Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters











Publication year range
1.
Bioorg Med Chem Lett ; 30(6): 126967, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32005415

ABSTRACT

Inhibition of the nuclear receptor Retinoic Acid Receptor-Related Orphan Receptor γt (RORγt) is a promising strategy for the treatment of autoimmune diseases. In this paper, we describe a series of allosteric, cysteine-dependent, inverse agonists of RORγt. Site-directed mutagenesis and molecular dynamics simulations are supportive of a mechanism of action through specific binding to Cys476 on alpha helix 11 of the ligand binding domain (LBD). Representative compounds in the series selectively inhibit RORγt, potently suppress interleukin-17A (IL-17A) production by human CD4+ T cells, and inhibit T helper 17 (Th17) differentiation from human naïve CD4+ T cells. The advanced compound 13 is orally bioavailable and active at a dose of 3 mg/kg in a murine collagen-induced model of rheumatoid arthritis. Collectively, these data are supportive of the development of compound 13 in autoimmune diseases.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Cysteine/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Pyrimidines/chemistry , Allosteric Site , Animals , Cell Differentiation/drug effects , Collagen/metabolism , Disease Models, Animal , Humans , Hydroxycholesterols/metabolism , Interleukin-17/metabolism , Ligands , Mice , Molecular Dynamics Simulation , Mutagenesis/drug effects , Protein Binding , Protein Conformation , Pyrimidines/metabolism , Pyrimidines/pharmacology , Structure-Activity Relationship , Th17 Cells
2.
Biochim Biophys Acta ; 1860(11 Pt A): 2537-2552, 2016 11.
Article in English | MEDLINE | ID: mdl-27474998

ABSTRACT

BACKGROUND: Semi-synthetic oleanane triterpenoid antioxidant inflammation modulators (tpAIMs) are small molecules that interact with KEAP1 cysteine residue 151 (C151) and activate NRF2. Exploration of the structure-activity relationship between the tpAIMs and KEAP1 is limited by the predominantly hydrocarbon nature of the oleanane triterpenoid pentacyclic ring structure. Therefore, we used novel, chemically-tractable, synthetic antioxidant inflammation modulators (sAIMs) to probe the stereoselectivity of the ligand-protein interaction. METHODS: We measured several parameters of NRF2 activation to assess the potency of sAIM enantiomers with natural (tpAIM-like) 4(S),5(S),10(R) or unnatural 4(R),5(R),10(S) configurations. Additionally, we determined the crystal structure of the KEAP1 BTB domain in complex with two different sAIMs. RESULTS: We found that the potencies of sAIM enantiomers in the natural configuration were similar to those of the tpAIM, RTA 405. Strikingly, sAIM enantiomers in the unnatural configuration were 10- to 40-fold less potent than their natural counterparts. Crystallographic studies of sAIMs in complex with the KEAP1 BTB domain demonstrated that these ligands form a covalent bond with C151 and revealed the presence of additional hydrogen bonds, Van der Waals interactions, and pi-stacking interactions. CONCLUSIONS: Although KEAP1 C151 is required for NRF2 activation by tpAIMs and sAIMs, interactions with other KEAP1 residues are critical for the stereospecific recognition and potency of these ligands. GENERAL SIGNIFICANCE: This work demonstrates that reversible cyanoenone Michael acceptors, such as the tpAIMs and sAIMs, can be specifically tuned to regulate redox sensitive cysteine residues on key signaling molecules, an approach with significant promise for innovative drug development.


Subject(s)
Antioxidants/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/chemistry , Quantitative Structure-Activity Relationship , Small Molecule Libraries/pharmacology , Animals , Antioxidants/chemistry , Binding Sites , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1/chemistry , Mice , Molecular Docking Simulation , NF-E2-Related Factor 2/chemistry , Small Molecule Libraries/chemistry
3.
PLoS One ; 10(4): e0122942, 2015.
Article in English | MEDLINE | ID: mdl-25897966

ABSTRACT

Semi-synthetic triterpenoids are antioxidant inflammation modulator (AIM) compounds that inhibit tumor cell growth and metastasis. Compounds in the AIM class bind to Keap1 and attenuate Nrf2 degradation. In the nucleus, Nrf2 increases antioxidant gene expression and reduces pro-inflammatory gene expression. By increasing Nrf2 activity, AIMs reduce reactive oxygen species and inflammation in the tumor microenvironment, which reverses tumor-mediated immune evasion and inhibits tumor growth and metastasis. AIMs also directly inhibit tumor cell growth by modulating oncogenic signaling pathways, such as IKKß/NF-κB. Here, we characterized the in vitro antioxidant, anti-inflammatory, and anticancer activities of RTA 408, a novel AIM that is currently being evaluated in patients with advanced malignancies. At low concentrations (≤ 25 nM), RTA 408 activated Nrf2 and suppressed nitric oxide and pro-inflammatory cytokine levels in interferon-γ-stimulated RAW 264.7 macrophage cells. At higher concentrations, RTA 408 inhibited tumor cell growth (GI50 = 260 ± 74 nM) and increased caspase activity in tumor cell lines, but not in normal primary human cells. Consistent with the direct effect of AIMs on IKKß, RTA 408 inhibited NF-κB signaling and decreased cyclin D1 levels at the same concentrations that inhibited cell growth and induced apoptosis. RTA 408 also increased CDKN1A (p21) levels and JNK phosphorylation. The in vitro activity profile of RTA 408 is similar to that of bardoxolone methyl, which was well-tolerated by patients at doses that demonstrated target engagement. Taken together, these data support clinical evaluation of RTA 408 for cancer treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Enzyme Activation , Gene Expression/drug effects , Humans , Inflammation Mediators/metabolism , Inhibitory Concentration 50 , MAP Kinase Kinase 4/metabolism , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism
4.
Proc Natl Acad Sci U S A ; 109(10): 3802-7, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22355143

ABSTRACT

Mint adaptor proteins bind to the amyloid precursor protein (APP) and regulate APP processing associated with Alzheimer's disease; however, the molecular mechanisms underlying Mint regulation in APP binding and processing remain unclear. Biochemical, biophysical, and cellular experiments now show that the Mint1 phosphotyrosine binding (PTB) domain that binds to APP is intramolecularly inhibited by the adjacent C-terminal linker region. The crystal structure of a C-terminally extended Mint1 PTB fragment reveals that the linker region forms a short α-helix that folds back onto the PTB domain and sterically hinders APP binding. This intramolecular interaction is disrupted by mutation of Tyr633 within the Mint1 autoinhibitory helix leading to enhanced APP binding and ß-amyloid production. Our findings suggest that an autoinhibitory mechanism in Mint1 is important for regulating APP processing and may provide novel therapies for Alzheimer's disease.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Amyloid beta-Protein Precursor/chemistry , Nerve Tissue Proteins/chemistry , Alzheimer Disease/metabolism , Biochemistry/methods , Biophysics/methods , Crystallography, X-Ray/methods , DNA Mutational Analysis , HEK293 Cells , Humans , Kinetics , Magnetic Resonance Spectroscopy/methods , Molecular Conformation , Neurons/metabolism , Protein Binding , Protein Structure, Tertiary , Tyrosine/chemistry
5.
Cell ; 144(2): 282-95, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241895

ABSTRACT

At a synapse, fast synchronous neurotransmitter release requires localization of Ca(2+) channels to presynaptic active zones. How Ca(2+) channels are recruited to active zones, however, remains unknown. Using unbiased yeast two-hybrid screens, we here identify a direct interaction of the central PDZ domain of the active-zone protein RIM with the C termini of presynaptic N- and P/Q-type Ca(2+) channels but not L-type Ca(2+) channels. To test the physiological significance of this interaction, we generated conditional knockout mice lacking all multidomain RIM isoforms. Deletion of RIM proteins ablated most neurotransmitter release by simultaneously impairing the priming of synaptic vesicles and by decreasing the presynaptic localization of Ca(2+) channels. Strikingly, rescue of the decreased Ca(2+)-channel localization required the RIM PDZ domain, whereas rescue of vesicle priming required the RIM N terminus. We propose that RIMs tether N- and P/Q-type Ca(2+) channels to presynaptic active zones via a direct PDZ-domain-mediated interaction, thereby enabling fast, synchronous triggering of neurotransmitter release at a synapse.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Calcium Channels/metabolism , GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , ATP-Binding Cassette Transporters/chemistry , Animals , Calcium/metabolism , Calcium Channels/chemistry , GTP-Binding Proteins/chemistry , Mice , Mice, Knockout , Nerve Tissue Proteins/chemistry , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Two-Hybrid System Techniques
6.
J Cell Biol ; 184(5): 751-64, 2009 Mar 09.
Article in English | MEDLINE | ID: mdl-19255244

ABSTRACT

Munc18-1 and soluble NSF attachment protein receptors (SNAREs) are critical for synaptic vesicle fusion. Munc18-1 binds to the SNARE syntaxin-1 folded into a closed conformation and to SNARE complexes containing open syntaxin-1. Understanding which steps in fusion depend on the latter interaction and whether Munc18-1 competes with other factors such as complexins for SNARE complex binding is critical to elucidate the mechanisms involved. In this study, we show that lentiviral expression of Munc18-1 rescues abrogation of release in Munc18-1 knockout mice. We describe point mutations in Munc18-1 that preserve tight binding to closed syntaxin-1 but markedly disrupt Munc18-1 binding to SNARE complexes containing open syntaxin-1. Lentiviral rescue experiments reveal that such disruption selectively impairs synaptic vesicle priming but not Ca(2+)-triggered fusion of primed vesicles. We also find that Munc18-1 and complexin-1 bind simultaneously to SNARE complexes. These results suggest that Munc18-1 binding to SNARE complexes mediates synaptic vesicle priming and that the resulting primed state involves a Munc18-1-SNARE-complexin macromolecular assembly that is poised for Ca(2+) triggering of fusion.


Subject(s)
Brain/metabolism , Munc18 Proteins/metabolism , Presynaptic Terminals/metabolism , SNARE Proteins/metabolism , Synaptic Membranes/metabolism , Synaptic Vesicles/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Brain/ultrastructure , Cells, Cultured , Genetic Vectors/genetics , Macromolecular Substances/metabolism , Membrane Fusion/physiology , Mice , Mice, Knockout , Munc18 Proteins/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Point Mutation/genetics , Presynaptic Terminals/ultrastructure , Protein Binding/genetics , Rats , SNARE Proteins/ultrastructure , Synaptic Membranes/ultrastructure , Synaptic Transmission/genetics , Synaptic Vesicles/ultrastructure , Syntaxin 1/genetics , Syntaxin 1/metabolism , Transfection
7.
Science ; 321(5895): 1507-10, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18703708

ABSTRACT

During synaptic vesicle fusion, the soluble N-ethylmaleimide-sensitive factor-attachment protein receptor (SNARE) protein syntaxin-1 exhibits two conformations that both bind to Munc18-1: a "closed" conformation outside the SNARE complex and an "open" conformation in the SNARE complex. Although SNARE complexes containing open syntaxin-1 and Munc18-1 are essential for exocytosis, the function of closed syntaxin-1 is unknown. We generated knockin/knockout mice that expressed only open syntaxin-1B. Syntaxin-1B(Open) mice were viable but succumbed to generalized seizures at 2 to 3 months of age. Binding of Munc18-1 to syntaxin-1 was impaired in syntaxin-1B(Open) synapses, and the size of the readily releasable vesicle pool was decreased; however, the rate of synaptic vesicle fusion was dramatically enhanced. Thus, the closed conformation of syntaxin-1 gates the initiation of the synaptic vesicle fusion reaction, which is then mediated by SNARE-complex/Munc18-1 assemblies.


Subject(s)
Synaptic Vesicles/physiology , Syntaxin 1/chemistry , Syntaxin 1/metabolism , Animals , Calcium/metabolism , Epilepsy/etiology , Excitatory Postsynaptic Potentials , Membrane Fusion , Mice , Mice, Knockout , Munc18 Proteins/metabolism , Mutation , Protein Conformation , Protein Structure, Tertiary , SNARE Proteins/metabolism , Sucrose/metabolism , Synapses/physiology , Synaptic Vesicles/ultrastructure , Syntaxin 1/genetics
8.
J Biomol NMR ; 41(1): 43-54, 2008 May.
Article in English | MEDLINE | ID: mdl-18458823

ABSTRACT

The Sec1/Munc18 (SM) protein Munc18-1 and the SNAREs syntaxin-1, SNAP-25 and synaptobrevin form the core of the membrane fusion machinery that triggers neurotransmitter release. Munc18-1 binds to syntaxin-1 folded into a closed conformation and to the SNARE complex formed by the three SNAREs, which involves an open syntaxin-1 conformation. The former interaction is likely specialized for neurotransmitter release, whereas SM protein/SNARE complex interactions are likely key for all types of intracellular membrane fusion. It is currently unclear whether the closed conformation is highly or only marginally populated in isolated syntaxin-1, and whether Munc18-1 stabilizes the close conformation or helps to open it to facilitate SNARE complex formation. A detailed NMR analysis now suggests that the closed conformation is almost quantitatively populated in isolated syntaxin-1 in the absence of oligomerization, and indicates that its structure is very similar to that observed previously in the crystal structure of the Munc18-1/syntaxin-1 complex. Moreover, we demonstrate that Munc18-1 binding prevents opening of the syntaxin-1 closed conformation. These results support a model whereby the closed conformation constitutes a key intrinsic property of isolated syntaxin-1 and Munc18-1 binding stabilizes this conformation; in this model, Munc18-1 plays in addition an active role in downstream events after another factor(s) helps to open the syntaxin-1 conformation.


Subject(s)
Syntaxin 1/chemistry , Models, Molecular , Munc18 Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation
9.
J Neurosci ; 27(45): 12147-55, 2007 Nov 07.
Article in English | MEDLINE | ID: mdl-17989281

ABSTRACT

The SM (Sec1/Munc18-like) protein Munc18-1 and the soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE) proteins syntaxin-1, SNAP-25, and synaptobrevin/VAMP (vesicle-associated membrane protein) constitute the core fusion machinery for synaptic vesicle exocytosis. Strikingly, Munc18-1 interacts with neuronal SNARE proteins in two distinct modes (i.e., with isolated syntaxin-1 alone in a "closed" conformation and with assembled SNARE complexes containing syntaxin-1 in an "open" conformation). However, it is unclear whether the two modes of Munc18/SNARE interactions are linked. We now show that both Munc18/SNARE interaction modes involve the same low-affinity binding of the extreme syntaxin-1 N terminus to Munc18-1, suggesting that this binding connects the two Munc18/SNARE interaction modes to each other. Using transfected cells as an in vitro assay system, we demonstrate that truncated syntaxins lacking a transmembrane region universally block exocytosis, but only if they contain a free intact N terminus. This block is enhanced by coexpression of either Munc18-1 or SNAP-25, suggesting that truncated syntaxins block exocytosis by forming an untethered inhibitory SNARE complex/Munc18-1 assembly in which the N-terminal syntaxin/Munc18 interaction is essential. Introduction of an N-terminal syntaxin peptide that disrupts this assembly blocks neurotransmitter release in the calyx of Held synapse, whereas a mutant peptide that does not disrupt the SNARE complex/Munc18 assembly has no effect. Viewed together, our data indicate that binding of Munc18 to the syntaxin N terminus unites different modes of Munc18/SNARE interactions and is essential for exocytic membrane fusion.


Subject(s)
Munc18 Proteins/metabolism , Peptide Fragments/metabolism , SNARE Proteins/metabolism , Syntaxin 1/metabolism , Animals , Exocytosis/physiology , HeLa Cells , Humans , Munc18 Proteins/chemistry , PC12 Cells , Peptide Fragments/chemistry , Protein Binding/physiology , Rats , SNARE Proteins/chemistry , Syntaxin 1/chemistry
10.
Biochemistry ; 46(31): 8988-98, 2007 Aug 07.
Article in English | MEDLINE | ID: mdl-17630786

ABSTRACT

RIM proteins play critical roles in synaptic vesicle priming and diverse forms of presynaptic plasticity. The C-terminal C2B domain is the only module that is common to all RIMs but is only distantly related to well-studied C2 domains, and its three-dimensional structure and interactions have not been characterized in detail. Using NMR spectroscopy, we now show that N- and C-terminal extensions beyond the predicted C2B domain core sequence are necessary to form a folded, stable RIM1alpha C2B domain. We also find that the isolated RIM1alpha C2B domain is not sufficient for previously described protein-protein interactions involving the RIM1alpha C-terminus, suggesting that additional sequences adjacent to the C2B domain might be required for these interactions. However, analytical ultracentrifugation shows that the RIM1alpha C2B domain forms weak dimers in solution. The crystal structure of the RIM1alpha C2B domain dimer at 1.7 A resolution reveals that it forms a beta-sandwich characteristic of C2 domains and that the unique N- and C-terminal extensions form a small subdomain that packs against the beta-sandwich and mediates dimerization. Our results provide a structural basis to understand the function of RIM C2B domains and suggest that dimerization may be a crucial aspect of RIM function.


Subject(s)
GTP-Binding Proteins/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Animals , Binding Sites/genetics , Calcium/chemistry , Calcium/metabolism , Crystallography, X-Ray , Dimerization , Fluorescence Resonance Energy Transfer , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Protein Binding , Protein Conformation , Protein Structure, Secondary , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Static Electricity , Structural Homology, Protein , Synaptotagmin I/chemistry , Synaptotagmin I/metabolism
11.
Proc Natl Acad Sci U S A ; 104(8): 2697-702, 2007 Feb 20.
Article in English | MEDLINE | ID: mdl-17301226

ABSTRACT

Both SM proteins (for Sec1/Munc18-like proteins) and SNARE proteins (for soluble NSF-attachment protein receptors) are essential for intracellular membrane fusion, but the general mechanism of coupling between their functions is unclear, in part because diverse SM protein/SNARE binding modes have been described. During synaptic vesicle exocytosis, the SM protein Munc18-1 is known to bind tightly to the SNARE protein syntaxin-1, but only when syntaxin-1 is in a closed conformation that is incompatible with SNARE complex formation. We now show that Munc18-1 also binds tightly to assembled SNARE complexes containing syntaxin-1. The newly discovered Munc18-1/SNARE complex interaction involves contacts of Munc18-1 with the N-terminal H(abc) domain of syntaxin-1 and the four-helical bundle of the assembled SNARE complex. Together with earlier studies, our results suggest that binding of Munc18-1 to closed syntaxin-1 is a specialization that evolved to meet the strict regulatory requirements of neuronal exocytosis, whereas binding of Munc18-1 to assembled SNARE complexes reflects a general function of SM proteins involved in executing membrane fusion.


Subject(s)
Munc18 Proteins/metabolism , Neurons/metabolism , SNARE Proteins/metabolism , Animals , Chromatography, Gel , Cross-Linking Reagents/pharmacology , Humans , Magnetic Resonance Spectroscopy , Models, Biological , Munc18 Proteins/isolation & purification , Protein Binding/drug effects , Rats
12.
PLoS Biol ; 4(7): e192, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16732694

ABSTRACT

C(2) domains are well characterized as Ca(2+)/phospholipid-binding modules, but little is known about how they mediate protein-protein interactions. In neurons, a Munc13-1 C(2)A-domain/RIM zinc-finger domain (ZF) heterodimer couples synaptic vesicle priming to presynaptic plasticity. We now show that the Munc13-1 C(2)A domain homodimerizes, and that homodimerization competes with Munc13-1/RIM heterodimerization. X-ray diffraction studies guided by nuclear magnetic resonance (NMR) experiments reveal the crystal structures of the Munc13-1 C(2)A-domain homodimer and the Munc13-1 C(2)A-domain/RIM ZF heterodimer at 1.44 A and 1.78 A resolution, respectively. The C(2)A domain adopts a beta-sandwich structure with a four-stranded concave side that mediates homodimerization, leading to the formation of an eight-stranded beta-barrel. In contrast, heterodimerization involves the bottom tip of the C(2)A-domain beta-sandwich and a C-terminal alpha-helical extension, which wrap around the RIM ZF domain. Our results describe the structural basis for a Munc13-1 homodimer-Munc13-1/RIM heterodimer switch that may be crucial for vesicle priming and presynaptic plasticity, uncovering at the same time an unexpected versatility of C(2) domains as protein-protein interaction modules, and illustrating the power of combining NMR spectroscopy and X-ray crystallography to study protein complexes.


Subject(s)
Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Amino Acid Sequence , Animals , Crystallography, X-Ray/methods , Dimerization , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Rats , Sequence Alignment , Synaptic Vesicles/metabolism , Zinc Fingers
13.
Nat Struct Mol Biol ; 12(11): 1017-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16228007

ABSTRACT

Munc13 proteins are essential in neurotransmitter release, controlling the priming of synaptic vesicles to a release-ready state. The sequences responsible for this priming activity are unknown. Here we identify a large alpha-helical domain of mammalian Munc13-1 that is autonomously folded and is sufficient to rescue the total arrest in neurotransmitter release observed in hippocampal neurons lacking Munc13s.


Subject(s)
Exocytosis/physiology , Gene Expression , Hippocampus/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism , Animals , Chromatography, Gel , Computational Biology , Excitatory Postsynaptic Potentials , Exocytosis/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurotransmitter Agents/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Folding , Protein Structure, Secondary/genetics , Protein Structure, Tertiary , Rats , Structure-Activity Relationship , Sucrose/pharmacology
14.
EMBO J ; 24(16): 2839-50, 2005 Aug 17.
Article in English | MEDLINE | ID: mdl-16052212

ABSTRACT

alpha-RIMs and Munc13s are active zone proteins that control priming of synaptic vesicles to a readily releasable state, and interact with each other via their N-terminal sequences. The alpha-RIM N-terminal sequence also binds to Rab3s (small synaptic vesicle GTPases), an interaction that regulates presynaptic plasticity. We now demonstrate that alpha-RIMs contain adjacent but separate Munc13- and Rab3-binding sites, allowing formation of a tripartite Rab3/RIM/Munc13 complex. Munc13 binding is mediated by the alpha-RIM zinc-finger domain. Elucidation of the three-dimensional structure of this domain by NMR spectroscopy facilitated the design of a mutation that abolishes alpha-RIM/Munc13 binding. Selective disruption of this interaction in the calyx of Held synapse decreased the size of the readily releasable vesicle pool. Our data suggest that the ternary Rab3/RIM/Munc13 interaction approximates synaptic vesicles to the priming machinery, providing a substrate for presynaptic plasticity. The modular architecture of alpha-RIMs, with nested binding sites for Rab3 and other targets, may be a general feature of Rab effectors that share homology with the alpha-RIM N-terminal sequence.


Subject(s)
GTP-Binding Proteins/metabolism , Models, Molecular , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Vesicles/metabolism , rab3 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Binding Sites/genetics , Calorimetry , Electrophysiology , Escherichia coli , GTP-Binding Proteins/genetics , Glutathione Transferase , Humans , Molecular Sequence Data , Multiprotein Complexes/genetics , Mutagenesis , Nerve Tissue Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Sequence Alignment
15.
J Mol Biol ; 346(2): 589-601, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15670607

ABSTRACT

Sec1/Mun18-like (SM) proteins and soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) play central roles in intracellular membrane fusion. Diverse modes of interaction between SM proteins and SNAREs from the syntaxin family have been described. However, the observation that the N-terminal domains of Sly1 and Vps45, the SM proteins involved in traffic at the endoplasmic reticulum, the Golgi, the trans-Golgi network and the endosomes, bind to similar N-terminal sequences of their cognate syntaxins suggested a unifying theme for SM protein/SNARE interactions in most internal membrane compartments. To further understand this mechanism of SM protein/SNARE coupling, we have elucidated the structure in solution of the isolated N-terminal domain of rat Sly1 (rSly1N) and analyzed its complex with an N-terminal peptide of rat syntaxin 5 by NMR spectroscopy. Comparison with the crystal structure of a complex between Sly1p and Sed5p, their yeast homologues, shows that syntaxin 5 binding requires a striking conformational change involving a two-residue shift in the register of the C-terminal beta-strand of rSly1N. This conformational change is likely to induce a significant alteration in the overall shape of full-length rSly1 and may be critical for its function. Sequence analyses indicate that this conformational change is conserved in the Sly1 family but not in other SM proteins, and that the four families represented by the four SM proteins found in yeast (Sec1p, Sly1p, Vps45p and Vps33p) diverged early in evolution. These results suggest that there are marked distinctions between the mechanisms of action of each of the four families of SM proteins, which may have arisen from different regulatory requirements of traffic in their corresponding membrane compartments.


Subject(s)
Carrier Proteins/chemistry , Immediate-Early Proteins/chemistry , Membrane Proteins/chemistry , Animals , Carrier Proteins/genetics , Conserved Sequence , Evolution, Molecular , Immediate-Early Proteins/genetics , Membrane Proteins/genetics , Munc18 Proteins , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Qa-SNARE Proteins , Rats , Sequence Alignment
16.
Biochemistry ; 43(30): 9583-8, 2004 Aug 03.
Article in English | MEDLINE | ID: mdl-15274612

ABSTRACT

Cleavage of amyloid-beta precursor protein (APP) by site-specific proteases generates amyloid-beta peptides (Abetas), which are thought to induce Alzheimer's disease. We have identified an independently folded extracellular domain of human APP localized proximal to the Abeta sequence, and determined the three-dimensional structure of this domain by NMR spectroscopy. The domain is composed of four alpha-helices, three of which form a tight antiparallel bundle, and constitutes the C-terminal half of the central extracellular region of APP that has been implicated in the regulation of APP cleavage. Sequence comparisons demonstrate that the domain is highly conserved among all members of the APP family, including invertebrate homologues, suggesting an important role for this region in the biological function of APP. The identification of this domain and the availability of its atomic structure will facilitate analysis of APP function and of the role of the extracellular region in the regulation of APP cleavage.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Extracellular Space/chemistry , Peptide Fragments/chemistry , Protein Folding , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Humans , Hydrolysis , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid
17.
Proc Natl Acad Sci U S A ; 100(1): 32-7, 2003 Jan 07.
Article in English | MEDLINE | ID: mdl-12506202

ABSTRACT

Sec1Munc18-like (SM) proteins functionally interact with soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) in membrane fusion, but the mechanisms of these interactions differ. In vertebrates, SM proteins that mediate exocytosis (Munc18-1, 18-2, and 18c) bind to the closed conformation of syntaxins 1-4, which requires the N-terminal H(abc) domains and SNARE motifs of these syntaxins. In contrast, SM proteins that mediate Golgi and endoplasmic reticulum fusion (Sly1 and Vps45) bind only to short N-terminal sequences of syntaxins 5, 16, or 18, independently of their H(abc) domains and SNARE motifs. We now show that Munc18-1, Sly1, and Vps45 interact with cognate syntaxins via similar, autonomously folded N-terminal domains, but the syntaxin 5-binding surface of the Sly1 N-terminal domain is opposite to the syntaxin 1-binding surface of the Munc18-1 N-terminal domain. In transfected cells, the N-terminal domain of Sly1 specifically disrupts the structure of the Golgi complex, supporting the notion that the interaction of Sly1 with syntaxin 5 is essential for fusion. These data, together with previous results, suggest that a relatively small N-terminal domain of SM proteins is dedicated to mechanistically distinct interactions with SNAREs, leaving the remaining large parts of SM proteins free to execute their as yet unknown function as effector domains.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins , Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Vesicular Transport Proteins , Animals , Binding Sites , Carrier Proteins/metabolism , Chlorocebus aethiops , Cloning, Molecular , Escherichia coli/genetics , Models, Molecular , Munc18 Proteins , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Secondary , Qa-SNARE Proteins , Recombinant Fusion Proteins/metabolism , SNARE Proteins , Saccharomyces cerevisiae Proteins/metabolism , Transfection , Vero Cells
18.
EMBO J ; 21(14): 3620-31, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12110575

ABSTRACT

Soluble N-ethylmaleimide sensitive factor-attachment protein receptors (SNAREs) and Sec1p/Munc18-homologs (SM proteins) play key roles in intracellular membrane fusion. The SNAREs form tight four-helix bundles (core complexes) that bring the membranes together, but it is unclear how this activity is coupled to SM protein function. Studies of the yeast trans-Golgi network (TGN)/endosomal SNARE complex, which includes the syntaxin-like SNARE Tlg2p, have suggested that its assembly requires activation by binding of the SM protein Vps45p to the cytoplasmic region of Tlg2p folded into a closed conformation. Nuclear magnetic resonance and biochemical experiments now show that Tlg2p and Pep12p, a late- endosomal syntaxin that interacts functionally but not directly with Vps45p, have a domain structure characteristic of syntaxins but do not adopt a closed conformation. Tlg2p binds tightly to Vps45p via a short N-terminal peptide motif that is absent in Pep12p. The Tlg2p/Vps45p binding mode is shared by the mammalian syntaxin 16, confirming that it is a Tlg2p homolog, and resembles the mode of interaction between the SM protein Sly1p and the syntaxins Ufe1p and Sed5p. Thus, this mechanism represents the most widespread mode of coupling between syntaxins and SM proteins.


Subject(s)
Fungal Proteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Saccharomyces cerevisiae Proteins , Vesicular Transport Proteins , Amino Acid Motifs , Amino Acid Sequence , Fungal Proteins/chemistry , Membrane Proteins/chemistry , Molecular Sequence Data , Protein Binding , Protein Conformation , Qa-SNARE Proteins , Sequence Homology, Amino Acid , Syntaxin 16
19.
J Biol Chem ; 277(39): 36449-56, 2002 Sep 27.
Article in English | MEDLINE | ID: mdl-12114520

ABSTRACT

The SNAREs syntaxin 7, syntaxin 8, vti1b, and endobrevin/VAMP8 function in the fusion of late endosomes. Although the core complex formed by these SNAREs is very similar to the neuronal SNARE complex, it differs from the neuronal complex in that three of the four SNAREs contain extended N-terminal regions of unknown structure and function. Here we show that the N-terminal regions of syntaxin 7, syntaxin 8, and vti1b contain well folded alpha-helical domains. Multidimensional NMR spectroscopy revealed that in syntaxin 7 and vti1b, the domains form three-helix bundles resembling those of syntaxin 1, Sso1p, and Vam3p. The three-helix bundle domain of vti1b is the first of its kind identified in a SNARE outside the syntaxin family. Only syntaxin 7 adopts a closed conformation, whereas in vti1b and syntaxin 8, the N-terminal domains do not interact with the adjacent SNARE motifs. Accordingly, the rate of SNARE complex assembly is retarded about 7-fold when syntaxin 7 contains its N-terminal domain, whereas the N-terminal domains of vti1b and syntaxin 8 have no influence on assembly kinetics. We conclude that three-helix bundles represent a common fold for SNARE N-terminal domains, not restricted to the syntaxin family. However, they differ in their ability to adopt closed conformations and thus to regulate the assembly of SNARE complexes.


Subject(s)
Carrier Proteins/chemistry , Membrane Proteins/chemistry , Saccharomyces cerevisiae Proteins , Vesicular Transport Proteins , Amino Acid Motifs , Amino Acid Sequence , Animals , Antigens, Surface/chemistry , Cloning, Molecular , Cytoplasm/metabolism , Electrophoresis, Polyacrylamide Gel , Fungal Proteins/chemistry , Glutathione Transferase/metabolism , Humans , Kinetics , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Qa-SNARE Proteins , Qb-SNARE Proteins , SNARE Proteins , Sequence Homology, Amino Acid , Syntaxin 1 , Temperature , Time Factors
20.
Biochemistry ; 41(19): 5956-62, 2002 May 14.
Article in English | MEDLINE | ID: mdl-11993989

ABSTRACT

PX domains have been recently found to act as phosphoinositide binding modules. In the yeast SNARE protein Vam7p, the PX domain binds to PtdIns(3)P and is required for vacuolar targeting. To gain insight into how PX domains function, the solution structure of the ligand-free Vam7p PX domain has been determined by NMR spectroscopy. The Vam7p PX domain has the same overall alpha/beta fold observed in the structures of the ligand-free p47(phox) PX domain and the PtdIns(3)P-bound p40(phox) PX domain, exhibiting several similarities and differences with these two PX domains. Most striking is the similarity between the Vam7p and p40(phox) PX domains in a subset of secondary structure elements despite the low level of sequence identity between them, suggesting that these elements form a conserved core in the PX domain fold. These similarities and the observation that a putative PtdIns(3)P binding site is already formed in the apo Vam7p PX domains suggest that ligand binding does not induce major conformational changes, contrary to what was previously thought. The proposed ligand binding site of the Vam7p PX domain includes basic side chains from the conserved structural core that also participate in PtdIns(3)P binding to the p40(phox) PX domain, and basic side chains from a variable loop that probably inserts into the membrane. These results indicate that PX domains contain a combination of conserved and variable features that allow them to have a common function and at the same time exhibit distinct specificities, mechanisms of regulation, or modes of interaction with effector molecules.


Subject(s)
Membrane Proteins/chemistry , Nerve Tissue Proteins/chemistry , Amino Acid Sequence , Binding Sites , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , NADPH Oxidases , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Phosphatidylinositols/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Protein Conformation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Solutions , Synaptosomal-Associated Protein 25 , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL