Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 3.922
Filtrar
Más filtros

Publication year range
1.
Annu Rev Biochem ; 90: 817-846, 2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-33823652

RESUMEN

Sulfonates include diverse natural products and anthropogenic chemicals and are widespread in the environment. Many bacteria can degrade sulfonates and obtain sulfur, carbon, and energy for growth, playing important roles in the biogeochemical sulfur cycle. Cleavage of the inert sulfonate C-S bond involves a variety of enzymes, cofactors, and oxygen-dependent and oxygen-independent catalytic mechanisms. Sulfonate degradation by strictly anaerobic bacteria was recently found to involve C-S bond cleavage through O2-sensitive free radical chemistry, catalyzed by glycyl radical enzymes (GREs). The associated discoveries of new enzymes and metabolic pathways for sulfonate metabolism in diverse anaerobic bacteria have enriched our understanding of sulfonate chemistry in the anaerobic biosphere. An anaerobic environment of particular interest is the human gut microbiome, where sulfonate degradation by sulfate- and sulfite-reducing bacteria (SSRB) produces H2S, a process linked to certain chronic diseases and conditions.


Asunto(s)
Liasas de Carbono-Carbono/metabolismo , Microbioma Gastrointestinal/fisiología , Ácidos Sulfónicos/metabolismo , Acetiltransferasas/química , Acetiltransferasas/metabolismo , Alcanosulfonatos/metabolismo , Anaerobiosis , Bacterias/metabolismo , Liasas de Carbono-Carbono/química , Glicina/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Ácido Isetiónico/metabolismo , Microbiota/fisiología , Taurina/metabolismo
2.
Cell ; 184(3): 615-627.e17, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33453153

RESUMEN

The microbiota shields the host against infections in a process known as colonization resistance. How infections themselves shape this fundamental process remains largely unknown. Here, we show that gut microbiota from previously infected hosts display enhanced resistance to infection. This long-term functional remodeling is associated with altered bile acid metabolism leading to the expansion of taxa that utilize the sulfonic acid taurine. Notably, supplying exogenous taurine alone is sufficient to induce this alteration in microbiota function and enhance resistance. Mechanistically, taurine potentiates the microbiota's production of sulfide, an inhibitor of cellular respiration, which is key to host invasion by numerous pathogens. As such, pharmaceutical sequestration of sulfide perturbs the microbiota's composition and promotes pathogen invasion. Together, this work reveals a process by which the host, triggered by infection, can deploy taurine as a nutrient to nourish and train the microbiota, promoting its resistance to subsequent infection.


Asunto(s)
Microbioma Gastrointestinal , Interacciones Huésped-Patógeno , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Recuento de Colonia Microbiana , Microbioma Gastrointestinal/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Inmunidad , Ratones Endogámicos C57BL , Sulfuros/metabolismo , Taurina/farmacología
3.
Cell ; 173(1): 74-89.e20, 2018 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-29570999

RESUMEN

A decline in capillary density and blood flow with age is a major cause of mortality and morbidity. Understanding why this occurs is key to future gains in human health. NAD precursors reverse aspects of aging, in part, by activating sirtuin deacylases (SIRT1-SIRT7) that mediate the benefits of exercise and dietary restriction (DR). We show that SIRT1 in endothelial cells is a key mediator of pro-angiogenic signals secreted from myocytes. Treatment of mice with the NAD+ booster nicotinamide mononucleotide (NMN) improves blood flow and increases endurance in elderly mice by promoting SIRT1-dependent increases in capillary density, an effect augmented by exercise or increasing the levels of hydrogen sulfide (H2S), a DR mimetic and regulator of endothelial NAD+ levels. These findings have implications for improving blood flow to organs and tissues, increasing human performance, and reestablishing a virtuous cycle of mobility in the elderly.


Asunto(s)
Envejecimiento , Sulfuro de Hidrógeno/metabolismo , NAD/metabolismo , Animales , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Noqueados , Microvasos/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Neovascularización Fisiológica , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Condicionamiento Físico Animal , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Sirtuina 1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Physiol Rev ; 103(1): 31-276, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35435014

RESUMEN

Over the last two decades, hydrogen sulfide (H2S) has emerged as an endogenous regulator of a broad range of physiological functions. H2S belongs to the class of molecules known as gasotransmitters, which typically include nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine γ-lyase (CSE), cystathionine ß-synthase (CBS), and 3-mercaptopyruvate sulfurtransferase (3-MST). The present article reviews the regulation of these enzymes as well as the pathways of their enzymatic and nonenzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g., NO) and reactive oxygen species are also outlined. Next, the various biological targets and signaling pathways are outlined, with special reference to H2S or oxidative posttranscriptional modification (persulfidation or sulfhydration) of proteins and the effect of H2S on various channels and intracellular second messenger pathways, the regulation of gene transcription and translation, and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed, including the regulation of membrane potential, endo- and exocytosis, regulation of various cell organelles (endoplasmic reticulum, Golgi, mitochondria), regulation of cell movement, cell cycle, cell differentiation, and physiological aspects of regulated cell death. Next, the physiological roles of H2S in various cell types and organ systems are overviewed, including the role of H2S in red blood cells, immune cells, the central and peripheral nervous systems (with focus on neuronal transmission, learning, and memory formation), and regulation of vascular function (including angiogenesis as well as its specialized roles in the cerebrovascular, renal, and pulmonary vascular beds) and the role of H2S in the regulation of special senses, vision, hearing, taste and smell, and pain-sensing. Finally, the roles of H2S in the regulation of various organ functions (lung, heart, liver, kidney, urogenital organs, reproductive system, bone and cartilage, skeletal muscle, and endocrine organs) are presented, with a focus on physiology (including physiological aging) but also extending to some common pathophysiological conditions. From these data, a wide array of significant roles of H2S in the physiological regulation of all organ functions emerges and the characteristic bell-shaped biphasic effects of H2S are highlighted. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified.


Asunto(s)
Gasotransmisores , Sulfuro de Hidrógeno , Animales , Monóxido de Carbono , Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Gasotransmisores/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Mamíferos/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno
5.
Mol Cell ; 81(18): 3820-3832.e7, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34233158

RESUMEN

A metabolic imbalance between lipid synthesis and degradation can lead to hepatic lipid accumulation, a characteristic of patients with non-alcoholic fatty liver disease (NAFLD). Here, we report that high-fat-diet-induced sterol regulatory element-binding protein (SREBP)-1c, a key transcription factor that regulates lipid biosynthesis, impairs autophagic lipid catabolism via altered H2S signaling. SREBP-1c reduced cystathionine gamma-lyase (CSE) via miR-216a, which in turn decreased hepatic H2S levels and sulfhydration-dependent activation of Unc-51-like autophagy-activating kinase 1 (ULK1). Furthermore, Cys951Ser mutation of ULK1 decreased autolysosome formation and promoted hepatic lipid accumulation in mice, suggesting that the loss of ULK1 sulfhydration was directly associated with the pathogenesis of NAFLD. Moreover, silencing of CSE in SREBP-1c knockout mice increased liver triglycerides, confirming the connection between CSE, autophagy, and SREBP-1c. Overall, our results uncover a 2-fold mechanism for SREBP-1c-driven hepatic lipid accumulation through reciprocal activation and inhibition of hepatic lipid biosynthesis and degradation, respectively.


Asunto(s)
Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Hígado Graso/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Autofagia , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/fisiología , Línea Celular Tumoral , Dieta Alta en Grasa/efectos adversos , Hígado Graso/fisiopatología , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Lipogénesis , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Transducción de Señal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Triglicéridos/metabolismo
6.
Proc Natl Acad Sci U S A ; 121(12): e2319473121, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38478695

RESUMEN

Hydrogen sulfide exposure in moderate doses can induce profound but reversible hypometabolism in mammals. At a cellular level, H2S inhibits the electron transport chain (ETC), augments aerobic glycolysis, and glutamine-dependent carbon utilization via reductive carboxylation; however, the durability of these changes is unknown. We report that despite its volatility, H2S preconditioning increases P50(O2), the O2 pressure for half-maximal cellular respiration, and has pleiotropic effects on oxidative metabolism that persist up to 24 to 48 h later. Notably, cyanide, another complex IV inhibitor, does not induce this type of metabolic memory. Sulfide-mediated prolonged fractional inhibition of complex IV by H2S is modulated by sulfide quinone oxidoreductase, which commits sulfide to oxidative catabolism. Since induced hypometabolism can be beneficial in disease settings that involve insufficient or interrupted blood flow, our study has important implications for attenuating reperfusion-induced ischemic injury and/or prolonging the shelf life of biologics like platelets.


Asunto(s)
Sulfuro de Hidrógeno , Daño por Reperfusión , Animales , Sulfuro de Hidrógeno/farmacología , Sulfuro de Hidrógeno/metabolismo , Sulfuros , Oxidación-Reducción , Mamíferos/metabolismo
7.
Trends Biochem Sci ; 47(8): 689-698, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35397924

RESUMEN

The electron transport chain (ETC) is a major currency converter that exchanges the chemical energy of fuel oxidation to proton motive force and, subsequently, ATP generation, using O2 as a terminal electron acceptor. Discussed herein, two new studies reveal that the mammalian ETC is forked. Hypoxia or H2S exposure promotes the use of fumarate as an alternate terminal electron acceptor. The fumarate/succinate and CoQH2/CoQ redox couples are nearly iso-potential, revealing that complex II is poised for facile reverse electron transfer, which is sensitive to CoQH2 and fumarate concentrations. The gas regulators, H2S and •NO, modulate O2 affinity and/or inhibit the electron transfer rate at complex IV. Their induction under hypoxia suggests a mechanism for how traffic at the ETC fork can be regulated.


Asunto(s)
Electrones , Fumaratos , Animales , Transporte de Electrón , Hipoxia , Mamíferos , Oxidación-Reducción
8.
Proc Natl Acad Sci U S A ; 120(3): e2205044120, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36630448

RESUMEN

Although hydrogen sulfide (H2S) is an endogenous signaling molecule with antioxidant properties, it is also cytotoxic by potently inhibiting cytochrome c oxidase and mitochondrial respiration. Paradoxically, the primary route of H2S detoxification is thought to occur inside the mitochondrial matrix via a series of relatively slow enzymatic reactions that are unlikely to compete with its rapid inhibition of cytochrome c oxidase. Therefore, alternative or complementary cellular mechanisms of H2S detoxification are predicted to exist. Here, superoxide dismutase [Cu-Zn] (SOD1) is shown to be an efficient H2S oxidase that has an essential role in limiting cytotoxicity from endogenous and exogenous sulfide. Decreased SOD1 expression resulted in increased sensitivity to H2S toxicity in yeast and human cells, while increased SOD1 expression enhanced tolerance to H2S. SOD1 rapidly converted H2S to sulfate under conditions of limiting sulfide; however, when sulfide was in molar excess, SOD1 catalyzed the formation of per- and polysulfides, which induce cellular thiol oxidation. Furthermore, in SOD1-deficient cells, elevated levels of reactive oxygen species catalyzed sulfide oxidation to per- and polysulfides. These data reveal that a fundamental function of SOD1 is to regulate H2S and related reactive sulfur species.


Asunto(s)
Complejo IV de Transporte de Electrones , Sulfuro de Hidrógeno , Superóxido Dismutasa-1 , Humanos , Complejo IV de Transporte de Electrones/metabolismo , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/toxicidad , Sulfuros/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
J Biol Chem ; 300(5): 107301, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38641068

RESUMEN

Ubiquinol or coenzyme Q (CoQ) is a lipid-soluble electron carrier in the respiratory chain and an electron acceptor for various enzymes in metabolic pathways that intersect at this cofactor hub in the mitochondrial inner membrane. The reduced form of CoQ is an antioxidant, which protects against lipid peroxidation. In this study, we have optimized a UV-detected HPLC method for CoQ analysis from biological materials, which involves a rapid single-step extraction into n-propanol followed by direct sample injection onto a column. Using this method, we have measured the oxidized, reduced, and total CoQ pools and monitored shifts in the CoQ redox status in response to cell culture conditions and bioenergetic perturbations. We find that hypoxia or sulfide exposure induces a reductive shift in the intracellular CoQ pool. The effect of hypoxia is, however, rapidly reversed by exposure to ambient air. Interventions at different loci in the electron transport chain can induce sizeable redox shifts in the oxidative or reductive direction, depending on whether they are up- or downstream of complex III. We have also used this method to confirm that CoQ levels are higher and more reduced in murine heart versus brain. In summary, the availability of a convenient HPLC-based method described herein will facilitate studies on CoQ redox dynamics in response to environmental, nutritional, and endogenous alterations.


Asunto(s)
Oxidación-Reducción , Ubiquinona , Animales , Humanos , Ratones , Cromatografía Líquida de Alta Presión/métodos , Ubiquinona/química , Ubiquinona/metabolismo , Miocardio/enzimología , Encéfalo/enzimología , Femenino , Ratones Endogámicos C57BL , Células HT29
10.
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
11.
EMBO J ; 40(11): e106771, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33909912

RESUMEN

Chemical compounds have recently been introduced as alternative and non-integrating inducers of pluripotent stem cell fate. However, chemical reprogramming is hampered by low efficiency and the molecular mechanisms remain poorly characterized. Here, we show that inhibition of spleen tyrosine kinase (Syk) by R406 significantly promotes mouse chemical reprogramming. Mechanistically, R406 alleviates Syk / calcineurin (Cn) / nuclear factor of activated T cells (NFAT) signaling-mediated suppression of glycine, serine, and threonine metabolic genes and dependent metabolites. Syk inhibition upregulates glycine level and downstream transsulfuration cysteine biosynthesis, promoting cysteine metabolism and cellular hydrogen sulfide (H2 S) production. This metabolic rewiring decreased oxidative phosphorylation and ROS levels, enhancing chemical reprogramming. In sum, our study identifies Syk-Cn-NFAT signaling axis as a new barrier of chemical reprogramming and suggests metabolic rewiring and redox homeostasis as important opportunities for controlling cell fates.


Asunto(s)
Fibroblastos/metabolismo , Sulfuro de Hidrógeno/metabolismo , Quinasa Syk/antagonistas & inhibidores , Animales , Calcineurina/metabolismo , Células Cultivadas , Cisteína/metabolismo , Fibroblastos/efectos de los fármacos , Glicina/metabolismo , Ratones , Factores de Transcripción NFATC/metabolismo , Oxazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
12.
Circ Res ; 132(2): 154-166, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36575984

RESUMEN

BACKGROUND: Hydrogen sulfide is a critical endogenous signaling molecule that exerts protective effects in the setting of heart failure. Cystathionine γ-lyase (CSE), 1 of 3 hydrogen-sulfide-producing enzyme, is predominantly localized in the vascular endothelium. The interaction between the endothelial CSE-hydrogen sulfide axis and endothelial-mesenchymal transition, an important pathological process contributing to the formation of fibrosis, has yet to be investigated. METHODS: Endothelial-cell-specific CSE knockout and Endothelial cell-CSE overexpressing mice were subjected to transverse aortic constriction to induce heart failure with reduced ejection fraction. Cardiac function, vascular reactivity, and treadmill exercise capacity were measured to determine the severity of heart failure. Histological and gene expression analyses were performed to investigate changes in cardiac fibrosis and the activation of endothelial-mesenchymal transition. RESULTS: Endothelial-cell-specific CSE knockout mice exhibited increased endothelial-mesenchymal transition and reduced nitric oxide bioavailability in the myocardium, which was associated with increased cardiac fibrosis, impaired cardiac and vascular function, and worsened exercise performance. In contrast, genetic overexpression of CSE in endothelial cells led to increased myocardial nitric oxide, decreased endothelial-mesenchymal transition and cardiac fibrosis, preserved cardiac and endothelial function, and improved exercise capacity. CONCLUSIONS: Our data demonstrate that endothelial CSE modulates endothelial-mesenchymal transition and ameliorate the severity of pressure-overload-induced heart failure, in part, through nitric oxide-related mechanisms. These data further suggest that endothelium-derived hydrogen sulfide is a potential therapeutic for the treatment of heart failure with reduced ejection fraction.


Asunto(s)
Insuficiencia Cardíaca , Sulfuro de Hidrógeno , Disfunción Ventricular Izquierda , Ratones , Animales , Sulfuro de Hidrógeno/metabolismo , Células Endoteliales/metabolismo , Óxido Nítrico/metabolismo , Ratones Noqueados , Endotelio Vascular/metabolismo , Fibrosis
13.
Biochem J ; 481(8): 569-585, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38563463

RESUMEN

Homocystinuria is a rare disease caused by mutations in the CBS gene that results in a deficiency of cystathionine ß-synthase (CBS). CBS is an essential pyridoxal 5'-phosphate (PLP)-dependent enzyme in the transsulfuration pathway, responsible for combining serine with homocysteine to produce cystathionine, whose activity is enhanced by the allosteric regulator S-adenosylmethionine (SAM). CBS also plays a role in generating hydrogen sulfide (H2S), a gaseous signaling molecule with diverse regulatory functions within the vascular, nervous, and immune systems. In this study, we present the clinical and biochemical characterization of two novel CBS missense mutations that do not respond to pyridoxine treatment, namely c.689T > A (L230Q) and 215A > T (K72I), identified in a Chinese patient. We observed that the disease-associated K72I genetic variant had no apparent effects on the spectroscopic and catalytic properties of the full-length enzyme. In contrast, the L230Q variant expressed in Escherichia coli did not fully retain heme and when compared with the wild-type enzyme, it exhibited more significant impairments in both the canonical cystathionine-synthesis and the alternative H2S-producing reactions. This reduced activity is consistent with both in vitro and in silico evidence, which indicates that the L230Q mutation significantly decreases the overall protein's stability, which in turn, may represent the underlying cause of its pathogenicity.


Asunto(s)
Cistationina betasintasa , Homocistinuria , Mutación Missense , Cistationina betasintasa/genética , Cistationina betasintasa/química , Cistationina betasintasa/metabolismo , Homocistinuria/genética , Homocistinuria/metabolismo , Homocistinuria/enzimología , Humanos , Masculino , Femenino
14.
Proc Natl Acad Sci U S A ; 119(11): e2118002119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35271389

RESUMEN

SignificanceYeiE has been identified as a master virulence factor of Cronobacter sakazakii. In this study, we determined the crystal structures of the regulatory domain of YeiE in complex with its physiological ligand sulfite ion (SO32-). The structure provides the basis for the molecular mechanisms for sulfite sensing and the ligand-dependent conformational changes of the regulatory domain. The genes under the control of YeiE in response to sulfite were investigated to reveal the functional roles of YeiE in the sulfite tolerance of the bacteria. We propose the molecular mechanism underlying the ability of gram-negative pathogens to defend against the innate immune response involving sulfite, thus providing a strategy to control the pathogenesis of bacteria.


Asunto(s)
Proteínas Bacterianas , Cronobacter sakazakii , Estrés Fisiológico , Sulfitos , Factores de Transcripción , Factores de Virulencia , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Cronobacter sakazakii/genética , Cronobacter sakazakii/metabolismo , Cronobacter sakazakii/patogenicidad , Cristalización , Ligandos , Dominios Proteicos , Sulfitos/metabolismo , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Virulencia/química , Factores de Virulencia/genética
15.
J Biol Chem ; 299(12): 105449, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37949228

RESUMEN

Cystathionine ß-synthase (CBS) catalyzes the committing step in the transsulfuration pathway, which is important for clearing homocysteine and furnishing cysteine. The transsulfuration pathway also generates H2S, a signaling molecule. CBS is a modular protein with a heme and pyridoxal phosphate-binding catalytic core, which is separated by a linker region from the C-terminal regulatory domain that binds S-adenosylmethionine (AdoMet), an allosteric activator. Recent cryo-EM structures reveal that CBS exists in a fibrillar form and undergoes a dramatic architectural rearrangement between the basal and AdoMet-bound states. CBS is the single most common locus of mutations associated with homocystinuria, and, in this study, we have characterized three clinical variants (K384E/N and M391I), which reside in the linker region. The native fibrillar form is destabilized in the variants, and differences in their limited proteolytic fingerprints also reveal conformational alterations. The crystal structure of the truncated K384N variant, lacking the regulatory domain, reveals that the overall fold of the catalytic core is unperturbed. M391I CBS exhibits a modest (1.4-fold) decrease while the K384E/N variants exhibit a significant (∼8-fold) decrease in basal activity, which is either unresponsive to or inhibited by AdoMet. Pre-steady state kinetic analyses reveal that the K384E/N substitutions exhibit pleiotropic effects and that the differences between them are expressed in the second half reaction, that is, homocysteine binding and reaction with the aminoacrylate intermediate. Together, these studies point to an important role for the linker in stabilizing the higher-order oligomeric structure of CBS and enabling AdoMet-dependent regulation.


Asunto(s)
Cistationina betasintasa , Mutación , Humanos , Regulación Alostérica/genética , Cristalografía por Rayos X , Cistationina betasintasa/química , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Homocisteína/metabolismo , Homocistinuria/enzimología , Homocistinuria/genética , Cinética , S-Adenosilmetionina/metabolismo , Conformación Proteica , Dominio Catalítico
16.
Plant J ; 114(6): 1369-1384, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36948886

RESUMEN

Hydrogen sulfide (H2 S) promotes plant tolerance against various environmental cues, and d-cysteine desulfhydrase (DCD) is an enzymatic source of H2 S to enhance abiotic stress resistance. However, the role of DCD-mediated H2 S production in root growth under abiotic stress remains to be further elucidated. Here, we report that DCD-mediated H2 S production alleviates osmotic stress-mediated root growth inhibition by promoting auxin homeostasis. Osmotic stress up-regulated DCD gene transcript and DCD protein levels and thus H2 S production in roots. When subjected to osmotic stress, a dcd mutant showed more severe root growth inhibition, whereas the transgenic lines DCDox overexpressing DCD exhibited less sensitivity to osmotic stress in terms of longer root compared to the wild-type. Moreover, osmotic stress inhibited root growth through repressing auxin signaling, whereas H2 S treatment significantly alleviated osmotic stress-mediated inhibition of auxin. Under osmotic stress, auxin accumulation was increased in DCDox but decreased in dcd mutant. H2 S promoted auxin biosynthesis gene expression and auxin efflux carrier PIN-FORMED 1 (PIN1) protein level under osmotic stress. Taken together, our results reveal that mannitol-induced DCD and H2 S in roots promote auxin homeostasis, contributing to alleviating the inhibition of root growth under osmotic stress.


Asunto(s)
Proteínas de Arabidopsis , Sulfuro de Hidrógeno , Sulfuro de Hidrógeno/metabolismo , Raíces de Plantas/metabolismo , Presión Osmótica , Homeostasis , Ácidos Indolacéticos/metabolismo , Regulación de la Expresión Génica de las Plantas , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo
17.
Apoptosis ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980600

RESUMEN

Ferroptosis is a programmed cell death that relies on iron and lipid peroxidation. It differs from other forms of programmed cell death such as necrosis, apoptosis and autophagy. More and more evidence indicates that ferroptosis participates in many types of diseases, such as neurodegenerative diseases, ischemia-reperfusion injury, cardiovascular diseases and so on. Hence, clarifying the role and mechanism of ferroptosis in diseases is of great significance for further understanding the pathogenesis and treatment of some diseases. Hydrogen sulfide (H2S) is a colorless and flammable gas with the smell of rotten eggs. Many years ago, H2S was considered as a toxic gas. however, in recent years, increasing evidence indicates that it is the third important gas signaling molecule after nitric oxide and carbon monoxide. H2S has various physiological and pathological functions such as antioxidant stress, anti-inflammatory, anti-apoptotic and anti-tumor, and can participate in various diseases. It has been reported that H2S regulation of ferroptosis plays an important role in many types of diseases, however, the related mechanisms are not fully clear. In this review, we reviewed the recent literature about the role of H2S regulation of ferroptosis in diseases, and analyzed the relevant mechanisms, hoping to provide references for future in-depth researches.

18.
Biochem Cell Biol ; 102(3): 252-261, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38417127

RESUMEN

Diabetic kidney disease (DKD) is a major contributor to chronic kidney disease. Hydrogen sulfide (H2S) serves as an endogenous gaseous signaling molecule capable of safeguarding renal function within the context of DKD. However, the underlying mechanisms need to be elucidated. This study was undertaken to unveil the mechanisms by which H2S counteracts against DKD. Utilizing mice and human renal tubular epithelial (HK-2) cells, we demonstrated a reduction in cystathionine-γ-lyase/H2S levels within renal tissues of db/db mice and in HK-2 cells subjected to hyperglycemic and hyperlipidemic environments. Notably, we observed that sodium hydrosulfide (NaHS) supplementation could serve as an exogenous source of H2S. Exogenous H2S exhibited the capacity to mitigate the accumulation of reactive oxygen species and attenuate the degradation of superoxide dismutase 2 (SOD2) by Lon protease homolog 1 induced by hyperglycemia and hyperlipidemia, thus affording cellular protection against mitochondrial apoptosis. Consequently, NaHS treatment led to decreased serum levels of blood urea nitrogen and serum creatinine, reflecting alleviated renal damage and thereby preserving renal function in db/db mice. Based on these findings, we propose that exogenous H2S exerts a protective role against DKD by inhibiting SOD2 degradation.


Asunto(s)
Nefropatías Diabéticas , Sulfuro de Hidrógeno , Superóxido Dismutasa , Animales , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Ratones , Humanos , Superóxido Dismutasa/metabolismo , Masculino , Riñón/metabolismo , Riñón/efectos de los fármacos , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico
19.
Biochem Biophys Res Commun ; 692: 149343, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38065000

RESUMEN

Wound healing difficulties in diabetes continue to be a clinical challenge, posing a considerable burden to patients and society. Recently, exploration of the mechanism of wound healing and associated treatment options in diabetes has become topical. Of note, the positive role of hydrogen sulfide in promoting wound healing has been demonstrated in recent studies. Hydrogen sulfide is a confirmed gas transmitter in mammals, playing an essential role in pathology and physiology. This review describes the mechanism underlying the role of hydrogen sulfide in the promotion of diabetic wound healing and the potential for hydrogen sulfide supplementation as a therapeutic application.


Asunto(s)
Diabetes Mellitus , Sulfuro de Hidrógeno , Animales , Humanos , Sulfuro de Hidrógeno/farmacología , Diabetes Mellitus/tratamiento farmacológico , Cicatrización de Heridas/fisiología , Mamíferos
20.
Biochem Biophys Res Commun ; 699: 149562, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38277726

RESUMEN

Hydrogen sulfide (H2S) acts as a gas-signaling agent in various tissues. Although it has been reported that endogenous enzymes that generate H2S are expressed abundantly in the kidney, few reports examine cellular responses to H2S in renal tubular epithelial cells. In this study, we investigated the effects of NaHS, an H2S donor, and l-cysteine, a substrate for H2S production, on the principal cells of rat cortical collecting ducts (CCDs). NaHS increased the intracellular Ca2+ concentration ([Ca2+]i) in the principal cells. The removal of extracellular Ca2+ largely attenuated the [Ca2+]i response. The TRPV4 channel blocker significantly inhibited the effect of NaHS. Extracellular administration of l-cysteine also elicited a rise in [Ca2+]i. Prior treatment of CCDs with AOAA, an inhibitor of H2S production enzyme, l-cysteine-induced [Ca2+]i response was significantly reduced. These results suggest that not only exogenous H2S but also endogenously produced H2S triggers the extracellular influx pathway of Ca2+ in the principal cells of rat CCDs.


Asunto(s)
Sulfuro de Hidrógeno , Ratas , Animales , Sulfuro de Hidrógeno/farmacología , Sulfuro de Hidrógeno/metabolismo , Cisteína/metabolismo , Sulfuros/farmacología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda