Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Biochemistry ; 62(20): 2934-2951, 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37776275

ABSTRACT

γ-herpesviruses (γHVs) encode BCL2 homologues (vBCL2) that bind the Bcl-2 homology 3 domains (BH3Ds) of diverse proteins, inhibiting apoptosis and promoting host cell and virus survival. vBCLs encoded by Kaposi sarcoma-associated HV (KSHV) and γHV68 downregulate autophagy, a degradative cellular process crucial for homeostasis and innate immune responses to pathogens, by binding to a BH3D in BECN1, a key autophagy protein. Epstein-Barr virus (EBV) encodes a vBCL2 called BHRF1. Here we show that unlike the KSHV and γHV68 vBCL2s, BHRF1 does not bind the isolated BECN1 BH3D. We use yeast two-hybrid assays to identify the minimal region of BECN1 required and sufficient for binding BHRF1. We confirm that this is a direct, albeit weak, interaction via affinity pull-down assays and isothermal titration calorimetry. To understand the structural bases of BHRF1 specificity, we determined the 2.6 Šcrystal structure of BHRF1 bound to the BID BH3D, which binds ∼400-times tighter to BHRF1 than does BECN1, and performed a detailed structural comparison with complexes of diverse BH3Ds bound to BHRF1 and to other antiapoptotic BCL2s. Lastly, we used mammalian cell autophagy assays to demonstrate that BHRF1 downregulates autophagy and that a cell-permeable peptide derived from the BID BH3D inhibits BHRF1-mediated downregulation of autophagy. In summary, our results suggest that BHRF1 downregulates autophagy by noncanonical binding of a flexible region of BECN1 that includes but is not limited to the BH3D and that BH3D-derived peptides that bind better to BHRF1 can block downregulation of autophagy by BHRF1.

2.
J Biol Chem ; 295(17): 5795-5806, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32107313

ABSTRACT

Cell-surface signaling (CSS) in Gram-negative bacteria involves highly conserved regulatory pathways that optimize gene expression by transducing extracellular environmental signals to the cytoplasm via inner-membrane sigma regulators. The molecular details of ferric siderophore-mediated activation of the iron import machinery through a sigma regulator are unclear. Here, we present the 1.56 Å resolution structure of the periplasmic complex of the C-terminal CSS domain (CCSSD) of PupR, the sigma regulator in the Pseudomonas capeferrum pseudobactin BN7/8 transport system, and the N-terminal signaling domain (NTSD) of PupB, an outer-membrane TonB-dependent transducer. The structure revealed that the CCSSD consists of two subdomains: a juxta-membrane subdomain, which has a novel all-ß-fold, followed by a secretin/TonB, short N-terminal subdomain at the C terminus of the CCSSD, a previously unobserved topological arrangement of this domain. Using affinity pulldown assays, isothermal titration calorimetry, and thermal denaturation CD spectroscopy, we show that both subdomains are required for binding the NTSD with micromolar affinity and that NTSD binding improves CCSSD stability. Our findings prompt us to present a revised model of CSS wherein the CCSSD:NTSD complex forms prior to ferric-siderophore binding. Upon siderophore binding, conformational changes in the CCSSD enable regulated intramembrane proteolysis of the sigma regulator, ultimately resulting in transcriptional regulation.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Pseudomonas/metabolism , Signal Transduction , Bacterial Outer Membrane Proteins/chemistry , Crystallography, X-Ray , Models, Molecular , Periplasm/metabolism , Protein Conformation , Protein Domains , Protein Interaction Maps , Pseudomonas/chemistry , Siderophores/metabolism
3.
J Biol Chem ; 292(39): 16235-16248, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28798234

ABSTRACT

Beclin 1 (BECN1) is a key regulator of autophagy, a critical catabolic homeostasis pathway that involves sequestration of selected cytoplasmic components by multilayered vesicles called autophagosomes, followed by lysosomal fusion and degradation. BECN1 is a core component of class III phosphatidylinositol-3-kinase complexes responsible for autophagosome nucleation. Without heterologous binding partners, BECN1 forms an antiparallel homodimer via its coiled-coil domain (CCD). However, the last 16 CCD residues, composing an "overlap helix" (OH), have been crystallized in two mutually exclusive states: either as part of the CCD or packed against the C-terminal ß-α repeated, autophagy-specific domain (BARAD). Here, using CD spectroscopy, isothermal titration calorimetry, and small-angle X-ray scattering, we show that in the homodimeric state, the OH transitions between these two different packing states, with the predominant state comprising the OH packed against the BARAD, contrary to expectations based on known BECN1 interactions with heterologous partners. We confirmed this observation by comparing the impact of mutating four residues that mediate packing of the OH against both the CCD and BARAD on structure and stability of the CCD, the OH+BARAD, and the two-domain CCD-BARAD. Last, we used cellular assays to demonstrate that mutation of these OH-interface residues abrogates starvation-induced up-regulation of autophagy but does not affect basal autophagy. In summary, we have identified a BECN1 helical region that transitions between packing as part of either one of two conserved domains (i.e. the CCD or the BARAD). Our findings have important implications for the relative stability of autophagy-inactive and autophagy-active BECN1 complexes.


Subject(s)
Autophagy , Beclin-1/metabolism , Models, Molecular , Amino Acid Substitution , Beclin-1/chemistry , Beclin-1/genetics , Circular Dichroism , Crystallography, X-Ray , Dimerization , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , MCF-7 Cells , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Conformation, alpha-Helical , Protein Folding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Stability , Protein Unfolding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Scattering, Small Angle
4.
Biochemistry ; 55(30): 4239-53, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27383850

ABSTRACT

Autophagy, an essential eukaryotic homeostasis pathway, allows the sequestration of unwanted, damaged, or harmful cytoplasmic components in vesicles called autophagosomes, permitting subsequent lysosomal degradation and nutrient recycling. Autophagosome nucleation is mediated by class III phosphatidylinositol-3-kinase complexes that include two key autophagy proteins, BECN1/Beclin 1 and ATG14/BARKOR, which form parallel heterodimers via their coiled-coil domains (CCDs). Here we present the 1.46 Å X-ray crystal structure of the antiparallel, human BECN1 CCD homodimer, which represents BECN1 oligomerization outside the autophagosome nucleation complex. We use circular dichroism and small-angle X-ray scattering (SAXS) to show that the ATG14 CCD is significantly disordered but becomes more helical in the BECN1:ATG14 heterodimer, although it is less well-folded than the BECN1 CCD homodimer. SAXS also indicates that the BECN1:ATG14 heterodimer is more curved than other BECN1-containing CCD dimers, which has important implications for the structure of the autophagosome nucleation complex. A model of the BECN1:ATG14 CCD heterodimer that agrees well with the SAXS data shows that BECN1 residues at the homodimer interface are also responsible for heterodimerization, allowing us to identify ATG14 interface residues. Finally, we verify the role of BECN1 and ATG14 interface residues in binding by assessing the impact of point mutations of these residues on co-immunoprecipitation of the partner and demonstrate that these mutations abrogate starvation-induced upregulation of autophagy but do not impact basal autophagy. Thus, this research provides insights into structures of the BECN1 CCD homodimer and the BECN1:ATG14 CCD heterodimer and identifies interface residues that are important for BECN1:ATG14 heterodimerization and for autophagy.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/physiology , Autophagy-Related Proteins/chemistry , Autophagy-Related Proteins/physiology , Autophagy/physiology , Beclin-1/chemistry , Beclin-1/physiology , Adaptor Proteins, Vesicular Transport/genetics , Autophagy-Related Proteins/genetics , Beclin-1/genetics , Circular Dichroism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Scattering, Small Angle , Starvation/physiopathology , X-Ray Diffraction
5.
Biochemistry ; 55(13): 1945-58, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26937551

ABSTRACT

BECN1 is essential for autophagy, a critical eukaryotic cellular homeostasis pathway. Here we delineate a highly conserved BECN1 domain located between previously characterized BH3 and coiled-coil domains and elucidate its structure and role in autophagy. The 2.0 Å sulfur-single-wavelength anomalous dispersion X-ray crystal structure of this domain demonstrates that its N-terminal half is unstructured while its C-terminal half is helical; hence, we name it the flexible helical domain (FHD). Circular dichroism spectroscopy, double electron-electron resonance-electron paramagnetic resonance, and small-angle X-ray scattering (SAXS) analyses confirm that the FHD is partially disordered, even in the context of adjacent BECN1 domains. Molecular dynamic simulations fitted to SAXS data indicate that the FHD transiently samples more helical conformations. FHD helicity increases in 2,2,2-trifluoroethanol, suggesting it may become more helical upon binding. Lastly, cellular studies show that conserved FHD residues are required for starvation-induced autophagy. Thus, the FHD likely undergoes a binding-associated disorder-to-helix transition, and conserved residues critical for this interaction are essential for starvation-induced autophagy.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Autophagy , Membrane Proteins/chemistry , Models, Molecular , Amino Acid Sequence , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Biomarkers/metabolism , Cell Line, Tumor , Conserved Sequence , Culture Media, Serum-Free , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Pliability , Point Mutation , Protein Conformation , Protein Refolding , Protein Stability , Protein Structure, Tertiary , Protein Unfolding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
6.
Biochemistry ; 54(38): 5867-77, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26313375

ABSTRACT

Gram-negative bacteria tightly regulate intracellular levels of iron, an essential nutrient. To ensure this strict control, some outer membrane TonB-dependent transporters (TBDTs) that are responsible for iron import stimulate their own transcription in response to extracellular binding by an iron-laden siderophore. This process is mediated by an inner membrane sigma regulator protein (an anti-sigma factor) that transduces an unknown periplasmic signal from the TBDT to release an intracellular sigma factor from the inner membrane, which ultimately upregulates TBDT transcription. Here, we use the Pseudomonas putida ferric-pseudobactin BN7/BN8 sigma regulator, PupR, as a model system to understand the molecular mechanism of this conserved class of sigma regulators. We have determined the X-ray crystal structure of the cytoplasmic anti-sigma domain (ASD) of PupR to 2.0 Å. Size exclusion chromatography, small-angle X-ray scattering, and sedimentation velocity analytical ultracentrifugation all indicate that, in contrast to other ASDs, the PupR-ASD exists as a dimer in solution. Mutagenesis of residues at the dimer interface identified from the crystal structure disrupts dimerization and protein stability, as determined by sedimentation velocity analytical ultracentrifugation and thermal denaturation circular dichroism spectroscopy. These combined results suggest that this type of inner membrane sigma regulator may utilize an unusual mechanism to sequester their cognate sigma factors and prevent transcription activation.


Subject(s)
Bacterial Proteins/chemistry , Membrane Proteins/chemistry , Pseudomonas putida/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Multimerization , Protein Stability , Protein Structure, Tertiary
7.
J Biol Chem ; 289(12): 8029-40, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24443581

ABSTRACT

γ-herpesviruses (γHVs) are common human pathogens that encode homologs of the anti-apoptotic cellular Bcl-2 proteins, which are critical to viral reactivation and oncogenic transformation. The murine γHV68 provides a tractable in vivo model for understanding general features of these important human pathogens. Bcl-XL, a cellular Bcl-2 homolog, and the murine γHV68 Bcl-2 homolog, M11, both bind to a BH3 domain within the key autophagy effector Beclin 1 with comparable affinities, resulting in the down-regulation of Beclin 1-mediated autophagy. Despite this similarity, differences in residues lining the binding site of M11 and Bcl-XL dictate varying affinities for the different BH3 domain-containing proteins. Here we delineate Beclin 1 differential specificity determinants for binding to M11 or Bcl-XL by quantifying autophagy levels in cells expressing different Beclin 1 mutants and either M11 or Bcl-XL, and we show that a G120E/D121A Beclin 1 mutant selectively prevents down-regulation of Beclin 1-mediated autophagy by Bcl-XL, but not by M11. We use isothermal titration calorimetry to identify a Beclin 1 BH3 domain-derived peptide that selectively binds to M11, but not to Bcl-XL. The x-ray crystal structure of this peptide bound to M11 reveals the mechanism by which the M11 BH3 domain-binding groove accommodates this M11-specific peptide. This information was used to develop a cell-permeable peptide inhibitor that selectively inhibits M11-mediated, but not Bcl-XL-mediated, down-regulation of autophagy.


Subject(s)
Autophagy/drug effects , Down-Regulation/drug effects , Gammaherpesvirinae/physiology , Host-Pathogen Interactions/drug effects , Peptides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Line, Tumor , Crystallography, X-Ray , Gammaherpesvirinae/chemistry , Herpesviridae Infections/drug therapy , Herpesviridae Infections/metabolism , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Protein Interaction Maps , Proto-Oncogene Proteins c-bcl-2/chemistry , Sequence Alignment , Viral Proteins/chemistry , bcl-X Protein/chemistry , bcl-X Protein/metabolism
8.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 5): 1176-83, 2015 May.
Article in English | MEDLINE | ID: mdl-25945582

ABSTRACT

S100B is a damage-associated molecular pattern protein that, when released into the extracellular milieu, triggers initiation of the inflammatory response through the receptor for advanced glycation end products (RAGE). Recognition of S100B is accomplished via the amino-terminal variable immunoglobulin domain (V-domain) of RAGE. To gain insights into this interaction, a complex between S100B and a 15-amino-acid peptide derived from residues 54-68 of the V-domain was crystallized. The X-ray crystal structure was solved to 2.55 Å resolution. There are two dimers of S100B and one peptide in the asymmetric unit. The binding interface of this peptide is compared with that found in the complex between S100B and the 12-amino-acid CapZ-derived peptide TRTK-12. This comparison reveals that although the peptides adopt completely different backbone structures, the residues buried at the interface interact with S100B in similar regions to form stable complexes. The binding affinities of S100B for the intact wild-type V-domain and a W61A V-domain mutant were determined to be 2.7 ± 0.5 and 1.3 ± 0.7 µM, respectively, using fluorescence titration experiments. These observations lead to a model whereby conformational flexibility in the RAGE receptor allows the adoption of a binding conformation for interaction with the stable hydrophobic groove on the surface of S100B.


Subject(s)
CapZ Actin Capping Protein/metabolism , Peptide Fragments/metabolism , Receptor for Advanced Glycation End Products/chemistry , Receptor for Advanced Glycation End Products/genetics , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein beta Subunit/chemistry , S100 Calcium Binding Protein beta Subunit/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding , Protein Structure, Tertiary
9.
Biochemistry ; 53(36): 5779-90, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25144868

ABSTRACT

Calcineurin is an essential serine/threonine phosphatase that plays vital roles in neuronal development and function, heart growth, and immune system activation. Calcineurin is unique in that it is the only phosphatase known to be activated by calmodulin in response to increasing intracellular calcium concentrations. Calcium-loaded calmodulin binds to the regulatory domain of calcineurin, resulting in a conformational change that removes an autoinhibitory domain from the active site of the phosphatase. We have determined a 1.95 Å crystal structure of calmodulin bound to a peptide corresponding to its binding region from calcineurin. In contrast to previous structures of this complex, our structure has a stoichiometry of 1:1 and has the canonical collapsed, wraparound conformation observed for many calmodulin-substrate complexes. In addition, we have used size-exclusion chromatography and time-resolved fluorescence to probe the stoichiometry of binding of calmodulin to a construct corresponding to almost the entire regulatory domain from calcineurin, again finding a 1:1 complex. Taken in sum, our data strongly suggest that a single calmodulin protein is necessary and sufficient to bind to and activate each calcineurin enzyme.


Subject(s)
Calcineurin/metabolism , Calmodulin/metabolism , Base Sequence , Calcineurin/chemistry , Calmodulin/chemistry , Chromatography, Gel , DNA Primers , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Conformation , Spectrometry, Fluorescence
10.
Proteins ; 82(4): 565-78, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24115198

ABSTRACT

Autophagy is an essential eukaryotic pathway required for cellular homeostasis. Numerous key autophagy effectors and regulators have been identified, but the mechanism by which they carry out their function in autophagy is not fully understood. Our rigorous bioinformatic analysis shows that the majority of key human autophagy proteins include intrinsically disordered regions (IDRs), which are sequences lacking stable secondary and tertiary structure; suggesting that IDRs play an important, yet hitherto uninvestigated, role in autophagy. Available crystal structures corroborate the absence of structure in some of these predicted IDRs. Regions of orthologs equivalent to the IDRs predicted in the human autophagy proteins are poorly conserved, indicating that these regions may have diverse functions in different homologs. We also show that IDRs predicted in human proteins contain several regions predicted to facilitate protein-protein interactions, and delineate the network of proteins that interact with each predicted IDR-containing autophagy protein, suggesting that many of these interactions may involve IDRs. Lastly, we experimentally show that a BCL2 homology 3 domain (BH3D), within the key autophagy effector BECN1 is an IDR. This BH3D undergoes a dramatic conformational change from coil to α-helix upon binding to BCL2s, with the C-terminal half of this BH3D constituting a binding motif, which serves to anchor the interaction of the BH3D to BCL2s. The information presented here will help inform future in-depth investigations of the biological role and mechanism of IDRs in autophagy proteins.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Intrinsically Disordered Proteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Autophagy , Beclin-1 , Humans , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Sequence Alignment
11.
Autophagy ; 20(2): 380-396, 2024 02.
Article in English | MEDLINE | ID: mdl-37791766

ABSTRACT

ABBREVIATIONS: AFM: aromatic finger mutant; BH3D: BCL2 homology 3 domain; CCD: coiled-coil domain; CD: circular dichroism spectroscopy; [CysDM1]: C18S and C21S double mutant; [CysDM2]: C137S, and C140S double mutant; [CysTM], C18S, C21S, C137S, and C140S tetrad mutant; Dmax: maximum particle diameter; dRI, differential refractive index; EFA: evolving factor analysis; FHD: flexible helical domain; FL: full length; GFP: green fluorescent protein; HDX-MS: hydrogen/deuterium exchange mass spectrometry; ICP-MS: inductively coupled plasma mass spectrometry; IDR: intrinsically disordered region; ITC, isothermal titration calorimetry; MALS, multi angle light scattering; MBP: maltose-binding protein; MoRFs: molecular recognition features; P(r): pairwise-distance distribution; PtdIns3K: class III phosphatidylinositol 3-kinase; Rg: radius of gyration; SASBDB: small angle scattering biological data bank; SEC: size-exclusion chromatography; SEC-SAXS: size-exclusion chromatography in tandem with small angle X-ray scattering; TEV: tobacco-etch virus; TFE: 2,2,2-trifluoroethanol; TPEN: N,N,N,N-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine; Vc: volume of correlation; WT: wild-type.


Subject(s)
Autophagy , Zinc , Scattering, Small Angle , X-Ray Diffraction , Autophagy/physiology , Protein Domains
12.
Biomolecules ; 14(9)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39334875

ABSTRACT

Cell surface signaling (CSS) is a means of rapidly adjusting transcription in response to extracellular stimuli in Gram-negative bacteria. The pseudobactin BN7/8 uptake (Pup) system not only imports iron but also upregulates its own transcription through CSS in Pseudomonas capeferrum. In the absence of ferric pseudobactin BN7/8, the signaling components are maintained in a resting state via the formation of a periplasmic complex between the N-terminal signaling domain (NTSD) of the outer membrane iron-transporter, PupB, and the C-terminal CSS domain (CCSSD) of the sigma regulator, PupR. The previously determined 1.6 Å crystal structure of this periplasmic complex has allowed us to probe the structural and thermodynamic consequences of mutating key interfacial residues. In this report, we describe the solution structure of the PupB NTSD and use Nuclear Magnetic Resonance spectroscopy, Isothermal Titration Calorimetry, and Circular Dichroism spectroscopy together with thermal denaturation to investigate whether three PupB point mutations, Q69K, H72D, and L74A, influence the interaction merely due to the chemical nature of the amino acid substitution or also cause changes in overall protein structure. Our results demonstrate that binding to the PupR CCSSD does not alter the structure of PupB NTSD and that the individual mutations have only minor effects on structure. The mutations generally lower thermodynamic stability of the NTSD and weaken binding to the CCSSD. These findings validate the X-ray crystal structure interface, emphasizing the importance of amino acid chemical nature at the interface.


Subject(s)
Bacterial Proteins , Pseudomonas , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Pseudomonas/metabolism , Pseudomonas/genetics , Protein Domains , Signal Transduction , Thermodynamics , Models, Molecular
13.
G3 (Bethesda) ; 14(10)2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39167608

ABSTRACT

Flavonoids are secondary metabolites associated with plant seed coat and flower color. These compounds provide health benefits to humans as anti-inflammatory and antioxidant compounds. The expression of the late biosynthetic genes in the flavonoid pathway is controlled by a ternary MBW protein complex consisting of interfacing MYB, beta-helix-loop-helix (bHLH), and WD40 Repeat (WDR) proteins. P, the master regulator gene of the flavonoid expression in common bean (Phaseolus vulgaris L.), was recently determined to encode a bHLH protein. The T and Z genes control the distribution of color in bean seeds and flowers and have historically been considered regulators of the flavonoid gene expression. T and Z candidates were identified using reverse genetics based on genetic mapping, phylogenetic analysis, and mutant analysis. Domain and AlphaFold2 structure analyses determined that T encodes a seven-bladed ß-propeller WDR protein, while Z encodes a R2R3 MYB protein. Deletions and SNPs in T and Z mutants, respectively, altered the 3D structure of these proteins. Modeling of the Z MYB/P bHLH/T WDR MBW complex identified interfacing sequence domains and motifs in all three genes that are conserved in dicots. One Z MYB motif is a possible beta-molecular recognition feature (ß-MoRF) that only appears in a structured state when Z MYB is modeled as a component of a MBW complex. Complexes containing mutant T and Z proteins changed the interaction of members of the complex in ways that would alter their role in regulating the expression of genes in the flavonoid pathway.


Subject(s)
Gene Expression Regulation, Plant , Phaseolus , Plant Proteins , Seeds , Plant Proteins/genetics , Plant Proteins/metabolism , Phaseolus/genetics , Phaseolus/metabolism , Seeds/metabolism , Seeds/genetics , Phylogeny , Mutation , Genes, Plant , Models, Molecular , Flavonoids/metabolism , Protein Binding , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
14.
Proc Natl Acad Sci U S A ; 107(44): 18820-5, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20952656

ABSTRACT

Spot 14 (S14) is a protein that is abundantly expressed in lipogenic tissues and is regulated in a manner similar to other enzymes involved in fatty acid synthesis. Deletion of S14 in mice decreased lipid synthesis in lactating mammary tissue, but the mechanism of S14's action is unknown. Here we present the crystal structure of S14 to 2.65 Å and biochemical data showing that S14 can form heterodimers with MIG12. MIG12 modulates fatty acid synthesis by inducing the polymerization and activity of acetyl-CoA carboxylase, the first committed enzymatic reaction in the fatty acid synthesis pathway. Coexpression of S14 and MIG12 leads to heterodimers and reduced acetyl-CoA carboxylase polymerization and activity. The structure of S14 suggests a mechanism whereby heterodimer formation with MIG12 attenuates the ability of MIG12 to activate ACC.


Subject(s)
Fatty Acids/biosynthesis , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Models, Molecular , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Crystallography, X-Ray , Fatty Acids/chemistry , Fatty Acids/genetics , Female , Mice , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Protein Structure, Tertiary , Structure-Activity Relationship , Transcription Factors/genetics
15.
Biosci Rep ; 40(1)2020 01 31.
Article in English | MEDLINE | ID: mdl-31912881

ABSTRACT

The receptor for advanced glycation end products (RAGE) recognizes damage-associated molecular patterns (DAMPs) and plays a critical role for the innate immune response and sterile tissue inflammation. RAGE overexpression is associated with diabetic complications, neurodegenerative diseases and certain cancers. Yet, the molecular mechanism of ligand recognition by RAGE is insufficiently understood to rationalize the binding of diverse ligands. The N-terminal V-type Ig-domain of RAGE contains a triad of tryptophan residue; Trp51, Trp61 and Trp72. The role of these three Trp residues for domain folding, stability and binding of the RAGE ligand S100B was investigated through site-directed mutagenesis, UV/VIS, CD and fluorescence spectrometry, protein-protein interaction studies, and X-ray crystallography. The data show that the Trp triad stabilizes the folded V-domain by maintaining a short helix in the structure. Mutation of any Trp residue increases the structural plasticity of the domain. Residues Trp61 and Trp72 are involved in the binding of S100B, yet they are not strictly required for S100B binding. The crystal structure of the RAGE-derived peptide W72 in complex with S100B showed that Trp72 is deeply buried in a hydrophobic depression on the S100B surface. The studies suggest that multiple binding modes between RAGE and S100B exist and point toward a not previously recognized role of the Trp residues for RAGE-ligand binding. The Trp triad of the V-domain appears to be a suitable target for novel RAGE inhibitors, either in the form of monoclonal antibodies targeting this epitope, or small organic molecules.


Subject(s)
Receptor for Advanced Glycation End Products/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Binding Sites , Hydrophobic and Hydrophilic Interactions , Ligands , Mutation , Protein Binding , Protein Folding , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Secondary , Receptor for Advanced Glycation End Products/chemistry , Receptor for Advanced Glycation End Products/genetics , S100 Calcium Binding Protein beta Subunit/chemistry , Structure-Activity Relationship , Tryptophan
16.
Biomol NMR Assign ; 12(1): 91-94, 2018 04.
Article in English | MEDLINE | ID: mdl-29071576

ABSTRACT

Outer membrane TonB-dependent transducers (TBDTs) actively transport ferric siderophore complexes from the extracellular environment into Gram-negative bacteria. They also participate in a cell-surface signaling regulatory pathway that results in upregulation of the transducer itself, in response to iron-deplete conditions. The TBDT PupB transports ferric pseudobactin, and signals through its N-terminal signaling domain (NTSD), while the TBDT homolog PupA is signaling-inactive. Here, we report the NMR chemical shift assignments of the PupB-NTSD. This information will provide the basis for structural characterization of the PupB-NTSD to further explore its signaling properties.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Signal Transduction , Protein Domains
17.
Protein Sci ; 26(5): 972-984, 2017 05.
Article in English | MEDLINE | ID: mdl-28218432

ABSTRACT

ATG14 binding to BECN/Beclin homologs is essential for autophagy, a critical catabolic homeostasis pathway. Here, we show that the α-helical, coiled-coil domain (CCD) of BECN2, a recently identified mammalian BECN1 paralog, forms an antiparallel, curved homodimer with seven pairs of nonideal packing interactions, while the BECN2 CCD and ATG14 CCD form a parallel, curved heterodimer stabilized by multiple, conserved polar interactions. Compared to BECN1, the BECN2 CCD forms a weaker homodimer, but binds more tightly to the ATG14 CCD. Mutation of nonideal BECN2 interface residues to more ideal pairs improves homodimer self-association and thermal stability. Unlike BECN1, all BECN2 CCD mutants bind ATG14, although more weakly than wild type. Thus, polar BECN2 CCD interface residues result in a metastable homodimer, facilitating dissociation, but enable better interactions with polar ATG14 residues stabilizing the BECN2:ATG14 heterodimer. These structure-based mechanistic differences in BECN1 and BECN2 homodimerization and heterodimerization likely dictate competitive ATG14 recruitment.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Autophagy-Related Proteins/chemistry , Autophagy , Intracellular Signaling Peptides and Proteins/chemistry , Protein Multimerization , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Structure, Quaternary , Protein Structure, Secondary
18.
Acta Crystallogr D Struct Biol ; 73(Pt 9): 775-792, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28876241

ABSTRACT

Mammalian Golgi-associated plant pathogenesis-related protein 1 (GAPR-1) is a negative autophagy regulator that binds Beclin 1, a key component of the autophagosome nucleation complex. Beclin 1 residues 267-284 are required for binding GAPR-1. Here, sequence analyses, structural modeling, mutagenesis combined with pull-down assays, X-ray crystal structure determination and small-angle X-ray scattering were used to investigate the Beclin 1-GAPR-1 interaction. Five conserved residues line an equatorial GAPR-1 surface groove that is large enough to bind a peptide. A model of a peptide comprising Beclin 1 residues 267-284 docked onto GAPR-1, built using the CABS-dock server, indicates that this peptide binds to this GAPR-1 groove. Mutation of the five conserved residues lining this groove, H54A/E86A/G102K/H103A/N138G, abrogates Beclin 1 binding. The 1.27 Šresolution X-ray crystal structure of this pentad mutant GAPR-1 was determined. Comparison with the wild-type (WT) GAPR-1 structure shows that the equatorial groove of the pentad mutant is shallower and more positively charged, and therefore may not efficiently bind Beclin 1 residues 267-284, which include many hydrophobic residues. Both WT and pentad mutant GAPR-1 crystallize as dimers, and in each case the equatorial groove of one subunit is partially occluded by the other subunit, indicating that dimeric GAPR-1 is unlikely to bind Beclin 1. SAXS analysis of WT and pentad mutant GAPR-1 indicates that in solution the WT forms monomers, while the pentad mutant is primarily dimeric. Thus, changes in the structure of the equatorial groove combined with the improved dimerization of pentad mutant GAPR-1 are likely to abrogate binding to Beclin 1.


Subject(s)
Beclin-1/metabolism , Membrane Proteins/metabolism , Protein Interaction Maps , Amino Acid Sequence , Animals , Autophagy , Beclin-1/chemistry , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Docking Simulation , Mutation , Protein Binding , Protein Conformation , Protein Multimerization , Scattering, Small Angle , Sequence Alignment , X-Ray Diffraction
19.
PLoS One ; 10(2): e0116512, 2015.
Article in English | MEDLINE | ID: mdl-25719602

ABSTRACT

Maintaining the integrity of the genome requires the high fidelity duplication of the genome and the ability of the cell to recognize and repair DNA lesions. The heterotrimeric single stranded DNA (ssDNA) binding complex Replication Protein A (RPA) is central to multiple DNA processes, which are coordinated by RPA through its ssDNA binding function and through multiple protein-protein interactions. Many RPA interacting proteins have been reported through large genetic and physical screens; however, the number of interactions that have been further characterized is limited. To gain a better understanding of how RPA functions in DNA replication, repair, and cell cycle regulation and to identify other potential functions of RPA, a yeast two hybrid screen was performed using the yeast 70 kDa subunit, Replication Factor A1 (Rfa1), as a bait protein. Analysis of 136 interaction candidates resulted in the identification of 37 potential interacting partners, including the cell cycle regulatory protein and DNA damage clamp loader Rad24. The Rfa1-Rad24 interaction is not dependent on ssDNA binding. However, this interaction appears affected by DNA damage. The regions of both Rfa1 and Rad24 important for this interaction were identified, and the region of Rad24 identified is distinct from the region reported to be important for its interaction with Rfc2 5. This suggests that Rad24-Rfc2-5 (Rad24-RFC) recruitment to DNA damage substrates by RPA occurs, at least partially, through an interaction between the N terminus of Rfa1 and the C terminus of Rad24. The predicted structure and location of the Rad24 C-terminus is consistent with a model in which RPA interacts with a damage substrate, loads Rad24-RFC at the 5' junction, and then releases the Rad24-RFC complex to allow for proper loading and function of the DNA damage clamp.


Subject(s)
Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Replication Protein A/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , DNA Damage/drug effects , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Methyl Methanesulfonate/toxicity , Molecular Sequence Data , Plasmids/genetics , Plasmids/metabolism , Protein Interaction Domains and Motifs/drug effects , Protein Structure, Tertiary , Replication Protein A/chemistry , Replication Protein A/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Alignment , Two-Hybrid System Techniques
20.
ACS Appl Mater Interfaces ; 5(9): 3500-3, 2013 May.
Article in English | MEDLINE | ID: mdl-23594364

ABSTRACT

PEG-derivatized corannulene compound has been found to be very effective in solubilizing single-walled carbon nanotubes in tetrahydrofuran. Solubilizing efficiency is close to the commonly used anionic surfactant, sodium dodecyl sulfate (SDS). Corannulene derivative has also been found to have a tendency to disperse metallic nanotubes more effectively than the SDS counterpart. Theoretical calculations predict higher dispersion interactions of corannulene backbone with the convex surface of nanotubes in comparison to those calculated with other commonly used polyaromatic hydrocarbon derivatives.


Subject(s)
Furans/chemistry , Nanotubes, Carbon/chemistry , Polycyclic Aromatic Hydrocarbons/chemistry , Polyethylene Glycols/chemistry , Models, Molecular , Solubility , Spectrum Analysis, Raman
SELECTION OF CITATIONS
SEARCH DETAIL