Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 298(4): 101808, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35271852

RESUMO

Iron is an essential element for nearly all organisms, and under anoxic and/or reducing conditions, Fe2+ is the dominant form of iron available to bacteria. The ferrous iron transport (Feo) system is the primary prokaryotic Fe2+ import machinery, and two constituent proteins (FeoA and FeoB) are conserved across most bacterial species. However, how FeoA and FeoB function relative to one another remains enigmatic. In this work, we explored the distribution of feoAB operons encoding a fusion of FeoA tethered to the N-terminal, G-protein domain of FeoB via a connecting linker region. We hypothesized that this fusion poises FeoA to interact with FeoB to affect function. To test this hypothesis, we characterized the soluble NFeoAB fusion protein from Bacteroides fragilis, a commensal organism implicated in drug-resistant infections. Using X-ray crystallography, we determined the 1.50-Å resolution structure of BfFeoA, which adopts an SH3-like fold implicated in protein-protein interactions. Using a combination of structural modeling, small-angle X-ray scattering, and hydrogen-deuterium exchange mass spectrometry, we show that FeoA and NFeoB interact in a nucleotide-dependent manner, and we mapped the protein-protein interaction interface. Finally, using guanosine triphosphate (GTP) hydrolysis assays, we demonstrate that BfNFeoAB exhibits one of the slowest known rates of Feo-mediated GTP hydrolysis that is not potassium-stimulated. Importantly, truncation of FeoA from this fusion demonstrates that FeoA-NFeoB interactions function to stabilize the GTP-bound form of FeoB. Taken together, our work reveals a role for FeoA function in the fused FeoAB system and suggests a function for FeoA among prokaryotes.


Assuntos
Proteínas de Bactérias , Bacteroides fragilis , Proteínas de Transporte de Cátions , Proteínas de Ligação ao Ferro , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Bacteroides fragilis/genética , Bacteroides fragilis/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cristalografia por Raios X , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Hidrólise , Ferro/metabolismo , Proteínas de Ligação ao Ferro/química , Proteínas de Ligação ao Ferro/metabolismo , Estabilidade Proteica
2.
Microlife ; 3: uqac002, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37223350

RESUMO

The recent discovery that giant viruses encode proteins related to sugar synthesis and processing paved the way for the study of their glycosylation machinery. We focused on the proposed Megavirinae subfamily, for which glycan-related genes were proposed to code for proteins involved in glycosylation of the layer of fibrils surrounding their icosahedral capsids. We compared sugar compositions and corresponding biosynthetic pathways among clade members using a combination of chemical and bioinformatics approaches. We first demonstrated that Megavirinae glycosylation differs in many aspects from what was previously reported for viruses, as they have complex glycosylation gene clusters made of six and up to 33 genes to synthetize their fibril glycans (biosynthetic pathways for nucleotide-sugars and glycosyltransferases). Second, they synthesize rare amino-sugars, usually restricted to bacteria and absent from their eukaryotic host. Finally, we showed that Megavirinae glycosylation is clade-specific and that Moumouvirus australiensis, a B-clade outsider, shares key features with Cotonvirus japonicus (clade E) and Tupanviruses (clade D). The existence of a glycosylation toolbox in this family could represent an advantageous strategy to survive in an environment where members of the same family are competing for the same amoeba host. This study expands the field of viral glycobiology and raises questions on how Megavirinae evolved such versatile glycosylation machinery.

3.
J Inorg Biochem ; 214: 111267, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33099233

RESUMO

Nitric oxide (NO), carbon monoxide (CO), and oxygen (O2) are important physiological messengers whose concentrations vary in a remarkable range, [NO] typically from nM to several µM while [O2] reaching to hundreds of µM. One of the machineries evolved in living organisms for gas sensing is sensor hemoproteins whose conformational change upon gas binding triggers downstream response cascades. The recently proposed "sliding scale rule" hypothesis provides a general interpretation for gaseous ligand selectivity of hemoproteins, identifying five factors that govern gaseous ligand selectivity. Hemoproteins have intrinsic selectivity for the three gases due to a neutral proximal histidine ligand while proximal strain of heme and distal steric hindrance indiscriminately adjust the affinity of these three gases for heme. On the other hand, multiple-step NO binding and distal hydrogen bond donor(s) specifically enhance affinity for NO and O2, respectively. The "sliding scale rule" hypothesis provides clear interpretation for dramatic selectivity for NO over O2 in soluble guanylate cyclase (sGC) which is an important example of sensor hemoproteins and plays vital roles in a wide range of physiological functions. The "sliding scale rule" hypothesis has so far been validated by all experimental data and it may guide future designs for heme-based gas sensors.


Assuntos
Monóxido de Carbono/metabolismo , Hemeproteínas/metabolismo , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Guanilil Ciclase Solúvel/metabolismo , Monóxido de Carbono/química , Hemeproteínas/química , Óxido Nítrico/química , Oxigênio/química , Guanilil Ciclase Solúvel/química
4.
J Biol Chem ; 294(48): 18451-18464, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31645439

RESUMO

Soluble guanylyl cyclase (sGC) is the main receptor for nitric oxide (NO) and a central component of the NO-cGMP pathway, critical to cardiovascular function. NO binding to the N-terminal sensor domain in sGC enhances the cyclase activity of the C-terminal catalytic domain. Our understanding of the structural elements regulating this signaling cascade is limited, hindering structure-based drug design efforts that target sGC to improve the management of cardiovascular diseases. Conformational changes are thought to propagate the NO-binding signal throughout the entire sGC heterodimer, via its coiled-coil domain, to reorient the catalytic domain into an active conformation. To identify the structural elements involved in this signal transduction cascade, here we optimized a cGMP-based luciferase assay that reports on heterologous sGC activity in Escherichia coli and identified several mutations that activate sGC. These mutations resided in the dorsal flaps, dimer interface, and GTP-binding regions of the catalytic domain. Combinations of mutations from these different elements synergized, resulting in even greater activity and indicating a complex cross-talk among these regions. Molecular dynamics simulations further revealed conformational changes underlying the functional impact of these mutations. We propose that the interfacial residues play a central role in the sGC activation mechanism by coupling the coiled-coil domain to the active site via a series of hot spots. Our results provide new mechanistic insights not only into the molecular pathway for sGC activation but also for other members of the larger nucleotidyl cyclase family.


Assuntos
GMP Cíclico/metabolismo , Simulação de Dinâmica Molecular , Mutação , Óxido Nítrico/metabolismo , Guanilil Ciclase Solúvel/genética , Sequência de Aminoácidos , Animais , Domínio Catalítico , GMP Cíclico/química , Ativação Enzimática/genética , Humanos , Cinética , Óxido Nítrico/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Guanilil Ciclase Solúvel/química , Guanilil Ciclase Solúvel/metabolismo
5.
PLoS One ; 14(7): e0218897, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31276567

RESUMO

CD47 is an immune checkpoint molecule that downregulates key aspects of both the innate and adaptive anti-tumor immune response via its counter receptor SIRPα, and it is expressed at high levels in a wide variety of tumor types. This has led to the development of biologics that inhibit SIRPα engagement including humanized CD47 antibodies and a soluble SIRPα decoy receptor that are currently undergoing clinical trials. Unfortunately, toxicological issues, including anemia related to on-target mechanisms, are barriers to their clinical advancement. Another potential issue with large biologics that bind CD47 is perturbation of CD47 signaling through its high-affinity interaction with the matricellular protein thrombospondin-1 (TSP1). One approach to avoid these shortcomings is to identify and develop small molecule molecular probes and pretherapeutic agents that would (1) selectively target SIRPα or TSP1 interactions with CD47, (2) provide a route to optimize pharmacokinetics, reduce on-target toxicity and maximize tissue penetration, and (3) allow more flexible routes of administration. As the first step toward this goal, we report the development of an automated quantitative high-throughput screening (qHTS) assay platform capable of screening large diverse drug-like chemical libraries to discover novel small molecules that inhibit CD47-SIRPα interaction. Using time-resolved Förster resonance energy transfer (TR-FRET) and bead-based luminescent oxygen channeling assay formats (AlphaScreen), we developed biochemical assays, optimized their performance, and individually tested them in small-molecule library screening. Based on performance and low false positive rate, the LANCE TR-FRET assay was employed in a ~90,000 compound library qHTS, while the AlphaScreen oxygen channeling assay served as a cross-validation orthogonal assay for follow-up characterization. With this multi-assay strategy, we successfully eliminated compounds that interfered with the assays and identified five compounds that inhibit the CD47-SIRPα interaction; these compounds will be further characterized and later disclosed. Importantly, our results validate the large library qHTS for antagonists of CD47-SIRPα interaction and suggest broad applicability of this approach to screen chemical libraries for other protein-protein interaction modulators.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antígenos de Diferenciação/metabolismo , Antígeno CD47/metabolismo , Descoberta de Drogas/métodos , Ensaios de Triagem em Larga Escala/métodos , Receptores Imunológicos/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Antígenos de Diferenciação/química , Biotina/química , Biotina/metabolismo , Antígeno CD47/química , Antígeno CD47/imunologia , Humanos , Modelos Moleculares , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Receptores Imunológicos/química , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos
6.
Semin Cell Dev Biol ; 86: 162-173, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29574117

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) plays a key role in glycolysis but is also known for its involvement in a myriad of extra-glycolytic functions. While GAPDH is not the only enzyme with established moonlighting roles, it shows great diversity in terms of its functions, cellular localizations, protein partners, and post-translational modifications. This review focuses on GAPDH's role as a non-canonical RNA binding protein to regulate the stability and translation of cellular mRNAs. Despite the clear involvement of GAPDH in gene expression regulation, how and where GAPDH binds to its RNA targets is still unknown. In addition, the mechanism by which GAPDH switches among its various cellular functions is also unknown. This review will summarize our current understanding of GAPDH-mediated regulation of RNA function.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , RNA/genética , RNA/metabolismo , Sequência Rica em At , Gliceraldeído-3-Fosfato Desidrogenases/química , Humanos , Modelos Moleculares
7.
Nitric Oxide ; 77: 53-64, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29702251

RESUMO

Soluble guanylyl cyclase (GC-1) is the primary receptor of nitric oxide (NO) in smooth muscle cells and maintains vascular function by inducing vasorelaxation in nearby blood vessels. GC-1 converts guanosine 5'-triphosphate (GTP) into cyclic guanosine 3',5'-monophosphate (cGMP), which acts as a second messenger to improve blood flow. While much work has been done to characterize this pathway, we lack a mechanistic understanding of how NO binding to the heme domain leads to a large increase in activity at the C-terminal catalytic domain. Recent structural evidence and activity measurements from multiple groups have revealed a low-activity cyclase domain that requires additional GC-1 domains to promote a catalytically-competent conformation. How the catalytic domain structurally transitions into the active conformation requires further characterization. This review focuses on structure/function studies of the GC-1 catalytic domain and recent advances various groups have made in understanding how catalytic activity is regulated including small molecules interactions, Cys-S-NO modifications and potential interactions with the NO-sensor domain and other proteins.


Assuntos
Domínio Catalítico , Guanilil Ciclase Solúvel/química , Guanilil Ciclase Solúvel/metabolismo , Animais , Biocatálise , Humanos , Óxido Nítrico/metabolismo , Conformação Proteica , Relação Estrutura-Atividade
8.
Subcell Biochem ; 83: 413-453, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28271485

RESUMO

Aside from its well-established role in glycolysis, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has been shown to possess many key functions in cells. These functions are regulated by protein oligomerization , biomolecular interactions, post-translational modifications , and variations in subcellular localization . Several GAPDH functions and regulatory mechanisms overlap with one another and converge around its role in intermediary metabolism. Several structural determinants of the protein dictate its function and regulation. GAPDH is ubiquitously expressed and is found in all domains of life. GAPDH has been implicated in many diseases, including those of pathogenic, cardiovascular, degenerative, diabetic, and tumorigenic origins. Understanding the mechanisms by which GAPDH can switch between its functions and how these functions are regulated can provide insights into ways the protein can be modulated for therapeutic outcomes.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/química , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Animais , Glicólise , Humanos , Processamento de Proteína Pós-Traducional
9.
Wiley Interdiscip Rev RNA ; 7(1): 53-70, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26564736

RESUMO

The glycolytic protein, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), has a vast array of extraglycolytic cellular functions, including interactions with nucleic acids. GAPDH has been implicated in the translocation of transfer RNA (tRNA), the regulation of cellular messenger RNA (mRNA) stability and translation, as well as the regulation of replication and gene expression of many single-stranded RNA viruses. A growing body of evidence supports GAPDH-RNA interactions serving as part of a larger coordination between intermediary metabolism and RNA biogenesis. Despite the established role of GAPDH in nucleic acid regulation, it is still unclear how and where GAPDH binds to its RNA targets, highlighted by the absence of any conserved RNA-binding sequences. This review will summarize our current understanding of GAPDH-mediated regulation of RNA function. WIREs RNA 2016, 7:53-70. doi: 10.1002/wrna.1315 For further resources related to this article, please visit the WIREs website.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Proteínas de Ligação a RNA/metabolismo , RNA/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/química , Humanos , Ligação Proteica , Conformação Proteica
10.
J Biol Chem ; 290(35): 21615-28, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26134567

RESUMO

Heat shock protein 90 (hsp90) drives heme insertion into the ß1 subunit of soluble guanylate cyclase (sGC) ß1, which enables it to associate with a partner sGCα1 subunit and mature into a nitric oxide (NO)-responsive active form. We utilized fluorescence polarization measurements and hydrogen-deuterium exchange mass spectrometry to define molecular interactions between the specific human isoforms hsp90ß and apo-sGCß1. hsp90ß and its isolated M domain, but not its isolated N and C domains, bind with low micromolar affinity to a heme-free, truncated version of sGCß1 (sGCß1(1-359)-H105F). Surprisingly, hsp90ß and its M domain bound to the Per-Arnt-Sim (PAS) domain of apo-sGC-ß1(1-359), which lies adjacent to its heme-binding (H-NOX) domain. The interaction specifically involved solvent-exposed regions in the hsp90ß M domain that are largely distinct from sites utilized by other hsp90 clients. The interaction strongly protected two regions of the sGCß1 PAS domain and caused local structural relaxation in other regions, including a PAS dimerization interface and a segment in the H-NOX domain. Our results suggest a means by which the hsp90ß interaction could prevent apo-sGCß1 from associating with its partner sGCα1 subunit while enabling structural changes to assist heme insertion into the H-NOX domain. This mechanism would parallel that in other clients like the aryl hydrocarbon receptor and HIF1α, which also interact with hsp90 through their PAS domains to control protein partner and small ligand binding interactions.


Assuntos
Guanilato Ciclase/química , Guanilato Ciclase/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Medição da Troca de Deutério , Proteínas de Choque Térmico HSP90/química , Heme/metabolismo , Humanos , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Guanilil Ciclase Solúvel
11.
Sci Rep ; 5: 12085, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-26160052

RESUMO

Characterizing biomolecular interactions is crucial to the understanding of biological processes. Existing characterization methods have low spatial resolution, poor specificity, and some lack the capability for deep tissue imaging. We describe a novel technique that relies on small-angle X-ray scattering signatures from high-contrast molecular probes that correlate with the presence of biomolecular interactions. We describe a proof-of-concept study that uses a model system consisting of mixtures of monomer solutions of gold nanoparticles (GNPs) as the non-interacting species and solutions of GNP dimers linked with an organic molecule (dimethyl suberimidate) as the interacting species. We report estimates of the interaction fraction obtained with the proposed small-angle X-ray scattering characterization method exhibiting strong correlation with the known relative concentration of interacting and non-interacting species.


Assuntos
Nanopartículas Metálicas/química , Ouro/química , Modelos Teóricos , Espalhamento a Baixo Ângulo , Soluções/química , Difração de Raios X/métodos , Raios X
13.
J Biol Chem ; 290(3): 1770-85, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25451934

RESUMO

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is an enzyme best known for its role in glycolysis. However, extra-glycolytic functions of GAPDH have been described, including regulation of protein expression via RNA binding. GAPDH binds to numerous adenine-uridine rich elements (AREs) from various mRNA 3'-untranslated regions in vitro and in vivo despite its lack of a canonical RNA binding motif. How GAPDH binds to these AREs is still unknown. Here we discovered that GAPDH binds with high affinity to the core ARE from tumor necrosis factor-α mRNA via a two-step binding mechanism. We demonstrate that a mutation at the GAPDH dimer interface impairs formation of the second RNA-GAPDH complex and leads to changes in the RNA structure. We investigated the effect of this interfacial mutation on GAPDH oligomerization by crystallography, small-angle x-ray scattering, nano-electrospray ionization native mass spectrometry, and hydrogen-deuterium exchange mass spectrometry. We show that the mutation does not significantly affect GAPDH tetramerization as previously proposed. Instead, the mutation promotes short-range and long-range dynamic changes in regions located at the dimer and tetramer interface and in the NAD(+) binding site. These dynamic changes are localized along the P axis of the GAPDH tetramer, suggesting that this region is important for RNA binding. Based on our results, we propose a model for sequential GAPDH binding to RNA via residues located at the dimer and tetramer interfaces.


Assuntos
Gliceraldeído-3-Fosfato Desidrogenases/química , RNA/química , Regiões 3' não Traduzidas , Motivos de Aminoácidos , Sequência de Aminoácidos , Anisotropia , Sítios de Ligação , DNA Complementar/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/genética , Glicólise , Humanos , Microscopia de Fluorescência , Dados de Sequência Molecular , Mutação , Peptídeos/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Espalhamento de Radiação , Homologia de Sequência de Aminoácidos , Espectrometria de Massas por Ionização por Electrospray , Fator de Necrose Tumoral alfa/metabolismo , Raios X
14.
Front Physiol ; 5: 134, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24772092

RESUMO

Nitric oxide (NO) maintains cardiovascular health by activating soluble guanylate cyclase (sGC) to increase cellular cGMP levels. Cardiovascular disease is characterized by decreased NO-sGC-cGMP signaling. Pharmacological activators and stimulators of sGC are being actively pursued as therapies for acute heart failure and pulmonary hypertension. Here we review molecular mechanisms that modulate sGC activity while emphasizing a novel biochemical pathway in which binding of the matricellular protein thrombospondin-1 (TSP1) to the cell surface receptor CD47 causes inhibition of sGC. We discuss the therapeutic implications of this pathway for blood flow, tissue perfusion, and cell survival under physiologic and disease conditions.

15.
Biochemistry ; 53(13): 2153-65, 2014 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-24669844

RESUMO

Soluble guanylate cyclase (sGC) plays a central role in the cardiovascular system and is a drug target for the treatment of pulmonary hypertension. While the three-dimensional structure of sGC is unknown, studies suggest that binding of the regulatory domain to the catalytic domain maintains sGC in an autoinhibited basal state. The activation signal, binding of NO to heme, is thought to be transmitted via the regulatory and dimerization domains to the cyclase domain and unleashes the full catalytic potential of sGC. Consequently, isolated catalytic domains should show catalytic turnover comparable to that of activated sGC. Using X-ray crystallography, activity measurements, and native mass spectrometry, we show unambiguously that human isolated catalytic domains are much less active than basal sGC, while still forming heterodimers. We identified key structural elements regulating the dimer interface and propose a novel role for residues located in an interfacial flap and a hydrogen bond network as key modulators of the orientation of the catalytic subunits. We demonstrate that even in the absence of the regulatory domain, additional sGC domains are required to guide the appropriate conformation of the catalytic subunits associated with high activity. Our data support a novel regulatory mechanism whereby sGC activity is tuned by distinct domain interactions that either promote or inhibit catalytic activity. These results further our understanding of heterodimerization and activation of sGC and open additional drug discovery routes for targeting the NO-sGC-cGMP pathway via the design of small molecules that promote a productive conformation of the catalytic subunits or disrupt inhibitory domain interactions.


Assuntos
Biocatálise , Domínio Catalítico , Guanilato Ciclase/química , Guanilato Ciclase/metabolismo , Multimerização Proteica , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Cristalografia por Raios X , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/genética , Humanos , Ligação de Hidrogênio , Espectrometria de Massas , Modelos Moleculares , Dados de Sequência Molecular , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/genética , Guanilil Ciclase Solúvel
16.
Biochemistry ; 53(1): 101-14, 2014 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-24328155

RESUMO

Soluble guanylate cyclase (sGC) is a heterodimeric heme protein and the primary nitric oxide receptor. NO binding stimulates cyclase activity, leading to regulation of cardiovascular physiology and making sGC an attractive target for drug discovery. YC-1 and related compounds stimulate sGC both independently and synergistically with NO and CO binding; however, where the compounds bind and how they work remain unknown. Using linked equilibrium binding measurements, surface plasmon resonance, and domain truncations in Manduca sexta and bovine sGC, we demonstrate that YC-1 binds near or directly to the heme-containing domain of the ß subunit. In the absence of CO, YC-1 binds with a Kd of 9-21 µM, depending on the construct. In the presence of CO, these values decrease to 0.6-1.1 µM. Pfizer compound 25 bound ∼10-fold weaker than YC-1 in the absence of CO, whereas compound BAY 41-2272 bound particularly tightly in the presence of CO (Kd = 30-90 nM). Additionally, we found that CO binds much more weakly to heterodimeric sGC proteins (Kd = 50-100 µM) than to the isolated heme domain (Kd = 0.2 µM for Manduca ß H-NOX/PAS). YC-1 greatly enhanced binding of CO to heterodimeric sGC, as expected (Kd ∼ 1 µM). These data indicate the α subunit induces a heme pocket conformation with a lower affinity for CO and NO. YC-1 family compounds bind near the heme domain, overcoming the α subunit effect and inducing a heme pocket conformation with high affinity. We propose this high-affinity conformation is required for the full-length protein to achieve high catalytic activity.


Assuntos
Guanilato Ciclase/metabolismo , Indazóis/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Regulação Alostérica/efeitos dos fármacos , Animais , Monóxido de Carbono/química , Bovinos , Heme/química , Manduca/enzimologia , Modelos Moleculares , Óxido Nítrico/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Guanilil Ciclase Solúvel , Ressonância de Plasmônio de Superfície
17.
J Biol Chem ; 285(5): 3064-75, 2010 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-19948738

RESUMO

Neuronal nitric-oxide synthase (nNOS) contains a unique autoinhibitory insert (AI) in its FMN subdomain that represses nNOS reductase activities and controls the calcium sensitivity of calmodulin (CaM) binding to nNOS. How the AI does this is unclear. A conserved charged residue (Lys(842)) lies within a putative CaM binding helix in the middle of the AI. We investigated its role by substituting residues that neutralize (Ala) or reverse (Glu) the charge at Lys(842). Compared with wild type nNOS, the mutant enzymes had greater cytochrome c reductase and NADPH oxidase activities in the CaM-free state, were able to bind CaM at lower calcium concentration, and had lower rates of heme reduction and NO synthesis in one case (K842A). Moreover, stopped-flow spectrophotometric experiments with the nNOS reductase domain indicate that the CaM-free mutants had faster flavin reduction kinetics and had less shielding of their FMN subdomains compared with wild type and no longer increased their level of FMN shielding in response to NADPH binding. Thus, Lys(842) is critical for the known functions of the AI and also enables two additional functions of the AI as newly identified here: suppression of electron transfer to FMN and control of the conformational equilibrium of the nNOS reductase domain. Its effect on the conformational equilibrium probably explains suppression of catalysis by the AI.


Assuntos
Calmodulina/química , Flavinas/química , Lisina/química , Neurônios/enzimologia , Óxido Nítrico Sintase/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Redutases do Citocromo/química , Humanos , Cinética , Dados de Sequência Molecular , Mutação , NADPH Oxidases/química , Ligação Proteica , Estrutura Terciária de Proteína , Ratos , Homologia de Sequência de Aminoácidos
18.
Nat Chem Biol ; 4(11): 700-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18849972

RESUMO

Nitric oxide synthase (NOS) enzymes synthesize nitric oxide, a signal for vasodilatation and neurotransmission at low concentrations and a defensive cytotoxin at higher concentrations. The high active site conservation among all three NOS isozymes hinders the design of selective NOS inhibitors to treat inflammation, arthritis, stroke, septic shock and cancer. Our crystal structures and mutagenesis results identified an isozyme-specific induced-fit binding mode linking a cascade of conformational changes to a new specificity pocket. Plasticity of an isozyme-specific triad of distant second- and third-shell residues modulates conformational changes of invariant first-shell residues to determine inhibitor selectivity. To design potent and selective NOS inhibitors, we developed the anchored plasticity approach: anchor an inhibitor core in a conserved binding pocket, then extend rigid bulky substituents toward remote specificity pockets, which become accessible upon conformational changes of flexible residues. This approach exemplifies general principles for the design of selective enzyme inhibitors that overcome strong active site conservation.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Óxido Nítrico Sintase/antagonistas & inibidores , Sequência de Aminoácidos , Aminopiridinas/química , Aminopiridinas/farmacologia , Animais , Bovinos , Cristalografia por Raios X , Modelos Animais de Doenças , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Humanos , Isoenzimas/antagonistas & inibidores , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Mutação , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Quinazolinas/química , Quinazolinas/farmacologia , Ratos
19.
Nat Struct Mol Biol ; 15(5): 515-22, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18408731

RESUMO

Escherichia coli endonuclease IV is an archetype for an abasic or apurinic-apyrimidinic endonuclease superfamily crucial for DNA base excision repair. Here biochemical, mutational and crystallographic characterizations reveal a three-metal ion mechanism for damage binding and incision. The 1.10-A resolution DNA-free and the 2.45-A resolution DNA-substrate complex structures capture substrate stabilization by Arg37 and reveal a distorted Zn3-ligand arrangement that reverts, after catalysis, to an ideal geometry suitable to hold rather than release cleaved DNA product. The 1.45-A resolution DNA-product complex structure shows how Tyr72 caps the active site, tunes its dielectric environment and promotes catalysis by Glu261-activated hydroxide, bound to two Zn2+ ions throughout catalysis. These structural, mutagenesis and biochemical results suggest general requirements for abasic site removal in contrast to features specific to the distinct endonuclease IV alpha-beta triose phosphate isomerase (TIM) barrel and APE1 four-layer alpha-beta folds of the apurinic-apyrimidinic endonuclease families.


Assuntos
Desoxirribonuclease IV (Fago T4-Induzido)/metabolismo , Escherichia coli/enzimologia , Sítios de Ligação , Cristalografia por Raios X , DNA/metabolismo , Desoxirribonuclease IV (Fago T4-Induzido)/química , Desoxirribonuclease IV (Fago T4-Induzido)/genética , Interações Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Estrutura Secundária de Proteína
20.
J Biol Chem ; 280(47): 39208-19, 2005 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-16150731

RESUMO

The neuronal nitric-oxide synthase (nNOS) flavoprotein domain (nNOSr) contains regulatory elements that repress its electron flux in the absence of bound calmodulin (CaM). The repression also requires bound NADP(H), but the mechanism is unclear. The crystal structure of a CaM-free nNOSr revealed an ionic interaction between Arg(1400) in the C-terminal tail regulatory element and the 2'-phosphate group of bound NADP(H). We tested the role of this interaction by substituting Ser and Glu for Arg(1400) in nNOSr and in the full-length nNOS enzyme. The CaM-free nNOSr mutants had cytochrome c reductase activities that were less repressed than in wild-type, and this effect could be mimicked in wild-type by using NADH instead of NADPH. The nNOSr mutants also had faster flavin reduction rates, greater apparent K(m) for NADPH, and greater rates of flavin auto-oxidation. Single-turnover cytochrome c reduction data linked these properties to an inability of NADP(H) to cause shielding of the FMN module in the CaM-free nNOSr mutants. The full-length nNOS mutants had no NO synthesis in the CaM-free state and had lower steady-state NO synthesis activities in the CaM-bound state compared with wild-type. However, the mutants had faster rates of ferric heme reduction and ferrous heme-NO complex formation. Slowing down heme reduction in R1400E nNOS with CaM analogues brought its NO synthesis activity back up to normal level. Our studies indicate that the Arg(1400)-2'-phosphate interaction is a means by which bound NADP(H) represses electron transfer into and out of CaM-free nNOSr. This interaction enables the C-terminal tail to regulate a conformational equilibrium of the FMN module that controls its electron transfer reactions in both the CaM-free and CaM-bound forms of nNOS.


Assuntos
NADP/metabolismo , Óxido Nítrico Sintase Tipo I/química , Óxido Nítrico Sintase Tipo I/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Arginina/química , Sequência de Bases , Calmodulina/metabolismo , DNA Complementar/genética , Transporte de Elétrons , Heme/metabolismo , Humanos , Técnicas In Vitro , Cinética , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo I/genética , Oxirredução , Conformação Proteica , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...