Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509349

RESUMEN

Angiogenic programming in the vascular endothelium is a tightly regulated process for maintaining tissue homeostasis and is activated in tissue injury and the tumor microenvironment. The metabolic basis of how gas signaling molecules regulate angiogenesis is elusive. Here, we report that hypoxic upregulation of ·NO in endothelial cells reprograms the transsulfuration pathway to increase biogenesis of hydrogen sulfide (H2S), a proangiogenic metabolite. However, decreased H2S oxidation due to sulfide quinone oxidoreductase (SQOR) deficiency synergizes with hypoxia, inducing a reductive shift and limiting endothelial proliferation that is attenuated by dissipation of the mitochondrial NADH pool. Tumor xenografts in whole-body (WBCreSqorfl/fl) and endothelial-specific (VE-cadherinCre-ERT2Sqorfl/fl) Sqor-knockout mice exhibit lower mass and angiogenesis than control mice. WBCreSqorfl/fl mice also exhibit decreased muscle angiogenesis following femoral artery ligation compared to control mice. Collectively, our data reveal the molecular intersections between H2S, O2 and ·NO metabolism and identify SQOR inhibition as a metabolic vulnerability for endothelial cell proliferation and neovascularization.

2.
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
3.
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
4.
Anal Biochem ; 673: 115191, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37207973

RESUMEN

H2S is a redox-active signaling molecule that exerts an array of cellular and physiological effects. While intracellular H2S concentrations are estimated to be in the low nanomolar range, intestinal luminal concentrations can be significantly higher due to microbial metabolism. Studies assessing H2S effects are typically conducted with a bolus treatment with sulfide salts or slow releasing sulfide donors, which are limited by the volatility of H2S, and by potential off-target effects of the donor molecules. To address these limitations, we describe the design and performance of a mammalian cell culture incubator for sustained exposure to 20-500 ppm H2S (corresponding to a dissolved sulfide concentrations of ∼4-120 µM in the cell culture medium). We report that colorectal adenocarcinoma HT29 cells tolerate prolonged exposure to H2S with no effect on cell viability after 24 h although ≥50 ppm H2S (∼10 µM) restricts cell proliferation. Even the lowest concentration of H2S used in this study (i.e. ∼4 µM) significantly enhanced glucose consumption and lactate production, revealing a much lower threshold for impacting cellular energy metabolism and activating aerobic glycolysis than has been previously appreciated from studies with bolus H2S treatment regimens.


Asunto(s)
Neoplasias Colorrectales , Sulfuro de Hidrógeno , Humanos , Animales , Sulfuro de Hidrógeno/metabolismo , Oxidación-Reducción , Proliferación Celular , Sulfuros/farmacología , Mamíferos/metabolismo
5.
Int J Mol Sci ; 24(12)2023 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-37373271

RESUMEN

A mathematical model of energy metabolism in erythrocyte-bioreactors loaded with alcohol dehydrogenase and acetaldehyde dehydrogenase was constructed and analyzed. Such erythrocytes can convert ethanol to acetate using intracellular NAD and can therefore be used to treat alcohol intoxication. Analysis of the model revealed that the rate of ethanol consumption by the erythrocyte-bioreactors increases proportionally to the activity of incorporated ethanol-consuming enzymes until their activity reaches a specific threshold level. When the ethanol-consuming enzyme activity exceeds this threshold, the steady state in the model becomes unstable and the model switches to an oscillation mode caused by the competition between glyceraldehyde phosphate dehydrogenase and ethanol-consuming enzymes for NAD. The amplitude and period of metabolite oscillations first increase with the increase in the activity of the encapsulated enzymes. A further increase in these activities leads to a loss of the glycolysis steady state, and a permanent accumulation of glycolytic intermediates. The oscillation mode and the loss of the steady state can lead to the osmotic destruction of erythrocyte-bioreactors due to an accumulation of intracellular metabolites. Our results demonstrate that the interaction of enzymes encapsulated in erythrocyte-bioreactors with erythrocyte metabolism should be taken into account in order to achieve the optimal efficacy of these bioreactors.


Asunto(s)
Etanol , NAD , Etanol/metabolismo , NAD/metabolismo , Eritrocitos/metabolismo , Glucólisis , Reactores Biológicos , Acetaldehído/metabolismo
6.
J Biol Chem ; 296: 100736, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33933447

RESUMEN

Hydrogen sulfide is synthesized by enzymes involved in sulfur metabolism and oxidized via a dedicated mitochondrial pathway that intersects with the electron transport chain at the level of complex III. Studies with H2S are challenging since it is volatile and also reacts with oxidized thiols in the culture medium, forming sulfane sulfur species. The half-life of exogenously added H2S to cultured cells is unknown. In this study, we first examined the half-life of exogenously added H2S to human colonic epithelial cells. In plate cultures, H2S disappeared with a t1/2 of 3 to 4 min at 37 °C with a small fraction being trapped as sulfane sulfur species. In suspension cultures, the rate of abiotic loss of H2S was slower, and we demonstrated that sulfide stimulated aerobic glycolysis, which was sensitive to the mitochondrial but not the cytoplasmic NADH pool. Oxidation of mitochondrial NADH using the genetically encoded mito-LbNOX tool blunted the cellular sensitivity to sulfide-stimulated aerobic glycolysis and enhanced its oxidation to thiosulfate. In contrast, sulfide did not affect flux through the oxidative pentose phosphate pathway or the TCA cycle. Knockdown of sulfide quinone oxidoreductase, which commits H2S to oxidation, sensitized cells to sulfide-stimulated aerobic glycolysis. Finally, we observed that sulfide decreased ATP levels in cells. The dual potential of H2S to activate oxidative phosphorylation at low concentrations, but inhibit it at high concentrations, suggests that it might play a role in tuning electron flux and, therefore, cellular energy metabolism, particularly during cell proliferation.


Asunto(s)
Glucólisis , Sulfuro de Hidrógeno/metabolismo , Mitocondrias/metabolismo , NAD/metabolismo , Transducción de Señal , Células HCT116 , Células HT29 , Humanos
7.
J Biol Chem ; 297(2): 100950, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34252456

RESUMEN

Mammalian cells synthesize H2S from sulfur-containing amino acids and are also exposed to exogenous sources of this signaling molecule, notably from gut microbes. As an inhibitor of complex IV in the electron transport chain, H2S can have a profound impact on metabolism, suggesting the hypothesis that metabolic reprogramming is a primary mechanism by which H2S signals. In this study, we report that H2S increases lipogenesis in many cell types, using carbon derived from glutamine rather than from glucose. H2S-stimulated lipid synthesis is sensitive to the mitochondrial NAD(P)H pools and is enabled by reductive carboxylation of α-ketoglutarate. Lipidomics analysis revealed that H2S elicits time-dependent changes across several lipid classes, e.g., upregulating triglycerides while downregulating phosphatidylcholine. Direct analysis of triglyceride concentration revealed that H2S induces a net increase in the size of this lipid pool. These results provide a mechanistic framework for understanding the effects of H2S on increasing lipid droplets in adipocytes and population studies that have pointed to a positive correlation between cysteine (a substrate for H2S synthesis) and fat mass.


Asunto(s)
Glutamina , Sulfuro de Hidrógeno , NAD , Metabolismo Energético , Lipogénesis , Mitocondrias/metabolismo , Transducción de Señal
8.
J Biol Chem ; 295(19): 6299-6311, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32179647

RESUMEN

3-Mercaptopyruvate sulfur transferase (MPST) catalyzes the desulfuration of 3-mercaptopyruvate (3-MP) and transfers sulfane sulfur from an enzyme-bound persulfide intermediate to thiophilic acceptors such as thioredoxin and cysteine. Hydrogen sulfide (H2S), a signaling molecule implicated in many physiological processes, can be released from the persulfide product of the MPST reaction. Two splice variants of MPST, differing by 20 amino acids at the N terminus, give rise to the cytosolic MPST1 and mitochondrial MPST2 isoforms. Here, we characterized the poorly-studied MPST1 variant and demonstrated that substitutions in its Ser-His-Asp triad, proposed to serve a general acid-base role, minimally affect catalytic activity. We estimated the 3-MP concentration in murine liver, kidney, and brain tissues, finding that it ranges from 0.4 µmol·kg-1 in brain to 1.4 µmol·kg-1 in kidney. We also show that N-acetylcysteine, a widely-used antioxidant, is a poor substrate for MPST and is unlikely to function as a thiophilic acceptor. Thioredoxin exhibits substrate inhibition, increasing the KM for 3-MP ∼15-fold compared with other sulfur acceptors. Kinetic simulations at physiologically-relevant substrate concentrations predicted that the proportion of sulfur transfer to thioredoxin increases ∼3.5-fold as its concentration decreases from 10 to 1 µm, whereas the total MPST reaction rate increases ∼7-fold. The simulations also predicted that cysteine is a quantitatively-significant sulfane sulfur acceptor, revealing MPST's potential to generate low-molecular-weight persulfides. We conclude that the MPST1 and MPST2 isoforms are kinetically indistinguishable and that thioredoxin modulates the MPST-catalyzed reaction in a physiologically-relevant concentration range.


Asunto(s)
Sulfurtransferasas , Tiorredoxinas , Animales , Catálisis , Células HEK293 , Células Hep G2 , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Especificidad de Órganos , Sulfurtransferasas/química , Sulfurtransferasas/metabolismo , Tiorredoxinas/química , Tiorredoxinas/metabolismo
9.
J Biol Chem ; 294(28): 11011-11022, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31160338

RESUMEN

Hydrogen sulfide (H2S) is a gaseous signaling molecule, which modulates a wide range of mammalian physiological processes. Cystathionine γ-lyase (CSE) catalyzes H2S synthesis and is a potential target for modulating H2S levels under pathophysiological conditions. CSE is inhibited by propargylglycine (PPG), a widely used mechanism-based inhibitor. In this study, we report that inhibition of H2S synthesis from cysteine, but not the canonical cystathionine cleavage reaction catalyzed by CSE in vitro, is sensitive to preincubation of the enzyme with PPG. In contrast, the efficacy of S-3-carboxpropyl-l-cysteine (CPC) a new inhibitor described herein, was not dependent on the order of substrate/inhibitor addition. We observed that CPC inhibited the γ-elimination reaction of cystathionine and H2S synthesis from cysteine by human CSE with Ki values of 50 ± 3 and 180 ± 15 µm, respectively. We noted that CPC spared the other enzymes involved either directly (cystathionine ß-synthase and mercaptopyruvate sulfurtransferase) or indirectly (cysteine aminotransferase) in H2S biogenesis. CPC also targeted CSE in cultured cells, inhibiting transsulfuration flux by 80-90%, as monitored by the transfer of radiolabel from [35S]methionine to GSH. The 2.5 Å resolution crystal structure of human CSE in complex with the CPC-derived aminoacrylate intermediate provided a structural framework for the molecular basis of its inhibitory effect. In summary, our study reveals a previously unknown confounding effect of PPG, widely used to inhibit CSE-dependent H2S synthesis, and reports on an alternative inhibitor, CPC, which could be used as a scaffold to develop more potent H2S biogenesis inhibitors.


Asunto(s)
Cistationina betasintasa/metabolismo , Cistationina gamma-Liasa/metabolismo , Sulfuro de Hidrógeno/metabolismo , Alquinos/metabolismo , Animales , Línea Celular , Cistationina gamma-Liasa/fisiología , Cisteína/farmacología , Glicina/análogos & derivados , Glicina/metabolismo , Humanos , Sulfuro de Hidrógeno/farmacología , Transducción de Señal/efectos de los fármacos , Sulfuros/farmacología
10.
J Biol Chem ; 294(32): 12077-12090, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31213529

RESUMEN

Unlike most other tissues, the colon epithelium is exposed to high levels of H2S derived from gut microbial metabolism. H2S is a signaling molecule that modulates various physiological effects. It is also a respiratory toxin that inhibits complex IV in the electron transfer chain (ETC). Colon epithelial cells are adapted to high environmental H2S exposure as they harbor an efficient mitochondrial H2S oxidation pathway, which is dedicated to its disposal. Herein, we report that the sulfide oxidation pathway enzymes are apically localized in human colonic crypts at the host-microbiome interface, but that the normal apical-to-crypt gradient is lost in colorectal cancer epithelium. We found that sulfide quinone oxidoreductase (SQR), which catalyzes the committing step in the mitochondrial sulfide oxidation pathway and couples to complex III, is a critical respiratory shield against H2S poisoning. H2S at concentrations ≤20 µm stimulated the oxygen consumption rate in colon epithelial cells, but, when SQR expression was ablated, H2S concentrations as low as 5 µm poisoned cells. Mitochondrial H2S oxidation altered cellular bioenergetics, inducing a reductive shift in the NAD+/NADH redox couple. The consequent electron acceptor insufficiency caused uridine and aspartate deficiency and enhanced glutamine-dependent reductive carboxylation. The metabolomic signature of this H2S-induced stress response mapped, in part, to redox-sensitive nodes in central carbon metabolism. Colorectal cancer tissues and cell lines appeared to counter the growth-restricting effects of H2S by overexpressing sulfide oxidation pathway enzymes. Our findings reveal an alternative mechanism for H2S signaling, arising from alterations in mitochondrial bioenergetics that drive metabolic reprogramming.


Asunto(s)
Metabolismo Energético , Sulfuro de Hidrógeno/metabolismo , Mitocondrias/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Colon/citología , Colon/metabolismo , Colon/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Cisteína/química , Cisteína/metabolismo , Metabolismo Energético/efectos de los fármacos , Humanos , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/farmacología , NAD/química , Oxidación-Reducción , Consumo de Oxígeno/efectos de los fármacos , Quinona Reductasas/antagonistas & inhibidores , Quinona Reductasas/genética , Quinona Reductasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
11.
J Biol Chem ; 292(13): 5584-5592, 2017 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-28213526

RESUMEN

Hydrogen sulfide is a cardioprotective signaling molecule but is toxic at elevated concentrations. Red blood cells can synthesize H2S but, lacking organelles, cannot dispose of H2S via the mitochondrial sulfide oxidation pathway. We have recently shown that at high sulfide concentrations, ferric hemoglobin oxidizes H2S to a mixture of thiosulfate and iron-bound polysulfides in which the latter species predominates. Here, we report the crystal structure of human hemoglobin containing low spin ferric sulfide, the first intermediate in heme-catalyzed sulfide oxidation. The structure provides molecular insights into why sulfide is susceptible to oxidation in human hemoglobin but is stabilized against it in HbI, a specialized sulfide-carrying hemoglobin from a mollusk adapted to life in a sulfide-rich environment. We have also captured a second sulfide bound at a postulated ligand entry/exit site in the α-subunit of hemoglobin, which, to the best of our knowledge, represents the first direct evidence for this site being used to access the heme iron. Hydrodisulfide, a postulated intermediate at the junction between thiosulfate and polysulfide formation, coordinates ferric hemoglobin and, in the presence of air, generated thiosulfate. At low sulfide/heme iron ratios, the product distribution between thiosulfate and iron-bound polysulfides was approximately equal. The iron-bound polysulfides were unstable at physiological glutathione concentrations and were reduced with concomitant formation of glutathione persulfide, glutathione disulfide, and H2S. Hence, although polysulfides are unlikely to be stable in the reducing intracellular milieu, glutathione persulfide could serve as a persulfide donor for protein persulfidation, a posttranslational modification by which H2S is postulated to signal.


Asunto(s)
Hemoglobinas/química , Hemoglobinas/metabolismo , Sulfuro de Hidrógeno/metabolismo , Sulfuros/metabolismo , Catálisis , Cristalografía por Rayos X , Humanos , Conformación Molecular , Oxidación-Reducción , Conformación Proteica , Sulfuros/química , Tiosulfatos/metabolismo
12.
Proc Natl Acad Sci U S A ; 111(16): E1610-9, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24706920

RESUMEN

Commensal and pathogenic bacteria must deal with many different stress conditions to survive in and colonize the human gastrointestinal tract. One major challenge that bacteria encounter in the gut is the high concentration of bile salts, which not only aid in food absorption but also act as effective physiological antimicrobials. The mechanism by which bile salts limit bacterial growth is still largely unknown. Here, we show that bile salts cause widespread protein unfolding and aggregation, affecting many essential proteins. Simultaneously, the bacterial cytosol becomes highly oxidizing, indicative of disulfide stress. Strains defective in reducing oxidative thiol modifications, restoring redox homeostasis, or preventing irreversible protein aggregation under disulfide stress conditions are sensitive to bile salt treatment. Surprisingly, cholate and deoxycholate, two of the most abundant and very closely related physiological bile salts, vary substantially in their destabilizing effects on proteins in vitro and cause protein unfolding of different subsets of proteins in vivo. Our results provide a potential mechanistic explanation for the antimicrobial effects of bile salts, help explain the beneficial effects of bile salt mixtures, and suggest that we have identified a physiological source of protein-unfolding disulfide stress conditions in bacteria.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Disulfuros/metabolismo , Desplegamiento Proteico/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Ácidos y Sales Biliares/química , Colatos/química , Colatos/farmacología , Citosol/efectos de los fármacos , Citosol/metabolismo , Ácido Desoxicólico/química , Ácido Desoxicólico/farmacología , Humanos , Oxidación-Reducción/efectos de los fármacos , Estructura Cuaternaria de Proteína
13.
J Biol Chem ; 290(13): 8310-20, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25688092

RESUMEN

A cardioprotectant at low concentrations, H2S is a toxin at high concentrations and inhibits cytochrome c oxidase. A conundrum in H2S homeostasis is its fate in red blood cells (RBCs), which produce H2S but lack the canonical mitochondrial sulfide oxidation pathway for its clearance. The sheer abundance of RBCs in circulation enhances the metabolic significance of their clearance strategy for H2S, necessary to avoid systemic toxicity. In this study, we demonstrate that H2S generation by RBCs is catalyzed by mercaptopyruvate sulfurtransferase. Furthermore, we have discovered the locus of sulfide oxidation in RBCs and describe a new role for an old protein, hemoglobin, which in the ferric or methemoglobin state binds H2S and oxidizes it to a mixture of thiosulfate and hydropolysulfides. Our study reveals a previously undescribed route for the biogenesis of hydropolysulfides, which are increasingly considered important for H2S-based signaling, but their origin in mammalian cells is unknown. An NADPH/flavoprotein oxidoreductase system restores polysulfide-carrying hemoglobin derivatives to ferrous hemoglobin, thus completing the methemoglobin-dependent sulfide oxidation cycle. Methemoglobin-dependent sulfide oxidation in mammals is complex and has similarities to chemistry reported for the dissolution of iron oxides in sulfidic waters and during bioleaching of metal sulfides. The catalytic oxidation of H2S by hemoglobin explains how RBCs maintain low steady-state H2S levels in circulation, and suggests that additional hemeproteins might be involved in sulfide homeostasis in other tissues.


Asunto(s)
Eritrocitos/metabolismo , Sulfuro de Hidrógeno/metabolismo , Sulfuros/metabolismo , Tiosulfatos/metabolismo , Anemia de Células Falciformes/genética , Ditiotreitol/farmacología , Hemoglobina Falciforme/química , Hemoglobina Falciforme/genética , Humanos , Sulfuro de Hidrógeno/química , Cinética , Metahemoglobina/química , Nitratos/farmacología , Oxidación-Reducción , Sustancias Reductoras/farmacología
14.
J Am Chem Soc ; 138(1): 289-99, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26667407

RESUMEN

Hydrogen sulfide (H2S) elicits pleiotropic physiological effects ranging from modulation of cardiovascular to CNS functions. A dominant method for transmission of sulfide-based signals is via posttranslational modification of reactive cysteine thiols to persulfides. However, the source of the persulfide donor and whether its relationship to H2S is as a product or precursor is controversial. The transsulfuration pathway enzymes can synthesize cysteine persulfide (Cys-SSH) from cystine and H2S from cysteine and/or homocysteine. Recently, Cys-SSH was proposed as the primary product of the transsulfuration pathway with H2S representing a decomposition product of Cys-SSH. Our detailed kinetic analyses demonstrate a robust capacity for Cys-SSH production by the human transsulfuration pathway enzymes, cystathionine beta-synthase and γ-cystathionase (CSE) and for homocysteine persulfide synthesis from homocystine by CSE only. However, in the reducing cytoplasmic milieu where the concentration of reduced thiols is significantly higher than of disulfides, substrate level regulation favors the synthesis of H2S over persulfides. Mathematical modeling at physiologically relevant hepatic substrate concentrations predicts that H2S rather than Cys-SSH is the primary product of the transsulfuration enzymes with CSE being the dominant producer. The half-life of the metastable Cys-SSH product is short and decomposition leads to a mixture of polysulfides (Cys-S-(S)n-S-Cys). These in vitro data, together with the intrinsic reactivity of Cys-SSH for cysteinyl versus sulfur transfer, are consistent with the absence of an observable increase in protein persulfidation in cells in response to exogenous cystine and evidence for the formation of polysulfides under these conditions.


Asunto(s)
Cisteína/análogos & derivados , Transducción de Señal , Células Cultivadas , Cromatografía Liquida , Cisteína/biosíntesis , Disulfuros , Cinética , Espectrometría de Masas
15.
J Am Chem Soc ; 138(27): 8476-88, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27310035

RESUMEN

Enzymes in the sulfur network generate the signaling molecule, hydrogen sulfide (H2S), from the amino acids cysteine and homocysteine. Since it is toxic at elevated concentrations, cells are equipped to clear H2S. A canonical sulfide oxidation pathway operates in mitochondria, converting H2S to thiosulfate and sulfate. We have recently discovered the ability of ferric hemoglobin to oxidize sulfide to thiosulfate and iron-bound hydropolysulfides. In this study, we report that myoglobin exhibits a similar capacity for sulfide oxidation. We have trapped and characterized iron-bound sulfur intermediates using cryo-mass spectrometry and X-ray absorption spectroscopy. Further support for the postulated intermediates in the chemically challenging conversion of H2S to thiosulfate and iron-bound catenated sulfur products is provided by EPR and resonance Raman spectroscopy in addition to density functional theory computational results. We speculate that the unusual sensitivity of skeletal muscle cytochrome c oxidase to sulfide poisoning in ethylmalonic encephalopathy, resulting from the deficiency in a mitochondrial sulfide oxidation enzyme, might be due to the concentration of H2S by myoglobin in this tissue.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Mioglobina/metabolismo , Animales , Caballos , Hierro/metabolismo , Cinética , Oxidación-Reducción , Unión Proteica
16.
J Biol Chem ; 289(45): 30901-10, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25225291

RESUMEN

Sulfide oxidation is expected to play an important role in cellular switching between low steady-state intracellular hydrogen sulfide levels and the higher concentrations where the physiological effects are elicited. Yet despite its significance, fundamental questions regarding how the sulfide oxidation pathway is wired remain unanswered, and competing proposals exist that diverge at the very first step catalyzed by sulfide quinone oxidoreductase (SQR). We demonstrate that, in addition to sulfite, glutathione functions as a persulfide acceptor for human SQR and that rhodanese preferentially synthesizes rather than utilizes thiosulfate. The kinetic behavior of these enzymes provides compelling evidence for the flow of sulfide via SQR to glutathione persulfide, which is then partitioned to thiosulfate or sulfite. Kinetic simulations at physiologically relevant metabolite concentrations provide additional support for the organizational logic of the sulfide oxidation pathway in which glutathione persulfide is the first intermediate formed.


Asunto(s)
Sulfuro de Hidrógeno/química , Mitocondrias/metabolismo , Quinona Reductasas/química , Catálisis , Cisteína/química , Citocromos c/química , Escherichia coli/enzimología , Glutatión/química , Homeostasis , Humanos , Concentración de Iones de Hidrógeno , Cinética , Oxidación-Reducción , Oxígeno/química , Espectrofotometría Ultravioleta , Sulfuros/química , Tiosulfato Azufretransferasa/química
17.
Annu Rev Nutr ; 34: 171-205, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25033061

RESUMEN

Hydrogen sulfide (H2S) has emerged as an important signaling molecule with beneficial effects on various cellular processes affecting, for example, cardiovascular and neurological functions. The physiological importance of H2S is motivating efforts to develop strategies for modulating its levels. However, advancement in the field of H2S-based therapeutics is hampered by fundamental gaps in our knowledge of how H2S is regulated, its mechanism of action, and its molecular targets. This review provides an overview of sulfur metabolism; describes recent progress that has shed light on the mechanism of H2S as a signaling molecule; and examines nutritional regulation of sulfur metabolism, which pertains to health and disease.


Asunto(s)
Dieta , Endotelio Vascular/metabolismo , Sulfuro de Hidrógeno/metabolismo , Modelos Biológicos , Transducción de Señal , Compuestos de Azufre/metabolismo , Azufre/metabolismo , Envejecimiento/metabolismo , Aminoácidos Sulfúricos/metabolismo , Animales , Sistema Cardiovascular/enzimología , Sistema Cardiovascular/metabolismo , Sistema Nervioso Central/irrigación sanguínea , Sistema Nervioso Central/enzimología , Sistema Nervioso Central/metabolismo , Estrés del Retículo Endoplásmico , Endotelio Vascular/enzimología , Humanos , Necesidades Nutricionales
18.
Biochim Biophys Acta ; 1822(11): 1671-81, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22820549

RESUMEN

Alzheimer's disease (AD) is associated with impaired glutamate clearance and depressed Na(+)/K(+) ATPase levels in AD brain that might lead to a cellular ion imbalance. To test this hypothesis, [Na(+)] and [K(+)] were analyzed in postmortem brain samples of 12 normal and 16 AD individuals, and in cerebrospinal fluid (CSF) from AD patients and matched controls. Statistically significant increases in [Na(+)] in frontal (25%) and parietal cortex (20%) and in cerebellar [K(+)] (15%) were observed in AD samples compared to controls. CSF from AD patients and matched controls exhibited no differences, suggesting that tissue ion imbalances reflected changes in the intracellular compartment. Differences in cation concentrations between normal and AD brain samples were modeled by a 2-fold increase in intracellular [Na(+)] and an 8-15% increase in intracellular [K(+)]. Since amyloid beta peptide (Aß) is an important contributor to AD brain pathology, we assessed how Aß affects ion homeostasis in primary murine astrocytes, the most abundant cells in brain tissue. We demonstrate that treatment of astrocytes with the Aß 25-35 peptide increases intracellular levels of Na(+) (~2-3-fold) and K(+) (~1.5-fold), which were associated with reduced levels of Na(+)/K(+) ATPase and the Na(+)-dependent glutamate transporters, GLAST and GLT-1. Similar increases in astrocytic Na(+) and K(+) levels were also caused by Aß 1-40, but not by Aß 1-42 treatment. Our study suggests a previously unrecognized impairment in AD brain cell ion homeostasis that might be triggered by Aß and could significantly affect electrophysiological activity of brain cells, contributing to the pathophysiology of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo , Potasio , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Sodio , Enfermedad de Alzheimer/líquido cefalorraquídeo , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiopatología , Transportador 2 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/líquido cefalorraquídeo , Ácido Glutámico/metabolismo , Células HEK293 , Humanos , Células Jurkat , Ratones , Potasio/líquido cefalorraquídeo , Potasio/metabolismo , Sodio/líquido cefalorraquídeo , Sodio/metabolismo
19.
Trends Biochem Sci ; 33(9): 413-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18703339

RESUMEN

Metabolic interdependence between specialized cells in an organ represents a strategy for energy economy by requiring expression of only a subset of pathway genes in a given cell type. In brain, sulfur metabolism exemplifies this principle of metabolic cooperation between glial and neuronal cells and furnishes three key reagents: S-adenosylmethionine, glutathione and taurine. The pathways for glutathione and taurine syntheses depend on metabolic integration between astrocytes and neurons and intersect with the glutamine-glutamate cycle, which underlies glutamatergic synaptic transmission and requires cooperation between these cell types. We propose that underlying waves of glutamate clearance by astrocytes are activation of cystine import and taurine efflux that result, respectively, from a shared transporter and an increase in solute concentration that triggers osmoregulatory responses.


Asunto(s)
Azufre/metabolismo , Transmisión Sináptica/fisiología , Animales , Astrocitos/fisiología , Encéfalo/fisiología , Cistina/metabolismo , Ácido Glutámico/fisiología , Glutatión/biosíntesis , Humanos , Modelos Neurológicos , Neuronas/fisiología , Taurina/metabolismo
20.
bioRxiv ; 2023 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-36993187

RESUMEN

Angiogenic programming in the vascular endothelium is a tightly regulated process to maintain tissue homeostasis and is activated in tissue injury and the tumor microenvironment. The metabolic basis of how gas signaling molecules regulate angiogenesis is elusive. Herein, we report that hypoxic upregulation of NO synthesis in endothelial cells reprograms the transsulfuration pathway and increases H 2 S biogenesis. Furthermore, H 2 S oxidation by mitochondrial sulfide quinone oxidoreductase (SQOR) rather than downstream persulfides, synergizes with hypoxia to induce a reductive shift, limiting endothelial cell proliferation that is attenuated by dissipation of the mitochondrial NADH pool. Tumor xenografts in whole-body WB Cre SQOR fl/fl knockout mice exhibit lower mass and reduced angiogenesis compared to SQOR fl/fl controls. WB Cre SQOR fl/fl mice also exhibit reduced muscle angiogenesis following femoral artery ligation, compared to controls. Collectively, our data reveal the molecular intersections between H 2 S, O 2 and NO metabolism and identify SQOR inhibition as a metabolic vulnerability for endothelial cell proliferation and neovascularization. Highlights: Hypoxic induction of •NO in endothelial cells inhibits CBS and switches CTH reaction specificity Hypoxic interruption of the canonical transsulfuration pathway promotes H 2 S synthesis Synergizing with hypoxia, SQOR deficiency induces a reductive shift in the ETC and restricts proliferationSQOR KO mice exhibit lower neovascularization in tumor xenograft and hind limb ischemia models.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA