatory complex. Finally, the effects of familial Mediterranean fever-associated mutations, R42W and A89T, on structural and functional properties of pyrin PYD were investigated. The R42W mutation had a significant effect on structure and increased stability. Although the R42W mutant exhibited reduced interaction with ASC, it also bound less to the pyrin B-box domain responsible for autoinhibition and hence may be constitutively active. Our data give new insights into the binding modes of PYDs and inflammasome architecture.


Subject(s)
Cytoskeletal Proteins/metabolism , Inflammasomes/metabolism , Amino Acid Sequence , Binding Sites , CARD Signaling Adaptor Proteins , Cytoskeletal Proteins/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Pyrin , Sequence Homology, Amino Acid
4.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 5): 774-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23633586

ABSTRACT

The caspase recruitment domain (CARD) is present in death-domain superfamily proteins involved in inflammation and apoptosis. BinCARD is named for its ability to interact with Bcl10 and inhibit downstream signalling. Human BinCARD is expressed as two isoforms that encode the same N-terminal CARD region but which differ considerably in their C-termini. Both isoforms are expressed in immune cells, although BinCARD-2 is much more highly expressed. Crystals of the CARD fold common to both had low symmetry (space group P1). Molecular replacement was unsuccessful in this low-symmetry space group and, as the construct contains no methionines, first one and then two residues were engineered to methionine for MAD phasing. The double-methionine variant was produced as a selenomethionine derivative, which was crystallized and the structure was solved using data measured at two wavelengths. The crystal structures of the native and selenomethionine double mutant were refined to high resolution (1.58 and 1.40 Šresolution, respectively), revealing the presence of a cis-peptide bond between Tyr39 and Pro40. Unexpectedly, the native crystal structure revealed that all three cysteines were oxidized. The mitochondrial localization of BinCARD-2 and the susceptibility of its CARD region to redox modification points to the intriguing possibility of a redox-regulatory role.


Subject(s)
CARD Signaling Adaptor Proteins/chemistry , Proteins/chemistry , Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Crystallography, X-Ray , Cysteine/metabolism , HeLa Cells , Humans , Mitochondria/metabolism , Models, Molecular , Mutation , Oxidation-Reduction , Proline/chemistry , Protein Conformation , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proteins/genetics , Selenomethionine
5.
Biomol NMR Assign ; 7(2): 117-20, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22618863

ABSTRACT

Proteins containing a domain of unknown function 59 (DUF59) appear to have a variety of physiological functions, ranging from iron-sulfur cluster assembly to DNA repair. DUF59 proteins have been found in bacteria, archaea and eukaryotes, however Fam96a and Fam96b are the only mammalian proteins predicted to contain a DUF59 domain. Fam96a is an 18 kDa protein comprised primarily of a DUF59 domain (residues 31-157) and an N-terminal signal peptide (residues 1-27). Interestingly, the DUF59 domain of Fam96a exists as monomeric and dimeric forms in solution, and X-ray crystallography studies of both forms unexpectedly revealed two different domain-swapped dimer structures. Here we report the backbone resonance assignments and secondary structure of the monomeric form of the 127 residue DUF59 domain of human Fam96a. This study provides the basis for further understanding the structural variability exhibited by Fam96a and the mechanism for domain swapping.


Subject(s)
Carrier Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Amino Acid Sequence , Humans , Metalloproteins , Molecular Sequence Data , Protein Multimerization , Protein Structure, Tertiary
6.
J Biol Chem ; 288(5): 3198-207, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23255593

ABSTRACT

The B3 DNA-binding domain is a plant-specific domain found throughout the plant kingdom from the alga Chlamydomonas to grasses and flowering plants. Over 100 B3 domain-containing proteins are found in the model plant Arabidopsis thaliana, and one of these is critical for accelerating flowering in response to prolonged cold treatment, an epigenetic process called vernalization. Despite the specific phenotype of genetic vrn1 mutants, the VERNALIZATION1 (VRN1) protein localizes throughout the nucleus and shows sequence-nonspecific binding in vitro. In this work, we used a dominant repressor tag that overcomes genetic redundancy to show that VRN1 is involved in processes beyond vernalization that are essential for Arabidopsis development. To understand its sequence-nonspecific binding, we crystallized VRN1(208-341) and solved its crystal structure to 1.6 Å resolution using selenium/single-wavelength anomalous diffraction methods. The crystallized construct comprises the second VRN1 B3 domain and a preceding region conserved among VRN1 orthologs but absent in other B3 domains. We established the DNA-binding face using NMR and then mutated positively charged residues on this surface with a series of 16 Ala and Glu substitutions, ensuring that the protein fold was not disturbed using heteronuclear single quantum correlation NMR spectra. The triple mutant R249E/R289E/R296E was almost completely incapable of DNA binding in vitro. Thus, we have revealed that although VRN1 is sequence-nonspecific in DNA binding, it has a defined DNA-binding surface.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/growth & development , Arabidopsis/metabolism , DNA, Plant/metabolism , Mutation/genetics , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Amino Acid Sequence , Arabidopsis Proteins/genetics , Binding Sites , Conserved Sequence , Crystallography, X-Ray , DNA Restriction Enzymes/chemistry , Kinetics , Models, Molecular , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phenotype , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Sequence Alignment
7.
J Biol Chem ; 287(52): 43810-24, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23105112

ABSTRACT

Cleavage of transmembrane receptors by γ-secretase is the final step in the process of regulated intramembrane proteolysis (RIP) and has a significant impact on receptor function. Although relatively little is known about the molecular mechanism of γ-secretase enzymatic activity, it is becoming clear that substrate dimerization and/or the α-helical structure of the substrate can regulate the site and rate of γ-secretase activity. Here we show that the transmembrane domain of the pan-neurotrophin receptor p75(NTR), best known for regulating neuronal death, is sufficient for its homodimerization. Although the p75(NTR) ligands NGF and pro-NGF do not induce homerdimerization or RIP, homodimers of p75(NTR) are γ-secretase substrates. However, dimerization is not a requirement for p75(NTR) cleavage, suggesting that γ-secretase has the ability to recognize and cleave each receptor molecule independently. The transmembrane cysteine 257, which mediates covalent p75(NTR) interactions, is not crucial for homodimerization, but this residue is required for normal rates of γ-secretase cleavage. Similarly, mutation of the residues alanine 262 and glycine 266 of an AXXXG dimerization motif flanking the γ-secretase cleavage site within the p75(NTR) transmembrane domain alters the orientation of the domain and inhibits γ-secretase cleavage of p75(NTR). Nonetheless, heteromer interactions of p75(NTR) with TrkA increase full-length p75(NTR) homodimerization, which in turn potentiates the rate of γ-cleavage following TrkA activation independently of rates of α-cleavage. These results provide support for the idea that the helical structure of the p75(NTR) transmembrane domain, which may be affected by co-receptor interactions, is a key element in γ-secretase-catalyzed cleavage.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Nerve Tissue Proteins/metabolism , Protein Multimerization/physiology , Proteolysis , Receptors, Growth Factor/metabolism , Receptors, Nerve Growth Factor/metabolism , Amino Acid Motifs , Amyloid Precursor Protein Secretases/genetics , Animals , Cell Death/physiology , Cysteine , Enzyme Activation , HEK293 Cells , Humans , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/genetics , PC12 Cells , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Structure, Tertiary , Rats , Receptor, trkA/genetics , Receptor, trkA/metabolism , Receptors, Growth Factor/genetics , Receptors, Nerve Growth Factor/genetics
8.
J Biol Chem ; 287(50): 41732-43, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23066025

ABSTRACT

A key process underlying an innate immune response to pathogens or cellular stress is activation of members of the NOD-like receptor family, such as NLRP3, to assemble caspase-1-activating inflammasome complexes. Activated caspase-1 processes proinflammatory cytokines into active forms that mediate inflammation. Activation of the NLRP3 inflammasome is also associated with common diseases including cardiovascular disease, diabetes, chronic kidney disease, and Alzheimer disease. However, the molecular details of NLRP3 inflammasome assembly are not established. The adaptor protein ASC plays a key role in inflammasome assembly. It is composed of an N-terminal pyrin domain (PYD) and a C-terminal caspase recruitment domain, which are protein interaction domains of the death fold superfamily. ASC interacts with NLRP3 via a homotypic PYD interaction and recruits procaspase-1 via a homotypic caspase recruitment domain interaction. Here we demonstrate that ASC PYD contains two distinct binding sites important for self-association and interaction with NLRP3 and the modulatory protein POP1. Modeling of the homodimeric ASC PYD complex formed via an asymmetric interaction using both sites resembles a type I interaction found in other death fold domain complexes. This interaction mode also permits assembly of ASC PYDs into filaments. Furthermore, a type I binding mode is likely conserved in interactions with NLRP3 and POP1, because residues critical for interaction of ASC PYD are conserved in these PYDs. We also demonstrate that ASC PYD can simultaneously self-associate and interact with NLRP3, rationalizing the model whereby ASC self-association upon recruitment to NLRP3 promotes clustering and activation of procaspase-1.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Models, Biological , Protein Multimerization/physiology , Binding Sites/physiology , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Binding , Protein Structure, Tertiary/physiology
9.
PLoS Pathog ; 8(10): e1002957, 2012.
Article in English | MEDLINE | ID: mdl-23071437

ABSTRACT

We have investigated the potential of the GTP synthesis pathways as chemotherapeutic targets in the human pathogen Cryptococcus neoformans, a common cause of fatal fungal meningoencephalitis. We find that de novo GTP biosynthesis, but not the alternate salvage pathway, is critical to cryptococcal dissemination and survival in vivo. Loss of inosine monophosphate dehydrogenase (IMPDH) in the de novo pathway results in slow growth and virulence factor defects, while loss of the cognate phosphoribosyltransferase in the salvage pathway yielded no phenotypes. Further, the Cryptococcus species complex displays variable sensitivity to the IMPDH inhibitor mycophenolic acid, and we uncover a rare drug-resistant subtype of C. gattii that suggests an adaptive response to microbial IMPDH inhibitors in its environmental niche. We report the structural and functional characterization of IMPDH from Cryptococcus, revealing insights into the basis for drug resistance and suggesting strategies for the development of fungal-specific inhibitors. The crystal structure reveals the position of the IMPDH moveable flap and catalytic arginine in the open conformation for the first time, plus unique, exploitable differences in the highly conserved active site. Treatment with mycophenolic acid led to significantly increased survival times in a nematode model, validating de novo GTP biosynthesis as an antifungal target in Cryptococcus.


Subject(s)
Cryptococcus neoformans/enzymology , Cryptococcus neoformans/pathogenicity , Guanosine Triphosphate/biosynthesis , IMP Dehydrogenase/chemistry , IMP Dehydrogenase/metabolism , Mycophenolic Acid/pharmacology , Animals , Antifungal Agents/pharmacology , Caenorhabditis elegans/microbiology , Cryptococcus gattii/drug effects , Cryptococcus gattii/genetics , Cryptococcus gattii/isolation & purification , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/metabolism , Crystallography, X-Ray , Drug Resistance, Fungal/genetics , Enzyme Inhibitors/pharmacology , IMP Dehydrogenase/antagonists & inhibitors , IMP Dehydrogenase/genetics , Meningoencephalitis/microbiology
10.
Biomol NMR Assign ; 6(1): 5-8, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21553305

ABSTRACT

VERNALIZATION1 (VRN1) is a multidomain DNA binding protein from Arabidopsis thaliana that is required for the acceleration of flowering time in response to prolonged cold treatment; a physiological process called vernalization. VRN1 is a 39 kDa protein comprised of two B3 domains flanking a putative nuclear localization sequence and two PEST domains. Here we report the (1)H, (13)C and (15)N resonance assignments of the 134 residue C-terminal region of VRN1, comprising a B3 DNA binding domain of the REM family and an upstream region that is highly conserved among VRN1 homologs from other dicotyledonous plant species.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis , DNA/metabolism , Nuclear Magnetic Resonance, Biomolecular , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Conserved Sequence , Protein Structure, Tertiary
11.
Proc Natl Acad Sci U S A ; 108(26): 10478-83, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21670253

ABSTRACT

The three-disulfide inhibitor cystine knot (ICK) motif is a fold common to venom peptides from spiders, scorpions, and aquatic cone snails. Over a decade ago it was proposed that the ICK motif is an elaboration of an ancestral two-disulfide fold coined the disulfide-directed ß-hairpin (DDH). Here we report the isolation, characterization, and structure of a novel toxin [U(1)-liotoxin-Lw1a (U(1)-LITX-Lw1a)] from the venom of the scorpion Liocheles waigiensis that is the first example of a native peptide that adopts the DDH fold. U(1)-LITX-Lw1a not only represents the discovery of a missing link in venom protein evolution, it is the first member of a fourth structural fold to be adopted by scorpion-venom peptides. Additionally, we show that U(1)-LITX-Lw1a has potent insecticidal activity across a broad range of insect pest species, thereby providing a unique structural scaffold for bioinsecticide development.


Subject(s)
Biological Evolution , Cystine/chemistry , Neurotoxins/chemistry , Scorpion Venoms/chemistry , Amino Acid Sequence , Animals , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Scorpions , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Article in English | MEDLINE | ID: mdl-19255487

ABSTRACT

VERNALIZATION1 (VRN1) is required in the model plant Arabidopsis thaliana for the epigenetic suppression of the floral repressor FLC by prolonged cold treatment. Stable suppression of FLC accelerates flowering, a physiological process known as vernalization. VRN1 is a 341-residue DNA-binding protein that contains two plant-specific B3 domains (B3a and B3b), a putative nuclear localization sequence (NLS) and two putative PEST domains. VRN1(208-341) includes the second B3 domain and a region upstream that is highly conserved in the VRN1 orthologues of other dicotyledonous plants. VRN1(208-341) was crystallized by the hanging-drop method in 0.05 M sodium acetate pH 6.0 containing 1.0 M NaCl and 18%(w/v) PEG 3350. Preliminary X-ray diffraction data analysis revealed that the VRN1(208-341) crystal diffracted to 2.1 A and belonged to space group C2, with unit-cell parameters a = 105.2, b = 47.9, c = 61.2 A, alpha = 90.0, beta = 115.4, gamma = 90.0 degrees . Assuming that two molecules occupy the asymmetric unit, a Matthews coefficient of 2.05 A(3) Da(-1) and a solvent content of 40.1% were calculated.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/isolation & purification , Arabidopsis/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , Repressor Proteins/chemistry , Repressor Proteins/isolation & purification , Arabidopsis Proteins/metabolism , Crystallography, X-Ray , DNA-Binding Proteins/metabolism , Repressor Proteins/metabolism
13.
Methods Mol Biol ; 426: 437-46, 2008.
Article in English | MEDLINE | ID: mdl-18542882

ABSTRACT

In the age of structural proteomics when protein structures are targeted on a genome-wide scale, the identification of proteins that are amenable to analysis using x-ray crystallography or NMR spectroscopy is the key to high throughput structure determination. NMR screening is a beneficial part of a structural proteomics pipeline because of its ability to provide detailed biophysical information about the protein targets under investigation at an early stage of the structure determination process. This chapter describes efficient methods for the production of uniformly (15)N-labeled proteins for NMR screening using both conventional IPTG induction and autoinduction approaches in E. coli. Details of sample preparation for NMR and the acquisition of 1D (1)H NMR and 2D (1)H-(15)N HSQC spectra to assess the structural characteristics and suitability of proteins for further structural studies are also provided.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Proteins/chemistry , Proteomics , Crystallization , Crystallography, X-Ray/methods , Escherichia coli/genetics , Escherichia coli/metabolism , Nitrogen Isotopes
14.
Methods Mol Biol ; 426: 577-87, 2008.
Article in English | MEDLINE | ID: mdl-18542891

ABSTRACT

This chapter describes the methodology adopted in a project aimed at structural and functional characterization of proteins that potentially play an important role in mammalian macrophages. The methodology that underpins this project is applicable to both small research groups and larger structural genomics consortia. Gene products with putative roles in macrophage function are identified using gene expression information obtained via DNA microarray technology. Specific targets for structural and functional characterization are then selected based on a set of criteria aimed at maximizing insight into function. The target proteins are cloned using a modification of Gateway cloning technology, expressed with hexa-histidine tags in E. coli, and purified to homogeneity using a combination of affinity and size exclusion chromatography. Purified proteins are finally subjected to crystallization trials and/or NMR-based screening to identify candidates for structure determination. Where crystallography and NMR approaches are unsuccessful, chemical cross-linking is employed to obtain structural information. This resulting structural information is used to guide cell biology experiments to further investigate the cellular and molecular function of the targets in macrophage biology. Jointly, the data sheds light on the molecular and cellular functions of macrophage proteins.


Subject(s)
Macrophages/metabolism , Proteins/chemistry , Proteomics/methods , Proteomics/organization & administration , Animals , Arthritis/genetics , Arthritis/metabolism , Computational Biology , Crystallography, X-Ray , Humans , Mice , Protein Conformation , Protein Folding , Proteins/genetics , Proteins/isolation & purification , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Queensland , Universities
15.
J Biol Chem ; 281(42): 31863-75, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16905547

ABSTRACT

Pyrin domain (PYD)-containing proteins are key components of pathways that regulate inflammation, apoptosis, and cytokine processing. Their importance is further evidenced by the consequences of mutations in these proteins that give rise to autoimmune and hyperinflammatory syndromes. PYDs, like other members of the death domain (DD) superfamily, are postulated to mediate homotypic interactions that assemble and regulate the activity of signaling complexes. However, PYDs are presently the least well characterized of all four DD subfamilies. Here we report the three-dimensional structure and dynamic properties of ASC2, a PYD-only protein that functions as a modulator of multidomain PYD-containing proteins involved in NF-kappaB and caspase-1 activation. ASC2 adopts a six-helix bundle structure with a prominent loop, comprising 13 amino acid residues, between helices two and three. This loop represents a divergent feature of PYDs from other domains with the DD fold. Detailed analysis of backbone 15N NMR relaxation data using both the Lipari-Szabo model-free and reduced spectral density function formalisms revealed no evidence of contiguous stretches of polypeptide chain with dramatically increased internal motion, except at the extreme N and C termini. Some mobility in the fast, picosecond to nanosecond timescale, was seen in helix 3 and the preceding alpha2-alpha3 loop, in stark contrast to the complete disorder seen in the corresponding region of the NALP1 PYD. Our results suggest that extensive conformational flexibility in helix 3 and the alpha2-alpha3 loop is not a general feature of pyrin domains. Further, a transition from complete disorder to order of the alpha2-alpha3 loop upon binding, as suggested for NALP1, is unlikely to be a common attribute of pyrin domain interactions.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Cytoskeletal Proteins/chemistry , Inflammation/metabolism , Ribonucleoproteins/chemistry , Amino Acid Sequence , Apoptosis , Apoptosis Regulatory Proteins/physiology , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Models, Statistical , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Pyrin , Ribonucleoproteins/physiology , Signal Transduction
16.
Mol Cell ; 22(5): 599-610, 2006 Jun 09.
Article in English | MEDLINE | ID: mdl-16762833

ABSTRACT

The structure of FADD has been solved in solution, revealing that the death effector domain (DED) and death domain (DD) are aligned with one another in an orthogonal, tail-to-tail fashion. Mutagenesis of FADD and functional reconstitution with its binding partners define the interaction with the intracellular domain of CD95 and the prodomain of procaspase-8 and reveal a self-association surface necessary to form a productive complex with an activated "death receptor." The identification of a procaspase-specific binding surface on the FADD DED suggests a preferential interaction with one, but not both, of the DEDs of procaspase-8 in a perpendicular arrangement. FADD self-association is mediated by a "hydrophobic patch" in the vicinity of F25 in the DED. The structure of FADD and its functional characterization, therefore, illustrate the architecture of key components in the death-inducing signaling complex.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Caspases/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Caspase 8 , Caspases/metabolism , Fas-Associated Death Domain Protein , Humans , Jurkat Cells , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Signal Transduction , Transfection , fas Receptor/metabolism
17.
J Biol Chem ; 279(2): 1474-81, 2004 Jan 09.
Article in English | MEDLINE | ID: mdl-14573612

ABSTRACT

The initiation of programmed cell death at CD95 (Fas, Apo-1) is achieved by forming a death-inducing signaling complex (DISC) at the cytoplasmic membrane surface. Assembly of the DISC has been proposed to occur via homotypic interactions between the death domain (DD) of FADD and the cytoplasmic domain of CD95. Previous analysis of the FADD/CD95 interaction led to the identification of a putative CD95 binding surface within FADD DD formed by alpha helices 2 and 3. More detailed analysis of the CD95/FADD DD interaction now demonstrates that a bimodal surface exists in the FADD DD for interaction with CD95. An expansive surface on one side of the domain is composed of elements in alpha helices 1, 2, 3, 5, and 6. This major surface is common to many proteins harboring this motif, whether or not they are associated with programmed cell death. A secondary surface resides on the opposite face of the domain and involves residues in helices 3 and 4. The major surface is topologically similar to the protein interaction surface identified in Drosophila Tube DD and the death effector domain of hamster PEA-15, two physiologically unrelated proteins which interact with structurally unrelated binding partners. These results demonstrate the presence of a structurally conserved surface within the DD which can mediate protein recognition with homo- and heterotypic binding partners, whereas a second surface may be responsible for stabilizing the higher order complex in the DISC.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/chemistry , fas Receptor/physiology , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Survival , Cytoplasm/metabolism , DNA, Complementary/metabolism , Drosophila/metabolism , Fas-Associated Death Domain Protein , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Precipitin Tests , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Time Factors , fas Receptor/chemistry , fas Receptor/metabolism
18.
J Biol Chem ; 278(52): 52587-97, 2003 Dec 26.
Article in English | MEDLINE | ID: mdl-14506247

ABSTRACT

Activation of Raf-1 suppresses integrin activation, potentially through the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2). However, bulk ERK1/2 activation does not correlate with suppression. PEA-15 reverses suppression of integrin activation and binds ERK1/2. Here we report that PEA-15 reversal of integrin suppression depends on its capacity to bind ERK1/2, indicating that ERK1/2 function is indeed required for suppression. Mutations in either the death effector domain or C-terminal tail of PEA-15 that block ERK1/2 binding abrogated the reversal of integrin suppression. Furthermore, we used ERK/p38 chimeras and site-directed mutagenesis to identify ERK1/2 residues required for binding PEA-15. Mutations of residues that precede the alphaG helix and within the mitogen-activated protein kinase insert blocked ERK2 binding to PEA-15, but not activation of ERK2. These ERK2 mutants blocked the ability of PEA-15 to reverse suppression of integrin activation. Thus, PEA-15 regulation of integrin activation depends on its binding to ERK1/2. To directly test the role of ERK1/2 localization in suppression, we enforced membrane association of ERK1 and 2 by joining a membrane-targeting CAAX box sequence to them. Both ERK1-CAAX and ERK2-CAAX were membrane-localized and suppressed integrin activation. In contrast to suppression by membrane-targeted Raf-CAAX, suppression by ERK1/2-CAAX was not reversed by PEA-15. Thus, ERK1/2 are the Raf effectors for suppression of integrin activation, and PEA-15 reverses suppression by binding ERK1/2.


Subject(s)
Integrins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Amino Acid Motifs , Animals , CHO Cells , Cell Membrane/metabolism , Cell Separation , Cricetinae , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Green Fluorescent Proteins , Immunoblotting , Ligands , Luminescent Proteins/metabolism , Mitogen-Activated Protein Kinase 3 , Models, Genetic , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Phosphoproteins/chemistry , Point Mutation , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases
19.
EMBO J ; 21(23): 6494-504, 2002 Dec 02.
Article in English | MEDLINE | ID: mdl-12456656

ABSTRACT

PEA-15 is a multifunctional protein that modulates signaling pathways which control cell proliferation and cell death. In particular, PEA-15 regulates the actions of the ERK MAP kinase cascade by binding to ERK and altering its subcellular localization. The three-dimensional structure of PEA-15 has been determined using NMR spectroscopy and its interaction with ERK defined by characterization of mutants that modulate ERK function. PEA-15 is composed of an N-terminal death effector domain (DED) and a C-terminal tail of irregular structure. NMR 'footprinting' and mutagenesis identified elements of both the DED and tail that are required for ERK binding. Comparison of the DED-binding surface for ERK2 with the death domain (DD)-binding surface of Drosophila Tube revealed an unexpected similarity between the interaction modes of the DD and DED motifs in these proteins. Despite a lack of functional or sequence similarity between PEA-15 and Tube, these proteins utilize a common surface of the structurally similar DD and DED to recognize functionally diverse targets.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins , Binding Sites , Epitopes , Magnetic Resonance Spectroscopy , Mice , Mitogen-Activated Protein Kinases/chemistry , Molecular Sequence Data , Phosphoproteins/chemistry , Protein Binding , Protein Structure, Tertiary , Sequence Alignment
20.
J Biol Chem ; 277(30): 27247-55, 2002 Jul 26.
Article in English | MEDLINE | ID: mdl-12006587

ABSTRACT

Mu-conotoxins are peptide inhibitors of voltage-sensitive sodium channels (VSSCs). Synthetic forms of mu-conotoxins PIIIA and PIIIA-(2-22) were found to inhibit tetrodotoxin (TTX)-sensitive VSSC current but had little effect on TTX-resistant VSSC current in sensory ganglion neurons. In rat brain neurons, these peptides preferentially inhibited the persistent over the transient VSSC current. Radioligand binding assays revealed that PIIIA, PIIIA-(2-22), and mu-conotoxins GIIIB discriminated among TTX-sensitive VSSCs in rat brain, that these and GIIIC discriminated among the corresponding VSSCs in human brain, and GIIIA had low affinity for neuronal VSSCs. (1)H NMR studies found that PIIIA adopts two conformations in solution due to cis/trans isomerization at hydroxyproline 8. The major trans conformation results in a three-dimensional structure that is significantly different from the previously identified conformation of mu-conotoxins GIIIA and GIIIB that selectively target TTX-sensitive muscle VSSCs. Comparison of the structures and activity of PIIIA to muscle-selective mu-conotoxins provides an insight into the structural requirements for inhibition of different TTX-sensitive sodium channels by mu-conotoxins.


Subject(s)
Conotoxins/chemistry , Sodium Channels/chemistry , Tetrodotoxin/pharmacology , Amino Acid Sequence , Animals , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Hippocampus/metabolism , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Neurons/metabolism , Peptide Biosynthesis , Protein Conformation , Radioligand Assay , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL