Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
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
2.
Int J Mol Sci ; 22(16)2021 Aug 22.
Article in English | MEDLINE | ID: mdl-34445757

ABSTRACT

Nitro-oleic acid (NO2-OA), a nitric oxide (NO)- and nitrite (NO2-)-derived electrophilic fatty acid metabolite, displays anti-inflammatory and anti-fibrotic signaling actions and therapeutic benefit in murine models of ischemia-reperfusion, atrial fibrillation, and pulmonary hypertension. Muscle LIM protein-deficient mice (Mlp-/-) develop dilated cardiomyopathy (DCM), characterized by impaired left ventricular function and increased ventricular fibrosis at the age of 8 weeks. This study investigated the effects of NO2-OA on cardiac function in Mlp-/- mice both in vivo and in vitro. Mlp-/- mice were treated with NO2-OA or vehicle for 4 weeks via subcutaneous osmotic minipumps. Wildtype (WT) littermates treated with vehicle served as controls. Mlp-/- mice exhibited enhanced TGFß signalling, fibrosis and severely reduced left ventricular systolic function. NO2-OA treatment attenuated interstitial myocardial fibrosis and substantially improved left ventricular systolic function in Mlp-/- mice. In vitro studies of TGFß-stimulated primary cardiac fibroblasts further revealed that the anti-fibrotic effects of NO2-OA rely on its capability to attenuate fibroblast to myofibroblast transdifferentiation by inhibiting phosphorylation of TGFß downstream targets. In conclusion, we demonstrate a substantial therapeutic benefit of NO2-OA in a murine model of DCM, mediated by interfering with endogenously activated TGFß signaling.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Nitro Compounds/therapeutic use , Oleic Acids/therapeutic use , Ventricular Function, Left/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Fibrosis , Heart/drug effects , LIM Domain Proteins/genetics , Mice , Muscle Proteins/genetics , Myocardium/metabolism , Nitro Compounds/pharmacology , Oleic Acids/pharmacology , Transforming Growth Factor beta/metabolism
3.
J Biol Chem ; 294(2): 397-404, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30478172

ABSTRACT

Homologous recombination (HR)-directed DNA double-strand break (DSB) repair enables template-directed DNA repair to maintain genomic stability. RAD51 recombinase (RAD51) is a critical component of HR and facilitates DNA strand exchange in DSB repair. We report here that treating triple-negative breast cancer (TNBC) cells with the fatty acid nitroalkene 10-nitro-octadec-9-enoic acid (OA-NO2) in combination with the antineoplastic DNA-damaging agents doxorubicin, cisplatin, olaparib, and γ-irradiation (IR) enhances the antiproliferative effects of these agents. OA-NO2 inhibited IR-induced RAD51 foci formation and enhanced H2A histone family member X (H2AX) phosphorylation in TNBC cells. Analyses of fluorescent DSB reporter activity with both static-flow cytometry and kinetic live-cell studies enabling temporal resolution of recombination revealed that OA-NO2 inhibits HR and not nonhomologous end joining (NHEJ). OA-NO2 alkylated Cys-319 in RAD51, and this alkylation depended on the Michael acceptor properties of OA-NO2 because nonnitrated and saturated nonelectrophilic analogs of OA-NO2, octadecanoic acid and 10-nitro-octadecanoic acid, did not react with Cys-319. Of note, OA-NO2 alkylation of RAD51 inhibited its binding to ssDNA. RAD51 Cys-319 resides within the SH3-binding site of ABL proto-oncogene 1, nonreceptor tyrosine kinase (ABL1), so we investigated the effect of OA-NO2-mediated Cys-319 alkylation on ABL1 binding and found that OA-NO2 inhibits RAD51-ABL1 complex formation both in vitro and in cell-based immunoprecipitation assays. The inhibition of the RAD51-ABL1 complex also suppressed downstream RAD51 Tyr-315 phosphorylation. In conclusion, RAD51 Cys-319 is a functionally significant site for adduction of soft electrophiles such as OA-NO2 and suggests further investigation of lipid electrophile-based combinational therapies for TNBC.


Subject(s)
Antineoplastic Agents/administration & dosage , DNA Damage/drug effects , Fatty Acids/administration & dosage , Rad51 Recombinase/metabolism , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/physiopathology , Alkylation , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cisplatin/administration & dosage , DNA Repair , Doxorubicin/administration & dosage , Drug Therapy, Combination , Fatty Acids/chemistry , Humans , Protein Binding/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Rad51 Recombinase/chemistry , Rad51 Recombinase/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
4.
Toxicol Appl Pharmacol ; 407: 115236, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32931793

ABSTRACT

Fatty acid nitroalkenes are reversibly-reactive electrophiles, endogenously detectable at nM concentrations, displaying anti-inflammatory actions. Nitroalkenes like 9- or 10-nitro-octadec-9-enoic acid (e.g. nitro-oleic acid, OA-NO2) pleiotropically suppress cardiovascular inflammatory responses, with pulmonary responses less well defined. C57BL/6 J male mice were intratracheally administered bleomycin (3 U/kg, ITB), to induce pulmonary inflammation and acute injury, or saline and were treated with 50 µL OA-NO2 (50 µg) or vehicle in the same instillation and 72 h post-exposure to assess anti-inflammatory properties. Bronchoalveolar lavage (BAL) and lung tissue were collected 7d later. ITB mice lost body weight, with OA-NO2 mitigating this loss (-2.3 ± 0.94 vs -0.4 ± 0.83 g). Histology revealed ITB induced cellular infiltration, proteinaceous debris deposition, and tissue injury, all significantly reduced by OA-NO2. Flow cytometry analysis of BAL demonstrated loss of Siglec F+/F4/80+/CD45+ alveolar macrophages with ITB (89 ± 3.5 vs 30 ± 3.7%). Analysis of CD11b/CD11c expressing cells showed ITB-induced non-resident macrophage infiltration (4 ± 2.3 vs 43 ± 2.4%) was decreased by OA-NO2 (24 ± 2.4%). Additionally, OA-NO2 attenuated increases in mature, activated interstitial macrophages (23 ± 4.8 vs. 43 ± 5.4%) in lung tissue digests. Flow analysis of CD31-/CD45-/Sca-1+ mesenchymal cells revealed ITB increased CD44+ populations (1 ± 0.4 vs 4 ± 0.4MFI), significantly reduced by OA-NO2 (3 ± 0.4MFI). Single cell analysis of mesenchymal cells by western blotting showed profibrotic ZEB1 protein expression induced by ITB. Lung digest CD45+ cells revealed ITB increased HMGB1+ cells, with OA-NO2 suppressing this response. Inhibition of HMGB1 expression correlated with increased basal phospholipid production and SP-B expression in the lung lining. These findings indicate OA-NO2 inhibits ITB-induced pro-inflammatory responses by modulating resident cell function.


Subject(s)
Acute Lung Injury/prevention & control , Alkenes/pharmacology , Bleomycin , Fatty Acids/pharmacology , Inflammation/prevention & control , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid , Inflammation/chemically induced , Inflammation/pathology , Leukocyte Common Antigens/metabolism , Lung/pathology , Macrophages, Alveolar/drug effects , Male , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Phospholipids/metabolism , Weight Loss/drug effects , Zinc Finger E-box-Binding Homeobox 1/biosynthesis , Zinc Finger E-box-Binding Homeobox 1/genetics
5.
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
6.
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
7.
J Biol Chem ; 293(4): 1120-1137, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29158255

ABSTRACT

Triple-negative breast cancer (TNBC) comprises ∼20% of all breast cancers and is the most aggressive mammary cancer subtype. Devoid of the estrogen and progesterone receptors, along with the receptor tyrosine kinase ERB2 (HER2), that define most mammary cancers, there are no targeted therapies for patients with TNBC. This, combined with a high metastatic rate and a lower 5-year survival rate than for other breast cancer phenotypes, means there is significant unmet need for new therapeutic strategies. Herein, the anti-neoplastic effects of the electrophilic fatty acid nitroalkene derivative, 10-nitro-octadec-9-enoic acid (nitro-oleic acid, NO2-OA), were investigated in multiple preclinical models of TNBC. NO2-OA reduced TNBC cell growth and viability in vitro, attenuated TNFα-induced TNBC cell migration and invasion, and inhibited the tumor growth of MDA-MB-231 TNBC cell xenografts in the mammary fat pads of female nude mice. The up-regulation of these aggressive tumor cell growth, migration, and invasion phenotypes is mediated in part by the constitutive activation of pro-inflammatory nuclear factor κB (NF-κB) signaling in TNBC. NO2-OA inhibited TNFα-induced NF-κB transcriptional activity in human TNBC cells and suppressed downstream NF-κB target gene expression, including the metastasis-related proteins intercellular adhesion molecule-1 and urokinase-type plasminogen activator. The mechanisms accounting for NF-κB signaling inhibition by NO2-OA in TNBC cells were multifaceted, as NO2-OA (a) inhibited the inhibitor of NF-κB subunit kinase ß phosphorylation and downstream inhibitor of NF-κB degradation, (b) alkylated the NF-κB RelA protein to prevent DNA binding, and (c) promoted RelA polyubiquitination and proteasomal degradation. Comparisons with non-tumorigenic human breast epithelial MCF-10A and MCF7 cells revealed that NO2-OA more selectively inhibited TNBC function. This was attributed to more facile mechanisms for maintaining redox homeostasis in normal breast epithelium, including a more favorable thiol/disulfide balance, greater extents of multidrug resistance protein-1 (MRP1) expression, and greater MRP1-mediated efflux of NO2-OA-glutathione conjugates. These observations reveal that electrophilic fatty acid nitroalkenes react with more alkylation-sensitive targets in TNBC cells to inhibit growth and viability.


Subject(s)
Cell Movement , Fatty Acids/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Survival , Fatty Acids/genetics , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
8.
Physiol Genomics ; 50(4): 287-295, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29473818

ABSTRACT

Nitro-conjugated linoleic acid (NO2-CLA) is formed by metabolic and inflammatory reactions of nitric oxide and nitrite, and represents the most abundant nitro-fatty acid species in humans. These electrophilic fatty acid nitroalkene derivatives mediate pleiotropic cell signaling responses. Here, we report a systematic approach to investigate the effect of NO2-CLA on human coronary artery smooth muscle cells (hCASMC), based on the RNA-Seq and bioinformatics analysis. There were extensive differentially expressed genes in NO2-CLA vs. control (510) and NO2-CLA vs. CLA (272) treatment groups, respectively. Notably, only minimal alterations were observed in CLA vs. control conditions, indicating that the electrophilic character of NO2-CLA is requited to induce differential gene expression responses independently from native CLA. Functional enrichment analysis of differentially expressed genes reveals multiple cellular processes to be affected under NO2-CLA treatment, including cell proliferation, lipid metabolism, antioxidant and inflammatory-related gene expression responses. These findings reveal that nitro-fatty acid derivatives such as NO2-CLA regulate a broad array of adaptive gene expression responses by hCASMC.


Subject(s)
Linoleic Acids, Conjugated/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Computational Biology/methods , Coronary Vessels/cytology , Coronary Vessels/drug effects , Gene Expression/drug effects , Gene Expression/genetics , Humans , Lipid Metabolism/genetics , Signal Transduction/drug effects
9.
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
10.
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
11.
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.

12.
Annu Rev Physiol ; 76: 79-105, 2014.
Article in English | MEDLINE | ID: mdl-24161076

ABSTRACT

Unsaturated fatty acids are metabolized to reactive products that can act as pro- or anti-inflammatory signaling mediators. Electrophilic fatty acid species, including nitro- and oxo-containing fatty acids, display salutary anti-inflammatory and metabolic actions. Electrophilicity can be conferred by both enzymatic and oxidative reactions, via the homolytic addition of nitrogen dioxide to a double bond or via the formation of α,ß-unsaturated carbonyl and epoxide substituents. The endogenous formation of electrophilic fatty acids is significant and influenced by diet, metabolic, and inflammatory reactions. Transcriptional regulatory proteins and enzymes can sense the redox status of the surrounding environment upon electrophilic fatty acid adduction of functionally significant, nucleophilic cysteines. Through this covalent and often reversible posttranslational modification, gene expression and metabolic responses are induced. At low concentrations, the pleiotropic signaling actions that are regulated by these protein targets suggest that some classes of electrophilic lipids may be useful for treating metabolic and inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents , Fatty Acids, Unsaturated/pharmacology , Animals , Epoxy Compounds/pharmacology , Fatty Acids/chemistry , Free Radicals , Humans , Keto Acids/pharmacology , Nitro Compounds/chemistry , Oxidation-Reduction , Prostaglandins/physiology , Signal Transduction/drug effects
13.
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
14.
Biochim Biophys Acta ; 1860(11 Pt A): 2428-2437, 2016 11.
Article in English | MEDLINE | ID: mdl-27431604

ABSTRACT

BACKGROUND: Inflammatory-mediated pathological processes in the endothelium arise as a consequence of the dysregulation of vascular homeostasis. Of particular importance are mediators produced by stimulated monocytes/macrophages inducing activation of endothelial cells (ECs). This is manifested by excessive soluble pro-inflammatory mediator production and cell surface adhesion molecule expression. Nitro-fatty acids are endogenous products of metabolic and inflammatory reactions that display immuno-regulatory potential and may represent a novel therapeutic strategy to treat inflammatory diseases. The purpose of our study was to characterize the effects of nitro-oleic acid (OA-NO2) on inflammatory responses and the endothelial-mesenchymal transition (EndMT) in ECs that is a consequence of the altered healing phase of the immune response. METHODS: The effect of OA-NO2 on inflammatory responses and EndMT was determined in murine macrophages and murine and human ECs using Western blotting, ELISA, immunostaining, and functional assays. RESULTS: OA-NO2 limited the activation of macrophages and ECs by reducing pro-inflammatory cytokine production and adhesion molecule expression through its modulation of STAT, MAPK and NF-κB-regulated signaling. OA-NO2 also decreased transforming growth factor-ß-stimulated EndMT and pro-fibrotic phenotype of ECs. These effects are related to the downregulation of Smad2/3. CONCLUSIONS: The study shows the pleiotropic effect of OA-NO2 on regulating EC-macrophage interactions during the immune response and suggests a role for OA-NO2 in the regulation of vascular endothelial immune and fibrotic responses arising during chronic inflammation. GENERAL SIGNIFICANCE: These findings propose the OA-NO2 may be useful as a novel therapeutic agent for treatment of cardiovascular disorders associated with dysregulation of the endothelial immune response.


Subject(s)
Endothelium, Vascular/drug effects , Epithelial-Mesenchymal Transition , Oleic Acids/pharmacology , Animals , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Inflammation/metabolism , MAP Kinase Signaling System , Macrophages/drug effects , Macrophages/metabolism , Mice , NF-kappa B/metabolism , STAT Transcription Factors/metabolism , Smad Proteins/metabolism , Transforming Growth Factor beta/pharmacology
15.
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
16.
Proc Natl Acad Sci U S A ; 111(22): 8167-72, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843165

ABSTRACT

Soluble epoxide hydrolase (sEH) is inhibited by electrophilic lipids by their adduction to Cys521 proximal to its catalytic center. This inhibition prevents hydrolysis of the enzymes' epoxyeicosatrienoic acid (EET) substrates, so they accumulate inducing vasodilation to lower blood pressure (BP). We generated a Cys521Ser sEH redox-dead knockin (KI) mouse model that was resistant to this mode of inhibition. The electrophilic lipid 10-nitro-oleic acid (NO2-OA) inhibited hydrolase activity and also lowered BP in an angiotensin II-induced hypertension model in wild-type (WT) but not KI mice. Furthermore, EET/dihydroxy-epoxyeicosatrienoic acid isomer ratios were elevated in plasma from WT but not KI mice following NO2-OA treatment, consistent with the redox-dead mutant being resistant to inhibition by lipid electrophiles. sEH was inhibited in WT mice fed linoleic acid and nitrite, key constituents of the Mediterranean diet that elevates electrophilic nitro fatty acid levels, whereas KIs were unaffected. These observations reveal that lipid electrophiles such as NO2-OA mediate antihypertensive signaling actions by inhibiting sEH and suggest a mechanism accounting for protection from hypertension afforded by the Mediterranean diet.


Subject(s)
Diet, Mediterranean , Epoxide Hydrolases/metabolism , Fatty Acids/metabolism , Hypertension/diet therapy , Hypertension/prevention & control , Angiotensin II/pharmacology , Animals , Blood Pressure , Cardiomegaly/diet therapy , Cardiomegaly/prevention & control , Cellulase , Disease Models, Animal , Epoxide Hydrolases/genetics , Gene Knock-In Techniques , Hypertension/chemically induced , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nitrates/metabolism , Nitrites/metabolism , Sulfhydryl Compounds/metabolism , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
17.
J Biol Chem ; 290(9): 5868-80, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25586183

ABSTRACT

15-Hydroxyprostaglandin dehydrogenase (15PGDH) is the primary enzyme catalyzing the conversion of hydroxylated arachidonic acid species to their corresponding oxidized metabolites. The oxidation of hydroxylated fatty acids, such as the conversion of prostaglandin (PG) E2 to 15-ketoPGE2, by 15PGDH is viewed to inactivate signaling responses. In contrast, the typically electrophilic products can also induce anti-inflammatory and anti-proliferative responses. This study determined that hydroxylated docosahexaenoic acid metabolites (HDoHEs) are substrates for 15PGDH. Examination of 15PGDH substrate specificity was conducted in cell culture (A549 and primary human airway epithelia and alveolar macrophages) using chemical inhibition and shRNA knockdown of 15PGDH. Substrate specificity is broad and relies on the carbon position of the acyl chain hydroxyl group. 14-HDoHE was determined to be the optimal DHA substrate for 15PGDH, resulting in the formation of its electrophilic metabolite, 14-oxoDHA. Consistent with this, 14-HDoHE was detected in bronchoalveolar lavage cells of mild to moderate asthmatics, and the exogenous addition of 14-oxoDHA to primary alveolar macrophages inhibited LPS-induced proinflammatory cytokine mRNA expression. These data reveal that 15PGDH-derived DHA metabolites are biologically active and can contribute to the salutary signaling actions of Ω-3 fatty acids.


Subject(s)
Fatty Acids, Omega-3/metabolism , Hydroxyprostaglandin Dehydrogenases/metabolism , Lipids/chemistry , Signal Transduction , Bronchoalveolar Lavage Fluid/cytology , Cell Line, Tumor , Cells, Cultured , Cytokines/genetics , Docosahexaenoic Acids/metabolism , Epithelial Cells/metabolism , Fatty Acids, Unsaturated/metabolism , Gene Expression/drug effects , Humans , Hydroxylation , Hydroxyprostaglandin Dehydrogenases/genetics , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Oxidation-Reduction , RNA Interference , Respiratory System/cytology , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity
18.
Cardiovasc Drugs Ther ; 30(6): 579-586, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27858190

ABSTRACT

RATIONALE: Pulmonary hypertension (PH) represents a serious health complication accompanied with hypoxic conditions, elevated levels of asymmetric dimethylarginine (ADMA), and overall dysfunction of pulmonary vascular endothelium. Since the prevention strategies for treatment of PH remain largely unknown, our study aimed to explore the effect of nitro-oleic acid (OA-NO2), an exemplary nitro-fatty acid (NO2-FA), in human pulmonary artery endothelial cells (HPAEC) under the influence of hypoxia or ADMA. METHODS: HPAEC were treated with OA-NO2 in the absence or presence of hypoxia and ADMA. The production of nitric oxide (NO) and interleukin-6 (IL-6) was monitored using the Griess method and ELISA, respectively. The expression or activation of different proteins (signal transducer and activator of transcription 3, STAT3; hypoxia inducible factor 1α, HIF-1α; endothelial nitric oxide synthase, eNOS; intercellular adhesion molecule-1, ICAM-1) was assessed by the Western blot technique. RESULTS: We discovered that OA-NO2 prevents development of endothelial dysfunction induced by either hypoxia or ADMA. OA-NO2 preserves normal cellular functions in HPAEC by increasing NO production and eNOS expression. Additionally, OA-NO2 inhibits IL-6 production as well as ICAM-1 expression, elevated by hypoxia and ADMA. Importantly, the effect of OA-NO2 is accompanied by prevention of STAT3 activation and HIF-1α stabilization. CONCLUSION: In summary, OA-NO2 eliminates the manifestation of hypoxia- and ADMA-mediated endothelial dysfunction in HPAEC via the STAT3/HIF-1α cascade. Importantly, our study is bringing a new perspective on molecular mechanisms of NO2-FAs action in pulmonary endothelial dysfunction, which represents a causal link in progression of PH. Graphical Abstract ᅟ.


Subject(s)
Cell Hypoxia/drug effects , Endothelial Cells/drug effects , Oleic Acids/pharmacology , Arginine/analogs & derivatives , Arginine/pharmacology , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Pulmonary Artery/cytology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism
19.
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
20.
Am J Respir Cell Mol Biol ; 51(1): 155-62, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24521348

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by adverse remodeling of pulmonary arteries. Although the origin of the disease and its underlying pathophysiology remain incompletely understood, inflammation has been identified as a central mediator of disease progression. Oxidative inflammatory conditions support the formation of electrophilic fatty acid nitroalkene derivatives, which exert potent anti-inflammatory effects. The current study investigated the role of 10-nitro-oleic acid (OA-NO2) in modulating the pathophysiology of PAH in mice. Mice were kept for 28 days under normoxic or hypoxic conditions, and OA-NO2 was infused subcutaneously. Right ventricular systolic pressure (RVPsys) was determined, and right ventricular and lung tissue was analyzed. The effect of OA-NO2 on cultured pulmonary artery smooth muscle cells (PASMCs) and macrophages was also investigated. Changes in RVPsys revealed increased pulmonary hypertension in mice on hypoxia, which was significantly decreased by OA-NO2 administration. Right ventricular hypertrophy and fibrosis were also attenuated by OA-NO2 treatment. The infiltration of macrophages and the generation of reactive oxygen species were elevated in lung tissue of mice on hypoxia and were diminished by OA-NO2 treatment. Moreover, OA-NO2 decreased superoxide production of activated macrophages and PASMCs in vitro. Vascular structural remodeling was also limited by OA-NO2. In support of these findings, proliferation and activation of extracellular signal-regulated kinases 1/2 in cultured PASMCs was less pronounced on application of OA-NO2.Our results show that the oleic acid nitroalkene derivative OA-NO2 attenuates hypoxia-induced pulmonary hypertension in mice. Thus, OA-NO2 represents a potential therapeutic agent for the treatment of PAH.


Subject(s)
Disease Models, Animal , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/physiopathology , Inflammation/prevention & control , Oleic Acids/therapeutic use , Animals , Blotting, Western , Cell Proliferation/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Familial Primary Pulmonary Hypertension , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/pathology , Inflammation/etiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL