Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Proc Natl Acad Sci U S A ; 120(3): e2205044120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36630448

ABSTRACT

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.


Subject(s)
Electron Transport Complex IV , Hydrogen Sulfide , Superoxide Dismutase-1 , Humans , Electron Transport Complex IV/metabolism , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/toxicity , Sulfides/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
2.
Anal Biochem ; 685: 115392, 2024 01 15.
Article in English | MEDLINE | ID: mdl-37967784

ABSTRACT

Sulfur is essential in the inception of life and crucial for maintaining human health. This mineral is primarily supplied through the intake of proteins and is used for synthesizing various sulfur-containing biomolecules. Recent research has highlighted the biological significance of endogenous supersulfides, which include reactive persulfide species and sulfur catenated residues in thiol and proteins. Ingestion of exogenous sulfur compounds is essential for endogenous supersulfide production. However, the content and composition of supersulfides in foods remain unclear. This study investigated the supersulfide profiles of protein-rich foods, including edible animal meat and beans. Quantification of the supersulfide content revealed that natto, chicken liver, and bean sprouts contained abundant supersulfides. In general, the supersulfide content in beans and their derivatives was higher than that in animal meat. The highest proportion (2.15 %) was detected in natto, a traditional Japanese fermented soybean dish. These results suggest that the abundance of supersulfides, especially in foods like natto and bean sprouts, may contribute to their health-promoting properties. Our findings may have significant biological implications and warrant developing novel dietary intervention for the human health-promoting effects of dietary supersulfides abundantly present in protein-rich foods such as natto and bean sprouts.


Subject(s)
Glycine max , Soy Foods , Humans , Meat , Sulfur
3.
Biochem Biophys Res Commun ; 668: 77-81, 2023 08 06.
Article in English | MEDLINE | ID: mdl-37244038

ABSTRACT

Carnosine and anserine were reported to inhibit tyrosine nitration. However, there are no reports on the nitration inhibitory activities of balenine, 2-oxo-carnosine, 2-oxo-anserine, and 2-oxo-balenine. We demonstrated for the first time that these compounds exhibit inhibitory activities against peroxynitrite-dependent tyrosine nitration. 2-Oxo-imidazole dipeptides (2-oxo-IDPs) showed higher inhibitory activity than their precursor IDPs, thereby suggesting that 2-oxo-IDPs may be effective against nitrative stress-related diseases.


Subject(s)
Carnosine , Carnosine/pharmacology , Carnosine/chemistry , Anserine , Peroxynitrous Acid , Dipeptides/pharmacology , Dipeptides/chemistry , Imidazoles/pharmacology , Imidazoles/chemistry , Tyrosine
4.
Int J Mol Sci ; 24(12)2023 Jun 10.
Article in English | MEDLINE | ID: mdl-37373128

ABSTRACT

Cystathionine γ-lyase (CSE) is an enzyme responsible for the biosynthesis of cysteine from cystathionine in the final step of the transsulfuration pathway. It also has ß-lyase activity toward cystine, generating cysteine persulfide (Cys-SSH). The chemical reactivity of Cys-SSH is thought to be involved in the catalytic activity of particular proteins via protein polysulfidation, the formation of -S-(S)n-H on their reactive cysteine residues. The Cys136/171 residues of CSE have been proposed to be redox-sensitive residues. Herein, we investigated whether CSE polysulfidation occurs at Cys136/171 during cystine metabolism. Transfection of wild-type CSE into COS-7 cells resulted in increased intracellular Cys-SSH production, which was significantly increased when Cys136Val or Cys136/171Val CSE mutants were transfected, instead of the wild-type enzyme. A biotin-polyethylene glycol-conjugated maleimide capture assay revealed that CSE polysulfidation occurs at Cys136 during cystine metabolism. In vitro incubation of CSE with CSE-enzymatically synthesized Cys-SSH resulted in the inhibition of Cys-SSH production. In contrast, the mutant CSEs (Cys136Val and Cys136/171Val) proved resistant to inhibition. The Cys-SSH-producing CSE activity of Cys136/171Val CSE was higher than that of the wild-type enzyme. Meanwhile, the cysteine-producing CSE activity of this mutant was equivalent to that of the wild-type enzyme. It is assumed that Cys-SSH-producing CSE activity could be auto-inactivated via the polysulfidation of the enzyme during cystine metabolism. Thus, the polysulfidation of CSE at the Cys136 residue may be an integral feature of cystine metabolism, which functions to down-regulate Cys-SSH synthesis by the enzyme.


Subject(s)
Cystathionine gamma-Lyase , Hydrogen Sulfide , Cystathionine gamma-Lyase/genetics , Cystathionine gamma-Lyase/metabolism , Cystine/metabolism , Cysteine/metabolism , Proteins/metabolism , Oxidation-Reduction , Hydrogen Sulfide/metabolism
5.
Nitric Oxide ; 120: 44-52, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35033681

ABSTRACT

We previously demonstrated different expression patterns of the neuronal nitric oxide synthase (nNOS) splicing variants, nNOS-µ and nNOS-α, in the rat brain; however, their exact functions have not been fully elucidated. In this study, we compared the enzymatic activities of nNOS-µ and nNOS-α and investigated intracellular redox signaling in nNOS-expressing PC12 cells, stimulated with a neurotoxicant, 1-methyl-4-phenylpyridinium ion (MPP+), to enhance the nNOS uncoupling reaction. Using in vitro studies, we show that nNOS-µ produced nitric oxide (NO), as did nNOS-α, in the presence of tetrahydrobiopterin (BH4), an important cofactor for the enzymatic activity. However, nNOS-µ generated more NO and less superoxide than nNOS-α in the absence of BH4. MPP + treatment induced more reactive oxygen species (ROS) production in nNOS-α-expressing PC12 cells than in those expressing nNOS-µ, which correlated with the intracellular production of 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), a downstream messenger of nNOS redox signaling, and apoptosis in these cells. Furthermore, post-treatment with 8-nitro-cGMP aggravated MPP+-induced cytotoxicity via activation of the H-Ras/extracellular signal-regulated kinase signaling pathway. In conclusion, our results provide strong evidence that nNOS-µ exhibits distinctive enzymatic properties of NO/ROS production, contributing to the regulation of intracellular redox signaling, including the downstream production of 8-nitro-cGMP.


Subject(s)
Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Superoxides/metabolism , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Apoptosis/drug effects , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidation-Reduction , PC12 Cells , Phosphorylation/drug effects , Protein Isoforms/metabolism , Rats
6.
Biosci Biotechnol Biochem ; 86(11): 1576-1580, 2022 Oct 20.
Article in English | MEDLINE | ID: mdl-35977395

ABSTRACT

2-Oxo-imidazole dipeptides (2-oxo-IDPs) are highly functional, but it is unclear whether 2-oxo-IDPs exist in meat. Here, we measured 2-oxo-IDPs levels in meat and observed that they varied according to animal species and body parts. In addition, 2-oxo-IDPs in chicken breast extract increased after aeration in the presence of CuSO4/ascorbate, suggesting the potential of elevated 2-oxo-IDPs in effective usage of meat.


Subject(s)
Carnosine , Dipeptides , Animals , Chickens , Meat/analysis , Imidazoles
7.
J Biol Chem ; 294(4): 1279-1289, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30504220

ABSTRACT

Imidazole-containing dipeptides (IDPs), such as carnosine and anserine, are found exclusively in various animal tissues, especially in the skeletal muscles and nerves. IDPs have antioxidant activity because of their metal-chelating and free radical-scavenging properties. However, the underlying mechanisms that would fully explain IDP antioxidant effects remain obscure. Here, using HPLC-electrospray ionization-tandem MS analyses, we comprehensively investigated carnosine and its related small peptides in the soluble fractions of mouse tissue homogenates and ubiquitously detected 2-oxo-histidine-containing dipeptides (2-oxo-IDPs) in all examined tissues. We noted enhanced production of the 2-oxo-IDPs in the brain of a mouse model of sepsis-associated encephalopathy. Moreover, in SH-SY5Y human neuroblastoma cells stably expressing carnosine synthase, H2O2 exposure resulted in the intracellular production of 2-oxo-carnosine, which was associated with significant inhibition of the H2O2 cytotoxicity. Notably, 2-oxo-carnosine showed a better antioxidant activity than endogenous antioxidants such as GSH and ascorbate. Mechanistic studies indicated that carnosine monooxygenation is mediated through the formation of a histidyl-imidazole radical, followed by the addition of molecular oxygen. Our findings reveal that 2-oxo-IDPs are metal-catalyzed oxidation products present in vivo and provide a revised paradigm for understanding the antioxidant effects of the IDPs.


Subject(s)
Antioxidants/pharmacology , Carnosine/pharmacology , Dipeptides/pharmacology , Histidine/chemistry , Neuroblastoma/pathology , Animals , Antioxidants/chemistry , Carnosine/chemistry , Cell Survival , Dipeptides/chemistry , Humans , Hydrogen Peroxide/pharmacology , Imidazoles/chemistry , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal , Neuroblastoma/drug therapy , Oxidants/pharmacology , Oxidation-Reduction , Tumor Cells, Cultured
8.
Biochem Biophys Res Commun ; 526(1): 225-230, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32201073

ABSTRACT

Nitric oxide (NO)-mediated production of cyclic guanosine 3',5'-monophosphate (cGMP) is a crucial signaling pathway that controls a wide array of neuronal functions, including exocytotic neurotransmitter release. A novel nitrated derivative of cGMP, 8-nitro-cGMP, not only activates cGMP-dependent protein kinase (PKG), but also has membrane permeability and redox activity to produce superoxide and S-guanylated protein. To date, no studies have addressed the effects of 8-nitro-cGMP on exocytotic kinetics. Here, we aimed to assess the 8-nitro-cGMP-mediated modulation of the depolarization-evoked catecholamine release from bovine chromaffin cells. 8-Nitro-cGMP was produced in bovine chromaffin cells dependent on NO donor. Amperometric analysis revealed that 8-nitro-cGMP modulated the kinetic parameters of secretory spikes from chromaffin cells, particularly decreased the speed of individual spikes, resulting in a reduced amperometric spike height, slope ß, and absolute value of slope γ. The modulatory effects were independent of the PKG signal and superoxide production. This is the first study to demonstrate that 8-nitro-cGMP modulates exocytosis and provide insights into a novel regulatory mechanism of exocytosis.


Subject(s)
Adrenal Glands/cytology , Chromaffin Cells/cytology , Cyclic GMP/analogs & derivatives , Exocytosis/drug effects , Animals , Catecholamines/metabolism , Cattle , Cerebellum/cytology , Chromaffin Cells/drug effects , Chromaffin Cells/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Free Radical Scavengers/pharmacology , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Protein Kinase Inhibitors/pharmacology , Superoxides/metabolism
9.
Biochem Biophys Res Commun ; 511(1): 141-147, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30773263

ABSTRACT

We previously reported that 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) is endogenously produced via nitric oxide/reactive oxygen species signaling pathways and it reacts with protein thiol residues to add cGMP structure to proteins through S-guanylation. S-Guanylation occurs on synaptosomal-associated protein 25 (SNAP-25), which is a part of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex that regulates exocytosis. However, the biological relevance of 8-nitro-cGMP in the nervous system remains unclear. Here, we investigated the effects of intracerebroventricular (icv) infusion of 8-nitro-cGMP on mouse brain functions. The results of an open-field test and fear-conditioning task revealed that icv infusion of 8-nitro-cGMP decreased the vertical activity and context-dependent fear memory of mice, which are both associated with the hippocampus. Immunohistochemical analysis revealed increased c-Fos-positive cells in the dentate gyrus in 8-nitro-cGMP-infused mice. Further, biochemical analyses showed that icv infusion of 8-nitro-cGMP increased S-guanylated proteins including SNAP-25 and SNARE complex formation as well as decreased complexes containing complexin, which regulates exocytosis by binding to the SNARE complex, in the hippocampus. These findings suggest that accumulation of 8-nitro-cGMP in the hippocampus affects its functions, including memory, via S-guanylation of hippocampal proteins such as SNAP-25.


Subject(s)
Cyclic GMP/analogs & derivatives , Fear , Memory , Animals , Brain/physiology , Conditioning, Classical , Cyclic GMP/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Synaptosomal-Associated Protein 25/metabolism
10.
Biochem Biophys Res Commun ; 495(3): 2165-2170, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29258821

ABSTRACT

To investigate the role of nitric oxide (NO)/reactive oxygen species (ROS) redox signaling in Parkinson's disease-like neurotoxicity, we used 1-methyl-4-phenylpyridinium (MPP+) treatment (a model of Parkinson's disease). We show that MPP+-induced neurotoxicity was dependent on ROS from neuronal NO synthase (nNOS) in nNOS-expressing PC12 cells (NPC12 cells) and rat cerebellar granule neurons (CGNs). Following MPP+ treatment, we found production of 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), a second messenger in the NO/ROS redox signaling pathway, in NPC12 cells and rat CGNs, that subsequently induced S-guanylation and activation of H-Ras. Additionally, following MPP+ treatment, extracellular signal-related kinase (ERK) phosphorylation was enhanced. Treatment with a mitogen-activated protein kinase (MAPK)/ERK kinase (MEK) inhibitor attenuated MPP+-induced ERK phosphorylation and neurotoxicity. In conclusion, we demonstrate for the first time that NO/ROS redox signaling via 8-nitro-cGMP is involved in MPP+-induced neurotoxicity and that 8-nitro-cGMP activates H-Ras/ERK signaling. Our results indicate a novel mechanism underlying MPP+-induced neurotoxicity, and therefore contribute novel insights to the mechanisms underlying Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium , Cerebellum/metabolism , Cyclic GMP/analogs & derivatives , Neurons/metabolism , Nitric Oxide/metabolism , Parkinsonian Disorders/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Cerebellum/drug effects , Cerebellum/pathology , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Neurons/drug effects , Neurons/pathology , Neurotoxins , PC12 Cells , Parkinsonian Disorders/chemically induced , Rats
11.
Biochem J ; 474(15): 2547-2562, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28637792

ABSTRACT

Reactive sulfur species (RSS) modulate protein functions via S-polysulfidation of reactive Cys residues. Here, we report that Ca2+/calmodulin (CaM)-dependent protein kinase IV (CaMKIV) was reversibly inactivated by RSS via polysulfidation of the active-site Cys residue. CaMKIV is phosphorylated at Thr196 by its upstream CaMK kinase (CaMKK), resulting in the induction of its full activity. In vitro incubation of CaMKIV with the exogenous RSS donors Na2S n (n = 2-4) resulted in dose-dependent inhibition of the CaMKK-induced phospho-Thr196 and consequent inactivation of the enzyme activity. Conversely, mutated CaMKIV (C198V) was refractory to the Na2S n -induced enzyme inhibition. A biotin-polyethylene glycol-conjugated maleimide capture assay revealed that Cys198 in CaMKIV represents a target for S-polysulfidation. Furthermore, phosho-Thr196 and CaMKIV activity were inhibited by incubation with cysteine hydropersulfide, a newly identified RSS that is generated from cystine by cystathionine-γ-lyase. In transfected cells expressing CaMKIV, ionomycin-induced CaMKIV phosphorylation at Thr196 was decreased upon treatment with either Na2S4 or the endoplasmic reticulum (ER) stress inducer thapsigargin, whereas cells expressing mutant CaMKIV (C198V) were resistant to this treatment. In addition, the ionomycin-induced phospho-Thr196 of endogenous CaMKIV was also inhibited by treatment either with Na2S4 or thapsigargin in Jurkat T lymphocytes. Taken together, these data define a novel signaling function for intracellular RSS in inhibiting CaMKIV activity via S-polysulfidation of its Cys198 during the response to ER stress.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cysteine/metabolism , Sulfides/metabolism , Sulfur/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4/antagonists & inhibitors , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation/drug effects , HEK293 Cells , Humans , Jurkat Cells , Mass Spectrometry , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphothreonine/metabolism , Rats , Thapsigargin/pharmacology
12.
Biochem J ; 474(7): 1149-1162, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28126743

ABSTRACT

We previously demonstrated different spacial expression profiles of the neuronal nitric oxide (NO) synthase (nNOS) splice variants nNOS-µ and nNOS-α in the brain; however, their exact functions are not fully understood. Here, we used electron paramagnetic resonance to compare the electron-uncoupling reactions of recombinant nNOS-µ and nNOS-α that generate reactive oxygen species (ROS), in this case superoxide. nNOS-µ generated 44% of the amount of superoxide that nNOS-α generated. We also evaluated the ROS production in HEK293 cells stably expressing nNOS-α and nNOS-µ by investigating these electron-uncoupling reactions as induced by calcium ionophore A23187. A23187 treatment induced greater ROS production in HEK293 cells expressing nNOS-α than those expressing nNOS-µ. Also, immunocytochemical analysis revealed that A23187-treated cells expressing nNOS-α produced more 8-nitroguanosine 3',5'-cyclic monophosphate, a second messenger in NO/ROS redox signaling, than did the cells expressing nNOS-µ. Molecular evolutionary analysis revealed that the ratio of nonsynonymous sites to synonymous sites for the nNOS-µ-specific region was higher than that for the complete gene, indicating that this region has fewer functional constraints than does the complete gene. These observations shed light on the physiological relevance of the nNOS-µ variant and may improve understanding of nNOS-dependent NO/ROS redox signaling and its pathophysiological consequences in neuronal systems.


Subject(s)
Alternative Splicing , Cyclic GMP/analogs & derivatives , Electrons , Nitric Oxide Synthase Type I/metabolism , Superoxides/metabolism , Amino Acid Sequence , Animals , Base Sequence , Calcimycin/pharmacology , Cloning, Molecular , Cyclic GMP/biosynthesis , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HEK293 Cells , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Nitric Oxide Synthase Type I/genetics , Oxidation-Reduction/drug effects , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Transfection
13.
Chem Res Toxicol ; 30(9): 1673-1684, 2017 09 18.
Article in English | MEDLINE | ID: mdl-28837763

ABSTRACT

Electrophiles such as methylmercury (MeHg) affect cellular functions by covalent modification with endogenous thiols. Reactive persulfide species were recently reported to mediate antioxidant responses and redox signaling because of their strong nucleophilicity. In this study, we used MeHg as an environmental electrophile and found that exposure of cells to the exogenous electrophile elevated intracellular concentrations of the endogenous electrophilic molecule 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), accompanied by depletion of reactive persulfide species and 8-SH-cGMP which is a metabolite of 8-nitro-cGMP. Exposure to MeHg also induced S-guanylation and activation of H-Ras followed by injury to cerebellar granule neurons. The electrophile-induced activation of redox signaling and the consequent cell damage were attenuated by pretreatment with a reactive persulfide species donor. In conclusion, exogenous electrophiles such as MeHg with strong electrophilicity impair the redox signaling regulatory mechanism, particularly of intracellular reactive persulfide species and therefore lead to cellular pathogenesis. Our results suggest that reactive persulfide species may be potential therapeutic targets for attenuating cell injury by electrophiles.


Subject(s)
Methylmercury Compounds/chemistry , Sulfides/chemistry , Animals , Antibodies/immunology , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cyclic GMP/analogs & derivatives , Cyclic GMP/chemistry , Cyclic GMP/immunology , Cyclic GMP/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Immunohistochemistry , Male , Methylmercury Compounds/analysis , Methylmercury Compounds/toxicity , Microscopy, Fluorescence , Naphthoquinones/chemistry , Naphthoquinones/toxicity , Nitric Oxide/analysis , Oxidation-Reduction , PC12 Cells , Rats , Rats, Wistar , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Spectrometry, Mass, Electrospray Ionization , Sulfides/pharmacology , ras Proteins/genetics , ras Proteins/metabolism
14.
Biosci Biotechnol Biochem ; 81(8): 1607-1611, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28608752

ABSTRACT

We recently reported that two water-soluble derivatives of ferulic acid (1-feruloyl glycerol, 1-feruloyl diglycerol) previously developed by our group exhibited protective effects against amyloid-ß-induced neurodegeneration in vitro and in vivo. In the current study, we aimed to further understand this process by examining the derivatives' ability to suppress abnormal activation of astrocytes, the key event of neurodegeneration. We investigated the effects of ferulic acid (FA) derivatives on nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression in rat primary astrocytes. The results showed that these compounds inhibited NO production and iNOS expression in a concentration-dependent manner and that the mechanism underlying these effects was the suppression of the nuclear factor-κB pathway. This evidence suggests that FA and its derivatives may be effective neuroprotective agents and could be useful in the treatment of neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease.


Subject(s)
Astrocytes/drug effects , Coumaric Acids/pharmacology , Monoglycerides/pharmacology , NF-kappa B/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/pharmacology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Coumaric Acids/chemistry , Dose-Response Relationship, Drug , Embryo, Mammalian , Gene Expression Regulation , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Monoglycerides/chemistry , NF-kappa B/genetics , NF-kappa B/metabolism , Neuroprotective Agents/chemistry , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Primary Cell Culture , Rats , Signal Transduction , Solubility
15.
Proc Natl Acad Sci U S A ; 111(21): 7606-11, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24733942

ABSTRACT

Using methodology developed herein, it is found that reactive persulfides and polysulfides are formed endogenously from both small molecule species and proteins in high amounts in mammalian cells and tissues. These reactive sulfur species were biosynthesized by two major sulfurtransferases: cystathionine ß-synthase and cystathionine γ-lyase. Quantitation of these species indicates that high concentrations of glutathione persulfide (perhydropersulfide >100 µM) and other cysteine persulfide and polysulfide derivatives in peptides/proteins were endogenously produced and maintained in the plasma, cells, and tissues of mammals (rodent and human). It is expected that persulfides are especially nucleophilic and reducing. This view was found to be the case, because they quickly react with H2O2 and a recently described biologically generated electrophile 8-nitroguanosine 3',5'-cyclic monophosphate. These results indicate that persulfides are potentially important signaling/effector species, and because H2S can be generated from persulfide degradation, much of the reported biological activity associated with H2S may actually be that of persulfides. That is, H2S may act primarily as a marker for the biologically active of persulfide species.


Subject(s)
Cysteine/analogs & derivatives , Disulfides/metabolism , Oxidative Stress/physiology , Signal Transduction/physiology , Sulfhydryl Compounds/metabolism , Animals , Chromatography, Liquid , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Cysteine/biosynthesis , Cysteine/metabolism , Humans , Hydrogen Peroxide/metabolism , Mice , Oxidation-Reduction , Tandem Mass Spectrometry
16.
J Biol Chem ; 290(23): 14493-503, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25882840

ABSTRACT

In addition to its role in DNA repair, nuclear poly(ADP-ribose) polymerase-1 (PARP-1) mediates brain damage when it is over-activated by oxidative/nitrosative stress. Nonetheless, it remains unclear how PARP-1 is activated in neuropathological contexts. Here we report that PARP-1 interacts with a pool of glyceradehyde-3-phosphate dehydrogenase (GAPDH) that translocates into the nucleus under oxidative/nitrosative stress both in vitro and in vivo. A well conserved amino acid at the N terminus of GAPDH determines its protein binding with PARP-1. Wild-type (WT) but not mutant GAPDH, that lacks the ability to bind PARP-1, can promote PARP-1 activation. Importantly, disrupting this interaction significantly diminishes PARP-1 overactivation and protects against both brain damage and neurological deficits induced by middle cerebral artery occlusion/reperfusion in a rat stroke model. Together, these findings suggest that nuclear GAPDH is a key regulator of PARP-1 activity, and its signaling underlies the pathology of oxidative/nitrosative stress-induced brain damage including stroke.


Subject(s)
Brain/pathology , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Oxidative Stress , Poly(ADP-ribose) Polymerases/metabolism , Amino Acid Sequence , Animals , Brain/blood supply , Brain/enzymology , Brain/metabolism , Cell Line , Cell Nucleus/enzymology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Enzyme Activation , Glyceraldehyde-3-Phosphate Dehydrogenases/analysis , Humans , Infarction, Middle Cerebral Artery/enzymology , Male , Models, Molecular , Molecular Sequence Data , Nitro Compounds/analysis , Nitro Compounds/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/analysis , Rats , Rats, Wistar
17.
Biochem Biophys Res Commun ; 478(1): 7-11, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27473654

ABSTRACT

8-Nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) is a nitrated cGMP derivative formed in response to nitric oxide (NO) and reactive oxygen species (ROS). It can cause a post-translational modification (PTM) of protein thiols through cGMP adduction (protein S-guanylation). Accumulating evidence has suggested that, in mammals, S-guanylation of redox-sensor proteins may implicate in regulation of adaptive responses against ROS-associated oxidative stress. Occurrence as well as protein targets of S-guanylation in bacteria remained unknown, however. Here we demonstrated, for the first time, the endogenous occurrence of protein S-guanylation in Escherichia coli (E. coli). Western blotting using anti-S-guanylation antibody clearly showed that multiple proteins were S-guanylated in E. coli. Interestingly, some of those proteins were more intensely S-guanylated when bacteria were cultured under static culture condition than shaking culture condition. It has been known that E. coli is deficient of guanylate cyclase, an enzyme indispensable for 8-nitro-cGMP formation in mammals. We found that adenylate cyclase from E. coli potentially catalyzed 8-nitro-cGMP formation from its precursor 8-nitroguanosine 5'-triphosphate. More importantly, E. coli lacking adenylate cyclase showed significantly reduced formation of S-guanylated proteins. Our S-guanylation proteomics successfully identified S-guanylation protein targets in E. coli, including chaperons, ribosomal proteins, and enzymes which associate with protein synthesis, redox regulation and metabolism. Understanding of functional impacts for protein S-guanylation in bacterial signal transduction is necessary basis for development of potential chemotherapy and new diagnostic strategy for control of pathogenic bacterial infections.


Subject(s)
Cyclic GMP/analogs & derivatives , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Adenylyl Cyclases/metabolism , Cyclic GMP/metabolism , Nitric Oxide/metabolism , Nitrite Reductases/metabolism , Oxidation-Reduction , Oxidative Stress , Protein Processing, Post-Translational , Proteomics , Reactive Oxygen Species/metabolism , Signal Transduction
18.
Arch Biochem Biophys ; 595: 140-6, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27095231

ABSTRACT

Reactive oxygen (oxidant) and free radical species are known to cause nonspecific damage of various biological molecules. The oxidant toxicology is developing an emerging concept of the physiological functions of reactive oxygen species in cell signaling regulation. Redox signaling is precisely modulated by endogenous electrophilic substances that are generated from reactive oxygen species during cellular oxidative stress responses. Among diverse electrophilic molecular species that are endogenously generated, 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP) is a unique second messenger whose formation, signaling, and metabolism in cells was recently clarified. Most important, our current studies revealed that reactive cysteine persulfides that are formed abundantly in cells are critically involved in the metabolism of 8-nitro-cGMP. Modern redox biology involves frontiers of cell research and stem cell research; medical and clinical investigations of infections, cancer, metabolic syndrome, aging, and neurodegenerative diseases; and other fields. 8-Nitro-cGMP-mediated signaling and metabolism in cells may therefore be potential targets for drug development, which may lead to discovery of new therapeutic agents for many diseases.


Subject(s)
Cysteine/metabolism , Nucleotides, Cyclic/metabolism , Signal Transduction , Sulfides/metabolism , Animals , Humans , Oxidation-Reduction , Second Messenger Systems
19.
Biosci Biotechnol Biochem ; 80(3): 547-53, 2016.
Article in English | MEDLINE | ID: mdl-26540606

ABSTRACT

Ferulic acid (FA) has been reported to exhibit protective effects against amyloid-ß (Aß)-induced neurodegeneration in vitro and in vivo. Recently, we developed two water-soluble FA derivatives: 1-feruloyl glycerol and 1-feruloyl diglycerol. In this study, we examined the neuroprotective effects of these water-soluble FA derivatives on Aß-induced neurodegeneration both in vitro and in vivo. FA and water-soluble FA derivatives inhibited Aß aggregation and destabilized pre-aggregated Aß to a similar extent. Furthermore, water-soluble FA derivatives, as well as FA, inhibited Aß-induced neuronal cell death in cultured neuronal cells. In in vivo experiments, oral administration of water-soluble FA derivatives to mice improved Aß-induced dysmnesia assessed by contextual fear conditioning test and protected hippocampal neurons against Aß-induced neurotoxicity. This study provides useful evidence suggesting that water-soluble FA derivatives are expected to be effective neuroprotective agents.


Subject(s)
Amyloid beta-Peptides/physiology , Cell Death/physiology , Coumaric Acids/chemistry , Neurons/cytology , Amyloid beta-Peptides/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Solubility , Water
20.
Biosci Biotechnol Biochem ; 80(7): 1403-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26967587

ABSTRACT

Naringin (Nar) has antioxidant and anti-inflammatory properties. It was recently reported that enzymatic modification of Nar enhanced its functions. Here, we acylated Nar with fatty acids of different sizes (C2-C18) using immobilized lipase from Rhizomucor miehei and investigated the anti-inflammatory effects of these molecules. Treatment of murine macrophage RAW264.7 cells with Nar alkyl esters inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO) production, with Nar lauroyl ester (Nar-C12) showing the strongest effect. Furthermore, Nar-C12 suppressed the LPS-induced expression of inducible NO synthase by blocking the phosphorylation of inhibitor of nuclear factor (NF)-κB-α as well as the nuclear translocation of NF-κB subunit p65 in macrophage cells. Analysis of Nar-C12 uptake in macrophage cells revealed that Nar-C12 ester bond was partially degraded in the cell membrane and free Nar was translocated to the cytosol. These results indicate that Nar released from Nar-C12 exerts anti-inflammatory effects by suppressing NF-κB signaling pathway.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Flavanones/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Macrophages/drug effects , Nitric Oxide/antagonists & inhibitors , Transcription Factor RelA/antagonists & inhibitors , Acylation , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Enzymes, Immobilized/chemistry , Esters/chemistry , Fatty Acids/chemistry , Flavanones/chemistry , Fungal Proteins/chemistry , Gene Expression Regulation , Lipase/chemistry , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Rhizomucor/chemistry , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL