Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Biochem ; 84: 923-46, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25784051

RESUMEN

Polysaccharide monooxygenases (PMOs), also known as lytic PMOs (LPMOs), enhance the depolymerization of recalcitrant polysaccharides by hydrolytic enzymes and are found in the majority of cellulolytic fungi and actinomycete bacteria. For more than a decade, PMOs were incorrectly annotated as family 61 glycoside hydrolases (GH61s) or family 33 carbohydrate-binding modules (CBM33s). PMOs have an unusual surface-exposed active site with a tightly bound Cu(II) ion that catalyzes the regioselective hydroxylation of crystalline cellulose, leading to glycosidic bond cleavage. The genomes of some cellulolytic fungi contain more than 20 genes encoding cellulose-active PMOs, suggesting a diversity of biological activities. PMOs show great promise in reducing the cost of conversion of lignocellulosic biomass to fermentable sugars; however, many questions remain about their reaction mechanism and biological function. This review addresses, in depth, the structural and mechanistic aspects of oxidative depolymerization of cellulose by PMOs and considers their biological function and phylogenetic diversity.


Asunto(s)
Celulosa/metabolismo , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/metabolismo , Bacterias/metabolismo , Hongos/enzimología , Hongos/metabolismo , Filogenia , Células Vegetales/química , Células Vegetales/metabolismo , Plantas/metabolismo , Polisacáridos/metabolismo
2.
Annu Rev Biochem ; 81: 533-59, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22404633

RESUMEN

Nitric oxide (NO) is an essential signaling molecule in biological systems. In mammals, the diatomic gas is critical to the cyclic guanosine monophosphate (cGMP) pathway as it functions as the primary activator of soluble guanylate cyclase (sGC). NO is synthesized from l-arginine and oxygen (O(2)) by the enzyme nitric oxide synthase (NOS). Once produced, NO rapidly diffuses across cell membranes and binds to the heme cofactor of sGC. sGC forms a stable complex with NO and carbon monoxide (CO), but not with O(2). The binding of NO to sGC leads to significant increases in cGMP levels. The second messenger then directly modulates phosphodiesterases (PDEs), ion-gated channels, or cGMP-dependent protein kinases to regulate physiological functions, including vasodilation, platelet aggregation, and neurotransmission. Many studies are focused on elucidating the molecular mechanism of sGC activation and deactivation with a goal of therapeutic intervention in diseases involving the NO/cGMP-signaling pathway. This review summarizes the current understanding of sGC structure and regulation as well as recent developments in NO signaling.


Asunto(s)
Guanilato Ciclasa/química , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos y Nucleares/química , Animales , GMP Cíclico/metabolismo , Guanilato Ciclasa/aislamiento & purificación , Guanilato Ciclasa/metabolismo , Humanos , Isoenzimas/metabolismo , Receptores Citoplasmáticos y Nucleares/aislamiento & purificación , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal , Guanilil Ciclasa Soluble
3.
Proc Natl Acad Sci U S A ; 120(8): e2215426120, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36791100

RESUMEN

Blast disease in cereal plants is caused by the fungus Magnaporthe oryzae and accounts for a significant loss in food crops. At the outset of infection, expression of a putative polysaccharide monooxygenase (MoPMO9A) is increased. MoPMO9A contains a catalytic domain predicted to act on cellulose and a carbohydrate-binding domain that binds chitin. A sequence similarity network of the MoPMO9A family AA9 showed that 220 of the 223 sequences in the MoPMO9A-containing cluster of sequences have a conserved unannotated region with no assigned function. Expression and purification of the full length and two MoPMO9A truncations, one containing the catalytic domain and the domain of unknown function (DUF) and one with only the catalytic domain, were carried out. In contrast to other AA9 polysaccharide monooxygenases (PMOs), MoPMO9A is not active on cellulose but showed activity on cereal-derived mixed (1→3, 1→4)-ß-D-glucans (MBG). Moreover, the DUF is required for activity. MoPMO9A exhibits activity consistent with C4 oxidation of the polysaccharide and can utilize either oxygen or hydrogen peroxide as a cosubstrate. It contains a predicted 3-dimensional fold characteristic of other PMOs. The DUF is predicted to form a coiled-coil with six absolutely conserved cysteines acting as a zipper between the two α-helices. MoPMO9A substrate specificity and domain architecture are different from previously characterized AA9 PMOs. The results, including a gene ontology analysis, support a role for MoPMO9A in MBG degradation during plant infection. Consistent with this analysis, deletion of MoPMO9A results in reduced pathogenicity.


Asunto(s)
Ascomicetos , Magnaporthe , Oryza , Oxigenasas de Función Mixta/metabolismo , Polisacáridos/metabolismo , Celulosa/metabolismo , Ascomicetos/metabolismo , Magnaporthe/genética , Enfermedades de las Plantas/microbiología , Proteínas Fúngicas/metabolismo , Oryza/metabolismo
4.
J Biol Chem ; 299(12): 105386, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37898401

RESUMEN

Aggregation behavior provides bacteria protection from harsh environments and threats to survival. Two uncharacterized proteases, LapX and Lap, are important for Vibrio cholerae liquid-based aggregation. Here, we determined that LapX is a serine protease with a preference for cleavage after glutamate and glutamine residues in the P1 position, which processes a physiologically based peptide substrate with a catalytic efficiency of 180 ± 80 M-1s-1. The activity with a LapX substrate identified by a multiplex substrate profiling by mass spectrometry screen was 590 ± 20 M-1s-1. Lap shares high sequence identity with an aminopeptidase (termed VpAP) from Vibrio proteolyticus and contains an inhibitory bacterial prepeptidase C-terminal domain that, when eliminated, increases catalytic efficiency on leucine p-nitroanilide nearly four-fold from 5.4 ± 4.1 × 104 M-1s-1 to 20.3 ± 4.3 × 104 M-1s-1. We demonstrate that LapX processes Lap to its mature form and thus amplifies Lap activity. The increase is approximately eighteen-fold for full-length Lap (95.7 ± 5.6 × 104 M-1s-1) and six-fold for Lap lacking the prepeptidase C-terminal domain (11.3 ± 1.9 × 105 M-1s-1). In addition, substrate profiling reveals preferences for these two proteases that could inform in vivo function. Furthermore, purified LapX and Lap restore the timing of the V. cholerae aggregation program to a mutant lacking the lapX and lap genes. Both proteases must be present to restore WT timing, and thus they appear to act sequentially: LapX acts on Lap, and Lap acts on the substrate involved in aggregation.


Asunto(s)
Proteínas Bacterianas , Leucil Aminopeptidasa , Serina Proteasas , Vibrio cholerae , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Leucil Aminopeptidasa/química , Leucil Aminopeptidasa/genética , Leucil Aminopeptidasa/fisiología , Péptidos , Serina Proteasas/química , Serina Proteasas/genética , Serina Proteasas/fisiología , Especificidad por Sustrato , Vibrio cholerae/enzimología , Vibrio cholerae/genética , Vibrio cholerae/fisiología , Catálisis
5.
Biochemistry ; 62(10): 1568-1576, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37129924

RESUMEN

Soluble guanylate cyclase (sGC) is the primary nitric oxide (NO) receptor in higher eukaryotes, including humans. NO-dependent signaling via sGC is associated with important physiological effects in the vascular, pulmonary, and neurological systems, and sGC itself is an established drug target for the treatment of pulmonary hypertension due to its central role in vasodilation. Despite isolation in the late 1970s, high-resolution structural information on full-length sGC remained elusive until recent cryo-electron microscopy structures were determined of the protein in both the basal unactivated state and the NO-activated state. These structures revealed large-scale conformational changes upon activation that appear to be centered on rearrangements within the coiled-coil (CC) domains in the enzyme. Here, a structure-guided approach was used to engineer constitutively unactivated and constitutively activated sGC variants through mutagenesis of the CC domains. These results demonstrate that the activation-induced conformational change in the CC domains is necessary and sufficient for determining the level of sGC activity.


Asunto(s)
Óxido Nítrico , Transducción de Señal , Humanos , Guanilil Ciclasa Soluble/metabolismo , Microscopía por Crioelectrón , Modelos Moleculares , Dominios Proteicos , Óxido Nítrico/metabolismo , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo
6.
Acc Chem Res ; 54(24): 4565-4575, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34890183

RESUMEN

Heme proteins have proven to be a convenient platform for the development of designer proteins with novel functionalities. This is achieved by substituting the native iron porphyrin cofactor with a heme analogue that possesses the desired properties. Replacing the iron center of the porphyrin with another metal provides one inroad to novel protein function. A less explored approach is substitution of the porphyrin cofactor with an alternative tetrapyrrole macrocycle or a related ligand. In general, these ligands exhibit chemical properties and reactivity that are distinct from those of porphyrins. While these techniques have most prominently been utilized to develop artificial metalloenzymes, there are many other applications of this methodology to problems in biochemistry, health, and medicine. Incorporation of synthetic cofactors into protein environments represents a facile way to impart water solubility and biocompatibility. It circumvents the laborious synthesis of water-soluble cofactors, which often introduces substantial charge that leads to undesired bioaccumulation. To this end, the incorporation of unnatural cofactors in heme proteins has enabled the development of designer proteins as optical oxygen sensors, MRI contrast agents, spectroscopic probes, tools to interrogate protein function, antibiotics, and fluorescent proteins.Incorporation of an artificial cofactor is frequently accomplished by denaturing the holoprotein with removal of the heme; the refolded apoprotein is then reconstituted with the artificial cofactor. This process often results in substantial protein loss and does not necessarily guarantee that the refolded protein adopts the native structure. To circumvent these issues, our laboratory has pioneered the use of the RP523 strain of E. coli to incorporate artificial cofactors into heme proteins using expression-based methods. This strain lacks the ability to biosynthesize heme, and the bacterial cell wall is permeable to heme and related molecules. In this way, heme analogues supplemented in the growth medium are incorporated into heme proteins. This approach can also be leveraged for the direct expression of the apoprotein for subsequent reconstitution.These methodologies have been exploited to incorporate non-native cofactors into heme proteins that are resistant to harsh environmental conditions: the heme nitric oxide/oxygen binding protein (H-NOX) from Caldanaerobacter subterraneus (Cs) and the heme acquisition system protein A (HasA) from Pseudomonas aeruginosa (Pa). The exceptional stability of these proteins makes them ideal scaffolds for biomedical applications. Optical oxygen sensing has been accomplished using a phosphorescent ruthenium porphyrin as the artificial heme cofactor. Paramagnetic manganese and gadolinium porphyrins yield high-relaxivity, protein-based MRI contrast agents. A fluorescent phosphorus corrole serves as a heme analogue to produce fluorescent proteins. Iron complexes of nonporphyrin cofactors bound to HasA inhibit the growth of pathogenic bacteria. Moreover, HasA can deliver a gallium phthalocyanine into the bacterial cytosol to serve as a sensitizer for photochemical sterilization. Together, these examples illustrate the potential for designer heme proteins to address burgeoning problems in the areas of health and medicine. The concepts and methodologies presented in this Account can be extended to the development of next-generation biomedical sensing and imaging agents to identify and quantify clinically relevant metabolites and other key disease biomarkers.


Asunto(s)
Hemoproteínas , Metaloproteínas , Escherichia coli , Hemo , Metales
7.
Inorg Chem ; 61(27): 10521-10532, 2022 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-35766625

RESUMEN

Ratiometric sensors are self-referencing constructs that are functional in cells and tissues, and the read-out is independent of sensor concentration. One strategy for ratiometric sensing is to utilize two-color emission, where one component possesses analyte-dependent emission and the other is independent of analyte concentration, serving as an internal standard. In this way, the intensity ratio of the two components is a quantitative measure of the analyte. In this study, protein-based ratiometric oxygen sensors are prepared using the heme nitric oxide/oxygen-binding protein (H-NOX) from the thermophilic bacterium Caldanaerobacter subterraneus. The native heme cofactor is replaced with a Pd(II) or Pt(II) porphyrin as the oxygen-responsive phosphor. Mutagenesis is performed to incorporate a cysteine residue on the protein surface for thiol/maleimide coupling of the oxygen-insensitive dye, which serves as a Förster resonance energy transfer (FRET) donor for the porphyrin. While both Pd(II)- and Pt(II)-based sensors are responsive over biologically relevant ranges, the Pd sensor exhibits greater sensitivity at lower oxygen concentrations. Together, these sensors represent a new class of protein-based ratiometric oxygen sensors, and the modular platform allows the oxygen sensitivity to be tailored for a specific application. This proof-of-principle study has identified the key considerations and optimal methodologies to develop and subsequently refine protein-based ratiometric oxygen sensors.


Asunto(s)
Hemoproteínas , Porfirinas , Transferencia Resonante de Energía de Fluorescencia , Hemo/metabolismo , Oxígeno/química , Porfirinas/química
8.
Biochemistry ; 60(46): 3491-3496, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34096266

RESUMEN

Nitric oxide (NO) has long been known to be an intermediate in bacterial pathways of denitrification. Only in the middle to late 1980s was it found to play a central role in a much broader biological context. For example, it is now well established that NO acts as a signaling agent in metazoans, including humans, yet NO is toxic and very reactive under biological conditions. How is the biology in which NO plays a role controlled? How is NO used and the inherent toxicity avoided? Looking back at the initial discovery time, to the present, and on to the future provides many answers to questions such as those listed above.


Asunto(s)
Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Aniones/metabolismo , Bacterias/metabolismo , Bioquímica/historia , Historia del Siglo XX , Humanos , Redes y Vías Metabólicas , Óxido Nítrico Sintasa/metabolismo , Guanilil Ciclasa Soluble/metabolismo
9.
Inorg Chem ; 60(4): 2716-2729, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33513009

RESUMEN

Although fluorescent proteins have been utilized for a variety of biological applications, they have several optical limitations, namely weak red and near-infrared emission and exceptionally broad (>200 nm) emission profiles. The photophysical properties of fluorescent proteins can be enhanced through the incorporation of novel cofactors with the desired properties into a stable protein scaffold. To this end, a fluorescent phosphorus corrole that is structurally similar to the native heme cofactor is incorporated into two exceptionally stable heme proteins: H-NOX from Caldanaerobacter subterraneus and heme acquisition system protein A (HasA) from Pseudomonas aeruginosa. These yellow-orange emitting protein conjugates are examined by steady-state and time-resolved optical spectroscopy. The HasA conjugate exhibits enhanced fluorescence, whereas emission from the H-NOX conjugate is quenched relative to the free corrole. Despite the low fluorescence quantum yields, these corrole-substituted proteins exhibit more intense fluorescence in a narrower spectral profile than traditional fluorescent proteins that emit in the same spectral window. This study demonstrates that fluorescent corrole complexes are readily incorporated into heme proteins and provides an inroad for the development of novel fluorescent proteins.


Asunto(s)
Hemoproteínas/química , Proteínas Luminiscentes/química , Porfirinas/química , Cristalografía por Rayos X
10.
Proc Natl Acad Sci U S A ; 115(19): 4915-4920, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29686097

RESUMEN

Enzymatic conversion of polysaccharides into lower-molecular-weight, soluble oligosaccharides is dependent on the action of hydrolytic and oxidative enzymes. Polysaccharide monooxygenases (PMOs) use an oxidative mechanism to break the glycosidic bond of polymeric carbohydrates, thereby disrupting the crystalline packing and creating new chain ends for hydrolases to depolymerize and degrade recalcitrant polysaccharides. PMOs contain a mononuclear Cu(II) center that is directly involved in C-H bond hydroxylation. Molecular oxygen was the accepted cosubstrate utilized by this family of enzymes until a recent report indicated reactivity was dependent on H2O2 Reported here is a detailed analysis of PMO reactivity with H2O2 and O2, in conjunction with high-resolution MS measurements. The cosubstrate utilized by the enzyme is dependent on the assay conditions. PMOs will directly reduce O2 in the coupled hydroxylation of substrate (monooxygenase activity) and will also utilize H2O2 (peroxygenase activity) produced from the uncoupled reduction of O2 Both cosubstrates require Cu reduction to Cu(I), but the reaction with H2O2 leads to nonspecific oxidation of the polysaccharide that is consistent with the generation of a hydroxyl radical-based mechanism in Fenton-like chemistry, while the O2 reaction leads to regioselective substrate oxidation using an enzyme-bound Cu/O2 reactive intermediate. Moreover, H2O2 does not influence the ability of secretome from Neurospora crassa to degrade Avicel, providing evidence that molecular oxygen is a physiologically relevant cosubstrate for PMOs.


Asunto(s)
Proteínas Fúngicas/química , Glicósido Hidrolasas/química , Peróxido de Hidrógeno/química , Oxigenasas de Función Mixta/química , Oxígeno/química , Sordariales/enzimología , Dominio Catalítico , Cobre/química
11.
Int J Mol Sci ; 22(11)2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34064029

RESUMEN

The enzyme soluble guanylate cyclase (sGC) is the prototypical nitric oxide (NO) receptor in humans and other higher eukaryotes and is responsible for transducing the initial NO signal to the secondary messenger cyclic guanosine monophosphate (cGMP). Generation of cGMP in turn leads to diverse physiological effects in the cardiopulmonary, vascular, and neurological systems. Given these important downstream effects, sGC has been biochemically characterized in great detail in the four decades since its discovery. Structures of full-length sGC, however, have proven elusive until very recently. In 2019, advances in single particle cryo-electron microscopy (cryo-EM) enabled visualization of full-length sGC for the first time. This review will summarize insights revealed by the structures of sGC in the unactivated and activated states and discuss their implications in the mechanism of sGC activation.


Asunto(s)
Guanilil Ciclasa Soluble/metabolismo , Animales , Microscopía por Crioelectrón/métodos , GMP Cíclico/metabolismo , Humanos , Óxido Nítrico/metabolismo , Transducción de Señal/fisiología
12.
J Biol Chem ; 294(32): 12157-12166, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31235519

RESUMEN

Degradation of polysaccharides is central to numerous biological and industrial processes. Starch-active polysaccharide monooxygenases (AA13 PMOs) oxidatively degrade starch and can potentially be used with industrial amylases to convert starch into a fermentable carbohydrate. The oxidative activities of the starch-active PMOs from the fungi Neurospora crassa and Myceliophthora thermophila, NcAA13 and MtAA13, respectively, on three different starch substrates are reported here. Using high-performance anion-exchange chromatography coupled with pulsed amperometry detection, we observed that both enzymes have significantly higher oxidative activity on amylose than on amylopectin and cornstarch. Analysis of the product distribution revealed that NcAA13 and MtAA13 more frequently oxidize glycosidic linkages separated by multiples of a helical turn consisting of six glucose units on the same amylose helix. Docking studies identified important residues that are involved in amylose binding and suggest that the shallow groove that spans the active-site surface of AA13 PMOs favors the binding of helical amylose substrates over nonhelical substrates. Truncations of NcAA13 that removed its native carbohydrate-binding module resulted in diminished binding to amylose, but truncated NcAA13 still favored amylose oxidation over other starch substrates. These findings establish that AA13 PMOs preferentially bind and oxidize the helical starch substrate amylose. Moreover, the product distributions of these two enzymes suggest a unique interaction with starch substrates.


Asunto(s)
Proteínas Fúngicas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Almidón/metabolismo , Amilosa/química , Amilosa/metabolismo , Sitios de Unión , Dominio Catalítico , Proteínas Fúngicas/química , Oxigenasas de Función Mixta/química , Simulación del Acoplamiento Molecular , Neurospora crassa/enzimología , Oxidación-Reducción , Conformación Proteica en Hélice alfa , Sordariales/enzimología , Almidón/química , Especificidad por Sustrato
13.
Mol Cell ; 46(4): 449-60, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22542454

RESUMEN

Nitric oxide (NO) signaling in vertebrates is well characterized and involves the heme-nitric oxide/oxygen-binding (H-NOX) domain of soluble guanylate cyclase as a selective NO sensor. In contrast, little is known about the biological role or signaling output of bacterial H-NOX proteins. Here, we describe a molecular pathway for H-NOX signaling in Shewanella oneidensis. NO stimulates biofilm formation by controlling the levels of the bacterial secondary messenger cyclic diguanosine monophosphate (c-di-GMP). Phosphotransfer profiling was used to map the connectivity of a multicomponent signaling network that involves integration from two histidine kinases and branching to three response regulators. A feed-forward loop between response regulators with phosphodiesterase domains and phosphorylation-mediated activation intricately regulated c-di-GMP levels. Phenotypic characterization established a link between NO signaling and biofilm formation. Cellular adhesion may provide a protection mechanism for bacteria against reactive and damaging NO. These results are broadly applicable to H-NOX-mediated NO signaling in bacteria.


Asunto(s)
Biopelículas/crecimiento & desarrollo , GMP Cíclico/análogos & derivados , Óxido Nítrico/metabolismo , Shewanella/fisiología , 3',5'-GMP Cíclico Fosfodiesterasas/química , 3',5'-GMP Cíclico Fosfodiesterasas/genética , 3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , GMP Cíclico/metabolismo , Gammaproteobacteria/genética , Gammaproteobacteria/fisiología , Guanilato Ciclasa/química , Guanilato Ciclasa/genética , Guanilato Ciclasa/metabolismo , Histidina Quinasa , Modelos Biológicos , Datos de Secuencia Molecular , Fosforilación , Proteínas Quinasas/química , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Sistemas de Mensajero Secundario , Homología de Secuencia de Aminoácido , Shewanella/genética , Transducción de Señal , Guanilil Ciclasa Soluble
14.
Biochemistry ; 58(17): 2250-2259, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-30946781

RESUMEN

Signaling pathways that involve diatomic gases in photosynthetic organisms are not well understood. Exposure to nitric oxide or carbon monoxide is known to elicit certain responses in some photosynthetic organisms. For example, Chlamydomonas reinhardtii grown in low-iron media responds to exogenous carbon monoxide by increasing cell growth and intracellular chlorophyll levels. Here, we characterize Cyg11, a gas-responsive soluble guanylate cyclase from the eukaryotic green alga C. reinhardtii that converts GTP to cGMP. Cyg11 transcription is upregulated when C. reinhardtii is grown in iron-limited media, suggesting its importance in nutrient-limited environments. Cyg11 is purified as a homodimer and is activated by nitric oxide (2.5-fold over basal activity) and carbon monoxide (6.3-fold). The heme binding stoichiometry of Cyg11 was found to be one heme per homodimer, an unexpected result based on the sequence and oligomerization state of the enzyme. Gas binding properties, the kinetics of gas binding, and the ligand-modulated activity of Cyg11 are consistent with CO as the relevant physiological ligand.


Asunto(s)
Proteínas Algáceas/metabolismo , Monóxido de Carbono/metabolismo , Chlamydomonas reinhardtii/enzimología , Guanilil Ciclasa Soluble/metabolismo , Proteínas Algáceas/química , Proteínas Algáceas/genética , Dióxido de Carbono/metabolismo , Chlamydomonas reinhardtii/genética , Hemo/química , Hemo/metabolismo , Cinética , Óxido Nítrico/metabolismo , Unión Proteica , Multimerización de Proteína , Transducción de Señal , Guanilil Ciclasa Soluble/química , Guanilil Ciclasa Soluble/genética , Regulación hacia Arriba
15.
J Biol Chem ; 293(17): 6282-6296, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29483187

RESUMEN

Cysteine S-nitrosation is a reversible post-translational modification mediated by nitric oxide (•NO)-derived agents. S-Nitrosation participates in cellular signaling and is associated with several diseases such as cancer, cardiovascular diseases, and neuronal disorders. Despite the physiological importance of this nonclassical •NO-signaling pathway, little is understood about how much S-nitrosation affects protein function. Moreover, identifying physiologically relevant targets of S-nitrosation is difficult because of the dynamics of transnitrosation and a limited understanding of the physiological mechanisms leading to selective protein S-nitrosation. To identify proteins whose activities are modulated by S-nitrosation, we performed a metabolomics study comparing WT and endothelial nitric-oxide synthase knockout mice. We integrated our results with those of a previous proteomics study that identified physiologically relevant S-nitrosated cysteines, and we found that the activity of at least 21 metabolic enzymes might be regulated by S-nitrosation. We cloned, expressed, and purified four of these enzymes and observed that S-nitrosation inhibits the metabolic enzymes 6-phosphogluconate dehydrogenase, Δ1-pyrroline-5-carboxylate dehydrogenase, catechol-O-methyltransferase, and d-3-phosphoglycerate dehydrogenase. Furthermore, using site-directed mutagenesis, we identified the predominant cysteine residue influencing the observed activity changes in each enzyme. In summary, using an integrated metabolomics approach, we have identified several physiologically relevant S-nitrosation targets, including metabolic enzymes, which are inhibited by this modification, and we have found the cysteines modified by S-nitrosation in each enzyme.


Asunto(s)
Metaboloma , Metabolómica , Óxido Nítrico/metabolismo , Oxidorreductasas/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Ratones , Ratones Noqueados , Nitrosación , Oxidorreductasas/genética
16.
Chembiochem ; 20(1): 7-19, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30320963

RESUMEN

Heme-nitric oxide/oxygen binding (H-NOX) proteins are a family of gas-binding hemoproteins that bind diatomic gas ligands such as nitric oxide (NO) and oxygen (O2 ). In bacteria, H-NOXs are often associated with signaling partners, including histidine kinases (HKs), diguanylate cyclases (DGCs) or methyl-accepting chemotaxis proteins (MCPs), either as a stand-alone protein or as a domain of a larger polypeptide. H-NOXs regulate the activity of cognate signaling proteins through ligand-induced conformational changes in the H-NOX domain and protein/protein interactions between the H-NOX and the cognate signaling partner. This review summarizes recent progress toward deciphering the molecular mechanism of bacterial H-NOX activation and the subsequent regulation of H-NOX-associated cognate sensor proteins from a structural and biochemical point of view.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hemoproteínas/metabolismo , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Bacterias/metabolismo , Proteínas de Escherichia coli/metabolismo , Histidina Quinasa/metabolismo , Liasas de Fósforo-Oxígeno/metabolismo , Unión Proteica , Dominios Proteicos , Transducción de Señal/fisiología
17.
Nat Chem Biol ; 13(12): 1216-1221, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28967923

RESUMEN

The binding of nitric oxide (NO) to the heme cofactor of heme-nitric oxide/oxygen binding (H-NOX) proteins can lead to the dissociation of the heme-ligating histidine residue and yield a five-coordinate nitrosyl complex, an important step for NO-dependent signaling. In the five-coordinate nitrosyl complex, NO can reside on either the distal or proximal side of the heme, which could have a profound influence over the lifetime of the in vivo signal. To investigate this central molecular question, we characterized the Shewanella oneidensis H-NOX (So H-NOX)-NO complex biophysically under limiting and excess NO conditions. The results show that So H-NOX preferably forms a distal NO species with both limiting and excess NO. Therefore, signal strength and complex lifetime in vivo will be dictated by the dissociation rate of NO from the distal complex and the rebinding of the histidine ligand to the heme.


Asunto(s)
Proteínas Bacterianas/metabolismo , Óxido Nítrico/metabolismo , Shewanella/metabolismo , Transducción de Señal , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Histidina Quinasa/antagonistas & inhibidores , Histidina Quinasa/metabolismo , Ligandos , Modelos Moleculares , Óxido Nítrico/química
18.
Biochemistry ; 57(22): 3191-3199, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29683313

RESUMEN

Polysaccharide monooxygenases (PMOs) are mononuclear copper enzymes that catalyze the hydroxylation of polysaccharides leading to the scission of the glycosidic bond. The mechanism, in which PMOs utilize molecular oxygen to oxidize the polysaccharide substrate, still remains largely unknown. Here, steady-state kinetics assays were used to probe the mechanism of oxygen-dependent cellohexaose oxidation catalyzed by MtPMO9E. Kinetic analysis indicated that both kcat/ KM(O2) and kcat/ KM(Glc6) were dependent on the concentration of the second substrate. Inhibition studies using carbon monoxide were also carried out. In addition, KD values for Glc6 were determined for the Cu(I) and Cu(II) forms of the enzyme. Taken together, PMOs follow a random-sequential kinetic mechanism to form a ternary ES-O2 complex. The optimal pH for MtPMO9E turnover was determined to be between pH 6.00 and pH 7.00. Furthermore, the kinetic parameters kcat, kcat/ KM(O2), and kcat/ KM(Glc6) demonstrate a decrease in PMO activity at a low pH and provide equivalent kinetic p Ka's of 5.10. This points to the protonation of a general base required for turnover. These results provide a basis for the initial chemical steps in the mechanism of PMOs.


Asunto(s)
Cobre/química , Oxigenasas de Función Mixta/química , Catálisis , Cobre/metabolismo , Concentración de Iones de Hidrógeno , Cinética , Oxigenasas de Función Mixta/metabolismo , Oxidación-Reducción , Oxígeno/química , Polisacáridos/química
19.
Biochemistry ; 57(47): 6570-6580, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30398342

RESUMEN

Nitric oxide (NO) is a critical signaling molecule involved in the regulation of a wide variety of physiological processes across every domain of life. In most aerobic and facultative anaerobic bacteria, heme-nitric oxide/oxygen binding (H-NOX) proteins selectively sense NO and inhibit the activity of a histidine kinase (HK) located on the same operon. This NO-dependent inhibition of the cognate HK alters the phosphorylation of the downstream response regulators. In the marine bacterium Saccharophagus degradans ( Sde), in addition to a typical H-NOX ( Sde 3804)/HK ( Sde 3803) pair, an orphan H-NOX ( Sde 3557) with no associated signaling protein has been identified distant from the H-NOX/HK pair in the genome. The characterization reported here elucidates the function of both H-NOX proteins. Sde 3557 exhibits a weaker binding affinity with the kinase, yet both Sde 3804 and Sde 3557 are functional H-NOXs with proper gas binding properties and kinase inhibition activity. Additionally, Sde 3557 has an NO dissociation rate that is significantly slower than that of Sde 3804, which may confer prolonged kinase inhibition in vivo. While it is still unclear whether Sde 3557 has another signaling partner or shares the histidine kinase with Sde 3804, Sde 3557 is the only orphan H-NOX characterized to date. S. degradans is likely using a dual-H-NOX system to fine-tune the downstream response of NO signaling.


Asunto(s)
Proteínas Bacterianas/metabolismo , Gammaproteobacteria/metabolismo , Hemoproteínas/metabolismo , Histidina Quinasa/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Proteínas Bacterianas/química , Hemoproteínas/química , Filogenia
20.
Biochemistry ; 57(11): 1779-1789, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29457883

RESUMEN

Heme-nitric oxide/oxygen binding (H-NOX) proteins are a group of hemoproteins that bind diatomic gas ligands such as nitric oxide (NO) and oxygen (O2). H-NOX proteins typically regulate histidine kinases (HK) located within the same operon. It has been reported that NO-bound H-NOXs inhibit cognate histidine kinase autophosphorylation in bacterial H-NOX/HK complexes; however, a detailed mechanism of NO-mediated regulation of the H-NOX/HK activity remains unknown. In this study, the binding interface of Vibrio cholerae ( Vc) H-NOX/HK complex was characterized by hydrogen/deuterium exchange mass spectrometry (HDX-MS) and further validated by mutagenesis, leading to a new model for NO-dependent kinase inhibition. A conformational change in Vc H-NOX introduced by NO generates a new kinase-binding interface, thus locking the kinase in an inhibitory conformation.


Asunto(s)
Proteínas Bacterianas/química , Hemoproteínas/química , Histidina Quinasa/química , Óxido Nítrico/química , Vibrio cholerae/química , Proteínas Bacterianas/metabolismo , Medición de Intercambio de Deuterio , Hemoproteínas/metabolismo , Histidina Quinasa/metabolismo , Espectrometría de Masas , Óxido Nítrico/metabolismo , Vibrio cholerae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA