Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Mol Cell ; 57(4): 735-749, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25661490

RESUMO

The nucleotide (p)ppGpp mediates bacterial stress responses, but its targets and underlying mechanisms of action vary among bacterial species and remain incompletely understood. Here, we characterize the molecular interaction between (p)ppGpp and guanylate kinase (GMK), revealing the importance of this interaction in adaptation to starvation. Combining structural and kinetic analyses, we show that (p)ppGpp binds the GMK active site and competitively inhibits the enzyme. The (p)ppGpp-GMK interaction prevents the conversion of GMP to GDP, resulting in GMP accumulation upon amino acid downshift. Abolishing this interaction leads to excess (p)ppGpp and defective adaptation to amino acid starvation. A survey of GMKs from phylogenetically diverse bacteria shows that the (p)ppGpp-GMK interaction is conserved in members of Firmicutes, Actinobacteria, and Deinococcus-Thermus, but not in Proteobacteria, where (p)ppGpp regulates RNA polymerase (RNAP). We propose that GMK is an ancestral (p)ppGpp target and RNAP evolved more recently as a direct target in Proteobacteria.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/metabolismo , Evolução Molecular , Guanosina Pentafosfato/metabolismo , Guanosina Tetrafosfato/metabolismo , Guanilato Quinases/metabolismo , Bactérias/genética , Bactérias/metabolismo , Ligação Competitiva , Domínio Catalítico , Cristalografia por Raios X , RNA Polimerases Dirigidas por DNA/metabolismo , Guanosina Pentafosfato/química , Guanosina Tetrafosfato/química , Guanosina Trifosfato/metabolismo , Guanilato Quinases/química , Modelos Biológicos , Especificidade da Espécie , Estresse Fisiológico
2.
Int J Mol Sci ; 24(4)2023 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-36834510

RESUMO

Both bioactive sphingolipids and Sigma-1 receptor (S1R) chaperones occur ubiquitously in mammalian cell membranes. Endogenous compounds that regulate the S1R are important for controlling S1R responses to cellular stress. Herein, we interrogated the S1R in intact Retinal Pigment Epithelial cells (ARPE-19) with the bioactive sphingoid base, sphingosine (SPH), or the pain-provoking dimethylated SPH derivative, N,N'-dimethylsphingosine (DMS). As informed by a modified native gel approach, the basal and antagonist (BD-1047)-stabilized S1R oligomers dissociated to protomeric forms in the presence of SPH or DMS (PRE-084 as control). We, thus, posited that SPH and DMS are endogenous S1R agonists. Consistently, in silico docking of SPH and DMS to the S1R protomer showed strong associations with Asp126 and Glu172 in the cupin beta barrel and extensive van der Waals interactions of the C18 alkyl chains with the binding site including residues in helices 4 and 5. Mean docking free energies were 8.73-8.93 kcal/mol for SPH and 8.56-8.15 kcal/mol for DMS, and calculated binding constants were ~40 nM for SPH and ~120 nM for DMS. We hypothesize that SPH, DMS, and similar sphingoid bases access the S1R beta barrel via a membrane bilayer pathway. We further propose that the enzymatic control of ceramide concentrations in intracellular membranes as the primary sources of SPH dictates availability of endogenous SPH and DMS to the S1R and the subsequent control of S1R activity within the same cell and/or in cellular environments.


Assuntos
Receptores sigma , Esfingosina , Animais , Esfingolipídeos , Ceramidas , Mamíferos/metabolismo , Receptor Sigma-1
3.
Proc Natl Acad Sci U S A ; 115(39): E9075-E9084, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30201718

RESUMO

DNA replication restart, the essential process that reinitiates prematurely terminated genome replication reactions, relies on exquisitely specific recognition of abandoned DNA replication-fork structures. The PriA DNA helicase mediates this process in bacteria through mechanisms that remain poorly defined. We report the crystal structure of a PriA/replication-fork complex, which resolves leading-strand duplex DNA bound to the protein. Interaction with PriA unpairs one end of the DNA and sequesters the 3'-most nucleotide from the nascent leading strand into a conserved protein pocket. Cross-linking studies reveal a surface on the winged-helix domain of PriA that binds to parental duplex DNA. Deleting the winged-helix domain alters PriA's structure-specific DNA unwinding properties and impairs its activity in vivo. Our observations lead to a model in which coordinated parental-, leading-, and lagging-strand DNA binding provide PriA with the structural specificity needed to act on abandoned DNA replication forks.


Assuntos
DNA Helicases/química , Replicação do DNA , DNA Bacteriano/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , Modelos Biológicos , Cristalografia por Raios X , Domínios Proteicos , Estrutura Secundária de Proteína , Relação Estrutura-Atividade
4.
J Biol Chem ; 293(6): 1994-2005, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29279331

RESUMO

Cellular metabolites act as important signaling cues, but are subject to complex unknown chemistry. Kynurenine is a tryptophan metabolite that plays a crucial role in cancer and the immune system. Despite its atypical, non-ligand-like, highly polar structure, kynurenine activates the aryl hydrocarbon receptor (AHR), a PER, ARNT, SIM (PAS) family transcription factor that responds to diverse environmental and cellular ligands. The activity of kynurenine is increased 100-1000-fold by incubation or long-term storage and relies on the hydrophobic ligand-binding pocket of AHR, with identical structural signatures for AHR induction before and after activation. We purified trace-active derivatives of kynurenine and identified two novel, closely related condensation products, named trace-extended aromatic condensation products (TEACOPs), which are active at low picomolar levels. The synthesized compound for one of the predicted structures matched the purified compound in both chemical structure and AHR pharmacology. Our study provides evidence that kynurenine acts as an AHR pro-ligand, which requires novel chemical conversions to act as a receptor agonist.


Assuntos
Cinurenina/química , Cinurenina/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Sítios de Ligação , Cinética , Ligantes , Camundongos , Estrutura Molecular , Receptores de Hidrocarboneto Arílico/química , Receptores de Hidrocarboneto Arílico/genética
5.
J Am Chem Soc ; 141(19): 7660-7664, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31045358

RESUMO

Phenol-soluble modulin α3 (PSMα3) is a cytotoxic peptide secreted by virulent strains of Staphylococcus aureus. We used a stereochemical strategy to examine the mechanism of PSMα3-mediated toxicity. One hypothesis is that PSMα3 toxicity requires fibril formation; an alternative is that toxicity is caused by soluble forms of PSMα3, possibly oligomeric. We find that the unnatural enantiomer (D residues) displays cytotoxicity comparable to that of L-PSMα3. Racemic PSMα3 is similarly toxic to enantiopure PSMα3 (L or D) under some conditions, but the toxicity is lost under conditions that cause racemic PSMα3 to aggregate. A crystal structure of racemic PSMα3-NH2 displays an α-helical secondary structure and a packing pattern that is reminiscent of the cross-α arrangement recently discovered in crystals of L-PSMα3. Our data suggest that the cytotoxicity of PSMα3 does not depend on stereospecific engagement of a target protein or other chiral macromolecule, an observation that supports a mechanism based on membrane disruption. In addition, our data support the hypothesis that toxicity is exerted by a soluble form rather than an insoluble fibrillar form.


Assuntos
Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidade , Células HEK293 , Humanos , Modelos Moleculares , Conformação Proteica , Estereoisomerismo , Relação Estrutura-Atividade
6.
Mol Cell ; 41(3): 331-42, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21292165

RESUMO

Proper formation of protein phosphatase 2A (PP2A) holoenzymes is essential for the fitness of all eukaryotic cells. Carboxyl methylation of the PP2A catalytic subunit plays a critical role in regulating holoenzyme assembly; methylation is catalyzed by PP2A-specific methyltransferase LCMT-1, an enzyme required for cell survival. We determined crystal structures of human LCMT-1 in isolation and in complex with PP2A stabilized by a cofactor mimic. The structures show that the LCMT-1 active-site pocket recognizes the carboxyl terminus of PP2A, and, interestingly, the PP2A active site makes extensive contacts to LCMT-1. We demonstrated that activation of the PP2A active site stimulates methylation, suggesting a mechanism for efficient conversion of activated PP2A into substrate-specific holoenzymes, thus minimizing unregulated phosphatase activity or formation of inactive holoenzymes. A dominant-negative LCMT-1 mutant attenuates the cell cycle without causing cell death, likely by inhibiting uncontrolled phosphatase activity. Our studies suggested mechanisms of LCMT-1 in tight control of PP2A function, important for the cell cycle and cell survival.


Assuntos
Proteína O-Metiltransferase/química , Proteína Fosfatase 2/química , Animais , Biocatálise , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Metilação , Modelos Moleculares , Mutação , Ligação Proteica , Proteína O-Metiltransferase/genética , Proteína O-Metiltransferase/metabolismo , Proteína Fosfatase 2/metabolismo , Estrutura Quaternária de Proteína , Ratos
7.
Mol Pharm ; 15(11): 5410-5426, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30285456

RESUMO

New tools and concepts are needed to combat antimicrobial resistance. Actinomycetes and firmicutes share several eukaryotic-like Ser/Thr kinases (eSTK) that offer antibiotic development opportunities, including PknB, an essential mycobacterial eSTK. Despite successful development of potent biochemical PknB inhibitors by many groups, clinically useful microbiologic activity has been elusive. Additionally, PknB kinetics are not fully described, nor are structures with specific inhibitors available to inform inhibitor design. We used computational modeling with available structural information to identify human kinase inhibitors predicted to bind PknB, and we selected hits based on drug-like characteristics intended to increase the likelihood of cell entry. The computational model suggested a family of inhibitors, the imidazopyridine aminofurazans (IPAs), bind PknB with high affinity. We performed an in-depth characterization of PknB and found that these inhibitors biochemically inhibit PknB, with potency roughly following the predicted models. A novel X-ray structure confirmed that the inhibitors bound as predicted and made favorable protein contacts with the target. These inhibitors also have antimicrobial activity toward mycobacteria and nocardia. We demonstrated that the inhibitors are uniquely potentiated by ß-lactams but not antibiotics traditionally used to treat mycobacteria, consistent with PknB's role in sensing cell wall stress. This is the first demonstration in the phylum actinobacteria that some ß-lactam antibiotics could be more effective if paired with a PknB inhibitor. Collectively, our data show that in silico modeling can be used as a tool to discover promising drug leads, and the inhibitors we discovered can act with clinically relevant antibiotics to restore their efficacy against bacteria with limited treatment options.


Assuntos
Descoberta de Drogas/métodos , Mycobacterium tuberculosis/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , beta-Lactamas/farmacologia , Cristalografia por Raios X , Sinergismo Farmacológico , Ensaios Enzimáticos , Concentração Inibidora 50 , Simulação de Acoplamento Molecular , Mycobacterium tuberculosis/metabolismo , Ligação Proteica , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/isolamento & purificação , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
9.
J Bacteriol ; 198(8): 1218-29, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26833410

RESUMO

UNLABELLED: Bacteriophytochrome photoreceptors (BphPs) and their cognate response regulators make up two-component signal transduction systems which direct bacteria to mount phenotypic responses to changes in environmental light quality. Most of these systems utilize single-domain response regulators to transduce signals through unknown pathways and mechanisms. Here we describe the photocycle and autophosphorylation kinetics of RtBphP1, a red light-regulated histidine kinase from the desert bacterium Ramlibacter tataouinensis RtBphP1 undergoes red to far-red photoconversion with rapid thermal reversion to the dark state. RtBphP1 is autophosphorylated in the dark; this activity is inhibited under red light. The RtBphP1 cognate response regulator, the R. tataouinensis bacteriophytochrome response regulator (RtBRR), and a homolog, AtBRR from Agrobacterium tumefaciens, crystallize unexpectedly as arm-in-arm dimers, reliant on a conserved hydrophobic motif, hFWAhL (where h is a hydrophobic M, V, L, or I residue). RtBRR and AtBRR dimerize distinctly from four structurally characterized phytochrome response regulators found in photosynthetic organisms and from all other receiver domain homodimers in the Protein Data Bank. A unique cacodylate-zinc-histidine tag metal organic framework yielded single-wavelength anomalous diffraction phases and may be of general interest. Examination of the effect of the BRR stoichiometry on signal transduction showed that phosphorylated RtBRR is accumulated more efficiently than the engineered monomeric RtBRR (RtBRRmon) in phosphotransfer reactions. Thus, we conclude that arm-in-arm dimers are a relevant signaling intermediate in this class of two-component regulatory systems. IMPORTANCE: BphP histidine kinases and their cognate response regulators comprise widespread red light-sensing two-component systems. Much work on BphPs has focused on structural understanding of light sensing and on enhancing the natural infrared fluorescence of these proteins, rather than on signal transduction or the resultant phenotypes. To begin to address this knowledge gap, we solved the crystal structures of two single-domain response regulators encoded by a region immediately downstream of that encoding BphPs. We observed a previously unknown arm-in-arm dimer linkage. Monomerization via deletion of the C-terminal dimerization motif had an inhibitory effect on net response regulator phosphorylation, underlining the importance of these unusual dimers for signal transduction.


Assuntos
Proteínas de Bactérias/metabolismo , Comamonadaceae/metabolismo , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Comamonadaceae/genética , Regulação Bacteriana da Expressão Gênica , Luz , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica
10.
PLoS Biol ; 11(11): e1001714, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24260024

RESUMO

Pentameric ligand-gated ion channels (pLGICs) are neurotransmitter-activated receptors that mediate fast synaptic transmission. In pLGICs, binding of agonist to the extracellular domain triggers a structural rearrangement that leads to the opening of an ion-conducting pore in the transmembrane domain and, in the continued presence of neurotransmitter, the channels desensitize (close). The flexible loops in each subunit that connect the extracellular binding domain (loops 2, 7, and 9) to the transmembrane channel domain (M2-M3 loop) are essential for coupling ligand binding to channel gating. Comparing the crystal structures of two bacterial pLGIC homologues, ELIC and the proton-activated GLIC, suggests channel gating is associated with rearrangements in these loops, but whether these motions accurately predict the motions in functional lipid-embedded pLGICs is unknown. Here, using site-directed spin labeling (SDSL) electron paramagnetic resonance (EPR) spectroscopy and functional GLIC channels reconstituted into liposomes, we examined if, and how far, the loops at the ECD/TMD gating interface move during proton-dependent gating transitions from the resting to desensitized state. Loop 9 moves ∼9 Šinward toward the channel lumen in response to proton-induced desensitization. Loop 9 motions were not observed when GLIC was in detergent micelles, suggesting detergent solubilization traps the protein in a nonactivatable state and lipids are required for functional gating transitions. Proton-induced desensitization immobilizes loop 2 with little change in position. Proton-induced motion of the M2-M3 loop was not observed, suggesting its conformation is nearly identical in closed and desensitized states. Our experimentally derived distance measurements of spin-labeled GLIC suggest ELIC is not a good model for the functional resting state of GLIC, and that the crystal structure of GLIC does not correspond to a desensitized state. These findings advance our understanding of the molecular mechanisms underlying pLGIC gating.


Assuntos
Proteínas de Bactérias/fisiologia , Cianobactérias , Ativação do Canal Iônico , Canais Iônicos de Abertura Ativada por Ligante/fisiologia , Substituição de Aminoácidos , Animais , Proteínas de Bactérias/química , Células Cultivadas , Espectroscopia de Ressonância de Spin Eletrônica , Canais Iônicos de Abertura Ativada por Ligante/química , Lipossomos/química , Potenciais da Membrana , Modelos Moleculares , Mutagênese Sítio-Dirigida , Técnicas de Patch-Clamp , Estrutura Quaternária de Proteína , Marcadores de Spin , Xenopus laevis
11.
J Am Chem Soc ; 137(37): 11884-7, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26369301

RESUMO

Quasiracemic crystallography has been used to explore the significance of homochiral and heterochiral associations in a set of host-defense peptide derivatives. The previously reported racemic crystal structure of a magainin 2 derivative displayed a homochiral antiparallel dimer association featuring a "phenylalanine zipper" notable for the dual roles of phenylalanines in mediating dimerization and formation of an exposed hydrophobic swath. This motif is seen as well in two new quasiracemate crystals that contain the d form of the magainin 2 derivative along with an l-peptide in which one Ala has been replaced by a ß-amino acid residue. This structural trend supports the hypothesis that the Phe zipper motif has functional significance.


Assuntos
Magaininas/química , Fenilalanina/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Cristalografia por Raios X , Modelos Moleculares , Dados de Sequência Molecular , Estereoisomerismo
12.
PLoS Pathog ; 9(7): e1003464, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23853587

RESUMO

Blastomyces adhesin-1 (BAD-1) is a 120-kD surface protein on B. dermatitidis yeast. We show here that BAD-1 contains 41 tandem repeats and that deleting even half of them impairs fungal pathogenicity. According to NMR, the repeats form tightly folded 17-amino acid loops constrained by a disulfide bond linking conserved cysteines. Each loop contains a highly conserved WxxWxxW motif found in thrombospondin-1 (TSP-1) type 1 heparin-binding repeats. BAD-1 binds heparin specifically and saturably, and is competitively inhibited by soluble heparin, but not related glycosaminoglycans. According to SPR analysis, the affinity of BAD-1 for heparin is 33 nM±14 nM. Putative heparin-binding motifs are found both at the N-terminus and within each tandem repeat loop. Like TSP-1, BAD-1 blocks activation of T cells in a manner requiring the heparan sulfate-modified surface molecule CD47, and impairs effector functions. The tandem repeats of BAD-1 thus confer pathogenicity, harbor motifs that bind heparin, and suppress T-cell activation via a CD47-dependent mechanism, mimicking mammalian TSP-1.


Assuntos
Antígenos de Fungos/metabolismo , Antígenos de Superfície/metabolismo , Blastomyces/imunologia , Proteínas Fúngicas/metabolismo , Heparina/metabolismo , Ativação Linfocitária , Linfócitos T/imunologia , Animais , Antígenos de Fungos/química , Antígenos de Fungos/genética , Antígenos de Superfície/química , Antígenos de Superfície/genética , Blastomyces/química , Blastomyces/metabolismo , Blastomyces/patogenicidade , Blastomicose/imunologia , Blastomicose/metabolismo , Blastomicose/microbiologia , Antígeno CD47/química , Antígeno CD47/genética , Antígeno CD47/metabolismo , Células Cultivadas , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Humanos , Células Jurkat , Masculino , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Linfócitos T/citologia , Linfócitos T/metabolismo , Sequências de Repetição em Tandem , Trombospondina 1/química , Trombospondina 1/metabolismo , Virulência
13.
Plant Cell ; 24(6): 2528-45, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22706284

RESUMO

Arbuscular mycorrhiza and the rhizobia-legume symbiosis are two major root endosymbioses that facilitate plant nutrition. In Lotus japonicus, two symbiotic cation channels, CASTOR and POLLUX, are indispensable for the induction of nuclear calcium spiking, one of the earliest plant responses to symbiotic partner recognition. During recent evolution, a single amino acid substitution in DOES NOT MAKE INFECTIONS1 (DMI1), the POLLUX putative ortholog in the closely related Medicago truncatula, rendered the channel solo sufficient for symbiosis; castor, pollux, and castor pollux double mutants of L. japonicus were rescued by DMI1 alone, while both Lj-CASTOR and Lj-POLLUX were required for rescuing a dmi1 mutant of M. truncatula. Experimental replacement of the critical serine by an alanine in the selectivity filter of Lj-POLLUX conferred a symbiotic performance indistinguishable from DMI1. Electrophysiological characterization of DMI1 and Lj-CASTOR (wild-type and mutants) by planar lipid bilayer experiments combined with calcium imaging in Human Embryonic Kidney-293 cells expressing DMI1 (the wild type and mutants) suggest that the serine-to-alanine substitution conferred reduced conductance with a long open state to DMI1 and improved its efficiency in mediating calcium oscillations. We propose that this single amino acid replacement in the selectivity filter made DMI1 solo sufficient for symbiosis, thus explaining the selective advantage of this allele at the mechanistic level.


Assuntos
Sinalização do Cálcio/fisiologia , Evolução Molecular , Canais Iônicos/metabolismo , Lotus/fisiologia , Medicago truncatula/fisiologia , Proteínas de Plantas/genética , Alanina/genética , Substituição de Aminoácidos , Evolução Biológica , Linhagem Celular , Fenômenos Eletrofisiológicos , Fabaceae/fisiologia , Teste de Complementação Genética , Humanos , Canais Iônicos/genética , Dados de Sequência Molecular , Mutação , Micorrizas/fisiologia , Filogenia , Proteínas de Plantas/metabolismo , Potássio/metabolismo , Serina/genética , Simbiose/fisiologia
14.
Biochemistry ; 53(18): 2956-65, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24730580

RESUMO

Indolethylamine-N-methyltransferase (INMT) is a Class 1 transmethylation enzyme known for its production of N,N-dimethyltryptamine (DMT), a hallucinogen with affinity for various serotonergic, adrenergic, histaminergic, dopaminergic, and sigma-1 receptors. DMT is produced via the action of INMT on the endogenous substrates tryptamine and S-adenosyl-l-methionine (SAM). The biological, biochemical, and selective small molecule regulation of INMT enzyme activity remain largely unknown. Kinetic mechanisms for inhibition of rabbit lung INMT (rabINMT) by the product, DMT, and by a new novel tryptamine derivative were determined. After Michaelis-Menten and Lineweaver-Burk analyses had been applied to study inhibition, DMT was found to be a mixed competitive and noncompetitive inhibitor when measured against tryptamine. The novel tryptamine derivative, N-[2-(1H-indol-3-yl)ethyl]-N',N'-dimethylpropane-1,3-diamine (propyl dimethyl amino tryptamine or PDAT), was shown to inhibit rabINMT by a pure noncompetitive mechanism when measured against tryptamine with a Ki of 84 µM. No inhibition by PDAT was observed at 2 mM when it was tested against structurally similar Class 1 methyltransferases, such as human phenylethanolamine-N-methyltransferase (hPNMT) and human nicotinamide-N-methyltransferase (hNNMT), indicating selectivity for INMT. The demonstration of noncompetitive mechanisms for INMT inhibition implies the presence of an inhibitory allosteric site. In silico analyses using the computer modeling software Autodock and the rabINMT sequence threaded onto the human INMT (hINMT) structure (Protein Data Bank entry 2A14 ) identified an N-terminal helix-loop-helix non-active site binding region of the enzyme. The energies for binding of DMT and PDAT to this region of rabINMT, as determined by Autodock, were -6.34 and -7.58 kcal/mol, respectively. Assessment of the allosteric control of INMT may illuminate new biochemical pathway(s) underlying the biology of INMT.


Assuntos
Metiltransferases/antagonistas & inibidores , N,N-Dimetiltriptamina/farmacologia , Triptaminas/farmacologia , Animais , Inibidores Enzimáticos/farmacologia , Cinética , Pulmão/enzimologia , Metiltransferases/química , Modelos Moleculares , Simulação de Acoplamento Molecular , Coelhos
15.
J Biol Chem ; 288(24): 17420-31, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23640880

RESUMO

General anesthetics exert many of their CNS actions by binding to and modulating membrane-embedded pentameric ligand-gated ion channels (pLGICs). The structural mechanisms underlying how anesthetics modulate pLGIC function remain largely unknown. GLIC, a prokaryotic pLGIC homologue, is inhibited by general anesthetics, suggesting anesthetics stabilize a closed channel state, but in anesthetic-bound GLIC crystal structures the channel appears open. Here, using functional GLIC channels expressed in oocytes, we examined whether propofol induces structural rearrangements in the GLIC transmembrane domain (TMD). Residues in the GLIC TMD that frame intrasubunit and intersubunit water-accessible cavities were individually mutated to cysteine. We measured and compared the rates of modification of the introduced cysteines by sulfhydryl-reactive reagents in the absence and presence of propofol. Propofol slowed the rate of modification of L240C (intersubunit) and increased the rate of modification of T254C (intrasubunit), indicating that propofol binding induces structural rearrangements in these cavities that alter the local environment near these residues. Propofol acceleration of T254C modification suggests that in the resting state propofol does not bind in the TMD intrasubunit cavity as observed in the crystal structure of GLIC with bound propofol (Nury, H., Van Renterghem, C., Weng, Y., Tran, A., Baaden, M., Dufresne, V., Changeux, J. P., Sonner, J. M., Delarue, M., and Corringer, P. J. (2011) Nature 469, 428-431). In silico docking using a GLIC closed channel homology model suggests propofol binds to intersubunit sites in the TMD in the resting state. Propofol-induced motions in the intersubunit cavity were distinct from motions associated with channel activation, indicating propofol stabilizes a novel closed state.


Assuntos
Anestésicos Intravenosos/metabolismo , Proteínas de Bactérias/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos de Abertura Ativada por Ligante/metabolismo , Propofol/metabolismo , Regulação Alostérica , Substituição de Aminoácidos , Anestésicos Intravenosos/farmacologia , Animais , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Cianobactérias , Cisteína/química , Cisteína/genética , Concentração de Íons de Hidrogênio , Cinética , Canais Iônicos de Abertura Ativada por Ligante/química , Canais Iônicos de Abertura Ativada por Ligante/genética , Potenciais da Membrana/efeitos dos fármacos , Metanossulfonato de Metila/análogos & derivados , Metanossulfonato de Metila/química , Simulação de Acoplamento Molecular , Mutagênese Sítio-Dirigida , Propofol/farmacologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Estrutura Secundária de Proteína , Subunidades Proteicas , Homologia Estrutural de Proteína , Xenopus laevis
16.
J Biol Chem ; 287(10): 7000-9, 2012 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-22210774

RESUMO

Phytochrome is a multidomain dimeric red light photoreceptor that utilizes a chromophore-binding domain (CBD), a PHY domain, and an output module to induce cellular changes in response to light. A promising biotechnology tool emerged when a structure-based substitution at Asp-207 was shown to be an infrared fluorophore that uses a biologically available tetrapyrrole chromophore. We report multiple crystal structures of this D207H variant of the Deinococcus radiodurans CBD, in which His-207 is observed to form a hydrogen bond with either the tetrapyrrole A-ring oxygen or the Tyr-263 hydroxyl. Based on the implications of this duality for fluorescence properties, Y263F was introduced and shown to have stronger fluorescence than the original D207H template. Our structures are consistent with the model that the Y263F change prevents a red light-induced far-red light absorbing phytochrome chromophore configuration. With the goal of decreasing size and thereby facilitating use as a fluorescent tag in vivo, we also engineered a monomeric form of the CBD. Unexpectedly, photoconversion was observed in the monomer despite the lack of a PHY domain. This observation underscores an interplay between dimerization and the photochemical properties of phytochrome and suggests that the monomeric CBD could be used for further studies of the photocycle. The D207H substitution on its own in the monomer did not result in fluorescence, whereas Y263F did. Combined, the D207H and Y263F substitutions in the monomeric CBD lead to the brightest of our variants, designated Wisconsin infrared phytofluor (Wi-Phy).


Assuntos
Substituição de Aminoácidos , Proteínas de Bactérias/química , Deinococcus/química , Fitocromo/química , Proteínas de Bactérias/genética , Cristalografia por Raios X , Deinococcus/genética , Fluorescência , Mutação de Sentido Incorreto , Fitocromo/genética , Estrutura Terciária de Proteína
17.
J Am Chem Soc ; 135(22): 8149-52, 2013 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-23701135

RESUMO

Cyclic constraints have proven to be very effective for preorganizing ß-amino acid residues and thereby stabilizing ß- and α/ß-peptide helices, but little is known about possible preorganization effects among γ residues. Here we assess and compare the impact of cyclic preorganization of ß and γ residues in the context of a specific α/ß/γ-peptide helix. The results show that ß residue preorganization is critical for helix stability but that γ residue preorganization is less important.


Assuntos
Peptídeos/química , Água/química , Cristalografia por Raios X , Modelos Moleculares , Estrutura Secundária de Proteína
18.
Proc Natl Acad Sci U S A ; 107(2): 633-8, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20018747

RESUMO

Bacterial single-stranded DNA-binding proteins (SSBs) help to recruit a diverse array of genome maintenance enzymes to their sites of action through direct protein interactions. For all cases examined to date, these interactions are mediated by the evolutionarily conserved C terminus of SSB (SSB-Ct). The essential nature of SSB protein interactions makes inhibitors that block SSB complex formation valuable biochemical tools and attractive potential antibacterial agents. Here, we identify four small molecules that disrupt complexes formed between Escherichia coli SSB and Exonuclease I (ExoI), a well-studied SSB-interacting enzyme. Each compound disrupts ExoI/SSB-Ct peptide complexes and abrogates SSB stimulation of ExoI nuclease activity. Structural and biochemical studies support a model for three of the compounds in which they compete with SSB for binding to ExoI. The fourth appears to rely on an allosteric mechanism to disrupt ExoI/SSB complexes. Subsets of the inhibitors block SSB-Ct complex formation with two other SSB-interaction partners as well, which highlights their utility as reagents for investigating the roles of SSB/protein interactions in diverse DNA replication, recombination, and repair reactions.


Assuntos
Proteínas de Ligação a DNA/genética , Exodesoxirribonucleases/genética , Proteínas de Bactérias/genética , Sítios de Ligação , Sequência Conservada , Cristalografia por Raios X , DNA de Cadeia Simples/genética , DNA de Cadeia Simples/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Exodesoxirribonucleases/antagonistas & inibidores , Exodesoxirribonucleases/química , Genoma Bacteriano , Cinética , Modelos Moleculares , Ligação Proteica , Conformação Proteica
19.
Proc Natl Acad Sci U S A ; 107(2): 559-64, 2010 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-20080719

RESUMO

Preorganization is shown to endow a protein with extraordinary conformational stability. This preorganization is achieved by installing side-chain substituents that impose stereoelectronic and steric effects that restrict main-chain torsion angles. Replacing proline residues in (ProProGly)(7) collagen strands with 4-fluoroproline and 4-methylproline leads to the most stable known triple helices, having T ( m ) values that are increased by > 50 degrees C. Differential scanning calorimetry data indicate an entropic basis to the hyperstability, as expected from an origin in preorganization. Structural data at a resolution of 1.21 A reveal a prototypical triple helix with insignificant deviations to its main chain, even though 2/3 of the residues are nonnatural. Thus, preorganization of a main chain by subtle changes to side chains can confer extraordinary conformational stability upon a protein without perturbing its structure.


Assuntos
Colágeno/química , Proteínas/química , Sequência de Aminoácidos , Cristalografia por Raios X , Estabilidade de Medicamentos , Eletrônica , Modelos Moleculares , Conformação Molecular , Prolina/análogos & derivados , Prolina/química , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Proteínas/metabolismo , Solubilidade , Estereoisomerismo
20.
Proc Natl Acad Sci U S A ; 107(47): 20275-80, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21057107

RESUMO

Most histone acetyltransferases (HATs) function as multisubunit complexes in which accessory proteins regulate substrate specificity and catalytic efficiency. Rtt109 is a particularly interesting example of a HAT whose specificity and catalytic activity require association with either of two histone chaperones, Vps75 or Asf1. Here, we utilize biochemical, structural, and genetic analyses to provide the detailed molecular mechanism for activation of a HAT (Rtt109) by its activating subunit Vps75. The rate-determining step of the activated complex is the transfer of the acetyl group from acetyl CoA to the acceptor lysine residue. Vps75 stimulates catalysis (> 250-fold), not by contributing a catalytic base, but by stabilizing the catalytically active conformation of Rtt109. To provide structural insight into the functional complex, we produced a molecular model of Rtt109-Vps75 based on X-ray diffraction of crystals of the complex. This model reveals distinct negative electrostatic surfaces on an Rtt109 molecule that interface with complementary electropositive ends of a symmetrical Vps75 dimer. Rtt109 variants with interface point substitutions lack the ability to be fully activated by Vps75, and one such variant displayed impaired Vps75-dependent histone acetylation functions in yeast, yet these variants showed no adverse effect on Asf1-dependent Rtt109 activities in vitro and in vivo. Finally, we provide evidence for a molecular model in which a 12 complex of Rtt109-Vps75 acetylates a heterodimer of H3-H4. The activation mechanism of Rtt109-Vps75 provides a valuable framework for understanding the molecular regulation of HATs within multisubunit complexes.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Western Blotting , Catálise , Cristalização , Dimerização , Eletroforese em Gel de Poliacrilamida , Espectrometria de Massas , Eletricidade Estática , Difração de Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA