Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Proc Natl Acad Sci U S A ; 120(4): e2208924120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36652486

ABSTRACT

Nitro-fatty acids (NO2-FAs) are unsaturated fatty acid nitration products that exhibit anti-inflammatory actions in experimental mouse models of autoimmune and allergic diseases. These electrophilic molecules interfere with intracellular signaling pathways by reversible post-translational modification of nucleophilic amino-acid residues. Several regulatory proteins have been identified as targets of NO2-FAs, modifying their activity and promoting gene expression changes that result in anti-inflammatory effects. Herein, we report the effects of nitro-oleic acid (NO2-OA) on pro-inflammatory T cell functions, showing that 9- and 10-NOA, but not their oleic acid precursor, decrease T cell proliferation, expression of activation markers CD25 and CD71 on the plasma membrane, and IL-2, IL-4, and IFN-γ cytokine gene expressions. Moreover, we have found that NO2-OA inhibits the transcriptional activity of nuclear factor of activated T cells (NFAT) and that this inhibition takes place through the regulation of the phosphatase activity of calcineurin (CaN), hindering NFAT dephosphorylation, and nuclear translocation in activated T cells. Finally, using mass spectrometry-based approaches, we have found that NO2-OA nitroalkylates CaNA on four Cys (Cys129, 228, 266, and 372), of which only nitroalkylation on Cys372 was of importance for the regulation of CaN phosphatase activity in cells, disturbing functional CaNA/CaNB heterodimer formation. These results provide evidence for an additional mechanism by which NO2-FAs exert their anti-inflammatory actions, pointing to their potential as therapeutic bioactive lipids for the modulation of harmful T cell-mediated immune responses.


Subject(s)
Calcineurin , Nitrogen Dioxide , Mice , Animals , Calcineurin/metabolism , Oleic Acid , Protein Processing, Post-Translational , Fatty Acids/metabolism
2.
Toxicol Appl Pharmacol ; 468: 116514, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37061008

ABSTRACT

BACKGROUND & AIMS: Vinyl chloride (VC) monomer is a volatile organic compound commonly used in industry. At high exposure levels, VC causes liver cancer and toxicant-associated steatohepatitis. However, lower exposure levels (i.e., sub-regulatory exposure limits) that do not directly damage the liver, enhance injury caused by Western diet (WD). It is still unknown if the long-term impact of transient low-concentration VC enhances the risk of liver cancer development. This is especially a concern given that fatty liver disease is in and of itself a risk factor for the development of liver cancer. METHODS: C57Bl/6 J mice were fed WD or control diet (CD) for 1 year. During the first 12 weeks of feeding only, mice were also exposed to VC via inhalation at sub-regulatory limit concentrations (<1 ppm) or air for 6 h/day, 5 days/week. RESULTS: Feeding WD for 1 year caused significant hepatic injury, which was exacerbated by VC. Additionally, VC increased the number of tumors which ranged from moderately to poorly differentiated hepatocellular carcinoma (HCC). Transcriptomic analysis demonstrated VC-induced changes in metabolic but also ribosomal processes. Epitranscriptomic analysis showed a VC-induced shift of the modification pattern that has been associated with metabolic disease, mitochondrial dysfunction, and cancer. CONCLUSIONS: These data indicate that VC sensitizes the liver to other stressors (e.g., WD), resulting in enhanced tumorigenesis. These data raise concerns about potential interactions between VC exposure and WD. It also emphasizes that current safety restrictions may be insufficient to account for other factors that can influence hepatotoxicity.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Vinyl Chloride , Mice , Animals , Vinyl Chloride/toxicity , Vinyl Chloride/metabolism , Transcriptome , Carcinoma, Hepatocellular/pathology , Diet, Western , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism
3.
Planta ; 254(6): 120, 2021 Nov 13.
Article in English | MEDLINE | ID: mdl-34773515

ABSTRACT

MAIN CONCLUSION: Nitro fatty acids (NO2-FA)have relevant physiological roles as signaling molecules in biotic and abiotic stress, growth, and development, but the mechanism of action remains controversial. The two main mechanisms involving nitric oxide release and thiol modification are discussed. Fatty acids (FAs) are major components of membranes and contribute to cellular energetic demands. Besides, FAs are precursors of signaling molecules, including oxylipins and other oxidized fatty acids derived from the activity of lipoxygenases. In addition, non-canonical modified fatty acids, such as nitro-fatty acids (NO2-FAs), are formed in animals and plants. The synthesis NO2-FAs involves a nitration reaction between unsaturated fatty acids and reactive nitrogen species (RNS). This review will focus on recent findings showing that, in plants, NO2-FAs such as nitro-linolenic acid (NO2-Ln) and nitro-oleic acid (NO2-OA) have relevant physiological roles as signaling molecules in biotic and abiotic stress, growth, and development. Moreover, since there is controversy on mechanisms of action of NO2-FAs as signaling molecules, we will provide evidence showing why this aspect needs further evaluation.


Subject(s)
Fatty Acids , Nitric Oxide , Animals , Plant Physiological Phenomena , Reactive Nitrogen Species , Signal Transduction
4.
Cardiovasc Drugs Ther ; 35(5): 939-951, 2021 10.
Article in English | MEDLINE | ID: mdl-32671602

ABSTRACT

PURPOSE: Abdominal aortic aneurysm (AAA) is one of the leading causes of death in the developed world and is currently undertreated due to the complicated nature of the disease. Herein, we aimed to address the therapeutic potential of a novel class of pleiotropic mediators, specifically a new drug candidate, nitro-oleic acid (NO2-OA), on AAA, in a well-characterized murine AAA model. METHODS: We generated AAA using a mouse model combining AAV.PCSK9-D377Y induced hypercholesterolemia with angiotensin II given by chronic infusion. Vehicle control (PEG-400), oleic acid (OA), or NO2-OA were subcutaneously delivered to mice using an osmotic minipump. We characterized the effects of NO2-OA on pathophysiological responses and dissected the underlying molecular mechanisms through various in vitro and ex vivo strategies. RESULTS: Subcutaneous administration of NO2-OA significantly decreased the AAA incidence (8/28 mice) and supra-renal aorta diameters compared to mice infused with either PEG-400 (13/19, p = 0.0117) or OA (16/23, p = 0.0078). In parallel, the infusion of NO2-OA in the AAA model drastically decreased extracellular matrix degradation, inflammatory cytokine levels, and leucocyte/macrophage infiltration in the vasculature. Administration of NO2-OA reduced inflammation, cytokine secretion, and cell migration triggered by various biological stimuli in primary and macrophage cell lines partially through activation of the peroxisome proliferator-activated receptor-gamma (PPARγ). Moreover, the protective effect of NO2-OA relies on the inhibition of macrophage prostaglandin E2 (PGE2)-induced PGE2 receptor 4 (EP4) cAMP signaling, known to participate in the development of AAA. CONCLUSION: Administration of NO2-OA protects against AAA formation and multifactorial macrophage activation. With NO2-OA currently undergoing FDA approved phase II clinical trials, these findings may expedite the use of this nitro-fatty acid for AAA therapy.


Subject(s)
Aortic Aneurysm, Abdominal/physiopathology , Macrophage Activation/drug effects , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Angiotensin II/pharmacology , Animals , Cell Movement/drug effects , Disease Models, Animal , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Signal Transduction/drug effects
5.
Proc Natl Acad Sci U S A ; 115(33): E7768-E7775, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061387

ABSTRACT

The adaptor molecule stimulator of IFN genes (STING) is central to production of type I IFNs in response to infection with DNA viruses and to presence of host DNA in the cytosol. Excessive release of type I IFNs through STING-dependent mechanisms has emerged as a central driver of several interferonopathies, including systemic lupus erythematosus (SLE), Aicardi-Goutières syndrome (AGS), and stimulator of IFN genes-associated vasculopathy with onset in infancy (SAVI). The involvement of STING in these diseases points to an unmet need for the development of agents that inhibit STING signaling. Here, we report that endogenously formed nitro-fatty acids can covalently modify STING by nitro-alkylation. These nitro-alkylations inhibit STING palmitoylation, STING signaling, and subsequently, the release of type I IFN in both human and murine cells. Furthermore, treatment with nitro-fatty acids was sufficient to inhibit production of type I IFN in fibroblasts derived from SAVI patients with a gain-of-function mutation in STING. In conclusion, we have identified nitro-fatty acids as endogenously formed inhibitors of STING signaling and propose for these lipids to be considered in the treatment of STING-dependent inflammatory diseases.


Subject(s)
Fatty Acids/metabolism , Herpes Simplex/metabolism , Herpesvirus 2, Human/metabolism , Membrane Proteins/metabolism , Signal Transduction , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/pathology , Herpes Simplex/genetics , Herpes Simplex/pathology , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Lipoylation , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Membrane Proteins/genetics , Mice , Mice, Knockout , Nervous System Malformations/genetics , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , RAW 264.7 Cells
6.
J Lipid Res ; 60(2): 388-399, 2019 02.
Article in English | MEDLINE | ID: mdl-30545956

ABSTRACT

Electrophilic nitro-fatty acids [NO2-FAs (fatty acid nitroalkenes)] showed beneficial signaling actions in preclinical studies and safety in phase 1 clinical trials. A detailed description of the pharmacokinetics (PK) of NO2-FAs is complicated by the capability of electrophilic fatty acids to alkylate thiols reversibly and become esterified in various complex lipids, and the instability of the nitroalkene moiety during enzymatic and base hydrolysis. Herein, we report the mechanism and kinetics of absorption, metabolism, and distribution of the endogenously detectable and prototypical NO2-FA, 10-nitro-oleic acid (10-NO2-OA), in dogs after oral administration. Supported by HPLC-high-resolution-MS/MS analysis of synthetic and plasma-derived 10-NO2-OA-containing triacylglycerides (TAGs), we show that a key mechanism of NO2-FA distribution is an initial esterification into complex lipids. Quantitative analysis of plasma free and esterified lipid fractions confirmed time-dependent preferential incorporation of 10-NO2-OA into TAGs when compared with its principal metabolite, 10-nitro-stearic acid. Finally, new isomers of 10-NO2-OA were identified in vivo, and their electrophilic reactivity and metabolism characterized. Overall, we reveal that NO2-FAs display unique PK, with the principal mechanism of tissue distribution involving complex lipid esterification, which serves to shield the electrophilic character of this mediator from plasma and hepatic inactivation and thus permits efficient distribution to target organs.


Subject(s)
Alkenes/chemistry , Fatty Acids/chemistry , Fatty Acids/metabolism , Lipid Metabolism , Nitro Compounds/chemistry , Animals , Biological Transport , Dogs , Electron Transport , Esterification , Fatty Acids/blood , Fatty Acids/pharmacokinetics , Hydrogen-Ion Concentration , Isomerism , Male , Tissue Distribution
7.
Physiol Genomics ; 51(6): 224-233, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31074702

ABSTRACT

Endothelial cell (EC) dysfunction is a crucial initiation event in the development of atherosclerosis and is associated with diabetes mellitus, hypertension, and heart failure. Both digestive and oxidative inflammatory conditions lead to the endogenous formation of nitrated derivatives of unsaturated fatty acids (FAs) upon generation of the proximal nitrating species nitrogen dioxide (·NO2) by nitric oxide (·NO) and nitrite-dependent reactions. Nitro-FAs (NO2-FAs) such as nitro-oleic acid (NO2-OA) and nitro-linoleic acid (NO2-LA) potently inhibit inflammation and oxidative stress, regulate cellular functions, and maintain cardiovascular homeostasis. Recently, conjugated linoleic acid (CLA) was identified as the preferential FA substrate of nitration in vivo. However, the functions of nitro-CLA (NO2-CLA) in ECs remain to be explored. In the present study, a distinct transcriptome regulated by NO2-CLA was revealed in primary human coronary artery endothelial cells (HCAECs) through RNA sequencing. Differential gene expression and pathway enrichment analysis identified numerous regulatory networks including those related to the modulation of inflammation, oxidative stress, cell cycle, and hypoxic responses by NO2-CLA, suggesting a diverse impact of NO2-CLA and other electrophilic nitrated FAs on cellular processes. These findings extend the understanding of the protective actions of NO2-CLA in cardiovascular diseases and provide new insight into the underlying mechanisms that mediate the pleiotropic cellular responses to NO2-CLA.


Subject(s)
Endothelial Cells/drug effects , Gene Regulatory Networks/drug effects , Linoleic Acids, Conjugated/pharmacology , Adult , Cardiovascular System/drug effects , Cells, Cultured , Gene Regulatory Networks/genetics , Homeostasis/drug effects , Homeostasis/genetics , Humans , Inflammation/genetics , Male , Nitric Oxide/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Transcriptome/drug effects , Transcriptome/genetics
8.
J Pharmacol Exp Ther ; 369(3): 503-510, 2019 06.
Article in English | MEDLINE | ID: mdl-30894457

ABSTRACT

Underlying pathogenic mechanisms in chronic kidney disease (CKD) include chronic inflammation, oxidant stress, and matrix remodeling associated with dysregulated nuclear factor-κ B, nuclear factor-κ B, and SMAD signaling pathways, respectively. Important cytoprotective mechanisms activated by oxidative inflammatory conditions are mediated by nitrated fatty acids that covalently modify proteins to limit inflammation and oxidant stress. In the present study, we evaluated the effects of chronic treatment with CXA-10 (10-nitro-9(E)-octadec-9-enoic acid) in the uninephrectomized deoxycorticosterone acetate-high-salt mouse model of CKD. After 4 weeks of treatment, CXA-10 [2.5 millligrams per kilogram (mpk), p.o.] significantly attenuated increases in plasma cholesterol, heart weight, and kidney weight observed in the model without impacting systemic arterial blood pressure. CXA-10 also reduced albuminuria, nephrinuria, glomerular hypertrophy, and glomerulosclerosis in the model. Inflammatory MCP-1 and fibrosis (collagen, fibronectin, plasminogen activator inhibitor-1, and osteopontin) renal biomarkers were significantly reduced in the CXA-10 (2.5 mpk) group. The anti-inflammatory and antifibrotic effects, as well as glomerular protection, were not observed in the enalapril-treated group. Also, CXA-10 appears to exhibit hormesis as all protective effects observed in the low-dose group were absent in the high-dose group (12.5 mpk). Taken together, these findings demonstrate that, at the appropriate dose, the nitrated fatty acid CXA-10 exhibits anti-inflammatory and antifibrotic effects in the kidney and limits renal injury in a model of CKD.


Subject(s)
Cytoprotection/drug effects , Desoxycorticosterone Acetate/pharmacology , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney/drug effects , Kidney/pathology , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Salts/adverse effects , Animals , Desoxycorticosterone Acetate/pharmacokinetics , Kidney/metabolism , Kidney Diseases/metabolism , Male , Mice , Nitro Compounds/pharmacokinetics , Oleic Acids/pharmacokinetics , Oxidative Stress/drug effects , Tissue Distribution
9.
Circ Res ; 121(2): 137-148, 2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28584062

ABSTRACT

RATIONALE: Soluble guanylate cyclase (sGC) heme iron, in its oxidized state (Fe3+), is desensitized to NO and limits cGMP production needed for downstream activation of protein kinase G-dependent signaling and blood vessel dilation. OBJECTIVE: Although reactive oxygen species are known to oxidize the sGC heme iron, the basic mechanism(s) governing sGC heme iron recycling to its NO-sensitive, reduced state remain poorly understood. METHODS AND RESULTS: Oxidant challenge studies show that vascular smooth muscle cells have an intrinsic ability to reduce oxidized sGC heme iron and form protein-protein complexes between cytochrome b5 reductase 3, also known as methemoglobin reductase, and oxidized sGC. Genetic knockdown and pharmacological inhibition in vascular smooth muscle cells reveal that cytochrome b5 reductase 3 expression and activity is critical for NO-stimulated cGMP production and vasodilation. Mechanistically, we show that cytochrome b5 reductase 3 directly reduces oxidized sGC required for NO sensitization as assessed by biochemical, cellular, and ex vivo assays. CONCLUSIONS: Together, these findings identify new insights into NO-sGC-cGMP signaling and reveal cytochrome b5 reductase 3 as the first identified physiological sGC heme iron reductase in vascular smooth muscle cells, serving as a critical regulator of cGMP production and protein kinase G-dependent signaling.


Subject(s)
Cyclic GMP/metabolism , Cytochrome-B(5) Reductase/physiology , Signal Transduction/physiology , Soluble Guanylyl Cyclase/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Benzoates/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Oxidation-Reduction/drug effects , Rats , Signal Transduction/drug effects , Vasodilation/drug effects , Vasodilation/physiology
10.
J Biol Chem ; 292(4): 1145-1159, 2017 01 27.
Article in English | MEDLINE | ID: mdl-27923813

ABSTRACT

Nitroalkene fatty acids are formed in vivo and exert protective and anti-inflammatory effects via reversible Michael addition to thiol-containing proteins in key signaling pathways. Nitro-conjugated linoleic acid (NO2-CLA) is preferentially formed, constitutes the most abundant nitrated fatty acid in humans, and contains two carbons that could potentially react with thiols, modulating signaling actions and levels. In this work, we examined the reactions of NO2-CLA with low molecular weight thiols (glutathione, cysteine, homocysteine, cysteinylglycine, and ß-mercaptoethanol) and human serum albumin. Reactions followed reversible biphasic kinetics, consistent with the presence of two electrophilic centers in NO2-CLA located on the ß- and δ-carbons with respect to the nitro group. The differential reactivity was confirmed by computational modeling of the electronic structure. The rates (kon and koff) and equilibrium constants for both reactions were determined for different thiols. LC-UV-Visible and LC-MS analyses showed that the fast reaction corresponds to ß-adduct formation (the kinetic product), while the slow reaction corresponds to the formation of the δ-adduct (the thermodynamic product). The pH dependence of the rate constants, the correlation between intrinsic reactivity and thiol pKa, and the absence of deuterium solvent kinetic isotope effects suggested stepwise mechanisms with thiolate attack on NO2-CLA as rate-controlling step. Computational modeling supported the mechanism and revealed additional features of the transition states, anionic intermediates, and final neutral products. Importantly, the detection of cysteine-δ-adducts in human urine provided evidence for the biological relevance of this reaction. Finally, human serum albumin was found to bind NO2-CLA both non-covalently and to form covalent adducts at Cys-34, suggesting potential modes for systemic distribution. These results provide new insights into the chemical basis of NO2-CLA signaling actions.


Subject(s)
Linoleic Acid/chemistry , Nitro Compounds/chemistry , Serum Albumin/chemistry , Signal Transduction , Sulfhydryl Compounds/chemistry , Humans
11.
Am J Physiol Endocrinol Metab ; 315(2): E180-E195, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29486138

ABSTRACT

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a canonical regulator of cytoprotective gene expression, but evidence of its cross talk with other pathways, including metabolic ones, is ever increasing. Pharmacologic or systemic genetic activation of the Nrf2 pathway partially protects from obesity in mice and ameliorates fasting hyperglycemia in mice and humans. However, systemic Nrf2 deletion also protects from diet-induced obesity and insulin resistance in mice. To further investigate the effect of the disruption of Nrf2 on obesity in a tissue-specific manner, we focused on adipocytes and hepatocytes with targeted deletion of Nrf2. To this end, mice with cell-specific deletion of Nrf2 in adipocytes (ANKO) or hepatocytes (HeNKO) were fed a high-fat diet (HFD) for 6 mo and showed similar increases in body weight and body fat content. ANKO mice showed a partially deteriorated glucose tolerance, higher fasting glucose levels, and higher levels of cholesterol and nonesterified fatty acids compared with their Control counterparts. The HeNKO mice, though, had lower insulin levels and trended toward improved insulin sensitivity without having any difference in liver triglyceride accumulation. This study compared for the first time two conditional Nrf2 knockout models in adipocytes and in hepatocytes during HFD-induced obesity. None of these models could completely recapitulate the unexpected protection against obesity observed in the whole body Nrf2 knockout mice, but this study points out the differential roles that Nrf2 may play, beyond cytoprotection, in different target tissues and rather suggests systemic activation of the Nrf2 pathway as an effective means of prevention and treatment of obesity and type 2 diabetes.


Subject(s)
Adipocytes/metabolism , Diet, High-Fat/adverse effects , Hepatocytes/metabolism , NF-E2-Related Factor 2/metabolism , Obesity/genetics , Obesity/metabolism , Adiposity/genetics , Animals , Blood Glucose/metabolism , Body Composition/genetics , Body Weight/genetics , Glucose Intolerance/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Triglycerides/blood
12.
Nitric Oxide ; 77: 106-111, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29742447

ABSTRACT

Foundational advances in eicosanoid signaling, the free radical biology of oxygen and nitric oxide and mass spectrometry all converged to enable the discovery of nitrated unsaturated fatty acids. Due to the unique biochemical characteristics of fatty acid nitroalkenes, these species undergo rapid and reversible Michael addition of biological nucleophiles such as cysteine, leading to the post-translational modification of low molecular weight and protein thiols. This capability has led to the present understanding that nitro-fatty acid reaction with the alkylation-sensitive cysteine proteome leads to physiologically-beneficial alterations in transcriptional regulatory protein function, gene expression and in vivo rodent model responses to metabolic and inflammatory stress. These findings motivated the preclinical and clinical development of nitro-fatty acids as new drug candidates for treating acute and chronic metabolic and inflammatory disorders.


Subject(s)
Endothelium, Vascular/metabolism , Fatty Acids/metabolism , Inflammation/metabolism , Muscle, Smooth, Vascular/metabolism , Nitro Compounds/metabolism , Signal Transduction , Alkylation , Fatty Acids/chemistry , Humans , Nitro Compounds/chemistry
13.
Nitric Oxide ; 79: 31-37, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29944935

ABSTRACT

Nitrated oleic acid (NO2-OA) was first identified in 2003, and after the characterization of its formation and thiol reactivity, it was used as a prototypical molecule to investigate the physiological actions of endogenous nitrated fatty acids (NO2-FA). Based on in vitro observations showing significant activation of cytoprotective and anti-inflammatory signaling responses by NO2-FA, experiments were designed to determine their pharmacological potential. Supported by strong intellectual protection and favorable pharmacokinetic and pharmacodynamic data, 10-NO2-OA (CXA-10) underwent pharmaceutical development as a drug to treat fibrotic and inflammatory diseases. NO2-FA are at the intersection of three unconventional drug candidate classes that include 1) fatty acids, 2) metabolic intermediates and 3) electrophilic molecules. These three groups use different scaffolds for drug development, are characterized by broad activities and are individually gaining traction as alternatives to mono-target drug therapies. In particular, NO2-FA share key characteristics with currently approved pharmacological agents regarding reactivity, distribution, and mechanism of action. This review first presents the characteristics, liabilities, and opportunities that these different drug candidate classes display, and then discusses these issues in the context of current progress in the preclinical and clinical development of NO2-FA as drugs. Lessons learned from the novel approaches presented herein were considered early on during development to structurally define and improve NO2-FA and their disease targets.


Subject(s)
Fatty Acids/therapeutic use , Fibrosis/drug therapy , Inflammation/drug therapy , Nitro Compounds/therapeutic use , Animals , Chronic Disease , Humans
14.
Tetrahedron Lett ; 59(39): 3524-3527, 2018 Sep 26.
Article in English | MEDLINE | ID: mdl-31379396

ABSTRACT

15-oxo-Lipoxin A4 (15-oxo- LXA4) has been identified as a natural metabolite of the fatty acid signaling mediator Lipoxin A4. Herein, we report a total synthesis of the methyl ester of 15-oxo-LXA4 to be used in investigations of potential electrophilic bioactivity of this metabolite. The methyl ester of 15-oxo-LXA4 was synthesized in a convergent 15 step (9 steps longest linear) sequence starting from 1-octyn-3-ol and 2-deoxy-D-ribose with Sonogashira and Suzuki cross-couplings of a MIDA boronate as key steps.

15.
J Lipid Res ; 58(2): 375-385, 2017 02.
Article in English | MEDLINE | ID: mdl-27913584

ABSTRACT

Electrophilic nitro-FAs (NO2-FAs) promote adaptive and anti-inflammatory cell signaling responses as a result of an electrophilic character that supports posttranslational protein modifications. A unique pharmacokinetic profile is expected for NO2-FAs because of an ability to undergo reversible reactions including Michael addition with cysteine-containing proteins and esterification into complex lipids. Herein, we report via quantitative whole-body autoradiography analysis of rats gavaged with radiolabeled 10-nitro-[14C]oleic acid, preferential accumulation in adipose tissue over 2 weeks. To better define the metabolism and incorporation of NO2-FAs and their metabolites in adipose tissue lipids, adipocyte cultures were supplemented with 10-nitro-oleic acid (10-NO2-OA), nitro-stearic acid, nitro-conjugated linoleic acid, and nitro-linolenic acid. Then, quantitative HPLC-MS/MS analysis was performed on adipocyte neutral and polar lipid fractions, both before and after acid hydrolysis of esterified FAs. NO2-FAs preferentially incorporated in monoacyl- and diacylglycerides, while reduced metabolites were highly enriched in triacylglycerides. This differential distribution profile was confirmed in vivo in the adipose tissue of NO2-OA-treated mice. This pattern of NO2-FA deposition lends new insight into the unique pharmacokinetics and pharmacologic actions that could be expected for this chemically-reactive class of endogenous signaling mediators and synthetic drug candidates.


Subject(s)
Adipose Tissue/metabolism , Fatty Acids/metabolism , Oleic Acids/administration & dosage , Oleic Acids/metabolism , Adipose Tissue/chemistry , Alkenes/chemistry , Animals , Carbon Radioisotopes/chemistry , Cysteine/chemistry , Esterification , Fatty Acids/chemistry , Mice , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Oleic Acids/chemistry , Protein Processing, Post-Translational , Rats , Signal Transduction/drug effects , Tandem Mass Spectrometry
16.
Nat Chem Biol ; 11(7): 504-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26006011

ABSTRACT

The current perspective holds that the generation of secondary signaling mediators from nitrite (NO2(-)) requires acidification to nitrous acid (HNO2) or metal catalysis. Herein, the use of stable isotope-labeled NO2(-) and LC-MS/MS analysis of products reveals that NO2(-) also participates in fatty acid nitration and thiol S-nitrosation at neutral pH. These reactions occur in the absence of metal centers and are stimulated by autoxidation of nitric oxide ((•)NO) via the formation of symmetrical dinitrogen trioxide (nitrous anhydride, symN2O3). Although theoretical models have predicted physiological symN2O3 formation, its generation is now demonstrated in aqueous reaction systems, cell models and in vivo, with the concerted reactions of (•)NO and NO2(-) shown to be critical for symN2O3 formation. These results reveal new mechanisms underlying the NO2(-) propagation of (•)NO signaling and the regulation of both biomolecule function and signaling network activity via NO2(-)-dependent nitrosation and nitration reactions.


Subject(s)
Macrophages/chemistry , Nitrates/chemistry , Nitric Oxide/chemistry , Nitrites/chemistry , Nitrogen Oxides/chemistry , Nitrous Acid/chemistry , Animals , Cell Line , Glutathione/chemistry , Glutathione/metabolism , Hydrogen-Ion Concentration , Inflammation/chemically induced , Inflammation/metabolism , Kinetics , Linoleic Acids, Conjugated/chemistry , Linoleic Acids, Conjugated/metabolism , Linoleic Acids, Conjugated/pharmacology , Lipopolysaccharides , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Nitrates/metabolism , Nitric Oxide/metabolism , Nitrites/metabolism , Nitrites/pharmacology , Nitrogen Isotopes , Nitrogen Oxides/metabolism , Nitrosation , Nitrous Acid/metabolism , Oxygen Isotopes
17.
J Neurosci ; 33(22): 9259-72, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23719795

ABSTRACT

Although it is well established that many glutamatergic neurons sequester Zn(2+) within their synaptic vesicles, the physiological significance of synaptic Zn(2+) remains poorly understood. In experiments performed in a Zn(2+)-enriched auditory brainstem nucleus--the dorsal cochlear nucleus--we discovered that synaptic Zn(2+) and GPR39, a putative metabotropic Zn(2+)-sensing receptor (mZnR), are necessary for triggering the synthesis of the endocannabinoid 2-arachidonoylglycerol (2-AG). The postsynaptic production of 2-AG, in turn, inhibits presynaptic probability of neurotransmitter release, thus shaping synaptic strength and short-term synaptic plasticity. Zn(2+)-induced inhibition of transmitter release is absent in mutant mice that lack either vesicular Zn(2+) or the mZnR. Moreover, mass spectrometry measurements of 2-AG levels reveal that Zn(2+)-mediated initiation of 2-AG synthesis is absent in mice lacking the mZnR. We reveal a previously unknown action of synaptic Zn(2+): synaptic Zn(2+) inhibits glutamate release by promoting 2-AG synthesis.


Subject(s)
Endocannabinoids/biosynthesis , Neurotransmitter Agents/metabolism , Synapses/physiology , Zinc/physiology , Animals , Arachidonic Acids/metabolism , Chromatography, Liquid , Dendrites/physiology , Endocannabinoids/metabolism , Female , Glutamic Acid/metabolism , Glycerides/metabolism , Male , Mass Spectrometry , Mice , Mice, Inbred ICR , Mice, Knockout , Microscopy, Fluorescence , Nerve Fibers/physiology , Patch-Clamp Techniques , Signal Transduction/physiology , Synaptic Transmission/physiology
18.
J Biol Chem ; 288(35): 25626-25637, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23878198

ABSTRACT

Inflammation, characterized by the activation of both resident and infiltrated immune cells, is accompanied by increased production of oxidizing and nitrating species. Nitrogen dioxide, the proximal nitrating species formed under these conditions, reacts with unsaturated fatty acids to yield nitroalkene derivatives. These electrophilic products modulate protein function via post-translational modification of susceptible nucleophilic amino acids. Nitroalkenes react with Keap1 to instigate Nrf2 signaling, activate heat shock response gene expression, and inhibit NF-κB-mediated signaling, inducing net anti-inflammatory and tissue-protective metabolic responses. We report the purification and characterization of a NADPH-dependent liver enzyme that reduces the nitroalkene moiety of nitro-oleic acid, yielding the inactive product nitro-stearic acid. Prostaglandin reductase-1 (PtGR-1) was identified as a nitroalkene reductase by protein purification and proteomic studies. Kinetic measurements, inhibition studies, immunological and molecular biology approaches as well as clinical analyses confirmed this identification. Overexpression of PtGR-1 in HEK293T cells promoted nitroalkene metabolism to inactive nitroalkanes, an effect that abrogated the Nrf2-dependent induction of heme oxygenase-1 expression by nitro-oleic acid. These results situate PtGR-1 as a critical modulator of both the steady state levels and signaling activities of fatty acid nitroalkenes in vivo.


Subject(s)
Alcohol Oxidoreductases/metabolism , Liver/metabolism , Nitro Compounds/metabolism , Oleic Acid/metabolism , Signal Transduction/physiology , Stearic Acids/metabolism , Alcohol Oxidoreductases/genetics , Animals , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oleic Acid/genetics , Rats
19.
J Org Chem ; 79(1): 25-33, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24350701

ABSTRACT

Nitro-conjugated linoleic acids (NO2-cLA), endogenous nitrodiene lipids which act as inflammatory signaling mediators, were isolated and single isomers purified from the biomimetic acidic nitration products of conjugated linoleic acid (CLA). Structures were elucidated by means of detailed NMR and HPLC-MS/MS spectroscopic analysis and the relative double bond configurations assigned. Additional synthetic methods produced useful quantities and similar isomeric distributions of these unusual and reactive compounds for biological studies and isotopic standards, and the potential conversion of nitro-linoleic to nitro-conjugated linoleic acids was explored via a facile base-catalyzed isomerization. This represents one of the few descriptions of naturally occurring conjugated nitro dienes (in particular, 1-nitro 1,3-diene), an unusual and highly reactive motif with few biological examples extant.


Subject(s)
Linoleic Acids, Conjugated/chemistry , Lipids/chemistry , Nitro Compounds/chemistry , Biomimetics , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Tandem Mass Spectrometry
20.
Food Chem ; 437(Pt 1): 137767, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-37879157

ABSTRACT

Nitrated fatty acids are important anti-inflammatory and protective lipids formed in the gastric compartment, with conjugated linoleic acid (rumenic acid, RA, 9Z,11E-18:2) being the primary substrate for lipid nitration. The recently reported identification of nitrated rumelenic acid (NO2-RLA) in human urine has led to hypothesize that rumelenic acid (RLA, 9Z,11E,15Z-18:3) from dairy fat is responsible for the formation of NO2-RLA. To evaluate the source and mechanism of NO2-RLA formation, 15N labeled standards of NO2-RLA were synthesized and characterized. Afterward, milk fat with different RA and RLA levels was administered to mice in the presence of nitrite, and the appearance of nitrated fatty acids in plasma and urine followed. We confirmed the formation of NO2-RLA and defined the main metabolites in plasma, urine, and tissues. In conclusion, RLA obtained from dairy products is the main substrate for forming this novel electrophilic lipid reported to be present in human urine.


Subject(s)
Linoleic Acids, Conjugated , Nitrates , Mice , Humans , Animals , Nitrates/chemistry , Nitrites/metabolism , Nitrogen Dioxide , Fatty Acids/chemistry , Dairy Products , Linolenic Acids
SELECTION OF CITATIONS
SEARCH DETAIL