Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 681
Filter
1.
Behav Brain Res ; 437: 114128, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36174841

ABSTRACT

While SSRIs are the current first-line pharmacotherapies against post-traumatic stress disorder (PTSD), they suffer from delayed onset of efficacy and low remission rates. One solution is to combine SSRIs with other treatments. Neuronal nitric oxide synthase (nNOS) has been shown to play a role in serotonergic signaling, and there is evidence of synergism between nNOS modulation and SSRIs in models of other psychiatric conditions. Therefore, in this study, we combined subchronic fluoxetine (Flx) with 7-nitroindazole (NI), a selective nNOS inhibitor, and evaluated their efficacy against anxiety-related behavior in an animal model of PTSD. We used the underwater trauma model to induce PTSD in rats. Animals underwent the open field (OFT) and elevated plus maze tests on days 14 (baseline) and 21 (post-treatment) after PTSD induction to assess anxiety-related behaviors. Between the two tests, the rats received daily intraperitoneal injections of 10 mg/kg Flx or saline, and were injected intraperitoneally before the second test with either 15 mg/kg NI or saline. The change in behaviors between the two tests was compared between treatment groups. Individual treatment with both Flx and NI had anxiogenic effects in the OFT. These effects were associated with modest increases in cFOS expression in the hippocampus. Combination therapy with Flx + NI did not show any anxiogenic effects, while causing even higher expression levels of cFOS. In conclusion, addition of NI treatment to subchronic Flx therapy accelerated the abrogation of Flx's anxiogenic properties. Furthermore, hippocampal activity, as evidenced by cFOS expression, was biphasically related to anxiety-related behavior.


Subject(s)
Anti-Anxiety Agents , Enzyme Inhibitors , Nitric Oxide Synthase Type I , Selective Serotonin Reuptake Inhibitors , Stress Disorders, Post-Traumatic , Animals , Rats , Anxiety/metabolism , Disease Models, Animal , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Hippocampus/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/metabolism , Drug Therapy, Combination , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use
2.
Circ Res ; 131(12): 952-961, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36349758

ABSTRACT

BACKGROUND: Neurovascular coupling (NVC) is a key process in cerebral blood flow regulation. NVC ensures adequate brain perfusion to changes in local metabolic demands. Neuronal nitric oxide synthase (nNOS) is suspected to be involved in NVC; however, this has not been tested in humans. Our objective was to investigate the effects of nNOS inhibition on NVC in humans. METHODS: We performed a 3-visit partially randomized, double-blinded, placebo-controlled, crossover study in 12 healthy subjects. On each visit, subjects received an intravenous infusion of either S-methyl-L-thiocitrulline (a selective nNOS-inhibitor), 0.9% saline (placebo control), or phenylephrine (pressor control). The NVC assessment involved eliciting posterior circulation hyperemia through visual stimulation while measuring posterior and middle cerebral arteries blood velocity. RESULTS: nNOS inhibition blunted the rapidity of the NVC response versus pressor control, evidenced by a reduced initial rise in mean posterior cerebral artery velocity (-3.3% [-6.5, -0.01], P=0.049), and a reduced rate of increase (ie, acceleration) in posterior cerebral artery velocity (slope reduced -4.3% [-8.5, -0.1], P=0.045). The overall magnitude of posterior cerebral artery response relative to placebo control or pressor control was not affected. Changes in BP parameters were well-matched between the S-methyl-L-thiocitrulline and pressor control arms. CONCLUSIONS: Neuronal NOS plays a role in dynamic cerebral blood flow control in healthy adults, particularly the rapidity of the NVC response to visual stimulation. This work opens the way to further investigation of the role of nNOS in conditions of impaired NVC, potentially revealing a therapeutic target.


Subject(s)
Enzyme Inhibitors , Neurovascular Coupling , Adult , Humans , Cerebrovascular Circulation , Cross-Over Studies , Enzyme Inhibitors/pharmacology , Nitric Oxide , Nitric Oxide Synthase Type I/antagonists & inhibitors
3.
Sci Rep ; 12(1): 1701, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35105915

ABSTRACT

Interferon-gamma (IFN-γ) is shown to stimulate melanoma development and progression. However, the underlying mechanism has not been completely defined. Our study aimed to determine the role of neuronal nitric oxide synthase (nNOS)-mediated signaling in IFN-γ-stimulated melanoma progression and the anti-melanoma effects of novel nNOS inhibitors. Our study shows that IFN-γ markedly induced the expression levels of nNOS in melanoma cells associated with increased intracellular nitric oxide (NO) levels. Co-treatment with novel nNOS inhibitors effectively alleviated IFN-γ-activated STAT1/3. Further, reverse phase protein array (RPPA) analysis demonstrated that IFN-γ induced the expression of HIF1α, c-Myc, and programmed death-ligand 1 (PD-L1), in contrast to IFN-α. Blocking the nNOS-mediated signaling pathway using nNOS-selective inhibitors was shown to effectively diminish IFN-γ-induced PD-L1 expression in melanoma cells. Using a human melanoma xenograft mouse model, the in vivo studies revealed that IFN-γ increased tumor growth compared to control, which was inhibited by the co-administration of nNOS inhibitor MAC-3-190. Another nNOS inhibitor, HH044, was shown to effectively inhibit in vivo tumor growth and was associated with reduced PD-L1 expression levels in melanoma xenografts. Our study demonstrates the important role of nNOS-mediated NO signaling in IFN-γ-stimulated melanoma progression. Targeting nNOS using highly selective small molecular inhibitors is a unique and effective strategy to improve melanoma treatment.


Subject(s)
Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Disease Progression , Enzyme Inhibitors/administration & dosage , Interferon-gamma/administration & dosage , Melanoma/drug therapy , Melanoma/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Animals , B7-H1 Antigen/metabolism , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Humans , Interferon-alpha/pharmacology , Melanoma/pathology , Mice , Mice, Nude , Nitric Oxide Synthase Type I/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Skin Neoplasms/pathology , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
4.
Psychopharmacology (Berl) ; 239(1): 267-276, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34661719

ABSTRACT

RATIONALE: Deregulated attack behaviors have devastating social consequences; however, satisfactory clinical management for the behavior is still an unmet need so far. Social isolation (SI) has been common during the COVID-19 pandemic and may have detrimental effects on mental health, including eliciting heightened attack behavior. OBJECTIVES: This study aims to explore whether injection of ZL006 can alleviate SI-induced escalation of attack behavior in mice. METHODS: Pharmacological tools, biochemical methods, and behavioral tests were used to explore the potential therapeutic effects of ZL006 targeting postsynaptic density 95 (PSD95)/neuronal nitric oxide synthase (nNOS) pathway on escalation of attack behavior induced by SI in mice. RESULTS: ZL006 mitigated SI-induced escalated attack behaviors and elevated nitric oxide (NO) level in the cortex of the SI mice. The beneficial effects of ZL006 lasted for at least 72 h after a single injection of ZL006. Potentiation of NO levels by L-arginine blocked the effects of ZL006. Moreover, a sub-effective dose of 7-NI in combination with a sub-effective dose of ZL006 decreased both SI-induced escalated attack behaviors and NO levels in mice subjected to SI. CONCLUSIONS: Our study highlights the importance of the PSD95/nNOS pathway in mediating SI-induced escalation of attack behavior. ZL006 may be a promising therapeutic strategy for treating aggressive behaviors.


Subject(s)
Aggression , Aminosalicylic Acids/pharmacology , Benzylamines/pharmacology , Disks Large Homolog 4 Protein/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Social Isolation , Animals , Mice
5.
Nitric Oxide ; 119: 29-40, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34896554

ABSTRACT

Nitric oxide is a small gaseous molecule that plays important roles in the majority of biological functions. Impairments of NO-related pathways contribute to the majority of neurological disorders, such as Alzheimer's disease (AD), and mental disorders, such as schizophrenia. Cognitive decline is one of the most serious impairments accompanying both AD and schizophrenia. In the present study, the activities of NO donors, slow (spermine NONOate) or fast (DETANONOate) releasers, and selective inhibitor of neuronal nitric oxide synthase N(ω)-propyl-l-arginine (NPLA) were investigated in pharmacological models of schizophrenia and AD. Cognitive impairments were induced by administration of MK-801 or scopolamine and were measured in novel object recognition (NOR) and Y-maze tests. The compounds were investigated at doses of 0.05-0.5 mg/kg. The dose-dependent effectiveness of all the compounds was observed in the NOR test, while only the highest doses of spermine NONOate and NPLA were active in the Y-maze test. DETANONOate was not active in the Y-maze test. The impact of the investigated compounds on motor coordination was tested at doses of 0.5 and 1 mg/kg. Only NPLA at a dose of 1 mg/kg slightly disturbed motor coordination in animals.


Subject(s)
Alzheimer Disease/drug therapy , Cognitive Dysfunction/drug therapy , Nitric Oxide Donors/therapeutic use , Nitric Oxide/metabolism , Nootropic Agents/therapeutic use , Schizophrenia/drug therapy , Alzheimer Disease/chemically induced , Animals , Arginine/analogs & derivatives , Arginine/therapeutic use , Cognitive Dysfunction/chemically induced , Dizocilpine Maleate , Enzyme Inhibitors/therapeutic use , Male , Mice , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitroso Compounds/therapeutic use , Open Field Test/drug effects , Rotarod Performance Test , Schizophrenia/chemically induced , Scopolamine , Spermine/analogs & derivatives , Spermine/therapeutic use
6.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638939

ABSTRACT

The adipokine leptin, which is best-known for its role in the control of metabolic function, is also a master regulator of cardiovascular function. While leptin has been approved for the treatment of metabolic disorders in patients with congenital generalized lipodystrophy (CGL), the effects of chronic leptin deficiency and the treatment on vascular contractility remain unknown. Herein, we investigated the effects of leptin deficiency and treatment (0.3 mg/day/7 days) on aortic contractility in male Berardinelli-Seip 2 gene deficient mice (gBscl2-/-, model of CGL) and their wild-type control (gBscl2+/+), as well as in mice with selective deficiency in endothelial leptin receptor (LepREC-/-). Lipodystrophy selectively increased vascular adrenergic contractility via NO-independent mechanisms and induced hypertrophic vascular remodeling. Leptin treatment and Nox1 inhibition blunted adrenergic hypercontractility in gBscl2-/- mice, however, leptin failed to rescue vascular media thickness. Selective deficiency in endothelial leptin receptor did not alter baseline adrenergic contractility but abolished leptin-mediated reduction in adrenergic contractility, supporting the contribution of endothelium-dependent mechanisms. These data reveal a new direct role for endothelial leptin receptors in the control of vascular contractility and homeostasis, and present leptin as a safe therapy for the treatment of vascular disease in CGL.


Subject(s)
Adrenergic Agents/metabolism , Aorta, Thoracic/metabolism , Endothelium, Vascular/metabolism , Leptin/metabolism , Lipodystrophy, Congenital Generalized/metabolism , Muscle Contraction/genetics , Muscle, Smooth, Vascular/metabolism , Signal Transduction/genetics , Adrenergic Agents/administration & dosage , Adrenergic Agents/adverse effects , Animals , Disease Models, Animal , GTP-Binding Protein gamma Subunits/genetics , GTP-Binding Protein gamma Subunits/metabolism , Leptin/administration & dosage , Leptin/adverse effects , Lipodystrophy, Congenital Generalized/drug therapy , Male , Mice , Mice, Knockout , Muscle Contraction/drug effects , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Treatment Outcome
7.
Neuropharmacology ; 198: 108762, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34437905

ABSTRACT

It was recently shown that kisspeptin neurons in the anteroventral periventricular area (AVPV) orchestrate female sexual behavior, including lordosis behavior and mate preference. A potential target of AVPV kisspeptin signaling could be neurons expressing the neuronal form of nitric oxide synthase (nNOS) in the ventrolateral part of the ventromedial hypothalamus (VMHvl). Therefore, in the present study, we further refined the role of the VHMvl in female sexual behavior. Adult female mice received a bilateral cannula aimed at the VMHvl. A single injection with kisspeptin (Kp-10) or SNAP/BAY, a nitric oxide donor, significantly increased lordosis, whereas the nNOS inhibitor l-NAME decreased it. None of these drugs affected mate preference. Interestingly, administration of GnRH into the VMHvl had no effect on lordosis or mate preference. To determine whether the stimulatory effect of Kp-10 on lordosis was specific to the VMHvl, an additional group of females received Kp-10 directly into the paraventricular nucleus (PVN). No effect was found on lordosis and mate preference. These results suggest that kisspeptin most likely modulates lordosis behavior through nNOS neurons in the VMHvl whereas mate preference is modulated by kisspeptin through a separate neuronal circuit not including the VMHvl.


Subject(s)
Kisspeptins/physiology , Mating Preference, Animal/physiology , Neurons/physiology , Nitric Oxide Synthase Type I/physiology , Sexual Behavior, Animal/physiology , Signal Transduction/physiology , Ventromedial Hypothalamic Nucleus/physiology , Animals , Female , Gonadotropin-Releasing Hormone/pharmacology , Kisspeptins/pharmacology , Male , Mice , Mice, Inbred C57BL , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors
8.
Bioorg Med Chem ; 44: 116294, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34218000

ABSTRACT

In search of new Nitric Oxide Synthase (NOS) inhibitor agents, two isosteric series of derivatives with an imidamide scaffold (one of them with a hydroxyl group and the other with a carbonyl one) were synthesized and evaluated on inducible (iNOS) and neuronal (nNOS) isoforms. These compounds have been designed by combining a kynurenamine framework with an amidine moiety in order to improve selectivity for the inducible isoform. In general, the in vitro inhibitory assays exhibited better inhibition values on the iNOS isoform, being the N-(3-(2-amino-5-methoxyphenyl)-3-hydroxypropyl)-4-(trifluoromethyl)benzimidamide 4i the most active inhibitor with the highest iNOS selectivity, without inhibiting eNOS. Docking studies on the two most active compounds suggest a different binding mode on both isozymes, supporting the experimentally observed selectivity towards the inducible isoform. Physicochemical in silico studies suggest that these compounds possess good drug-likeness properties.


Subject(s)
Amidines/pharmacology , Enzyme Inhibitors/pharmacology , Molecular Docking Simulation , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Amidines/chemical synthesis , Amidines/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type II/metabolism , Structure-Activity Relationship
9.
Arch Biochem Biophys ; 709: 108985, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34252390

ABSTRACT

The protein deacetylase sirtuin 1 (SIRT1) and adenosine monophosphate-dependent protein kinase (AMPK) play important roles in the development of insulin resistance. In glomerular podocytes, crosstalk between these two enzymes may be altered under hyperglycemic conditions. SIRT1 protein levels and activity and AMPK phosphorylation decrease under hyperglycemic conditions, with concomitant inhibition of the effect of insulin on glucose uptake into these cells. Nitric oxide (NO)-dependent regulatory signaling pathways have been shown to be downregulated under diabetic conditions. The present study examined the involvement of the NO synthase (NOS)/NO pathway in the regulation of SIRT1-AMPK signaling and glucose uptake in podocytes. We examined the effects of NOS/NO pathway alterations on SIRT1/AMPK signaling and glucose uptake using pharmacological tools and a small-interfering transfection approach. We also examined the ability of the NOS/NO pathway to protect podocytes against high glucose-induced alterations of SIRT1/AMPK signaling and insulin-dependent glucose uptake. Inhibition of the NOS/NO pathway reduced SIRT1 protein levels and activity, leading to a decrease in AMPK phosphorylation and blockade of the effect of insulin on glucose uptake. Treatment with the NO donor S-nitroso-N-acetylpenicillamine (SNAP) prevented high glucose-induced decreases in SIRT1 and AMPK activity and increased GLUT4 protein expression, thereby improving glucose uptake in podocytes. These findings suggest that inhibition of the NOS/NO pathway may result in alterations of the effects of insulin on glucose uptake in podocytes. In turn, the enhancement of NOS/NO pathway activity may prevent these deleterious effects of high glucose concentrations, thus bidirectionally stimulating the SIRT1-AMPK reciprocal activation loop.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide/metabolism , Podocytes/metabolism , Sirtuin 1/metabolism , Animals , Down-Regulation/physiology , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Glucose Transporter Type 4/metabolism , Insulin/metabolism , Insulin Resistance/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Phosphorylation/drug effects , Rats , S-Nitroso-N-Acetylpenicillamine/pharmacology , Signal Transduction , Sirtuin 1/genetics
10.
Inorg Chem ; 60(13): 9345-9358, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34137256

ABSTRACT

Neuronal nitric oxide synthase (nNOS) is one of the three isoforms of nitric oxide synthase (NOS). The other two isoforms include inducible NOS (iNOS) and endothelial NOS (eNOS). These three isoforms of NOS are widely present in both human and other mammals and are responsible for the biosynthesis of NO. As an essential biological molecule, NO plays an essential role in neurotransmission, immune response, and vasodilation; however, the overproduction of NO can cause a series of diseases. Thus, the selective inhibition of three isoforms of NOS has been considered to be important in treating related diseases. The active sites of the three enzymes are highly conserved, causing the selective inhibition of the three enzymes to be a great challenge. (S)-2-Amino-5-(2-(methylthio)acetimidamido)pentanoic acid (1) has been experimentally proved to be a selective and time-dependent irreversible inhibitor of nNOS, and three pathways, including sulfide oxidation, oxidative dethiolation, and oxidative demethylation, have been suggested. In this work, we performed quantum mechanics/molecular mechanics calculations to verify the chemical conversion of inactivator 1. Although we agree with the previously suggested chemical transformation process, our calculations demonstrated that there are lower energy pathways to accomplish both oxidative dethiolation and oxidative demethylation. These three branching reactions are competitive, but only dethiolation and demethylation reactions can generate inhibitory intermediates. As a powerful time-dependent irreversible inhibitor of nNOS, the key sulfur atom and middle imine are all necessary. Our calculation results not only verified the chemical reaction of inhibitor 1 occurring in the enzymatic active site but also explained the inactivation mechanism of inhibitor 1. This is also the first verified example of the heme-enzyme-catalyzed S-demethylation mechanism.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Pentanoic Acids/pharmacology , Crystallography, X-Ray , Density Functional Theory , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Conformation , Molecular Dynamics Simulation , Nitric Oxide Synthase Type I/metabolism , Pentanoic Acids/chemical synthesis , Pentanoic Acids/chemistry
11.
Cell Mol Neurobiol ; 41(6): 1373-1387, 2021 Aug.
Article in English | MEDLINE | ID: mdl-32594381

ABSTRACT

The imbalance between excess reactive oxygen species (ROS) generation and insufficient antioxidant defenses contribute to a range of neurodegenerative diseases. High ROS levels damage cellular macromolecules such as DNA, proteins and lipids, leading to neuron vulnerability and eventual death. However, the underlying molecular mechanism of the ROS regulation is not fully elucidated. Recently, an increasing number of studies suggest that microRNAs (miRNAs) emerge as the targets in regulating oxidative stress. We recently reported the neuroprotective effect of miR-137-3p for brachial plexus avulsion-induced motoneuron death. The present study is sought to investigate whether miR-137-3p also could protect PC12 cells against hydrogen peroxide (H2O2) induced neurotoxicity. By using cell viability assay, ROS assay, gene and protein expression assay, we found that PC-12 cells exposed to H2O2 exhibited decreased cell viability, increased expression levels of calpain-2 and neuronal nitric oxide synthase (nNOS), whereas a decreased miR-137-3p expression. Importantly, restoring the miR-137-3p levels in H2O2 exposure robustly inhibited the elevated nNOS, calpain-2 and ROS expression levels, which subsequently improved the cell viability. Furthermore, the suppressive effect of miR-137-3p on the elevated ROS level under oxidative stress was considerably blunted when we mutated the binding site of calpain-2 targted by miR-137-3p, suggesting the critical role of calpain-2 involving the neuroprotective effect of miR-137-3p. Collectively, these findings highlight the neuroprotective role of miR-137-3p through down-regulating calpain and NOS activity, suggesting its potential role for combating oxidative stress insults in the neurodegenerative diseases.


Subject(s)
Calpain/biosynthesis , Down-Regulation/physiology , MicroRNAs/biosynthesis , Nitric Oxide Synthase Type I/biosynthesis , Oxidative Stress/physiology , Animals , Calpain/antagonists & inhibitors , Cell Survival/drug effects , Cell Survival/physiology , Down-Regulation/drug effects , Hydrogen Peroxide/toxicity , Nitric Oxide Synthase Type I/antagonists & inhibitors , Oxidative Stress/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
12.
Pflugers Arch ; 472(12): 1743-1755, 2020 12.
Article in English | MEDLINE | ID: mdl-32940784

ABSTRACT

Nitric oxide (NO) affects mitochondrial activity through its interactions with complexes. Here, we investigated regulations of complex I (C-I) and complex II (C-II) by neuronal NO synthase (nNOS) in the presence of fatty acid supplementation and the impact on left ventricular (LV) mitochondrial activity from sham and angiotensin II (Ang-II)-induced hypertensive (HTN) rats. Our results showed that nNOS protein was expressed in sham and HTN LV mitochondrial enriched fraction. In sham, oxygen consumption rate (OCR) and intracellular ATP were increased by palmitic acid (PA) or palmitoyl-carnitine (PC). nNOS inhibitor, S-methyl-l-thiocitrulline (SMTC), did not affect OCR or cellular ATP increment by PA or PC. However, SMTC increased OCR with PA + malonate (a C-II inhibitor), but not with PA + rotenone (a C-I inhibitor), indicating that nNOS attenuates C-I with fatty acid supplementation. Indeed, SMTC increased C-I activity but not that of C-II. Conversely, nNOS-derived NO was increased by rotenone + PA in LV myocytes. In HTN, PC increased the activity of C-I but reduced that of C-II, consequently OCR was reduced. SMTC increased both C-I and C-II activities with PC, resulted in OCR enhancement in the mitochondria. Notably, SMTC increased OCR only with rotenone, suggesting that nNOS modulates C-II-mediated OCR in HTN. nNOS-derived NO was partially reduced by malonate + PA. Taken together, nNOS attenuates C-I-mediated mitochondrial OCR in the presence of fatty acid in sham and C-I modulates nNOS activity. In HTN, nNOS attenuates C-I and C-II activities whereas interactions between nNOS and C-II maintain mitochondrial activity.


Subject(s)
Electron Transport Complex II/metabolism , Electron Transport Complex I/metabolism , Hypertension/metabolism , Mitochondria, Heart/metabolism , Nitric Oxide Synthase Type I/metabolism , Angiotensin II/toxicity , Animals , Cells, Cultured , Citrulline/analogs & derivatives , Citrulline/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex II/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypertension/etiology , Hypertension/physiopathology , Male , Malonates/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Oxygen Consumption , Rats , Rats, Sprague-Dawley , Rotenone/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology
13.
Neurochem Res ; 45(11): 2775-2785, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32930947

ABSTRACT

Hypothyroidism-associated learning and memory impairment is reported to be connected to oxidative stress and reduced levels of brain-derived neurotrophic factor (BDNF). The effects of neuronal nitric oxide inhibitor 7-nitroindazole (7NI) on brain tissues oxidative damage, nitric oxide (NO), BDNF and memory impairments in hypothyroid juvenile rats were investigated. Male Wistar juvenile rats (20 days old) were divided into five groups, including Martinez et al. (J Neurochem 78 (5):1054-1063, 2001). Control in which vehicle was injected instead of 7NI, (Jackson in Thyroid 8 (10):951-956, 1998) Propylthiouracil (PTU) where 0.05% PTU was added in drinking water and vehicle was injected instead of 7NI, (Gong et al. in BMC Neurosci 11 (1):50, 2010; Alva-Sánchez et al. in Brain Res 1271:27-35, 2009; Anaeigoudari et al. in Pharmacol Rep 68 (2): 243-249, 2016) PTU-7NI 5, PTU-7NI 10 and PTU-7NI 20 in which 5, 10, or 20 mg/kg7NI was injected intraperitoneally (i.p.). Following 6 weeks, Morris water maze (MMW) and passive avoidance learning (PAL) tests were used to evaluate the memory. Finally, the hippocampus and the cortex of the rats were removed after anesthesia by urethane to be used for future analysis. The escape latency and traveled path in MWM test was increased in PTU group (P < 0.001). PTU also reduced the latency to enter the dark box of PAL and the time spent and the distance in the target quadrant in MWM test (P < 0.001 and P < 0.01). Treatment with 7NI attenuated all adverse effects of PTU (P < 0.05 to P < 0.001). PTU lowered BDNF and thiol content and superoxide dismutase (SOD) and catalase (CAT) activities in the brain but increased malondialdehyde (MDA) and nitric oxide (NO) metabolites. In addition, 7NI improved thiol, SOD, CAT, thiol, and BDNF but attenuated MDA and NO metabolites. The results of the current study showed that 7NI improvement in the learning and memory of the hypothyroid juvenile rats, which was accompanied with improving of BDNF and attenuation of NO and brain tissues oxidative damage.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hypothyroidism/metabolism , Indazoles/therapeutic use , Learning Disabilities/drug therapy , Memory Disorders/drug therapy , Nitric Oxide Synthase Type I/antagonists & inhibitors , Animals , Enzyme Inhibitors/therapeutic use , Hippocampus/drug effects , Hypothyroidism/chemically induced , Hypothyroidism/complications , Learning Disabilities/etiology , Male , Maze Learning/drug effects , Memory/drug effects , Memory Disorders/etiology , Morris Water Maze Test/drug effects , Oxidative Stress/drug effects , Propylthiouracil , Rats, Wistar
14.
Brain Res Bull ; 164: 29-36, 2020 11.
Article in English | MEDLINE | ID: mdl-32814090

ABSTRACT

Nitric oxide plays a role in the long term potentiation mechanisms produced in the mammalian hippocampus during spatial learning. A great deal of data has demonstrated that the dorsolateral telencephalon of fish could be homologous to the mammalian hippocampus sharing functional similarities. In the present study, we analyzed the role of nitric oxide in spatial learning in teleost fish. In Experiment 1, we studied the effects of the inhibition of telencephalic nitric oxide in goldfish during the acquisition of a spatial task. The results showed that nitric oxide is involved in the learning of a spatial task. Experiment 2 evaluated the effects of the inhibition of telencephalic nitric oxide in goldfish for the retrieval of a learned spatial response. The results indicated that the retrieval of the information previously stored is not dependent of the nitric oxide. The last experiment analyzed the role of the telencephalic nitric oxide in place and cue learning. Results showed a clear impairment in place but not in cue learning. As a whole, these results indicate that fish and mammals, could have a relational memory system mediated by similar biochemical mechanisms.


Subject(s)
Enzyme Inhibitors/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Spatial Learning/drug effects , Telencephalon/drug effects , Animals , Goldfish , Hippocampus/drug effects
15.
ChemMedChem ; 15(22): 2157-2163, 2020 11 18.
Article in English | MEDLINE | ID: mdl-32783298

ABSTRACT

Under different pathological conditions, aberrant induction of neuronal nitric oxide synthase (nNOS) generates overproduction of NO that can cause irreversible cell damage. The aim of this study was to develop an amidoxime prodrug of a potent nNOS inhibitor, the benzhydryl acetamidine. We synthesized the benzhydryl acetamidoxime, which was evaluated in vitro to ascertain the potential NOS inhibitory activity, as well as conducting bioconversion into the parent acetamidine. The prodrug was also profiled for in vitro physicochemical properties, by determining the lipophilicity, passive permeation through the human gastrointestinal tract and across the blood-brain barrier by PAMPA, and chemical, enzymatic, and plasma stability. The obtained data demonstrate that the amidoxime prodrug shows an improved pharmacokinetic profile with respect to the acetamidine nNOS inhibitor, thus suggesting that it could be a promising lead compound to treat all those pathological conditions in which nNOS activity is dysregulated.


Subject(s)
Amidines/pharmacology , Benzhydryl Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Prodrugs/pharmacology , Amidines/chemical synthesis , Amidines/chemistry , Benzhydryl Compounds/chemical synthesis , Benzhydryl Compounds/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Nitric Oxide Synthase Type I/metabolism , Prodrugs/chemical synthesis , Prodrugs/chemistry , Recombinant Proteins/metabolism
16.
Pflugers Arch ; 472(11): 1643-1654, 2020 11.
Article in English | MEDLINE | ID: mdl-32656734

ABSTRACT

Fatty acid (FA)-dependent mitochondrial activities of atrial myocardium in hypertension (HTN) and its regulation by nitric oxide (NO) remain unidentified. Here, we have studied palmitic acid (PA) regulation of cardiac mitochondrial oxygen consumption rate (OCR) in left atrial (LA) myocardium of sham and angiotensin II-induced HTN rats and their regulations by endothelial NO synthase (eNOS) and neuronal NO synthase (nNOS). The effects were compared with those of left ventricular (LV) myocytes. Our results showed that OCR was greater in HTN-LA compared with that in sham-LA. PA increased OCR in sham-LA, sham-LV, and HTN-LV but reduced it in HTN-LA. Inhibition of nNOS (S-methyl-L-thiocitrulline, SMTC) or eNOS/nNOS (Nω-nitro-L-arginine methyl ester hydrochloride, L-NAME) reduced PA increment of OCR in sham-LA but exerted no effect on OCR in HTN-LA. SMTC reduced OCR in HTN-LV and L-NAME reduced OCR in sham-LV. nNOS was the predominant source of NO in LA and LV. nNOS-derived NO was increased in HTN-LA and HTN-LV. PA reduced eNOSSer1177, nNOSSer1417, and NO level in HTN-LA but exerted no effect in sham-LA. In contrast, PA increased NO in HTN-LV and enhanced nNOSSer1417 but reduced NO level in sham-LV without affecting eNOSSer1177, eNOSThr495, or nNOSSer1417. 2-Bromopalmitate (2BP), which blocks the S-palmitoylation of target proteins, prevented PA-dependent decrease of nNOSSer1417 and OCR in HTN-LA. In HTN-LV, 2BP prevented PA-induced OCR without affecting nNOSSer1417. Our results reveal that FA-induced mitochondrial activity in atrial myocardium is impaired in HTN which is mediated by reduced nNOS activity and NO bioavailability. Metabolic dysregulation may underlie diastolic dysfunction of atrial myocardium in HTN.


Subject(s)
Heart Atria/metabolism , Hypertension/metabolism , Mitochondria, Heart/metabolism , Nitric Oxide Synthase Type I/metabolism , Oxygen/metabolism , Palmitic Acid/metabolism , Animals , Cell Respiration , Cells, Cultured , Heart Atria/cytology , Male , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/genetics , Rats , Rats, Sprague-Dawley
17.
J Med Chem ; 63(9): 4528-4554, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32302123

ABSTRACT

Inhibition of neuronal nitric oxide synthase (nNOS), an enzyme implicated in neurodegenerative disorders, is an attractive strategy for treating or preventing these diseases. We previously developed several classes of 2-aminoquinoline-based nNOS inhibitors, but these compounds had drawbacks including off-target promiscuity, low activity against human nNOS, and only modest selectivity for nNOS over related enzymes. In this study, we synthesized new nNOS inhibitors based on 7-phenyl-2-aminoquinoline and assayed them against rat and human nNOS, human eNOS, and murine and (in some cases) human iNOS. Compounds with a meta-relationship between the aminoquinoline and a positively charged tail moiety were potent and had up to nearly 900-fold selectivity for human nNOS over human eNOS. X-ray crystallography indicates that the amino groups of some compounds occupy a water-filled pocket surrounding an nNOS-specific aspartate residue (absent in eNOS). This interaction was confirmed by mutagenesis studies, making 7-phenyl-2-aminoquinolines the first aminoquinolines to interact with this residue.


Subject(s)
Aminoquinolines/pharmacology , Aspartic Acid/chemistry , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Aminoquinolines/chemical synthesis , Aminoquinolines/metabolism , Aminoquinolines/pharmacokinetics , Animals , Blood-Brain Barrier/metabolism , Catalytic Domain , Crystallography, X-Ray , Enzyme Assays , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Microsomes, Liver/metabolism , Molecular Structure , Mutagenesis, Site-Directed , Mutation , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Permeability , Protein Binding , Rats , Structure-Activity Relationship
18.
Nitric Oxide ; 100-101: 50-56, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32278831

ABSTRACT

The cerebellum is widely known as a motor structure because it regulates and controls motor learning, coordination, and balance. However, it is also critical for non-motor functions such as cognitive processing, sensory discrimination, addictive behaviors and mental disorders. The cerebellum has the highest relative abundance of neuronal nitric oxide synthase (nNos) and is sensitive to ethanol. Although it has been demonstrated that the interaction of γ-aminobutyric acid (GABA) and nitric oxide (NO) might play an important role in the regulation of ethanol-induced cerebellar ataxia, the molecular mechanisms through which ethanol regulates nNos function to elicit this behavioral effect have not been studied extensively. Here, we investigated the dose-dependent effects of acute ethanol treatment on motor impairment using the rotarod behavioral paradigm and the alterations of nNos mRNA expression in cerebellum, frontal cortex (FC), hippocampus and striatum. We also examined the link between acute ethanol-induced motor impairment and nNos by pharmacological manipulation of nNos function. We found that acute ethanol induced a dose-dependent elevation of ethanol blood levels which was associated with the impairment of motor coordination performance and decreased expression of cerebellar nNos. In contrast, acute ethanol increased nNos expression in FC but did not to change the expression for this enzyme in striatum and hippocampus. The effects of acute ethanol were attenuated by l-arginine, a precursor for NO and potentiated by 7-nitroindazole (7-NI), a selective inhibitor of nNos. Our data suggests that differential regulation of nNos mRNA expression in cerebellum and frontal cortex might be involved in acute ethanol-induced motor impairment.


Subject(s)
Alcohol-Induced Disorders, Nervous System/metabolism , Cerebellar Ataxia/metabolism , Ethanol/adverse effects , Nitric Oxide Synthase Type I/metabolism , Psychomotor Disorders/metabolism , Alcohol-Induced Disorders, Nervous System/chemically induced , Animals , Arginine/pharmacology , Cerebellar Ataxia/chemically induced , Cerebellum/drug effects , Cerebellum/metabolism , Enzyme Inhibitors/pharmacology , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Indazoles/pharmacology , Male , Nitric Oxide Synthase Type I/antagonists & inhibitors , Psychomotor Disorders/chemically induced , Rats, Sprague-Dawley
19.
Int Immunopharmacol ; 83: 106382, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32193098

ABSTRACT

Vascular inflammation plays a decisive role in the formation of foam cells and in the pathophysiology of atherosclerosis. However, the underlying mechanisms of these processes are not clearly understood. Macrophages engulf oxidized low-density lipoproteins (OxLDLs) via a scavenger receptor (SR), an event that mediates the elaboration of proinflammatory cytokines to initiate necrotic core formation in atherogenic plaques. In this study, we demonstrate that Nitric oxide synthase 1 (NOS1)-derived nitric oxide (NO) promotes OxLDL uptake and enhances the release of proinflammatory cytokines by macrophages. Conversely, we show that NOS1 inhibition by N(G)-nitro-L-arginine methyl ester (L-NAME) suppresses OxLDL uptake and proinflammatory cytokine expression. Current studies indicate that NOS1 plays a crucial role in vascular inflammation and in the progression of atherosclerosis. Therefore, interference with NOS1 enzymatic activity should serve as an effective strategy to reduce foam cell formation and limit the extent of atherosclerotic plaque expansion.


Subject(s)
Atherosclerosis/immunology , Foam Cells/immunology , Inflammation/immunology , Nitric Oxide Synthase Type I/metabolism , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Disease Progression , Humans , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors
20.
J Neurosci ; 40(17): 3348-3359, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32169968

ABSTRACT

Nitric oxide (NO) is an important signaling molecule that fulfills diverse functional roles as a neurotransmitter or diffusible second messenger in the developing and adult CNS. Although the impact of NO on different behaviors such as movement, sleep, learning, and memory has been well documented, the identity of its molecular and cellular targets is still an area of ongoing investigation. Here, we identify a novel role for NO in strengthening inhibitory GABAA receptor-mediated transmission in molecular layer interneurons of the mouse cerebellum. NO levels are elevated by the activity of neuronal NO synthase (nNOS) following Ca2+ entry through extrasynaptic NMDA-type ionotropic glutamate receptors (NMDARs). NO activates protein kinase G with the subsequent production of cGMP, which prompts the stimulation of NADPH oxidase and protein kinase C (PKC). The activation of PKC promotes the selective strengthening of α3-containing GABAARs synapses through a GΑΒΑ receptor-associated protein-dependent mechanism. Given the widespread but cell type-specific expression of the NMDAR/nNOS complex in the mammalian brain, our data suggest that NMDARs may uniquely strengthen inhibitory GABAergic transmission in these cells through a novel NO-mediated pathway.SIGNIFICANCE STATEMENT Long-term changes in the efficacy of GABAergic transmission is mediated by multiple presynaptic and postsynaptic mechanisms. A prominent pathway involves crosstalk between excitatory and inhibitory synapses whereby Ca2+-entering through postsynaptic NMDARs promotes the recruitment and strengthening of GABAA receptor synapses via Ca2+/calmodulin-dependent protein kinase II. Although Ca2+ transport by NMDARs is also tightly coupled to nNOS activity and NO production, it has yet to be determined whether this pathway affects inhibitory synapses. Here, we show that activation of NMDARs trigger a NO-dependent pathway that strengthens inhibitory GABAergic synapses of cerebellar molecular layer interneurons. Given the widespread expression of NMDARs and nNOS in the mammalian brain, we speculate that NO control of GABAergic synapse efficacy may be more widespread than has been appreciated.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cerebellum/metabolism , Interneurons/metabolism , Microtubule-Associated Proteins/metabolism , Nitric Oxide/metabolism , Signal Transduction/physiology , Animals , Cerebellum/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Interneurons/drug effects , Male , Mice , Neural Inhibition/drug effects , Neural Inhibition/physiology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Patch-Clamp Techniques , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...