Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 975
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(5): 107149, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38479599

RESUMEN

Persulfides (RSSH/RSS-) participate in sulfur metabolism and are proposed to transduce hydrogen sulfide (H2S) signaling. Their biochemical properties are poorly understood. Herein, we studied the acidity and nucleophilicity of several low molecular weight persulfides using the alkylating agent, monobromobimane. The different persulfides presented similar pKa values (4.6-6.3) and pH-independent rate constants (3.2-9.0 × 103 M-1 s-1), indicating that the substituents in persulfides affect properties to a lesser extent than in thiols because of the larger distance to the outer sulfur. The persulfides had higher reactivity with monobromobimane than analogous thiols and putative thiols with the same pKa, providing evidence for the alpha effect (enhanced nucleophilicity by the presence of a contiguous atom with high electron density). Additionally, we investigated two enzymes from the human mitochondrial H2S oxidation pathway that form catalytic persulfide intermediates, sulfide quinone oxidoreductase and thiosulfate sulfurtransferase (TST, rhodanese). The pH dependence of the activities of both enzymes was measured using sulfite and/or cyanide as sulfur acceptors. The TST half-reactions were also studied by stopped-flow fluorescence spectroscopy. Both persulfidated enzymes relied on protonated groups for reaction with the acceptors. Persulfidated sulfide quinone oxidoreductase appeared to have a pKa of 7.8 ± 0.2. Persulfidated TST presented a pKa of 9.38 ± 0.04, probably due to a critical active site residue rather than the persulfide itself. The TST thiol reacted in the anionic state with thiosulfate, with an apparent pKa of 6.5 ± 0.1. Overall, our study contributes to a fundamental understanding of persulfide properties and their modulation by protein environments.


Asunto(s)
Sulfuros , Tiosulfato Azufretransferasa , Humanos , Compuestos Bicíclicos con Puentes , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/química , Concentración de Iones de Hidrógeno , Oxidación-Reducción , Quinona Reductasas/metabolismo , Quinona Reductasas/química , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/metabolismo , Sulfuros/química , Sulfuros/metabolismo , Tiosulfato Azufretransferasa/metabolismo , Tiosulfato Azufretransferasa/química , Quinonas/química , Quinonas/metabolismo , Especificidad por Sustrato
2.
Immunity ; 45(4): 761-773, 2016 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-27692612

RESUMEN

Imiquimod is a small-molecule ligand of Toll-like receptor-7 (TLR7) that is licensed for the treatment of viral infections and cancers of the skin. Imiquimod has TLR7-independent activities that are mechanistically unexplained, including NLRP3 inflammasome activation in myeloid cells and apoptosis induction in cancer cells. We investigated the mechanism of inflammasome activation by imiquimod and the related molecule CL097 and determined that K+ efflux was dispensable for NLRP3 activation by these compounds. Imiquimod and CL097 inhibited the quinone oxidoreductases NQO2 and mitochondrial Complex I. This induced a burst of reactive oxygen species (ROS) and thiol oxidation, and led to NLRP3 activation via NEK7, a recently identified component of this inflammasome. Metabolic consequences of Complex I inhibition and endolysosomal effects of imiquimod might also contribute to NLRP3 activation. Our results reveal a K+ efflux-independent mechanism for NLRP3 activation and identify targets of imiquimod that might be clinically relevant.


Asunto(s)
Inflamasomas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Potasio/metabolismo , ARN Nuclear Pequeño/farmacología , Animales , Complejo I de Transporte de Electrón/metabolismo , Ratones , Quinasas Relacionadas con NIMA/metabolismo , Quinona Reductasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 7/metabolismo
3.
Biochem Biophys Res Commun ; 690: 149096, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37988924

RESUMEN

Electron-driven process helps the living organism in the generations of energy, biomass production and detoxification of synthetic compounds. Soluble quinone oxidoreductases (QORs) mediate the transfer of an electron from NADPH to various quinone and other compounds, helping in the detoxification of quinones. QORs play a crucial role in cellular metabolism and are thus potential targets for drug development. Here we report the crystal structure of the NADPH-dependent QOR from Leishmania donovani (LdQOR) at 2.05 Å. The enzyme exists as a homo-dimer, with each protomer consisting of two domains, responsible for binding NADPH cofactor and the substrate. Interestingly, the human QOR exists as a tetramer. Comparative analysis of the oligomeric interfaces of LdQOR with HsQOR shows no significant differences in the protomer/dimer assembly. The tetrameric interface of HsQOR is stabilized by salt bridges formed between Arg 169 and Glu 271 which is non-existent in LdQOR, with an Alanine replacing the glutamate. This distinct feature is conserved across other dimeric QORs, indicating the importance of this interaction for tetramer association. Among the homologs, the sequences of the loop region involved in the stabilization and binding of the adenine ring of the NADPH shows significant differences except for an Arginine & glycine residues. In dimer QORs, this Arginine acts as a gate to the co-factor, while the NADPH binding mode in the human homolog is distinct, stabilized by His 200 and Asn 229, which are not conserved in LdQOR. These distinct features have the potential to be utilized for therapeutic interventions.


Asunto(s)
NAD(P)H Deshidrogenasa (Quinona) , Quinona Reductasas , Humanos , NADP/metabolismo , Subunidades de Proteína , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Quinona Reductasas/química , Quinona Reductasas/metabolismo , Quinonas , Arginina , Sitios de Unión , Cristalografía por Rayos X
4.
Nitric Oxide ; 146: 19-23, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38521487

RESUMEN

The mammalian brain is exquisitely vulnerable to lack of oxygen. However, the mechanism underlying the brain's sensitivity to hypoxia is incompletely understood. In this narrative review, we present a case for sulfide catabolism as a key defense mechanism of the brain against acute oxygen shortage. We will examine literature on the role of sulfide in hypoxia/ischemia, deep hibernation, and leigh syndrome patients, and present our recent data that support the neuroprotective effects of sulfide catabolism and persulfide production. When oxygen levels become low, hydrogen sulfide (H2S) accumulates in brain cells and impairs the ability of these cells to use the remaining, available oxygen to produce energy. In recent studies, we found that hibernating ground squirrels, which can withstand very low levels of oxygen, have high levels of sulfide:quinone oxidoreductase (SQOR) and the capacity to catabolize hydrogen sulfide in the brain. Silencing SQOR increased the sensitivity of the brain of squirrels and mice to hypoxia, whereas neuron-specific SQOR expression prevented hypoxia-induced sulfide accumulation, bioenergetic failure, and ischemic brain injury in mice. Excluding SQOR from mitochondria increased sensitivity to hypoxia not only in the brain but also in heart and liver. Pharmacological agents that scavenge sulfide and/or increase persulfide maintained mitochondrial respiration in hypoxic neurons and made mice resistant to ischemic injury to the brain or spinal cord. Drugs that oxidize hydrogen sulfide and/or increase persulfide may prove to be an effective approach to the treatment of patients experiencing brain injury caused by oxygen deprivation or mitochondrial dysfunction.


Asunto(s)
Hibernación , Neuroprotección , Hibernación/fisiología , Animales , Humanos , Sulfuros/metabolismo , Sulfuros/farmacología , Sulfuro de Hidrógeno/metabolismo , Encéfalo/metabolismo , Ratones , Sciuridae/metabolismo , Enfermedad de Leigh/metabolismo , Quinona Reductasas/metabolismo
5.
J Appl Microbiol ; 135(9)2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39264044

RESUMEN

AIMS: Heterologous expression of sulfur: quinone oxidoreductase (Sqr) from Halomonas mongoliensis JS01, which is responsible for oxidizing sulfide to elemental sulfur, in Thioalkalivibrio versutus (T. versutus) D301 improves desulfurization. METHODS AND RESULTS: We expressed sqr in T. versutus D301 by conjugative transfer and then assayed its desulfurization capacity in an airlift reactor and analyzed its transcriptome at -380 mV ORP. Our findings demonstrate that the D301-sqr+ strain, utilizing sodium sulfide as a sulfur source under optimal ORP conditions (-380 mV), achieved an elemental sulfur yield of 95%. This represents an 8% increase over the T. versutus D301. Moreover, the sodium sulfide utilization rate for D301-sqr+ showed a marked improvement [0.741 vs. 0.651 mmol∙(l·h)-1], with a concurrent increase in the rate of elemental sulfur production when compared to the T. versutus D301 (0.716 vs. 0.518 mmol ∙(l·h)-1). Transcriptome analysis revealed that the flavocytochrome c (fcc) and the sox system were differentially transcriptionally down-regulated in D301-sqr+ compared with the T. versutus D301. CONCLUSIONS: Heterologous expression of the gene sqr altered the transcription of related genes in T. versutus D301 sulfur oxidation pathway, increasing the yield of elemental sulfur and the rate of sulfur oxidation, and making D301-sqr+ more potential for industrial applications.


Asunto(s)
Azufre , Azufre/metabolismo , Halomonas/genética , Halomonas/metabolismo , Halomonas/enzimología , Sulfuros/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Oxidación-Reducción , Quinona Reductasas/metabolismo , Quinona Reductasas/genética , Reactores Biológicos
6.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33372143

RESUMEN

The electron-transferring flavoprotein-menaquinone oxidoreductase ABCX (EtfABCX), also known as FixABCX for its role in nitrogen-fixing organisms, is a member of a family of electron-transferring flavoproteins that catalyze electron bifurcation. EtfABCX enables endergonic reduction of ferredoxin (E°' ∼-450 mV) using NADH (E°' -320 mV) as the electron donor by coupling this reaction to the exergonic reduction of menaquinone (E°' -80 mV). Here we report the 2.9 Å structure of EtfABCX, a membrane-associated flavin-based electron bifurcation (FBEB) complex, from a thermophilic bacterium. EtfABCX forms a superdimer with two membrane-associated EtfCs at the dimer interface that contain two bound menaquinones. The structure reveals that, in contrast to previous predictions, the low-potential electrons bifurcated from EtfAB are most likely directly transferred to ferredoxin, while high-potential electrons reduce the quinone via two [4Fe-4S] clusters in EtfX. Surprisingly, EtfX shares remarkable structural similarity with mammalian [4Fe-4S] cluster-containing ETF ubiquinone oxidoreductase (ETF-QO), suggesting an unexpected evolutionary link between bifurcating and nonbifurcating systems. Based on this structure and spectroscopic studies of a closely related EtfABCX, we propose a detailed mechanism of the catalytic cycle and the accompanying structural changes in this membrane-associated FBEB system.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/metabolismo , Quinona Reductasas/metabolismo , Quinona Reductasas/ultraestructura , Proteínas Bacterianas/metabolismo , Catálisis , Microscopía por Crioelectrón/métodos , Transporte de Electrón , Electrones , Ferredoxinas/metabolismo , Flavinas/metabolismo , Flavoproteínas/metabolismo , Proteínas Hierro-Azufre/metabolismo , Modelos Moleculares , NAD/metabolismo , Fijación del Nitrógeno/fisiología , Oxidación-Reducción , Pyrococcus furiosus/metabolismo , Quinona Reductasas/fisiología , Vitamina K 2/metabolismo
7.
J Biol Chem ; 298(1): 101435, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34808207

RESUMEN

The dual roles of H2S as an endogenously synthesized respiratory substrate and as a toxin raise questions as to how it is cleared when the electron transport chain is inhibited. Sulfide quinone oxidoreductase (SQOR) catalyzes the first step in the mitochondrial H2S oxidation pathway, using CoQ as an electron acceptor, and connects to the electron transport chain at the level of complex III. We have discovered that at high H2S concentrations, which are known to inhibit complex IV, a new redox cycle is established between SQOR and complex II, operating in reverse. Under these conditions, the purine nucleotide cycle and the malate aspartate shuttle furnish fumarate, which supports complex II reversal and leads to succinate accumulation. Complex II knockdown in colonocytes decreases the efficiency of H2S clearance while targeted knockout of complex II in intestinal epithelial cells significantly decreases the levels of thiosulfate, a biomarker of H2S oxidation, to approximately one-third of the values seen in serum and urine samples from control mice. These data establish the physiological relevance of this newly discovered redox circuitry between SQOR and complex II for prioritizing H2S oxidation and reveal the quantitatively significant contribution of intestinal epithelial cells to systemic H2S metabolism.


Asunto(s)
Sulfuro de Hidrógeno , Quinona Reductasas , Animales , Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/metabolismo , Sulfuro de Hidrógeno/metabolismo , Ratones , Oxidación-Reducción , Quinona Reductasas/genética , Quinona Reductasas/metabolismo
8.
Nat Chem Biol ; 17(1): 65-70, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33106663

RESUMEN

Cysteine thiol-based transcriptional regulators orchestrate the coordinated regulation of redox homeostasis and other cellular processes by 'sensing' or detecting a specific redox-active molecule, which in turn activates the transcription of a specific detoxification pathway. The extent to which these sensors are truly specific in cells for a singular class of reactive small-molecule stressors, for example, reactive oxygen or sulfur species, is largely unknown. Here, we report structural and mechanistic insights into the thiol-based transcriptional repressor SqrR, which reacts exclusively with oxidized sulfur species such as persulfides, to yield a tetrasulfide bridge that inhibits DNA operator-promoter binding. Evaluation of crystallographic structures of SqrR in various derivatized states, coupled with the results of a mass spectrometry-based kinetic profiling strategy, suggest that persulfide selectivity is determined by structural frustration of the disulfide form. These findings led to the identification of an uncharacterized repressor from the bacterial pathogen Acinetobacter baumannii as a persulfide sensor.


Asunto(s)
Acinetobacter baumannii/genética , Proteínas Bacterianas/química , Regulación Bacteriana de la Expresión Génica , Quinona Reductasas/química , Sulfuros/química , Transcripción Genética , Acinetobacter baumannii/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Cisteína/química , Cisteína/metabolismo , Disulfuros/química , Disulfuros/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Glutatión/química , Glutatión/metabolismo , Cinética , Modelos Moleculares , Oxidación-Reducción , Regiones Promotoras Genéticas , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Quinona Reductasas/genética , Quinona Reductasas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Sulfuros/metabolismo , Azufre/química , Azufre/metabolismo , Termodinámica
9.
Biochem J ; 479(1): 111-127, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-34981811

RESUMEN

The cytochrome b6f complex (b6f) has been initially considered as the ferredoxin-plastoquinone reductase (FQR) during cyclic electron flow (CEF) with photosystem I that is inhibited by antimycin A (AA). The binding of AA to the b6f Qi-site is aggravated by heme-ci, which challenged the FQR function of b6f during CEF. Alternative models suggest that PROTON GRADIENT REGULATION5 (PGR5) is involved in a b6f-independent, AA-sensitive FQR. Here, we show in Chlamydomonas reinhardtii that the b6f is conditionally inhibited by AA in vivo and that the inhibition did not require PGR5. Instead, activation of the STT7 kinase upon anaerobic treatment induced the AA sensitivity of b6f which was absent from stt7-1. However, a lock in State 2 due to persisting phosphorylation in the phosphatase double mutant pph1;pbcp did not increase AA sensitivity of electron transfer. The latter required a redox poise, supporting the view that state transitions and CEF are not coercively coupled. This suggests that the b6f-interacting kinase is required for structure-function modulation of the Qi-site under CEF favoring conditions. We propose that PGR5 and STT7 independently sustain AA-sensitive FQR activity of the b6f. Accordingly, PGR5-mediated electron injection into an STT7-modulated Qi-site drives a Mitchellian Q cycle in CEF conditions.


Asunto(s)
Antimicina A/farmacología , Chlamydomonas reinhardtii/enzimología , Complejo de Citocromo b6f/metabolismo , Electrones , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Tilacoides/enzimología , Antimicina A/metabolismo , Complejo de Citocromo b6f/antagonistas & inhibidores , Transporte de Electrón/efectos de los fármacos , Activación Enzimática , Ferredoxinas/metabolismo , Complejos de Proteína Captadores de Luz/metabolismo , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/metabolismo , Fosforilación/efectos de los fármacos , Fotosíntesis/fisiología , Complejo de Proteína del Fotosistema I/metabolismo , Plastoquinona/metabolismo , Quinona Reductasas/metabolismo
10.
Appl Environ Microbiol ; 88(3): e0194121, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-34878813

RESUMEN

Sulfur-oxidizing bacteria can oxidize hydrogen sulfide (H2S) to produce sulfur globules. Although the process is common, the pathway is unclear. In recombinant Escherichia coli and wild-type Corynebacterium vitaeruminis DSM 20294 with sulfide:quinone oxidoreductase (SQR) but no enzymes to oxidize zero valence sulfur, SQR oxidized H2S into short-chain inorganic polysulfide (H2Sn, n ≥ 2) and organic polysulfide (RSnH, n ≥ 2), which reacted with each other to form long-chain GSnH (n ≥ 2) and H2Sn before producing octasulfur (S8), the main component of elemental sulfur. GSnH also reacted with glutathione (GSH) to form GSnG (n ≥ 2) and H2S; H2S was again oxidized by SQR. After GSH was depleted, SQR simply oxidized H2S to H2Sn, which spontaneously generated S8. S8 aggregated into sulfur globules in the cytoplasm. The results highlight the process of sulfide oxidation to S8 globules in the bacterial cytoplasm and demonstrate the potential of using heterotrophic bacteria with SQR to convert toxic H2S into relatively benign S8 globules. IMPORTANCE Our results provide evidence of H2S oxidation producing octasulfur globules via sulfide:quinone oxidoreductase (SQR) catalysis and spontaneous reactions in the bacterial cytoplasm. Since the process is an important event in geochemical cycling, a better understanding facilitates further studies and provides theoretical support for using heterotrophic bacteria with SQR to oxidize toxic H2S into sulfur globules for recovery.


Asunto(s)
Sulfuro de Hidrógeno , Quinona Reductasas , Bacterias Aerobias/metabolismo , Citoplasma/metabolismo , Sulfuro de Hidrógeno/metabolismo , Oxidación-Reducción , Quinona Reductasas/metabolismo , Sulfuros/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA