Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 189
Filter
1.
Int J Mol Sci ; 23(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35054786

ABSTRACT

Antioxidant N-tert-Butyl-α-phenylnitron (PBN) partly protected embryos from the negative effects of a DNA demethylating drug 5-azacytidine during pregnancy. Our aim was to investigate PBN's impact on the placenta. Fischer rat dams were treated on gestation days (GD) 12 and 13 by PBN (40 mg/kg), followed by 5azaC (5 mg/kg) after one hour. Global methylation was assessed by pyrosequencing. Numerical density was calculated from immunohistochemical expression in single cells for proliferating (PCNA), oxidative (oxoguanosine) and nitrosative (nitrotyrosine) activity. Results were compared with the PBN-treated and control rats. PBN-pretreatment significantly increased placental weight at GD15 and GD20, diminished by 5azaC, and diminished apoptosis in GD 20 placentas caused by 5azaC. Oxoguanosine expression in placentas of 5azaC-treated dams was especially high in the placental labyrinth on GD 15, while PBN-pretreatment lowered its expression on GD 15 and GD 20 in both the labyrinth and basal layer. 5azaC enhanced nitrotyrosine level in the labyrinth of both gestational stages, while PBN-pretreatment lowered it. We conclude that PBN exerted its prophylactic activity against DNA hypomethylating agent 5azaC in the placenta through free radical scavenging, especially in the labyrinthine part of the placenta until the last day of pregnancy.


Subject(s)
Azacitidine/toxicity , Cyclic N-Oxides/pharmacology , DNA Methylation/drug effects , Oxidative Stress , Placenta/pathology , 8-Hydroxy-2'-Deoxyguanosine/metabolism , Animals , Biomarkers/metabolism , Cell Proliferation/drug effects , Female , Nitrosation/drug effects , Organ Size/drug effects , Oxidative Stress/drug effects , Placenta/drug effects , Pregnancy , Proliferating Cell Nuclear Antigen/metabolism , Rats, Inbred F344 , Tyrosine/analogs & derivatives , Tyrosine/metabolism
2.
J Pharmacol Exp Ther ; 376(3): 444-453, 2021 03.
Article in English | MEDLINE | ID: mdl-33384302

ABSTRACT

Currently available tocolytics are ineffective at significantly delaying preterm birth. This is due in part to our failure to better understand the mechanisms that drive spontaneous preterm labor (sPTL). Cyclic nucleotides are not the primary contributors to myometrial quiescence, but instead nitric oxide (NO)-mediated protein S-nitrosation (SNO) is integral to the relaxation of the tissue. Connexin-43 (Cx43), a myometrial "contractile-associated protein" that functions as either a gap junction channel or an hemichannel (HC), was the focus of this study. Protein analysis determined that Cx43 is downregulated in sPTL myometrium. Furthermore, Cx43 is S-nitrosated by NO, which correlates with an increase of phosphorylated Cx43 at serine 368 (Cx43-pS368 -gap junction inhibition) as well as an increase in the HC open-state probability (quiescence). Pharmacologic inhibition of Cx43 with 18ß-glycyrrhetinic acid (18ß-GA) exhibits a negative inotropic effect on the myometrium in a dose-dependent manner, as does administration of nebivolol, an NO synthase activator that increases total protein SNOs. When 18ß-GA and nebivolol were coadministered at their IC50 values, the effect on contractile dynamics was additive and all but eliminated contractions. The development of new tocolytics demands a better understanding of the underlying mechanisms of sPTL. Here it has been shown that 18ß-GA and nebivolol leverage dysregulated pathways in the myometrium, resulting in a novel approach for the treatment of sPTL. SIGNIFICANCE STATEMENT: Although there are many known causes of preterm labor (PTL), the mechanisms of "spontaneous" PTL (sPTL) remain obfuscated, which is why treating this condition is so challenging. Here we have identified that connexin-43 (Cx43), an important contractile-associated protein, is dysregulated in sPTL myometrium and that the pharmacologic inhibition of Cx43 and its S-nitrosation with 18ß-glycyrrhetinic acid and nebivolol, respectively, significantly blunts contraction in human myometrial tissue, presenting a novel approach to tocolysis that leverages maladjusted pathways in women who experience sPTL.


Subject(s)
Connexin 43/metabolism , Nitrosation/drug effects , Tocolytic Agents/pharmacology , Animals , Drug Discovery , Drug Interactions , Enzyme Inhibitors/pharmacology , Female , Gap Junctions/drug effects , Guinea Pigs , HEK293 Cells , Humans , Nitric Oxide/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Pregnancy
3.
J Mol Cell Cardiol ; 134: 40-50, 2019 09.
Article in English | MEDLINE | ID: mdl-31226341

ABSTRACT

Although nitrite improves vascular function and lowers blood pressure, its cardiac effects are not completely known. We investigated whether nitrite improves the cardiac function in normotensive and in hypertensive rats. Two-kidney, one-clip hypertension model (2K1C) was induced in Wistar rats. Blood pressure was evaluated by tail-cuff plethysmography over 6 weeks. By the end of week 2, hypertensive and normotensive rats received nitrite (daily dose of 1 or 15 mg/kg) by gavage for 4 weeks. Cardiac morphology and function were performed by transthoracic echocardiography. Intrinsic heart function was evaluated using the isolated heart model (Langendorff's preparation). Starling curves were generated under nitrite (1 µmol/L) and/or ascorbate (1 mmol/L) or vehicle. Cardiac tissue was collected and snap frozen for biochemical analysis. Nitrite treatment (15 mg/kg) lowered both systolic blood pressure and the increases in left ventricular (LV) mass found in 2K1C rats (P < .05). In addition, nitrite treatment restored the decreased cardiac output in 2K1C rats (P < .05) and improved the cardiac function. These findings were associated with increased nitrite, S-nitrosothiols, and protein S-nitrosylation (all P < .05) assessed in heart tissue. The cardiac effects of nitrite were further investigated in the isolated heart model, and nitrite infusion (1 µmol/L) enhanced cardiac contractility and relaxation. This infusion increased S-nitrosothiols concentrations and protein S-nitrosylation in the heart. Ascorbate completely blunted all nitrite-induced effects. These findings show that treatment with oral nitrite improves cardiac function by mechanisms involving increased S-nitrosothiols generation and S-nitrosylation of cardiac proteins. Pharmacological strategies promoting cardiac S-nitrosylation may be useful to improve myocardial function in heart diseases.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/prevention & control , Hypertension/complications , Myocardium/metabolism , Nitrates/metabolism , Sodium Nitrite/pharmacology , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Cardiomyopathies/metabolism , Heart/drug effects , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Male , Myocardium/pathology , Nitrosation/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sodium Nitrite/therapeutic use
4.
Br J Pharmacol ; 176(12): 2049-2062, 2019 06.
Article in English | MEDLINE | ID: mdl-30822355

ABSTRACT

BACKGROUND AND PURPOSE: Angiotensin II (AngII) and NO regulate the cerebral circulation. AngII AT1 receptors exert ligand-dependent and ligand-independent (myogenic tone [MT]) vasoconstriction of cerebral vessels. NO induces post-translational modifications of proteins such as S-nitrosation (redox modification of cysteine residues). In cultured cells, S-nitrosation decreases AngII's affinity for the AT1 receptor. The present work evaluated the functional consequences of S-nitrosation on both AngII-dependent and AngII-independent cerebrovascular responses. EXPERIMENTAL APPROACH: S-Nitrosation was induced in rat isolated middle cerebral arteries by pretreatment with the NO donors, S-nitrosoglutathione (GSNO) or sodium nitroprusside (SNP). Agonist-dependent activation of AT1 receptors was evaluated by obtaining concentration-response curves to AngII. Ligand-independent activation of AT1 receptors was evaluated by calculating MT (active vs. passive diameter) at pressures ranging from 20 to 200 mmHg in the presence or not of a selective AT1 receptor inverse agonist. KEY RESULTS: GSNO or SNP completely abolished the AngII-dependent AT1 receptor-mediated vasoconstriction of cerebral arteries. GSNO had no impact on responses to other vasoconstrictors sharing (phenylephrine, U46619) or not (5-HT) the same signalling pathway. MT was reduced by GSNO, and the addition of losartan did not further decrease MT, suggesting that GSNO blocks AT1 receptor-dependent MT. Ascorbate (which reduces S-nitrosated compounds) restored the response to AngII but not the soluble GC inhibitor ODQ, suggesting that these effects are mediated by S-nitrosation rather than by S-nitrosylation. CONCLUSIONS AND IMPLICATIONS: In rat middle cerebral arteries, GSNO pretreatment specifically affects the AT1 receptor and reduces both AngII-dependent and AngII-independent activation, most likely through AT1 receptor S-nitrosation.


Subject(s)
Cerebral Arteries/drug effects , Receptor, Angiotensin, Type 1/metabolism , S-Nitrosoglutathione/pharmacology , Angiotensin II/pharmacology , Animals , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Ligands , Male , Nitric Oxide/metabolism , Nitrosation/drug effects , Rats , Rats, Wistar , S-Nitrosoglutathione/administration & dosage , Signal Transduction/drug effects , Structure-Activity Relationship
5.
Oxid Med Cell Longev ; 2018: 3509091, 2018.
Article in English | MEDLINE | ID: mdl-30405876

ABSTRACT

The Toll-like receptor-4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway is vital in the pathogenesis of hepatic ischemia/reperfusion (HIR) injury. Dipeptidyl peptidase-4 (DPP4) inhibitors exert protective effects on IR injury of the kidney, heart, and lung; however, their effect on the liver is still unknown. Thus, the purpose of this study was to examine whether pretreatment with vildagliptin (Vilda), a DPP4 inhibitor, produces hepatic protection against IR injury and to investigate its influence on TLR4/NF-κB signaling in a rat model. Thirty male Wistar rats were divided into 3 groups: the sham group: subjected to a sham operation and received normal saline; the HIR group: subjected to HIR and received normal saline; and the Vilda + HIR group: subjected to HIR with pretreatment of 10 mg/kg/day Vilda for 10 days intraperitoneally. Hepatic ischemia lasted for 45 minutes followed by 3-hour reperfusion; then blood and liver samples were collected for biochemical and histopathological examination. The HIR group produced a significant increase in serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), hepatic malondialdehyde (MDA), nitric oxide (NO), and tumor necrosis factor alpha (TNF-α) levels and a significant reduction in the hepatic catalase level in comparison to the sham group. Moreover, a significant upregulation of gene and protein expressions of TLR4, NF-κB, and high-mobility group box-1 (HMGB1) along with caspase-3 protein expression was observed in the HIR group when compared with the sham group. Histopathological examination of the liver from the HIR group showed necrosis, sinusoidal congestion, hemorrhage, and hepatocyte degeneration. Administration of Vilda ameliorated the biochemical and histopathological changes caused by HIR. Vildagliptin showed for the first time a hepatoprotective effect in HIR injury through downregulation of TLR4/NF-κB/HMGB1 and caspase-3 hepatic expressions.


Subject(s)
Adamantane/analogs & derivatives , Liver/blood supply , NF-kappa B/metabolism , Nitriles/therapeutic use , Pyrrolidines/therapeutic use , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Toll-Like Receptor 4/metabolism , Adamantane/pharmacology , Adamantane/therapeutic use , Animals , Apoptosis/drug effects , Biomarkers/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Inflammation Mediators/metabolism , Liver/pathology , Liver/physiopathology , Liver Function Tests , Male , NF-kappa B/genetics , Nitriles/pharmacology , Nitrosation/drug effects , Oxidative Stress/drug effects , Pyrrolidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Reperfusion Injury/genetics , Reperfusion Injury/physiopathology , Toll-Like Receptor 4/genetics , Vildagliptin
6.
J Cell Mol Med ; 22(12): 6391-6395, 2018 12.
Article in English | MEDLINE | ID: mdl-30188599

ABSTRACT

Preterm birth before 37 weeks of completed gestation results in numerous health consequences for the foetus. Preterm labour leads to preterm birth in over 50% of cases, and no FDA-approved treatment can prevent labour or help a foetus remain in the womb until term. Examination of nitric oxide mediated relaxation signaling in the uterine smooth muscle reveals a role for protein S-nitrosation. The recent discovery of upregulated S-nitrosoglutathione reductase (GSNOR) in spontaneously preterm labouring women has emphasized the need to explore the function of S-nitrosation regulation in the maintenance of uterine quiescence. Here we have examined the ability of nebivolol to relax uterine smooth muscle and tested recent claims that nebivolol is a GSNOR inhibitor. In uterine smooth muscle strips from both mouse and human, nebivolol relaxes oxytocin-induced contractions in a dose dependent manner. Our data indicates that nebivolol has no effect on GSNOR activity, nor does nebivolol inhibit thioredoxin reductase, two of the major protein denitrosylases. The ability of nebivolol to relax uterine smooth muscle is likely the combined effects of increased nitric oxide synthase activity and ß3-adregnegic stimulation.


Subject(s)
Aldehyde Oxidoreductases/genetics , Nebivolol/administration & dosage , Obstetric Labor, Premature/drug therapy , Tocolytic Agents/administration & dosage , Aldehyde Oxidoreductases/antagonists & inhibitors , Animals , Female , Humans , Labor, Obstetric/drug effects , Mice , Muscle, Smooth/drug effects , Myometrium/drug effects , Nitric Oxide/genetics , Nitric Oxide/metabolism , Nitrosation/drug effects , Obstetric Labor, Premature/genetics , Obstetric Labor, Premature/physiopathology , Oxidation-Reduction/drug effects , Pregnancy , Premature Birth/drug therapy , Premature Birth/physiopathology , Signal Transduction/genetics , Uterus/drug effects , Uterus/physiopathology
7.
J Pharmacol Sci ; 138(1): 16-22, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30197059

ABSTRACT

Nitrosative/oxidative stress plays an important role in neuronal death following cerebral ischemia/reperfusion (I/R). Chrysophanol (CHR) has been shown to afford significant neuroprotection on ischemic stroke, however, whether its mechanism is related to attenuating nitrosative/oxidative stress is not clear. In the present study, we investigated the effect of CHR on neuronal injury related to nitric oxide (NO) production by using mouse middle cerebral artery occlusion (MCAO) model. Our results revealed that nitrite plus nitrate (NOx-) and 3-nitrotyrosine (3-NT) levels increased in ischemic brain 14 days after reperfusion, and were subsequently attenuated by CHR treatment. Moreover, 3-NT is colocalized with NeuN and TUNEL, suggesting that neuronal apoptosis following I/R is associated with 3-NT and CHR suppresses NO-associated neuronal cell death. Accordingly, cleaved caspase-3 expression in ischemic brain was decreased by CHR treatment. I/R also decreased the activity of total superoxide dismutase (SOD) and manganese-dependent SOD (MnSOD), whilst increased reactive oxygen species (ROS) production significantly. Interestingly, CHR reversed this decrease in total SOD, and MnSOD activity, and inhibited ROS generation in the ischemic brain. Taken together, our results provide direct evidence suggesting that CHR attenuates nitrosative/oxidative stress injury induced by I/R, providing a novel therapeutic target in the treatment of acute ischemic stroke.


Subject(s)
Anthraquinones/pharmacology , Anthraquinones/therapeutic use , Brain Ischemia/metabolism , Neuroprotective Agents , Nitrosation/drug effects , Oxidative Stress/drug effects , Reperfusion Injury/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Caspase 3/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/etiology , Superoxide Dismutase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
8.
Free Radic Biol Med ; 124: 517-524, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30037775

ABSTRACT

BACKGROUND: Establishing sustained reoxygenation/reperfusion ensures not only the recovery, but may initiate a reperfusion injury in which oxidative stress plays a major role. This study offers the mechanism and this mechanism-specific therapeutic strategy against excessive release of reactive oxygen species (ROS) associated with reperfusion-driven recovery of mitochondrial metabolism. AIMS AND METHODS: In neonatal mice subjected to cerebral hypoxia-ischaemia (HI) and reperfusion, we examined conformational changes and activity of mitochondrial complex I with and without post-HI administration of S-nitrosating agent, MitoSNO. Assessment of mitochondrial ROS production, oxidative brain damage, neuropathological and neurofunctional outcomes were used to define neuroprotective strength of MitoSNO. A specificity of reperfusion-driven mitochondrial ROS production to conformational changes in complex I was examined in-vitro. RESULTS: HI deactivated complex I, changing its conformation from active form (A) into the catalytically dormant, de-active form (D). Reperfusion rapidly converted the D-form into the A-form and increased ROS generation. Administration of MitoSNO at the onset of reperfusion, decelerated D→A transition of complex I, attenuated oxidative stress, and significantly improved neurological recovery. In cultured neurons, after simulated ischaemia-reperfusion injury, MitoSNO significantly reduced ROS generation and neuronal mortality. In isolated mitochondria subjected to anoxia-reoxygenation, MitoSNO restricted ROS release during D→A transitions. CONCLUSION: Rapid D→A conformation in response to reperfusion reactivates complex I. This is essential not only for metabolic recovery, but also contributes to excessive release of mitochondrial ROS and reperfusion injury. We propose that the initiation of reperfusion should be followed by pharmacologically-controlled gradual reactivation of complex I.


Subject(s)
Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Hypoxia-Ischemia, Brain/metabolism , Neuroprotective Agents/pharmacology , Reperfusion Injury/metabolism , Animals , Animals, Newborn , Mice , Mice, Inbred C57BL , Nitrosation/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
9.
Biomed Pharmacother ; 99: 1014-1021, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29665642

ABSTRACT

Estrogen plays aprotective effect on the cardiovascular system. Abnormal vascular smooth muscle cells (VSMCs) are involvedin the progress of atherosclerosis. However, the effect of estrogen on VSMCs is still unclear. The purpose of this study is to investigate the effect of 17ß-estradiol (E2), a potent endogeneous estrogen, on the development of atherosclerosis and its potential mechanisms. In vivo, the formation of atherosclerotic lesions and the extent of protein S-nitrosylation were compared between female and male apoE-/- mice to assess the effect of estrogen. It showedthat the gross lesion area of the aorta tree, the lesion area atthe aortic root and the contents of lipids, macrophages and VSMCs were significantly less in female apoE-/- mice than those in male mice (p < 0.05). In addition, compared with male mice, plasma NO level and protein S-nitrosylation level of VSMCs were significantly higher in female mice (p < 0.01). Rat VSMCs (rVSMCs) were successfully isolated. In vitro, the levels of NO and protein S-nitrosylation in rVSMCs with E2 treatment were also assessed. The result revealed that E2 treatment reversed the ox-LDL-induced superoxide anion level increaseand the ox-LDL-induced NO and protein S-nitrosylation levels decrease in rVSMCs. Our data indicated that female is less susceptible to atherosclerosis.It might be becauseestrogenpromotes NO production and down-regulatessuperoxide anion level, which further upregulates the protein S-nitrosylation level of VSMCs against the occurrence and development of atherosclerosis.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/pathology , Estradiol/pharmacology , Proteins/metabolism , Sex Characteristics , Animals , Aorta/pathology , Apolipoproteins E/metabolism , Atherosclerosis/blood , Culture Media, Conditioned/pharmacology , Female , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/blood , Nitrosation/drug effects , Rats, Sprague-Dawley , Superoxides/metabolism
10.
Sci Rep ; 8(1): 5614, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29618799

ABSTRACT

Tocolytics show limited efficacy to prevent preterm delivery. In uterine smooth muscle cGMP accumulation following addition of nitric oxide (NO) has little effect on relaxation suggesting a role for protein S-nitrosation. In human myometrial tissues from women in labor at term (TL), or spontaneously in labor preterm (sPTL), direct stimulation of soluble guanylyl cyclase (sGC) fails to relax myometrium, while the same treatment relaxes vascular smooth muscle completely. Unlike term myometrium, effects of NO are not only blunted in sPTL, but global protein S-nitrosation is also diminished, suggesting a dysfunctional response to NO-mediated protein S-nitrosation. Examination of the enzymatic regulator of endogenous S-nitrosoglutathione availability, S-nitrosoglutathione reductase, reveals increased expression of the reductase in preterm myometrium associated with decreased total protein S-nitrosation. Blockade of S-nitrosoglutathione reductase relaxes sPTL tissue. Addition of NO donor to the actin motility assay attenuates force. Failure of sGC activation to mediate relaxation in sPTL tissues, together with the ability of NO to relax TL, but not sPTL myometrium, suggests a unique pathway for NO-mediated relaxation in myometrium. Our results suggest that examining the action of S-nitrosation on critical contraction associated proteins central to the regulation of uterine smooth muscle contraction can reveal new tocolytic targets.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Nitric Oxide/metabolism , Obstetric Labor, Premature , Actins/metabolism , Aldehyde Oxidoreductases/antagonists & inhibitors , Benzamides/pharmacology , Cyclic GMP/metabolism , Female , Guanylate Cyclase/metabolism , Humans , Muscle, Smooth/physiology , Myometrium/metabolism , Myosins/metabolism , Nitrosation/drug effects , Pregnancy , Pyrroles/pharmacology , S-Nitrosoglutathione/metabolism , Uterine Contraction/drug effects
11.
Life Sci ; 195: 25-32, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29307523

ABSTRACT

AIMS: Propofol can be applied as an anesthetic or sedative agent for septic patients. Our previous studies showed that propofol ameliorated inflammation- and nitrosative stress-induced cellular insults. This study further evaluated effects of propofol on cecal ligation and puncture (CLP)-induced septic insults to rats and its possible mechanisms. MAIN METHODS: Wistar rats were administered with CLP and effects of propofol on CLP-induced liver dysfunction and rat death were evaluated. Levels of hepatic or systemic nitrogen oxides (NOx) and interleukin (IL)-6 were quantified. Sequentially, inducible nitric oxide synthase (iNOS) and IL-6 gene expressions, toll-like receptor 4 (TLR4) protein levels, and nuclear factor (NF)-κB translocation were determined. KEY FINDINGS: Subjecting rats to CLP led to body weight loss, liver weight gain, and death. Administration of propofol lessened CLP-induced augmentations of serum and hepatic nitrosative stress and IL-6 levels. Additionally, propofol suppressed CLP-induced enhancements in levels of hepatic iNOS protein. Furthermore, the CLP-induced iNOS and IL-6 mRNA expressions in the liver were inhibited following propofol administration. Sequentially, subjecting rats to CLP enhanced hepatic TLR4 protein levels and NF-κB translocation to nuclei, but propofol inhibited these augmentations. SIGNIFICANCE: Consequently, exposure to propofol protected against CLP-induced liver dysfunction and increased the survival rates of the animals. This study shows that propofol can protect rats against septic insults through suppression of systemic and hepatic nitrosative and inflammatory stress due to inhibition of TLR4/NF-κB-mediated iNOS and IL-6 mRNA and protein expressions.


Subject(s)
Hepatitis/drug therapy , Hypnotics and Sedatives/therapeutic use , Interleukin-6/biosynthesis , Liver/metabolism , Liver/pathology , NF-kappa B/drug effects , Nitric Oxide Synthase Type II/biosynthesis , Nitrosation/drug effects , Propofol/therapeutic use , Sepsis/drug therapy , Toll-Like Receptor 4/drug effects , Actins/metabolism , Animals , Cecal Diseases/metabolism , Cecal Diseases/pathology , Down-Regulation , Gene Expression Regulation/drug effects , Hepatitis/metabolism , Hepatitis/pathology , Interleukin-6/genetics , Male , Nitric Oxide Synthase Type II/genetics , Rats , Rats, Wistar , Sepsis/metabolism , Sepsis/pathology , Translocation, Genetic/drug effects
12.
Food Chem Toxicol ; 112: 282-289, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29277703

ABSTRACT

Endogenous and dietary nitrite produces reactive nitrogen species (RNS) that react with DNA causing mutations. The nitrosation of 2'-deoxyguanosine (dGuo) and DNA with nitrite was studied under different conditions, and the reaction and degradation products identified and analysed by HPLC-DAD-MS. Nitrosative deamination of dGuo produced xanthine along with 2'-deoxyxanthosine whereas DNA afforded xanthine. Formation of xanthine increased with nitrite concentration and in low pH such as that of stomach. Xanthine was measured as a marker of nitrosation of dGuo and DNA, and it was subsequently used to study the antinitrosating activity of ß-carboline alkaloids, and selected antioxidants. Food-occurring tetrahydro-ß-carbolines (THßCs) decreased nitrosative deamination of dGuo and DNA under conditions simulating the stomach. Antinitrosating activity was also evidenced for flavonoids (catechin, quercetin) and indole (melatonin) antioxidants. Among THßCs the most active antinitrosating compounds were 1,2,3,4-tetrahydro-ß-carboline-3-carboxylic acids (THßC-3-COOHs) that reacted with nitrite to give N-nitroso derivatives as main products along with 3,4-dihydro-ß-carboline-3-carboxylic acids and aromatic ß-carbolines (norharman and harman). Antinitrosating activity of THßCs correlated well with the formation of N-nitroso-THßC-3-COOHs. These N-nitroso derivatives were stable at pH 7 but degraded in acid conditions affording nitrosating species.


Subject(s)
Alkaloids/pharmacology , Antioxidants/pharmacology , Carbolines/pharmacology , DNA/chemistry , Deoxyguanosine/chemistry , Nitrites/chemistry , Alkaloids/chemistry , Antioxidants/chemistry , Carbolines/chemistry , DNA/genetics , Nitrites/toxicity , Nitrosation/drug effects
13.
J Biol Chem ; 292(35): 14486-14495, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28710281

ABSTRACT

Nitrate (NO3-) and nitrite (NO2-) are known to be cardioprotective and to alter energy metabolism in vivo NO3- action results from its conversion to NO2- by salivary bacteria, but the mechanism(s) by which NO2- affects metabolism remains obscure. NO2- may act by S-nitrosating protein thiols, thereby altering protein activity. But how this occurs, and the functional importance of S-nitrosation sites across the mammalian proteome, remain largely uncharacterized. Here we analyzed protein thiols within mouse hearts in vivo using quantitative proteomics to determine S-nitrosation site occupancy. We extended the thiol-redox proteomic technique, isotope-coded affinity tag labeling, to quantify the extent of NO2--dependent S-nitrosation of proteins thiols in vivo Using this approach, called SNOxICAT (S-nitrosothiol redox isotope-coded affinity tag), we found that exposure to NO2- under normoxic conditions or exposure to ischemia alone results in minimal S-nitrosation of protein thiols. However, exposure to NO2- in conjunction with ischemia led to extensive S-nitrosation of protein thiols across all cellular compartments. Several mitochondrial protein thiols exposed to the mitochondrial matrix were selectively S-nitrosated under these conditions, potentially contributing to the beneficial effects of NO2- on mitochondrial metabolism. The permeability of the mitochondrial inner membrane to HNO2, but not to NO2-, combined with the lack of S-nitrosation during anoxia alone or by NO2- during normoxia places constraints on how S-nitrosation occurs in vivo and on its mechanisms of cardioprotection and modulation of energy metabolism. Quantifying S-nitrosated protein thiols now allows determination of modified cysteines across the proteome and identification of those most likely responsible for the functional consequences of NO2- exposure.


Subject(s)
Disease Models, Animal , Mitochondria, Heart/metabolism , Myocardial Ischemia/metabolism , Myocardium/metabolism , Nitrites/metabolism , Protein Processing, Post-Translational , Up-Regulation , Affinity Labels/metabolism , Animals , Cardiotonic Agents/pharmacology , Cell Membrane Permeability/drug effects , Cysteine/metabolism , Female , Heart/drug effects , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Mitochondrial Swelling/drug effects , Myocardial Ischemia/drug therapy , Nitrates/pharmacology , Nitrites/pharmacology , Nitrosation/drug effects , Potassium Compounds/pharmacology , Proteomics/methods , Rats, Wistar , Up-Regulation/drug effects
14.
Mol Cell Biol ; 37(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28320874

ABSTRACT

Nitric oxide (NO) produced by endothelial NO synthase (eNOS) modulates many functions in endothelial cells. S-nitrosylation (SNO) of cysteine residues on ß-catenin by eNOS-derived NO has been shown to influence intercellular contacts between endothelial cells. However, the implication of SNO in the regulation of ß-catenin transcriptional activity is ill defined. Here, we report that NO inhibits the transcriptional activity of ß-catenin and endothelial cell proliferation induced by activation of Wnt/ß-catenin signaling. Interestingly, induction by Wnt3a of ß-catenin target genes, such as the axin2 gene, is repressed in an eNOS-dependent manner by vascular endothelial growth factor (VEGF). We identified Cys466 of ß-catenin as a target for SNO by eNOS-derived NO and as the critical residue for the repressive effects of NO on ß-catenin transcriptional activity. Furthermore, we observed that Cys466 of ß-catenin, located at the binding interface of the ß-catenin-TCF4 transcriptional complex, is essential for disruption of this complex by NO. Importantly, Cys466 of ß-catenin is necessary for the inhibitory effects of NO on Wnt3a-stimulated proliferation of endothelial cells. Thus, our data define the mechanism responsible for the repressive effects of NO on the transcriptional activity of ß-catenin and link eNOS-derived NO to the modulation by VEGF of Wnt/ß-catenin-induced endothelial cell proliferation.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/enzymology , Nitric Oxide Synthase Type III/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Wnt3A Protein/pharmacology , beta Catenin/metabolism , Animals , Cattle , Cell Proliferation/drug effects , Cysteine/metabolism , Endothelial Cells/drug effects , HEK293 Cells , Humans , Mice , Nitric Oxide/metabolism , Nitrosation/drug effects , Transcription, Genetic , Vascular Endothelial Growth Factor A/pharmacology , Wnt Signaling Pathway
15.
PLoS One ; 12(3): e0174542, 2017.
Article in English | MEDLINE | ID: mdl-28350848

ABSTRACT

Cataract, a leading cause of blindness, is of special concern in diabetics as it occurs at earlier onset. Polyol accumulation and increased oxidative-nitrosative stress in cataractogenesis are associated with NFκB activation, iNOS expression, ATP depletion, loss of ATPase functions, calpain activation and proteolysis of soluble to insoluble proteins. Tocotrienol was previously shown to reduce lens oxidative stress and inhibit cataractogenesis in galactose-fed rats. In current study, we investigated anticataract effects of topical tocotrienol and possible mechanisms involved in streptozotocin-induced diabetic rats. Diabetes was induced in Sprague Dawley rats by intraperitoneal injection of streptozotocin. Diabetic rats were treated with vehicle (DV) or tocotrienol (DT). A third group consists of normal, non-diabetic rats were treated with vehicle (NV). All treatments were given topically, bilaterally, twice daily for 8 weeks with weekly slit lamp monitoring. Subsequently, rats were euthanized and lenses were subjected to estimation of polyol accumulation, oxidative-nitrosative stress, NFκB activation, iNOS expression, ATP levels, ATPase activities, calpain activity and total protein levels. Cataract progression was delayed from the fifth week onwards in DT with lower mean of cataract stages compared to DV group (p<0.01) despite persistent hyperglycemia. Reduced cataractogenesis in DT group was accompanied with lower aldose reductase activity and sorbitol level compared to DV group (p<0.01). DT group also showed reduced NFκB activation, lower iNOS expression and reduced oxidative-nitrosative stress compared to DV group. Lenticular ATP and ATPase and calpain 2 activities in DT group were restored to normal. Consequently, soluble to insoluble protein ratio in DT group was higher compared to DV (p<0.05). In conclusion, preventive effect of topical tocotrienol on development of cataract in STZ-induced diabetic rats could be attributed to reduced lens aldose reductase activity, polyol levels and oxidative-nitrosative stress. These effects of tocotrienol invlove reduced NFκB activation, lower iNOS expression, restoration of ATP level, ATPase activities, calpain activity and lens protein levels.


Subject(s)
Cataract/prevention & control , Diabetes Mellitus, Experimental/complications , Lens, Crystalline/drug effects , Oxidative Stress/drug effects , Tocotrienols/pharmacology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Administration, Topical , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Blood Glucose/metabolism , Body Weight/drug effects , Calpain , Catalase/metabolism , Cataract/complications , Diabetes Mellitus, Experimental/blood , Gene Expression/drug effects , Glutathione/metabolism , Lens, Crystalline/metabolism , Lens, Crystalline/pathology , Male , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitrosation/drug effects , Pilot Projects , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Time Factors , Tocotrienols/administration & dosage
16.
Neurotoxicology ; 59: 105-109, 2017 03.
Article in English | MEDLINE | ID: mdl-28163087

ABSTRACT

BACKGROUND: L-ß-N-methylamino-l-alanine (BMAA) is a non-proteinic amino acid, that is neurotoxic in vitro and in animals, and is implicated in the causation of amyotrophic lateral sclerosis and parkinsonism-dementia complex (ALS-PDC) on Guam. Given that natural amino acids can be N-nitrosated to form toxic alkylating agents and the structural similarity of BMAA to other amino acids, our hypothesis was that N-nitrosation of BMAA might result in a toxic alkylating agent, providing a novel mechanistic hypothesis for BMAA action. FINDINGS: We have chemically nitrosated BMAA with sodium nitrite to produce nitrosated BMAA (N-BMAA) which was shown to react with the alkyl-trapping agent, 4-(p-nitrobenzyl)pyridine, cause DNA strand breaks in vitro and was toxic to the human neuroblastoma cell line SH-SY5Y under conditions in which BMAA itself was minimally toxic. CONCLUSIONS: Our results indicate that N-BMAA is an alkylating agent and toxin suggesting a plausible and previously unrecognised mechanism for the neurotoxic effects of BMAA.


Subject(s)
Alkylating Agents/toxicity , Amino Acids, Diamino/chemistry , DNA Damage/drug effects , Nitrates , Pyridines/toxicity , Cell Line, Tumor , Cyanobacteria Toxins , DNA Breaks, Single-Stranded/drug effects , Dose-Response Relationship, Drug , Humans , Neuroblastoma , Nitrosation/drug effects
17.
Biochem Biophys Res Commun ; 486(1): 29-35, 2017 04 22.
Article in English | MEDLINE | ID: mdl-28223216

ABSTRACT

Endothelial dysfunction played an important role in the progression of diabetes mellitus (DM). miR-181c has been implicated in many diseases, including DM. However, the molecular mechanisms of miR-181c regulate this process remained poorly understood. Healthy ICR mice were divided into control group (n = 10) and db/db DM group (n = 10). The expression of miR-181c and FoxO1 were both investigated in diabetic db/db mice or high glucose-induced endothelial cells (MAECs and END-D). Here we found that down-regulation of miR-181c and the activation of FoxO1/iNOS were observed in mice and endothelial cells. Furthermore, we verified that miR-181c directly targeted and inhibited FoxO1 gene expression by targeting its 3'-UTR through luciferase reporter assay. Knockdown of FoxO1 reversed the up-regulation of iNOS, nitrotyrosine and the down-regulation of p-eNOSSer1177/eNOS in high glucose (30 mM)-induced MAECs cells. In addition, over-expression of miR-181c could reverse the enhanced nitration stress induced by high glucose, while this effect could be attenuated by pcDNA-FoxO1 in MAECs. These results shown that miR-181c attenuated nitration stress through regulating FoxO1 expression and affecting endothelial cell function, which offering a new target for the development of preventive or therapeutic agents against DM.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Endothelial Cells/metabolism , Forkhead Box Protein O1/genetics , Gene Expression Regulation , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Blotting, Western , Cell Line , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Endothelial Cells/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Forkhead Box Protein O1/metabolism , Glucose/pharmacology , Male , Mice, Inbred ICR , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nitrosation/drug effects , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vasodilation/genetics
18.
Food Chem Toxicol ; 102: 176-185, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28219701

ABSTRACT

Microcystins are a group of cyclic heptapeptide toxins produced by cyanobacteria. More than 100 microcystin analogues have been detected, among which microcystin-LR is the most abundant and toxic variant. Present study was designed to reveal whether potential human carcinogen microcystin-LR could imbalance the glycolytic-oxidative-nitrosative status of heart, kidney and spleen of mice and also to explore the amelioratory effect of coenzyme Q10 on microcystin-LR induced toxicity. Microcystin-LR was administered at a dose of 10 µg/kg bw/day, ip for 14 days in male mice. In microcystin-LR treated mice as compared to control, significant increase in the level of lipid peroxidation, hydrogen peroxide, lactate dehydrogenase, nitric oxide with a concomitant decrease in the level of glutathione was observed, suggesting microcystin-LR induced toxicity via induction of oxidative-nitrosative-glycolytic pathway. Although several studies have evaluated numerous antioxidants but still there is no effective chemoprotectant against microcystin-LR induced toxicity. When microcystin-LR treated mice were co-administered coenzyme Q10 (10 mg/kg bw/day, im) for 14 days, it was observed that coenzyme Q10 ameliorates microcystin-LR induced toxicity via modulation of glycolytic-oxidative-nitrosative stress pathway. Thus, the results suggest that coenzyme Q10 has a potential to be developed as preventive agent against microcystin-LR induced toxicity.


Subject(s)
Carcinogens/toxicity , Microcystins/toxicity , Protective Agents/pharmacology , Ubiquinone/analogs & derivatives , Animals , Heart/drug effects , Kidney/drug effects , Kidney/metabolism , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Marine Toxins , Mice, Inbred Strains , Nitrosation/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Spleen/drug effects , Spleen/metabolism , Ubiquinone/pharmacology
19.
Apoptosis ; 22(6): 800-815, 2017 06.
Article in English | MEDLINE | ID: mdl-28213701

ABSTRACT

Plant-derived pentacyclic triterpenotids with multiple biological activities are considered as promising candidates for cancer therapy and prevention. However, their mechanisms of action are not fully understood. In the present study, we have analyzed the effects of low dose treatment (5-20 µM) of ursolic acid (UA) and betulinic acid (BA) on breast cancer cells of different receptor status, namely MCF-7 (ER+, PR+/-, HER2-), MDA-MB-231 (ER-, PR-, HER2-) and SK-BR-3 (ER-, PR-, HER2+). UA-mediated response was more potent than BA-mediated response. Triterpenotids (5-10 µM) caused G0/G1 cell cycle arrest, an increase in p21 levels and SA-beta-galactosidase staining that was accompanied by oxidative stress and DNA damage. UA (20 µM) also diminished AKT signaling that affected glycolysis as judged by decreased levels of HK2, PKM2, ATP and lactate. UA-induced energy stress activated AMPK that resulted in cytotoxic autophagy and apoptosis. UA-mediated elevation in nitric oxide levels and ATM activation may also account for AMPK activation-mediated cytotoxic response. Moreover, UA-promoted apoptosis was associated with decreased pERK1/2 signals and the depolarization of mitochondrial membrane potential. Taken together, we have shown for the first time that UA at low micromolar range may promote its anticancer action by targeting glycolysis in phenotypically distinct breast cancer cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Breast Neoplasms/pathology , Glycolysis/drug effects , Triterpenes/pharmacology , Breast Neoplasms/enzymology , Cell Line, Tumor , DNA Damage , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Models, Biological , Nitrosation/drug effects , Oxidative Stress/drug effects , Pentacyclic Triterpenes , Phenotype , Signal Transduction/drug effects , Betulinic Acid , Ursolic Acid
20.
Cell Mol Neurobiol ; 37(1): 65-81, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26886752

ABSTRACT

Chronic stress exposure can produce deleterious effects on the hippocampus (HC) which eventually leads to cognitive impairment and depression. Endoplasmic reticulum (ER) stress has been reported as one of the major culprits in the development of stress-induced cognitive impairment and depression. We investigated the neuroprotective efficacy of sodium phenylbutyrate (SPB), an ER stress inhibitor, and edaravone, a free radical scavenger, against chronic restraint stress (CRS)-induced cognitive deficits and anxiety- and depressive-like behavior in mice. Adult male Swiss albino mice were restrained for 6 h/day for 28 days and injected (i.p.) with SPB (40 and 120 mg/kg) or edaravone (3 and 10 mg/kg) for the last seven days. After stress cessation, the anxiety- and depressive-like behavior along with spatial learning and memory were examined. Furthermore, oxido-nitrosative stress, proinflammatory cytokines, and gene expression level of ER stress-related genes were assessed in HC and prefrontal cortex (PFC). CRS-exposed mice showed anxiety- and depressive-like behavior, which was significantly improved by SPB and edaravone treatment. In addition, SPB and edaravone treatment significantly alleviated CRS-induced spatial learning and memory impairment. Furthermore, CRS-evoked oxido-nitrosative stress, neuroinflammation, and depletion of Brain-derived neurotrophic factor were significantly ameliorated by SPB and edaravone treatment. We found significant up-regulation of ER stress-related genes in both HC and PFC regions, which were suppressed by SPB and edaravone treatment in CRS mice. Our study provides evidence that SPB and edaravone exerted neuroprotective effects on CRS-induced cognitive deficits and anxiety- and depressive-like behavior, which is possibly coupled with inhibition of oxido-nitrosative stress, neuroinflammation, and ER stress cascade.


Subject(s)
Antipyrine/analogs & derivatives , Depression/drug therapy , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects , Phenylbutyrates/therapeutic use , Stress, Psychological/drug therapy , Animals , Antipyrine/pharmacology , Antipyrine/therapeutic use , Chronic Disease , Depression/etiology , Depression/metabolism , Dose-Response Relationship, Drug , Edaravone , Endoplasmic Reticulum Stress/physiology , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Inflammation/drug therapy , Inflammation/metabolism , Male , Mice , Nitrosation/drug effects , Nitrosation/physiology , Oxidative Stress/physiology , Phenylbutyrates/pharmacology , Restraint, Physical , Stress, Psychological/complications , Stress, Psychological/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...