Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 40
Filtrer
1.
Nat Commun ; 12(1): 2743, 2021 05 12.
Article de Anglais | MEDLINE | ID: mdl-33980829

RÉSUMÉ

INI1/SMARCB1 binds to HIV-1 integrase (IN) through its Rpt1 domain and exhibits multifaceted role in HIV-1 replication. Determining the NMR structure of INI1-Rpt1 and modeling its interaction with the IN-C-terminal domain (IN-CTD) reveal that INI1-Rpt1/IN-CTD interface residues overlap with those required for IN/RNA interaction. Mutational analyses validate our model and indicate that the same IN residues are involved in both INI1 and RNA binding. INI1-Rpt1 and TAR RNA compete with each other for IN binding with similar IC50 values. INI1-interaction-defective IN mutant viruses are impaired for incorporation of INI1 into virions and for particle morphogenesis. Computational modeling of IN-CTD/TAR complex indicates that the TAR interface phosphates overlap with negatively charged surface residues of INI1-Rpt1 in three-dimensional space, suggesting that INI1-Rpt1 domain structurally mimics TAR. This possible mimicry between INI1-Rpt1 and TAR explains the mechanism by which INI1/SMARCB1 influences HIV-1 late events and suggests additional strategies to inhibit HIV-1 replication.


Sujet(s)
Intégrase du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , ARN viral/métabolisme , Protéine SMARCB1/métabolisme , Réplication virale , Génome viral , Intégrase du VIH/composition chimique , Intégrase du VIH/génétique , Interactions hôte-pathogène , Humains , Spectroscopie par résonance magnétique , Modèles moléculaires , Simulation de docking moléculaire , Liaison aux protéines , Domaines protéiques , ARN viral/composition chimique , Protéine SMARCB1/composition chimique , Protéine SMARCB1/génétique , Virion/croissance et développement , Virion/métabolisme
2.
Heliyon ; 5(12): e03018, 2019 Dec.
Article de Anglais | MEDLINE | ID: mdl-31890962

RÉSUMÉ

Fusion of host and viral membranes is a crucial step during infection by enveloped viruses. In the structurally-defined "class I″ viral glycoproteins, the formation of a highly stable α-helical bundle by the ectodomain of the fusion subunit (e.g., GP2 for Marburg virus, MARV) is postulated to provide the energetic driving force to overcome barriers associated with membrane fusion. Upon cell binding, the fusion subunit is proposed to form an extended intermediate that bridges both the viral and host membranes, and collapse of this extended intermediate brings the two membranes into proximity. While there is much high-resolution structural data available for prefusion and post-fusion structures of viral glycoproteins, little information is available about intermediate conformations especially in the context of the fusion loop/peptide (FL or FP) and membrane-proximal external region (MPER)/transmembrane (TM) segments. We present structural and functional studies on segments of MARV GP2 that encompass the FL and MPER/TM in detergent micelles and lipid bicelles. A protein that contains most elements of GP2 ("MGP2-full") is α-helical in membrane-mimicking environments and has pH-dependent membrane lytic activity. MGP2-full is monomeric under such conditions, contrasting with the trimeric species that has been described previously for MARV GP2 ectodomain in aqueous buffer. Variants of MARV GP2 containing the N- and C-terminal halves ("MGP2-FNL" and "MGP2-CMT", respectively) have similar properties. This work provides novel insight into conformational and membrane-perturbing properties of the MARV fusion subunit and how they may relate to viral membrane fusion.

3.
J Virol ; 92(18)2018 09 15.
Article de Anglais | MEDLINE | ID: mdl-29976679

RÉSUMÉ

Dengue virus is the most globally prevalent mosquito-transmitted virus. Primary infection with one of four cocirculating serotypes (DENV-1 to -4) causes a febrile illness, but secondary infection with a heterologous serotype can result in severe disease, due in part to antibody-dependent enhancement of infection (ADE). In ADE, cross-reactive but nonneutralizing antibodies, or subprotective levels of neutralizing antibodies, promote uptake of antibody-opsonized virus in Fc-γ receptor-positive cells. Thus, elicitation of broadly neutralizing antibodies (bNAbs), but not nonneutralizing antibodies, is desirable for dengue vaccine development. Domain III of the envelope glycoprotein (EDIII) is targeted by bNAbs and thus is an attractive immunogen. However, immunization with EDIII results in sera with limited neutralization breadth. We developed "resurfaced" EDIII immunogens (rsDIIIs) in which the A/G strand epitope that is targeted by bNAb 4E11 is maintained but less desirable epitopes are masked. RsDIIIs bound 4E11, but not serotype-specific or nonneutralizing antibodies. One rsDIII and, unexpectedly, wild-type (WT) DENV-2 EDIII elicited cross-neutralizing antibody responses against DENV-1 to -3 in mice. While these sera were cross-neutralizing, they were not sufficiently potent to protect AG129 immunocompromised mice at a dose of 200 µl (50% focus reduction neutralization titer [FRNT50], ∼1:60 to 1:130) against mouse-adapted DENV-2. Our results provide insight into immunogen design strategies based on EDIII.IMPORTANCE Dengue virus causes approximately 390 million infections per year. Primary infection by one serotype causes a self-limiting febrile illness, but secondary infection by a heterologous serotype can result in severe dengue syndrome, which is characterized by hemorrhagic fever and shock syndrome. This severe disease is thought to arise because of cross-reactive, non- or poorly neutralizing antibodies from the primary infection that are present in serum at the time of secondary infection. These cross-reactive antibodies enhance the infection rather than controlling it. Therefore, induction of a broadly and potently neutralizing antibody response is desirable for dengue vaccine development. Here, we explore a novel strategy for developing immunogens based on domain III of the E glycoprotein, where undesirable epitopes (nonneutralizing or nonconserved) are masked by mutation. This work provides fundamental insight into the immune response to domain III that can be leveraged for future immunogen design.


Sujet(s)
Anticorps neutralisants/biosynthèse , Anticorps antiviraux/immunologie , Virus de la dengue/génétique , Domaines protéiques/génétique , Protéines virales/immunologie , Animaux , Anticorps neutralisants/immunologie , Anticorps antiviraux/administration et posologie , Anticorps antiviraux/effets indésirables , Facilitation dépendante des anticorps , Techniques d'exposition à la surface cellulaire , Réactions croisées , Dengue/virologie , Vaccins contre la dengue/immunologie , Virus de la dengue/composition chimique , Virus de la dengue/immunologie , Épitopes/immunologie , Souris , Domaines protéiques/immunologie , Ingénierie des protéines/méthodes , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/immunologie , Protéines virales/composition chimique , Protéines virales/génétique
4.
Planta Med ; 84(16): 1201-1212, 2018 Nov.
Article de Anglais | MEDLINE | ID: mdl-29742762

RÉSUMÉ

Native mass spectrometry detection of ligand-protein complexes allowed rapid detection of natural product binders of apo and calcium-bound S100A4 (a member of the metal binding protein S100 family), T cell/transmembrane, immunoglobulin (Ig), and mucin protein 3, and T cell immunoreceptor with Ig and ITIM (immunoreceptor tyrosine-based inhibitory motif) domains precursor protein from extracts and fractions. Based on molecular weight common hits were detected binding to all four proteins. Seven common hits were identified as apigenin 6-C-ß-D-glucoside 8-C-α-L-arabinoside, sweroside, 4',5-dihydroxy-7-methoxyflavanone-6-C-rutinoside, loganin acid, 6-C-glucosylnaringenin, biochanin A 7-O-rutinoside and quercetin 3-O-rutinoside. Mass guided isolation and NMR identification of hits confirmed the mass accuracy of the ligand in the ligand-protein MS complexes. Thus, molecular weight ID from ligand-protein complexes by electrospray ionization Fourier transform mass spectrometry allowed rapid dereplication. Native mass spectrometry using electrospray ionization Fourier transform mass spectrometry is a tool for dereplication and metabolomics analysis.


Sujet(s)
Évaluation préclinique de médicament/méthodes , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Récepteurs immunologiques/métabolisme , Protéine S100A4 liant le calcium/métabolisme , Spectrométrie de masse ESI/méthodes , Calcium/composition chimique , Calcium/métabolisme , Analyse de Fourier , Récepteur cellulaire-2 du virus de l'hépatite A/analyse , Récepteur cellulaire-2 du virus de l'hépatite A/composition chimique , Spectroscopie par résonance magnétique , Masse moléculaire , Extraits de plantes/analyse , Extraits de plantes/métabolisme , Récepteurs immunologiques/analyse , Récepteurs immunologiques/composition chimique , Protéine S100A4 liant le calcium/analyse , Protéine S100A4 liant le calcium/composition chimique
5.
Nat Commun ; 8(1): 2215, 2017 12 20.
Article de Anglais | MEDLINE | ID: mdl-29263320

RÉSUMÉ

Nucleoplasmin (Npm) is a highly conserved histone chaperone responsible for the maternal storage and zygotic release of histones H2A/H2B. Npm contains a pentameric N-terminal core domain and an intrinsically disordered C-terminal tail domain. Though intrinsically disordered regions are common among histone chaperones, their roles in histone binding and chaperoning remain unclear. Using an NMR-based approach, here we demonstrate that the Xenopus laevis Npm tail domain controls the binding of histones at its largest acidic stretch (A2) via direct competition with both the C-terminal basic stretch and basic nuclear localization signal. NMR and small-angle X-ray scattering (SAXS) structural analyses allowed us to construct models of both the tail domain and the pentameric complex. Functional analyses demonstrate that these competitive intramolecular interactions negatively regulate Npm histone chaperone activity in vitro. Together these data establish a potentially generalizable mechanism of histone chaperone regulation via dynamic and specific intramolecular shielding of histone interaction sites.


Sujet(s)
Histone/métabolisme , Nucléoplasmines/métabolisme , Protéines de Xénope/métabolisme , Animaux , Chromatine , Cristallographie aux rayons X , Chaperons d'histones/métabolisme , Spectroscopie par résonance magnétique , Simulation de dynamique moléculaire , Nucléosomes/métabolisme , Liaison aux protéines , Diffusion aux petits angles , Xenopus laevis
6.
Proc Natl Acad Sci U S A ; 113(50): 14312-14317, 2016 12 13.
Article de Anglais | MEDLINE | ID: mdl-27911811

RÉSUMÉ

We are just beginning to understand the allosteric regulation of the human cytosolic sulfotransferase (SULTs) family-13 disease-relevant enzymes that regulate the activities of hundreds, if not thousands, of signaling small molecules. SULT1A1, the predominant isoform in adult liver, harbors two noninteracting allosteric sites, each of which binds a different molecular family: the catechins (naturally occurring flavonols) and nonsteroidal antiinflammatory drugs (NSAIDs). Here, we present the structure of an SULT allosteric binding site-the catechin-binding site of SULT1A1 bound to epigallocatechin gallate (EGCG). The allosteric pocket resides in a dynamic region of the protein that enables EGCG to control opening and closure of the enzyme's active-site cap. Furthermore, the structure offers a molecular explanation for the isozyme specificity of EGCG, which is corroborated experimentally. The binding-site structure was obtained without X-ray crystallography or multidimensional NMR. Instead, a SULT1A1 apoprotein structure was used to guide positioning of a small number of spin-labeled single-Cys mutants that coat the entire enzyme surface with a paramagnetic field of sufficient strength to determine its contribution to the bound ligand's transverse (T2) relaxation from its 1D solution spectrum. EGCG protons were mapped to the protein surface by triangulation using the T2 values to calculate their distances to a trio of spin-labeled Cys mutants. The final structure was obtained using distance-constrained molecular dynamics docking. This approach, which is readily extensible to other systems, is applicable over a wide range of ligand affinities, requires little protein, avoids the need for isotopically labeled protein, and has no protein molecular weight limitations.


Sujet(s)
Arylsulfotransferase/composition chimique , Arylsulfotransferase/métabolisme , Catéchine/métabolisme , Site allostérique , Arylsulfotransferase/génétique , Catéchine/analogues et dérivés , Humains , Isoenzymes/composition chimique , Isoenzymes/génétique , Isoenzymes/métabolisme , Cinétique , Modèles moléculaires , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Mutagenèse dirigée , Résonance magnétique nucléaire biomoléculaire , Liaison aux protéines , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Marqueurs de spin , Spécificité du substrat
7.
Biochemistry ; 55(31): 4275-85, 2016 08 09.
Article de Anglais | MEDLINE | ID: mdl-27420643

RÉSUMÉ

Methyl-CpG binding protein 2 (MeCP2) is a multifunctional protein that guides neuronal development through its binding to DNA, recognition of sites of methyl-CpG (mCpG) DNA modification, and interaction with other regulatory proteins. Our study explores the relationship between mCpG and hydroxymethyl-CpG (hmCpG) recognition mediated by its mCpG binding domain (MBD) and binding cooperativity mediated by its C-terminal polypeptide. Previous study of the isolated MBD of MeCP2 documented an unusual mechanism by which ion uptake is required for discrimination of mCpG and hmCpG from CpG. MeCP2 binding cooperativity suppresses discrimination of modified DNA and is highly sensitive to both the total ion concentration and the type of counterions. Higher than physiological total ion concentrations completely suppress MeCP2 binding cooperativity, indicating a dominant electrostatic component to the interaction. Substitution of SO4(2-) for Cl(-) at physiological total ion concentrations also suppresses MeCP2 binding cooperativity, This effect is of particular note as the intracellular Cl(-) concentration changes during neuronal development. A related effect is that the protein-stabilizing solutes, TMAO and glutamate, reduce MeCP2 (but not isolated MBD) binding affinity by 2 orders of magnitude without affecting the apparent binding cooperativity. These observations suggest that polypeptide flexibility facilitates DNA binding by MeCP2. Consistent with this view, nuclear magnetic resonance (NMR) analyses show that ions have discrete effects on the structure of MeCP2, both MBD and the C-terminal domains. Notably, anion substitution results in changes in the NMR chemical shifts of residues, including some whose mutation causes the autism spectrum disorder Rett syndrome. Binding cooperativity makes MeCP2 an effective competitor with histone H1 for accessible DNA sites. The relationship between MeCP2 binding specificity and cooperativity is discussed in the context of chromatin binding, neuronal function, and neuronal development.


Sujet(s)
ADN/métabolisme , Protéine-2 de liaison au CpG méthylé/métabolisme , ADN/génétique , Épigenèse génétique , Histone/métabolisme , Humains , Cinétique , Domaine de liaison aux méthyl-CpG/génétique , Protéine-2 de liaison au CpG méthylé/composition chimique , Protéine-2 de liaison au CpG méthylé/génétique , Neurogenèse , Liaison aux protéines , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Syndrome de Rett/génétique , Syndrome de Rett/métabolisme
8.
J Infect Dis ; 212 Suppl 2: S146-53, 2015 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-25786917

RÉSUMÉ

Marburg virus (MARV) and the ebolaviruses belong to the family Filoviridae (the members of which are filoviruses) that cause severe hemorrhagic fever. Infection requires fusion of the host and viral membranes, a process that occurs in the host cell endosomal compartment and is facilitated by the envelope glycoprotein fusion subunit, GP2. The N-terminal fusion loop (FL) of GP2 is a hydrophobic disulfide-bonded loop that is postulated to insert and disrupt the host endosomal membrane during fusion. Here, we describe the first structural and functional studies of a protein corresponding to the MARV GP2 FL. We found that this protein undergoes a pH-dependent conformational change, as monitored by circular dichroism and nuclear magnetic resonance. Furthermore, we report that, under low pH conditions, the MARV GP2 FL can induce content leakage from liposomes. The general aspects of this pH-dependent structure and lipid-perturbing behavior are consistent with previous reports on Ebola virus GP2 FL. However, nuclear magnetic resonance studies in lipid bicelles and mutational analysis indicate differences in structure exist between MARV and Ebola virus GP2 FL. These results provide new insight into the mechanism of MARV GP2-mediated cell entry.


Sujet(s)
Marburgvirus/composition chimique , Marburgvirus/métabolisme , Protéines de l'enveloppe virale/composition chimique , Protéines de l'enveloppe virale/métabolisme , Animaux , Escherichia coli/virologie , Concentration en ions d'hydrogène , Lipides/composition chimique , Liposomes/métabolisme , Maladie de Marbourg/virologie , Conformation des protéines , Pénétration virale
9.
Biochemistry ; 54(8): 1589-99, 2015 Mar 03.
Article de Anglais | MEDLINE | ID: mdl-25658332

RÉSUMÉ

Fusion of host and viral membranes is a critical step during infection by membrane-bound viruses. The HIV-1 glycoproteins gp120 (surface subunit) and gp41 (fusion subunit) represent the prototypic system for studying this process; in the prevailing model, the gp41 ectodomain forms a trimeric six-helix bundle that constitutes a critical intermediate and provides the energetic driving force for overcoming barriers associated with membrane fusion. However, most structural studies of gp41 variants have been performed either on ectodomain constructs lacking one or more of the membrane-associated segments (the fusion peptide, FP, the membrane-proximal external region, MPER, and the transmembrane domain, TM) or on variants consisting of these isolated segments alone without the ectodomain. Several recent reports have suggested that the HIV-1 ectodomain, as well as larger construct containing the membrane-bound segments, dissociates from a trimer to a monomer in detergent micelles. Here we compare the properties of a series of gp41 variants to delineate the roles of the ectodomain, FP, and MPER and TM, all in membrane-mimicking environments. We find that these proteins are prone to formation of a monomer in detergent micelles. In one case, we observed exclusive monomer formation at pH 4 but conditional trimerization at pH 7 even at low micromolar (∼5 µM) protein concentrations. Liposome release assays demonstrate that these gp41-related proteins have the capacity to induce content leakage but that this activity is also strongly modulated by pH with much higher activity at pH 4. Circular dichroism, nuclear magnetic resonance, and binding assays with antibodies specific to the MPER provide insight into the structural and functional roles of the FP, MPER, and TM and their effect on structure within the larger context of the fusion subunit.


Sujet(s)
Protéine d'enveloppe gp41 du VIH/composition chimique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/composition chimique , Multimérisation de protéines , Membrane cellulaire/composition chimique , Membrane cellulaire/génétique , Membrane cellulaire/métabolisme , Protéine d'enveloppe gp41 du VIH/génétique , Protéine d'enveloppe gp41 du VIH/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Concentration en ions d'hydrogène , Micelles , Structure quaternaire des protéines , Structure secondaire des protéines , Structure tertiaire des protéines , Protéines recombinantes/composition chimique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme
10.
J Am Chem Soc ; 137(6): 2171-4, 2015 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-25634639

RÉSUMÉ

Lipid-based micelles provide an attractive option for therapeutic and diagnostic applications because of their small size (<20 nm) and ability to self-assemble and improve the solubility of both hydrophobic drugs and dyes. Their use, however, has been challenged by the fact that these particles are inherently unstable in serum becaue of interactions with protein components, which drives the micelle equilibrium to the monomeric state. We have engineered serum stabilized micelles using short quadruplex forming oligonucleotide extensions as the lipid headgroup. Quadruplex formation on the surface of the particles, confirmed by (1)H NMR, results in slight distortion of the otherwise spherical micelles and renders them resistant to disassembly by serum proteins for >24 h. Using antisense oligonucleotides we demonstrated that disruption of the quadruplex leads to micelle destabilization and cargo release. The ability to use oligonucleotide interactions to control lipid particle stability represents a new approach in the design of programmed nanoscale devices.


Sujet(s)
Lipides/composition chimique , Micelles , Oligonucléotides/composition chimique , Microscopie électronique à transmission
11.
RNA ; 20(4): 447-61, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24570482

RÉSUMÉ

RNA aptamers are being developed as inhibitors of macromolecular and cellular function, diagnostic tools, and potential therapeutics. Our understanding of the physical nature of this emerging class of nucleic acid-protein complexes is limited; few atomic resolution structures have been reported for aptamers bound to their protein target. Guided by chemical mapping, we systematically minimized an RNA aptamer (Lys1) selected against hen egg white lysozyme. The resultant 59-nucleotide compact aptamer (Lys1.2minE) retains nanomolar binding affinity and the ability to inhibit lysozyme's catalytic activity. Our 2.0-Å crystal structure of the aptamer-protein complex reveals a helical stem stabilizing two loops to form a protein binding platform that binds lysozyme distal to the catalytic cleft. This structure along with complementary solution analyses illuminate a novel protein-nucleic acid interface; (1) only 410 Å(2) of solvent accessible surface are buried by aptamer binding; (2) an unusually small fraction (∼18%) of the RNA-protein interaction is electrostatic, consistent with the limited protein phosphate backbone contacts observed in the structure; (3) a single Na(+) stabilizes the loops that constitute the protein-binding platform, and consistent with this observation, Lys1.2minE-lysozyme complex formation takes up rather than displaces cations at low ionic strength; (4) Lys1.2minE inhibits catalysis of large cell wall substrates but not catalysis of small model substrates; and (5) the helical stem of Lys1.2minE can be shortened to four base pairs (Lys1.2minF) without compromising binding affinity, yielding a 45-nucleotide aptamer whose structure may be an adaptable protein binding platform.


Sujet(s)
Aptamères nucléotidiques/pharmacologie , Lysozyme/antagonistes et inhibiteurs , Lysozyme/métabolisme , Sodium/métabolisme , Animaux , Aptamères nucléotidiques/composition chimique , Appariement de bases , Séquence nucléotidique , Catalyse , Poulets , Spectroscopie par résonance magnétique , Micrococcus/enzymologie , Modèles moléculaires , Données de séquences moléculaires , Lysozyme/génétique , Conformation d'acide nucléique , Concentration osmolaire , Liaison aux protéines , Conformation des protéines , Sodium/composition chimique , Électricité statique , Spécificité du substrat , Ultracentrifugation , Diffraction des rayons X
12.
Biomol NMR Assign ; 8(1): 103-8, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-23315339

RÉSUMÉ

The type I phosphoribosyltransferase OMP synthase (EC 2.4.2.10) is involved in de novo synthesis of pyrimidine nucleotides forming the UMP precursor orotidine 5'-monophosphate (OMP). The homodimeric enzyme has a Rossman α/ß core topped by a base-enclosing "hood" domain and a flexible domain-swapped catalytic loop. High-resolution X-ray structures of the homologous Salmonella typhimurium and yeast enzymes show that a general compacting of the core as well as movement of the hood and a major disorder-to-order transition of the loop occur upon binding of ligands MgPRPP and orotate. Here we present backbone NMR assignments for the unliganded yeast enzyme (49 kDa) and its complex with product OMP. We were able to assign 212-213 of the 225 non-proline backbone (15)N and amide proton resonances. Significant difference in chemical shifts of the amide cross peaks occur in regions of the structure that undergo movement upon ligand occupancy in the S. typhimurium enzyme.


Sujet(s)
Résonance magnétique nucléaire biomoléculaire , Orotate phosphoribosyltransferase/composition chimique , Saccharomyces cerevisiae/composition chimique , Saccharomyces cerevisiae/enzymologie , Séquence d'acides aminés , Biocatalyse , Isotopes du carbone , Hydrogène , Ligands , Isotopes de l'azote , Structure secondaire des protéines , Uridine monophosphate/analogues et dérivés , Uridine monophosphate/composition chimique , Uridine monophosphate/métabolisme
13.
Biochemistry ; 52(20): 3393-404, 2013 May 21.
Article de Anglais | MEDLINE | ID: mdl-23650881

RÉSUMÉ

Ebola virus and Sudan virus are members of the family Filoviridae of nonsegmented negative-strand RNA viruses ("filoviruses") that cause severe hemorrhagic fever with fatality rates as high as 90%. Infection by filoviruses requires membrane fusion between the host and the virus; this process is facilitated by the two subunits of the envelope glycoprotein, GP1 (the surface subunit) and GP2 (the transmembrane subunit). The membrane-proximal external region (MPER) is a Trp-rich segment that immediately precedes the transmembrane domain of GP2. In the analogous glycoprotein for HIV-1 (gp41), the MPER is critical for membrane fusion and is the target of several neutralizing antibodies. However, the role of the MPER in filovirus GP2 and its importance in membrane fusion have not been established. Here, we characterize the conformational properties of peptides representing the GP MPER segments of Ebola virus and Sudan virus in the presence of micelle-forming surfactants and lipids, at pH 7 and 4.6. Circular dichroism spectroscopy and tryptophan fluorescence indicate that the GP2 MPER peptides bind to micelles of sodium dodecyl sulfate and dodecylphosphocholine (DPC). Nuclear magnetic resonance spectroscopy of the Sudan virus MPER peptide revealed that residues 644-651 interact directly with DPC, and that this interaction enhances the helical conformation of the peptide. The Sudan virus MPER peptide was found to moderately inhibit cell entry by a GP-pseudotyped vesicular stomatitis virus but did not induce leakage of a fluorescent molecule from a large unilammellar vesicle comprised of 1-palmitoyl-2-oleoylphosphatidylcholine or cause hemolysis. Taken together, this analysis suggests the filovirus GP2 MPER binds and inserts shallowly into lipid membranes.


Sujet(s)
Ebolavirus/métabolisme , Peptides/composition chimique , Protéines de l'enveloppe virale/composition chimique , Séquence d'acides aminés , Ebolavirus/génétique , Humains , Micelles , Données de séquences moléculaires , Résonance magnétique nucléaire biomoléculaire , Peptides/métabolisme , Phosphoryl-choline/analogues et dérivés , Phosphoryl-choline/composition chimique , Phosphoryl-choline/métabolisme , Conformation des protéines , Tensioactifs/composition chimique , Tensioactifs/métabolisme , Tryptophane/génétique , Tryptophane/métabolisme , Protéines de l'enveloppe virale/métabolisme
14.
Genes Dev ; 26(1): 43-53, 2012 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-22215810

RÉSUMÉ

How RNA-binding proteins recognize specific sets of target mRNAs remains poorly understood because current approaches depend primarily on sequence information. In this study, we demonstrate that specific recognition of messenger RNAs (mRNAs) by RNA-binding proteins requires the correct spatial positioning of these sequences. We characterized both the cis-acting sequence elements and the spatial restraints that define the mode of RNA binding of the zipcode-binding protein 1 (ZBP1/IMP1/IGF2BP1) to the ß-actin zipcode. The third and fourth KH (hnRNP K homology) domains of ZBP1 specifically recognize a bipartite RNA element comprised of a 5' element (CGGAC) followed by a variable 3' element (C/A-CA-C/U) that must be appropriately spaced. Remarkably, the orientation of these elements is interchangeable within target transcripts bound by ZBP1. The spatial relationship of this consensus binding site identified conserved transcripts that were verified to associate with ZBP1 in vivo. The dendritic localization of one of these transcripts, spinophilin, was found to be dependent on both ZBP1 and the RNA elements recognized by ZBP1 KH34.


Sujet(s)
Glycoprotéines/métabolisme , ARN messager/métabolisme , Actines/métabolisme , Animaux , Séquence nucléotidique , Cellules cultivées , Séquence consensus , Dendrites/métabolisme , Régulation de l'expression des gènes , Glycoprotéines/composition chimique , Humains , Ligands , Souris , Protéines des microfilaments/métabolisme , Modèles moléculaires , Données de séquences moléculaires , Protéines de tissu nerveux/métabolisme , Liaison aux protéines , Structure tertiaire des protéines , ARN messager/composition chimique , Protéines de liaison à l'ARN
15.
Biochemistry ; 50(21): 4568-78, 2011 May 31.
Article de Anglais | MEDLINE | ID: mdl-21506533

RÉSUMÉ

The extracellular signal-regulated protein kinase, ERK2, fully activated by phosphorylation and without a His(6) tag, shows little tendency to dimerize with or without either calcium or magnesium ions when analyzed by light scattering or analytical ultracentrifugation. Light scattering shows that ~90% of ERK2 is monomeric. Sedimentation equilibrium data (obtained at 4.8-11.2 µM ERK2) with or without magnesium (10 mM) are well described by an ideal one-component model with a fitted molar mass of 40180 ± 240 Da (without Mg(2+) ions) or 41290 ± 330 Da (with Mg(2+) ions). These values, close to the sequence-derived mass of 41711 Da, indicate that no significant dimerization of ERK2 occurs in solution. Analysis of sedimentation velocity data for a 15 µM solution of ERK2 with an enhanced van Holde-Weischet method determined the sedimentation coefficient (s) to be ~3.22 S for activated ERK2 with or without 10 mM MgCl(2). The frictional coefficient ratio (f/f(0)) of 1.28 calculated from the sedimentation velocity and equilibrium data is close to that expected for an ~42 kDa globular protein. The translational diffusion coefficient of ~8.3 × 10(-7) cm(2) s(-1) calculated from the experimentally determined molar mass and sedimentation coefficient agrees with the value determined by dynamic light scattering in the absence and presence of calcium or magnesium ions and a value determined by NMR spectrometry. ERK2 has been proposed to homodimerize and bind only to cytoplasmic but not nuclear proteins [Casar, B., et al. (2008) Mol. Cell 31, 708-721]. Our light scattering data show, however, that ERK2 forms a strong 1:1 complex of ~57 kDa with the cytoplasmic scaffold protein PEA-15. Thus, ERK2 binds PEA-15 as a monomer. Our data provide strong evidence that ERK2 is monomeric under physiological conditions. Analysis of the same ERK2 construct with the nonphysiological His(6) tag shows substantial dimerization under the same ionic conditions.


Sujet(s)
Cations divalents/métabolisme , Cytoplasme/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Mitogen-Activated Protein Kinase 1/métabolisme , Phosphoprotéines/métabolisme , Protéines régulatrices de l'apoptose , Activation enzymatique , Humains , Lumière , Diffusion de rayonnements , Ultracentrifugation
16.
Biomol NMR Assign ; 5(1): 11-4, 2011 Apr.
Article de Anglais | MEDLINE | ID: mdl-20737255

RÉSUMÉ

Mevalonate diphosphate decarboxylase (MDD) catalyzes the ATP-dependent decarboxylation of diphosphomevalonate (DPM) to produce isopentenyl diphosphate (IPP), the molecular "building block" for more than 25,000 distinct isoprenoids, including cholesterol, steroid hormones and terpenoids. Here, we present the first backbone assignment of Streptococcus pneumoniae MDD in the unliganded state and in a ternary complex with DPM and AMPPCP--a nucleotide analogue unable to transfer the γ-phosphoryl group. The secondary chemical shifts for the unliganded form are in good agreement with the crystal structure of Streptococcus pyogenes (~70% sequence identity). The addition of substrate and nucleotide to the enzyme results in chemical shift changes of cross peaks that correspond to residues in the binding pocket.


Sujet(s)
Carboxy-lyases/composition chimique , Résonance magnétique nucléaire biomoléculaire , Streptococcus pneumoniae/enzymologie , Isotopes du carbone , Hydrogène , Ligands , Isotopes de l'azote , Structure secondaire des protéines , Spécificité du substrat
17.
J Struct Funct Genomics ; 11(3): 191-9, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20690043

RÉSUMÉ

The New York Consortium on Membrane Protein Structure (NYCOMPS) was formed to accelerate the acquisition of structural information on membrane proteins by applying a structural genomics approach. NYCOMPS comprises a bioinformatics group, a centralized facility operating a high-throughput cloning and screening pipeline, a set of associated wet labs that perform high-level protein production and structure determination by x-ray crystallography and NMR, and a set of investigators focused on methods development. In the first three years of operation, the NYCOMPS pipeline has so far produced and screened 7,250 expression constructs for 8,045 target proteins. Approximately 600 of these verified targets were scaled up to levels required for structural studies, so far yielding 24 membrane protein crystals. Here we describe the overall structure of NYCOMPS and provide details on the high-throughput pipeline.


Sujet(s)
Génomique/méthodes , Protéines membranaires/génétique , Cellules cultivées , Biologie informatique , Cristallographie aux rayons X , Génome , État de New York , Structure secondaire des protéines
18.
Biomol NMR Assign ; 4(2): 139-42, 2010 Oct.
Article de Anglais | MEDLINE | ID: mdl-20407887

RÉSUMÉ

The plasmid-encoded small multidrug resistance pump from S. aureus transports a variety of quaternary ammonium and other hydrophobic compounds, enhancing the bacterial host's resistance to common hospital disinfectants. The protein folds as a homo-dimer of four transmembrane helices each, and appears to be fully functional only in lipid bilayers. Here we report the backbone resonance assignments and implied secondary structure for (2)H(13)C(15)N Smr reconstituted into lipid bicelles. Significant changes were observed between the chemical shifts of the protein in lipid bicelles compared to those in detergent micelles.


Sujet(s)
Antiports/composition chimique , Résonance magnétique nucléaire biomoléculaire , Staphylococcus aureus/composition chimique , Isotopes du carbone , Hydrogène , Isotopes de l'azote , Structure secondaire des protéines
19.
Proc Natl Acad Sci U S A ; 106(48): 20258-63, 2009 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-19915146

RÉSUMÉ

We previously proposed a model of Class IA PI3K regulation in which p85 inhibition of p110alpha requires (i) an inhibitory contact between the p85 nSH2 domain and the p110alpha helical domain, and (ii) a contact between the p85 nSH2 and iSH2 domains that orients the nSH2 so as to inhibit p110alpha. We proposed that oncogenic truncations of p85 fail to inhibit p110 due to a loss of the iSH2-nSH2 contact. However, we now find that within the context of a minimal regulatory fragment of p85 (the nSH2-iSH2 fragment, termed p85ni), the nSH2 domain rotates much more freely (tau(c) approximately 12.7 ns) than it could if it were interacting rigidly with the iSH2 domain. These data are not compatible with our previous model. We therefore tested an alternative model in which oncogenic p85 truncations destabilize an interface between the p110alpha C2 domain (residue N345) and the p85 iSH2 domain (residues D560 and N564). p85ni-D560K/N564K shows reduced inhibition of p110alpha, similar to the truncated p85ni-572(STOP). Conversely, wild-type p85ni poorly inhibits p110alphaN345K. Strikingly, the p110alphaN345K mutant is inhibited to the same extent by the wild-type or truncated p85ni, suggesting that mutation of p110alpha-N345 is not additive with the p85ni-572(STOP) mutation. Similarly, the D560K/N564K mutation is not additive with the p85ni-572(STOP) mutant for downstream signaling or cellular transformation. Thus, our data suggests that mutations at the C2-iSH2 domain contact and truncations of the iSH2 domain, which are found in human tumors, both act by disrupting the C2-iSH2 domain interface.


Sujet(s)
Phosphatidylinositol 3-kinases/métabolisme , Structure tertiaire des protéines/génétique , Transduction du signal/physiologie , Analyse de variance , Animaux , Technique de Western , Lignée cellulaire , Cellules HeLa , Humains , Spectroscopie par résonance magnétique , Souris , Modèles biologiques , Mutation/génétique , Phosphatidylinositol 3-kinases/génétique , Transduction du signal/génétique
20.
J Struct Funct Genomics ; 10(2): 101-6, 2009 Apr.
Article de Anglais | MEDLINE | ID: mdl-19288278

RÉSUMÉ

This Perspective, arising from a workshop held in July 2008 in Buffalo NY, provides an overview of the role NMR has played in the United States Protein Structure Initiative (PSI), and a vision of how NMR will contribute to the forthcoming PSI-Biology program. NMR has contributed in key ways to structure production by the PSI, and new methods have been developed which are impacting the broader protein NMR community.


Sujet(s)
Génomique/méthodes , Résonance magnétique nucléaire biomoléculaire , Protéines/composition chimique , Cristallographie aux rayons X , Génomique/tendances , Conformation des protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...