Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Am J Physiol Heart Circ Physiol ; 325(6): H1446-H1460, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37889254

ABSTRACT

Epidemiological evidence shows that residential proximity to greenspaces is associated with lower risk of all-cause and cardiovascular mortality; however, the mechanism(s) underlying this link remains unclear. Plants emit biogenic volatile organic compounds such as α-pinene that could elicit beneficial cardiovascular effects. To explore the role of α-pinene more directly, we studied the metabolism and the vascular effects of α-pinene. We found that exposure of mice to α-pinene (1 ppm, 6 h) generated two phase I oxidation metabolites, cis- and trans-verbenol [(1R,2R,5R)-verbenol and (1 R,2S,5R)-verbenol)] and myrtenol [(1S,5R)-(+)-myrtenol] that were identified in urine by GC-MS. Precontracted naïve murine male and female aorta and superior mesenteric artery (SMA) were relaxed robustly (60% tension reduction) by increasing concentrations of α-pinene, myrtenol, and verbenol to 0.3 mM, whereas 1 mM α-pinene was vasotoxic. The SMA was six times more sensitive than the aorta to α-pinene. Both myrtenol and verbenol were equally potent and efficacious as parent α-pinene in male and female SMA. The sensitive portion of the α-pinene-, myrtenol-, and verbenol-induced relaxations in male SMA was mediated by 1) endothelium, 2) eNOS-derived NO, and 3) guanylyl cyclase (GC) activity. Moreover, α-pinene activated the transient receptor potential ankyrin-1 (TRPA1) channel whereas the metabolites did not. Endothelial-derived NO regulates blood flow, blood pressure, and thrombosis, and it is plausible that inhaled (and ingested) α-pinene (or its metabolites) augments NO release to mediate the cardiovascular benefits of exposure to greenness.NEW & NOTEWORTHY A common plant-derived biogenic volatile organic compound, α-pinene, and two of its metabolites, myrtenol and verbenol, stimulate vasorelaxation in murine superior mesenteric artery. Both α-pinene- and its metabolites induce vasorelaxation by activation of the endothelium, nitric oxide, and guanylyl cyclase. α-Pinene also activates the transient receptor potential ankyrin-1. Positive associations between greenness exposure and human cardiovascular health may be a result of the vascular action of α-pinene and its metabolites, a novel consideration.


Subject(s)
Ankyrins , Monoterpenes , Humans , Animals , Mice , Monoterpenes/pharmacology , Monoterpenes/metabolism , Endothelium/metabolism , Guanylate Cyclase
2.
Toxicol Appl Pharmacol ; 426: 115647, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34271065

ABSTRACT

Acrolein, an electrophilic α,ß-unsaturated aldehyde, is present in foods and beverages, and is a product of incomplete combustion, and thus, reaches high ppm levels in tobacco smoke and structural fires. Exposure to acrolein is linked with cardiopulmonary toxicity and cardiovascular disease risk. The hypothesis of this study is the direct effects of acrolein in isolated murine blood vessels (aorta and superior mesenteric artery, SMA) are transient receptor potential ankyrin-1 (TRPA1) dependent. Using isometric myography, isolated aorta and SMA were exposed to increasing levels of acrolein. Acrolein inhibited phenylephrine (PE)-induced contractions (approximately 90%) in aorta and SMA of male and female mice in a concentration-dependent (0.01-100 µM) manner. The major metabolite of acrolein, 3-hydroxypropylmercapturic acid (3HPMA), also relaxed PE-precontracted SMA. As the SMA was 20× more sensitive to acrolein than aorta (SMA EC50 0.8 ± 0.2 µM; aorta EC50 > 29.4 ± 4.4 µM), the mechanisms of acrolein-induced relaxation were studied in SMA. The potency of acrolein-induced relaxation was inhibited significantly by: 1) mechanically-impaired endothelium; 2) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME); 3) guanylyl cyclase (GC) inhibitor (ODQ); and, 4) a TRPA1 antagonist (A967079). TRPA1 positive immunofluorescence was present in the endothelium. Compared with other known TRPA1 agonists, including allyl isothiocyanate (AITC), cinnamaldehyde, crotonaldehyde, and formaldehyde, acrolein stimulated a more potent TRPA1-dependent relaxation. Acrolein, at high concentration [100 µM], induced tension oscillations (spasms) independent of TRPA1 in precontracted SMA but not in aorta. In conclusion, acrolein is vasorelaxant at low levels (physiological) yet vasotoxic at high levels (toxicological).


Subject(s)
Acetylcysteine/analogs & derivatives , Acrolein/pharmacology , Aorta, Thoracic/drug effects , Mesenteric Artery, Superior/drug effects , TRPA1 Cation Channel/physiology , Acetylcysteine/blood , Acetylcysteine/pharmacology , Acrolein/blood , Animals , Aorta, Thoracic/physiology , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/physiology , Glutathione S-Transferase pi/genetics , Glutathione S-Transferase pi/physiology , Male , Mesenteric Artery, Superior/physiology , Mice, Inbred C57BL , Mice, Knockout , TRPA1 Cation Channel/genetics
4.
Toxicol Appl Pharmacol ; 398: 115012, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32320793

ABSTRACT

INTRODUCTION: Crotonaldehyde (CR) is an electrophilic α,ß-unsaturated aldehyde present in foods and beverages and is a minor metabolite of 1,3-butadiene. CR is a product of incomplete combustion, and is at high levels in smoke of cigarettes and structural fires. Exposure to CR has been linked to cardiopulmonary toxicity and cardiovascular disease. OBJECTIVE: The purpose of this study was to examine the direct effects of CR in murine blood vessels (aorta and superior mesenteric artery, SMA) using an in vitro system. METHODS AND RESULTS: CR induced concentration-dependent (1-300 µM) relaxations (75-80%) in phenylephrine (PE) precontracted aorta and SMA. Because the SMA was 20× more sensitive to CR than aorta (SMA EC50 3.8 ± 0.5 µM; aorta EC50 76.0 ± 2.0 µM), mechanisms of CR relaxation were studied in SMA. The CR-induced relaxation at low concentrations (1-30 µM) was inhibited by: 1) mechanically-impaired endothelium; 2) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME); 3) guanylyl cyclase (GC) inhibitor (ODQ); 4) transient receptor potential ankyrin-1 (TRPA1) antagonist (A967079); and, 5) by non-vasoactive level of nicotine (1 µM). Similarly, a TRPA1 agonist, allyl isothiocyanate (AITC; mustard oil), stimulated SMA relaxation dependent on TRPA1, endothelium, NO, and GC. Consistent with these mechanisms, TRPA1 was present in the SMA endothelium. CR, at higher concentrations (100-300 µM), induced tension oscillations (spasms) and irreversibly impaired contractility (a vasotoxic effect enhanced by impaired endothelium). CONCLUSIONS: CR relaxation depends on a functional endothelium and TRPA1, whereas vasotoxicity is enhanced by endothelium dysfunction. Thus, CR is both vasoactive and vasotoxic along a concentration continuum.


Subject(s)
Aldehydes/pharmacology , Aorta, Thoracic/drug effects , Endothelium, Vascular/drug effects , TRPA1 Cation Channel/metabolism , Vasodilation/drug effects , Animals , Aorta, Thoracic/metabolism , Endothelium, Vascular/metabolism , Female , Male , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Phenylephrine/metabolism
5.
Front Physiol ; 10: 277, 2019.
Article in English | MEDLINE | ID: mdl-30984013

ABSTRACT

Formaldehyde (FA), the smallest aldehyde, is generated endogenously, and is widespread in the environment in foods, beverages and as a gas phase product of incomplete combustion. The main metabolite of FA, formate, was increased significantly in murine urine (∼3×) after overnight feeding. Because feeding increases mesenteric blood flow, we explored the direct effects of FA in isolated murine superior mesenteric artery (SMA). Over the concentration range of 30-1,200 µM, FA strongly and reversibly relaxed contractions of SMA induced by three different agonists: phenylephrine (PE), thromboxane A2 analog (U46,619) and high potassium (60K, 60 mM K+). Formate (to 1.5 mM) induced a modest relaxation. FA (>1,500 µM) irreversibly depressed vascular function in SMA indicating vasotoxicity. The sensitivity (EC50) but not the efficacy (% relaxation) of FA-induced relaxations was dependent on blood vessel type (SMA << aorta) and contractile agonist (PE, EC50= 52 ± 14 µM; U46,619, EC50= 514 ± 129 µM; 60K, EC50= 1,093 ± 87 µM). The most sensitive component of FA vasorelaxation was within physiological levels (30-150 µM) and was inhibited significantly by: (1) mechanically impaired endothelium; (2) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME); (3) transient receptor potential ankyrin-1 (TRPA1) antagonist (A967079); (4) guanylyl cyclase (GC) inhibitor (ODQ); and, (5) K+ channel inhibitor (BaCl2). A similar mechanism of SMA vasorelaxation was stimulated by the TRPA1 agonist cinnamaldehyde. Positive TRPA1 immunofluorescent staining and gene-specific sequence were present in SMA but not in aorta. These data indicate FA, but not formate, robustly relaxes SMA via a sensitive TRPA1- and endothelium-dependent mechanism that is absent in aorta. Thus, as FA levels increase with feeding, FA likely contributes to the physiological reflex of post-prandial hyperemia via SMA vasodilatation.

6.
J Stat Comput Simul ; 85(9): 1902-1916, 2015.
Article in English | MEDLINE | ID: mdl-26412909

ABSTRACT

The statistical methods for variable selection and prediction could be challenging when missing covariates exist. Although multiple imputation (MI) is a universally accepted technique for solving missing data problem, how to combine the MI results for variable selection is not quite clear, because different imputations may result in different selections. The widely applied variable selection methods include the sparse partial least-squares (SPLS) method and the penalized least-squares method, e.g. the elastic net (ENet) method. In this paper, we propose an MI-based weighted elastic net (MI-WENet) method that is based on stacked MI data and a weighting scheme for each observation in the stacked data set. In the MI-WENet method, MI accounts for sampling and imputation uncertainty for missing values, and the weight accounts for the observed information. Extensive numerical simulations are carried out to compare the proposed MI-WENet method with the other competing alternatives, such as the SPLS and ENet. In addition, we applied the MIWENet method to examine the predictor variables for the endothelial function that can be characterized by median effective dose (ED50) and maximum effect (Emax) in an ex-vivo phenylephrine-induced extension and acetylcholine-induced relaxation experiment.

7.
Mol Pharmacol ; 82(4): 601-13, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22740640

ABSTRACT

Previously we demonstrated that aldehyde dehydrogenase (ALDH) 1a1 is the major ALDH expressed in mouse liver and is an effective catalyst in metabolism of lipid aldehydes. Quantitative real-time polymerase chain reaction analysis revealed a ≈2.5- to 3-fold induction of the hepatic ALDH1A1 mRNA in mice administered either acrolein (5 mg/kg acrolein p.o.) or butylated hydroxylanisole (BHA) (0.45% in the diet) and of cytosolic NAD⁺-dependent ALDH activity. We observed ≈2-fold increases in ALDH1A1 mRNA levels in both Nrf2⁺/⁺ and Nrf2⁻/⁻ mice treated with BHA compared with controls, suggesting that BHA-induced expression is independent of nuclear factor E2-related factor 2 (Nrf2). The levels of activator protein-1 (AP-1) mRNA and protein, as well as the amount of phosphorylated c-Jun were significantly increased in mouse liver or Hepa1c1c7 cells treated with either BHA or acrolein. With use of luciferase reporters containing the 5'-flanking sequence of Aldh1a1 (-1963/+27), overexpression of c-Jun resulted in an ≈4-fold induction in luciferase activity, suggesting that c-Jun transactivates the Aldh1a1 promoter as a homodimer and not as a c-Jun/c-Fos heterodimer. Promoter deletion and mutagenesis analyses demonstrated that the AP-1 site at position -758 and possibly -1069 relative to the transcription start site was responsible for c-Jun-mediated transactivation. Electrophoretic mobility shift assay analysis with antibodies against c-Jun and c-Fos showed that c-Jun binds to the proximal AP-1 site at position -758 but not at -1069. Recruitment of c-Jun to this proximal AP-1 site by BHA was confirmed by chromatin immunoprecipitation analysis, indicating that recruitment of c-Jun to the mouse Aldh1a1 gene promoter results in increased transcription. This mode of regulation of an ALDH has not been described before.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Transcription Factor AP-1/metabolism , Acrolein/toxicity , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Butylated Hydroxyanisole/toxicity , Cell Line, Tumor , Gene Expression Regulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/cytology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , NF-E2-Related Factor 2/genetics , Nuclear Proteins/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , Retinal Dehydrogenase , Transcription Factor AP-1/genetics , Transcription, Genetic
8.
Toxicol Appl Pharmacol ; 217(3): 277-88, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17095030

ABSTRACT

Increased risk of vasospasm, a spontaneous hyperconstriction, is associated with atherosclerosis, cigarette smoking, and hypertension-all conditions involving oxidative stress, lipid peroxidation, and inflammation. To test the role of the lipid peroxidation- and inflammation-derived aldehyde, acrolein, in human vasospasm, we developed an ex vivo model using human coronary artery bypass graft (CABG) blood vessels and a demonstrated acrolein precursor, allylamine. Allylamine induces hypercontraction in isolated rat coronary artery in a semicarbazide-sensitive amine oxidase activity (SSAO) dependent manner. Isolated human CABG blood vessels (internal mammary artery, radial artery, saphenous vein) were used to determine: (1) vessel responses and sensitivity to acrolein, allylamine, and H(2)O(2) exposure (1 microM-1 mM), (2) SSAO dependence of allylamine-induced effects using SSAO inhibitors (semicarbazide, 1 mM; MDL 72274-E, active isomer; MDL 72274-Z, inactive isomer; 100 microM), (3) the vasoactive effects of two other SSAO amine substrates, benzylamine and methylamine, and (4) the contribution of extracellular Ca(2+) to hypercontraction. Acrolein or allylamine but not H(2)O(2), benzylamine, or methylamine stimulated spontaneous and pharmacologically intractable hypercontraction in CABG blood vessels that was similar to clinical vasospasm. Allylamine-induced hypercontraction and blood vessel SSAO activity were abolished by pretreatment with semicarbazide or MDL 72274-E but not by MDL 72274-Z. Allylamine-induced hypercontraction also was significantly attenuated in Ca(2+)-free buffer. In isolated aorta of spontaneously hypertensive rat, allylamine-induced an SSAO-dependent contraction and enhanced norepinephrine sensitivity but not in Sprague-Dawley rat aorta. We conclude that acrolein generation in the blood vessel wall increases human susceptibility to vasospasm, an event that is enhanced in hypertension.


Subject(s)
Acrolein/pharmacology , Allylamine/pharmacology , Blood Vessels/drug effects , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Acrolein/metabolism , Adult , Aged , Aged, 80 and over , Allyl Compounds/pharmacology , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Amine Oxidase (Copper-Containing)/metabolism , Animals , Aorta/drug effects , Aorta/physiology , Blood Vessels/metabolism , Dose-Response Relationship, Drug , Drug Antagonism , Female , Humans , In Vitro Techniques , Male , Middle Aged , Propylamines/pharmacology , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Semicarbazides/pharmacology
9.
Toxicol Appl Pharmacol ; 217(3): 266-76, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17069868

ABSTRACT

Acrolein is generated endogenously during lipid peroxidation and inflammation and is an environmental pollutant. Protein adducts of acrolein are detected in atherosclerotic plaques and neurons of patients with Alzheimer's disease. To understand vascular effects of acrolein exposure, we studied acrolein vasoreactivity in perfused rodent mesenteric bed. Acrolein induced endothelium-dependent vasodilatation that was more robust and more sensitive than dilation induced by 4-hydroxy-trans-2-nonenal, trans-2-hexenal, or propionaldehyde. Acrolein-induced vasodilatation was mediated by K(+)-sensitive components, e.g., it was abolished in 0 [K(+)](o) buffer or in 3 mM tetrabutylammonium, inhibited 75% in 50 microM ouabain, and inhibited 64% in 20 mM K(+) buffer. Moreover, combined treatment with the Ca(2+)-activated K(+) channel inhibitors 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34, 100 nM) and apamin (5 microM) significantly reduced vasodilatation without altering sensitivity to acrolein. However, acrolein-induced % dilation was unaffected by l-NAME or indomethacin pretreatment indicating mechanistic independence of NO and prostaglandins. Moreover, acrolein induced vasodilatation in cirazoline-precontracted mesenteric bed of eNOS-null mice confirming eNOS independence. Pretreatment with 6-(2-propargyloxyphenyl) hexanoic acid (PPOH 50 microM), an epoxygenase inhibitor, or the superoxide dismutase mimetic Tempol (100 microM) significantly attenuated acrolein-induced vasodilatation. Collectively, these data indicate that acrolein stimulates mesenteric bed vasodilatation due to endothelium-derived signal(s) that is K(+)-, ouabain-, PPOH-, and Tempol-sensitive, and thus, a likely endothelium-derived hyperpolarizing factor (EDHF). These data indicate that low level acrolein exposure associated with vascular oxidative stress or inflammation stimulates vasodilatation via EDHF release in medium-sized arteries--a novel function.


Subject(s)
Acrolein/toxicity , Biological Factors/metabolism , Blood Vessels/drug effects , Environmental Pollutants/toxicity , Mesentery/blood supply , Vasodilation/drug effects , Animals , Apamin/pharmacology , Blood Vessels/metabolism , Drug Interactions , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Male , Mesentery/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley
10.
Am J Physiol Heart Circ Physiol ; 286(2): H667-76, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14715500

ABSTRACT

It is hypothesized that methylamine (MA) and semicarbazide-sensitive amine oxidase (SSAO) activity are involved in the cardiovascular complications in human diabetics. To test this, we 1) determined the acute vasoactive effects of MA (1-1,000 micromol/l) in uncontracted and norepinephrine (NE; 1 micromol/l)-precontracted human blood vessels used for coronary artery bypass grafts [left internal mammary artery (LIMA), radial artery (RA), and right saphenous vein (RSV)]; 2) tested whether MA effects in LIMA and RSV were dependent on SSAO activity using the SSAO inhibitor semicarbazide (1 mmol/l, 15 min); 3) determined the effects of MA metabolites formaldehyde and hydrogen peroxide in LIMA and RSV; 4) tested whether the MA response was nitric oxide, prostaglandin, or hyperpolarization dependent; 5) measured the LIMA and RSV cGMP levels after MA exposure; and 6) quantified SSAO activity in LIMA, RA, and RSV. In NE-precontracted vessels, MA stimulated a biphasic response in RA and RSV (rapid contraction followed by prolonged relaxation) and dominant relaxation in LIMA (mean +/- SE, %relaxation: 55.4 +/- 3.9, n = 30). The MA-induced relaxation in LIMA was repeatable, nontoxic, and age independent. Semicarbazide significantly blocked MA-induced relaxation (%inhibition: 82.5 +/- 4.8, n = 7) and SSAO activity (%inhibition: 98.1 +/- 1.3, n = 26) in LIMA. Formaldehyde (%relaxation: 37.3 +/- 18.6, n = 3) and H(2)O(2) (%relaxation: 55.6 +/- 9.0, n = 9) at 1 mmol/l relaxed NE-precontracted LIMA comparable with MA. MA-induced relaxation in LIMA was nitric oxide, prostaglandin, and possibly cGMP independent and blocked by hyperpolarization. We conclude that vascular SSAO activity may convert endogenous amines, like MA, to vasoactive metabolites.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Formaldehyde/pharmacology , Hydrogen Peroxide/pharmacology , Mammary Arteries/physiology , Methylamines/pharmacology , Muscle, Smooth, Vascular/physiology , Vasoconstrictor Agents/pharmacology , Adult , Aged , Aged, 80 and over , Aging/physiology , Animals , Female , Humans , In Vitro Techniques , Kinetics , Male , Mammary Arteries/drug effects , Mammary Arteries/growth & development , Middle Aged , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/growth & development , Rats , Regression Analysis , Vasoconstriction/drug effects , Vasodilation/drug effects
11.
In Vitro Cell Dev Biol Anim ; 38(9): 523-8, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12703980

ABSTRACT

Components of fetal calf serum (FCS) are known to contribute to growth and maintenance of cultured cells. Fetal calf serum supplementation of media also may contribute to the cytotoxicity of other substances to cells grown in vitro. Semicarbazide-sensitive amine oxidase (SSAO) enzyme, present in FCS, metabolizes primary amines and contributes to amine cytotoxicity in vascular smooth muscle cells (VSMC). In cell culture experiments, the media used may greatly affect enzymic activities such as SSAO. In these studies, the SSAO activity in FCS, cultured rat aortic VSMC, and rat plasma was determined in the presence and absence of various culture media. Semicarbazide-sensitive amine oxidase activity in FCS (5-20 microl) was significantly enhanced (approximately 1.5- to 2-fold) in the presence of various culture media, with Dulbecco modified Eagle medium (DMEM), causing the greatest enhancement. Dulbecco modified Eagle medium enhanced the SSAO activity of cultured VSMC in two of the four passages but reduced activity in two passages. Activity in rat plasma was reduced by approximately 25% in the presence of DMEM. The concentrations of various media components, such as glucose, sodium pyruvate, pyridoxine.HCl, and L-glutamine, were not correlated with enhancement. This study identifies an important enhancement effect of culture media on the FCS enzyme, SSAO, although the media components responsible for the enhancement are yet to be identified.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Culture Media , Muscle, Smooth, Vascular/enzymology , Semicarbazides/pharmacology , Amine Oxidase (Copper-Containing)/drug effects , Animals , Aorta , Cell Culture Techniques/methods , Cell Survival/drug effects , Glucose/pharmacology , Glutamine/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Rats , Rats, Sprague-Dawley
12.
Atherosclerosis ; 158(2): 339-50, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11583712

ABSTRACT

Oxidation of low-density lipoproteins (LDL) generates high concentrations of unsaturated aldehydes, such as 4-hydroxy trans-2-nonenal (HNE). These aldehydes are mitogenic to vascular smooth muscle cells and sustain a vascular inflammation. Nevertheless, the processes that mediate and regulate the vascular metabolism of these aldehydes have not been examined. In this communication, we report the identification of the major metabolic pathways and products of [(3)H]-HNE in rat aortic smooth muscle cells in culture. High-performance liquid chromatography separation of the radioactivity recovered from these cells revealed that a large (60-65%) proportion of the metabolism was linked to glutathione (GSH). Electrospray mass spectrometry showed that glutathionyl-1,4 dihydroxynonene (GS-DHN) was the major metabolite of HNE in these cells. The formation of GS-DHN appears to be due aldose reductase (AR)-catalyzed reduction of glutathionyl 4-hydroxynonanal (GS-HNE), since inhibitors of AR (tolrestat or sorbinil) prevented GS-DHN formation, and increased the fraction of the glutathione conjugate remaining as GS-HNE. Gas chromatography-chemical ionization mass spectroscopy of the metabolites identified a subsidiary route of HNE metabolism leading to the formation of 4-hydroxynonanoic acid (HNA). Oxidation to HNA accounted for 25-30% of HNE metabolism. The formation of HNA was inhibited by cyanamide, indicating that the acid is derived from an aldehyde dehydrogenase (ALDH)-catalyzed pathway. The overall rate of HNE metabolism was insensitive to inhibition of AR or ALDH, although inhibition of HNA formation by cyanamide led to a corresponding increase in the fraction of HNE metabolized by the GSH-linked pathway, indicating that ALDH-catalyzed oxidation competes with glutathione conjugation. These metabolic pathways may be the key regulators of the vascular effects of HNE and oxidized LDL.


Subject(s)
Aldehydes/metabolism , Imidazolidines , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/metabolism , Aldehyde Reductase/antagonists & inhibitors , Aldehyde Reductase/metabolism , Alkenes/metabolism , Animals , Aorta/metabolism , Cells, Cultured , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Imidazoles/pharmacology , Male , Mass Spectrometry , Naphthalenes , Rats , Rats, Sprague-Dawley
13.
Toxicol Appl Pharmacol ; 175(2): 149-59, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11543647

ABSTRACT

We hypothesized that allylamine (AA) induces subendocardial necrosis in mammals via coronary artery (CA) vasospasm. Additionally, AA toxicity is likely dependent on the enzyme semicarbazide-sensitive amine oxidase (SSAO), which is highly expressed in the aorta of rats and humans. We tested whether AA or acrolein (1, 10, 100, and 1000 microM), a highly reactive product of AA metabolism by SSAO, could contract CA or thoracic aorta (TA) in vitro and if the AA effects involved SSAO. AA or acrolein produced a similar pattern of responses in both CA and TA rings at 100 and 1000 microM, including (1) increased basal tension, (2) enhanced agonist-induced contraction (hypercontractility or vasospasm), (3) remarkable, agonist-induced slow wave vasomotion (vasospasm), and (4) irreversible reduction in vessel contractility after 1 mM exposure. Endothelium-dependent acetylcholine-induced relaxation was not altered during vasospasm in either vessel. Pretreatment with the SSAO inhibitor semicarbazide (1 mM; 10 min) prevented or significantly reduced the majority of AA's effects in both CA and TA rings and inhibited 100% of the SSAO activity present in rat TA and human CA and TA. We propose a two-step model for AA induction of CA vasospasm and resultant myocardial necrosis: (1) metabolism of AA to acrolein by coronary arterial SSAO activity and (2) acrolein induction of CA vasospasm independent of endothelial injury-a novel path.


Subject(s)
Acrolein/toxicity , Allylamine/toxicity , Amine Oxidase (Copper-Containing)/metabolism , Coronary Vasospasm/chemically induced , Muscle, Smooth, Vascular/drug effects , Semicarbazides/pharmacology , Acrolein/antagonists & inhibitors , Allylamine/antagonists & inhibitors , Analysis of Variance , Animals , Coronary Vasospasm/metabolism , Coronary Vessels/drug effects , Drug Interactions , Humans , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Sprague-Dawley , Thoracic Arteries/drug effects
14.
Chem Biol Interact ; 130-132(1-3): 563-71, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11306075

ABSTRACT

Phospholipid peroxidation generates a variety of aldehydes, which includes free saturated and unsaturated aldehydes, and aldehydes that remain esterified to the phosphoglyceride backbone - the so-called 'core' aldehydes. However, little is known in regarding the vascular metabolism of these aldehydes. To identify biochemical pathways that metabolize free aldehydes, we examined the metabolism of 4-hydroxy-trans-2-nonenal in human aortic endothelial cells. Incubation of these cells with [3H]-HNE led to the generation of four main metabolites, i.e. glutathionyl HNE (GS-HNE), glutathionyl dihydroxynonene (GS-DHN), DHN and 4-hydroxynonanoic acid (HNA), which accounted for 5, 50, 6, and 23% of the total HNE metabolized. The conversion of GS-HNE to GS-DHN was inhibited by tolrestat, indicating that it is catalyzed by aldose reductase (AR). The AR was also found to be an efficient catalyst for the reduction of the core aldehyde - 1-palmitoyl-2- (5-oxovaleroyl)-sn-glycero-3-phosphorylcholine, which is generated in minimally modified low-density lipoprotein, and activates the endothelium to bind monocytes. As determined by electrospray mass spectrometry, reduction of POVPC (m/z=594) by AR led to the formation of 1-palmitoyl-2- (5)-hydrovaleryl-sn-glycero-3-phosphorylcholine (PHVPC; m/z=596). These observations suggest that due to its ability to catalyze the reduction of lipid-derived aldehydes AR may be involved in preventing inflammation and diminishing oxidative stress during the early phases of atherogenesis.


Subject(s)
Aldehyde Reductase/metabolism , Aldehydes/metabolism , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Cells, Cultured , Endothelium, Vascular/metabolism , Glutathione/metabolism , Humans , Lipoproteins, LDL/metabolism , Oxidation-Reduction , Phospholipid Ethers/metabolism
15.
Toxicology ; 138(3): 137-54, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10593505

ABSTRACT

Allylamine (AA; 3-aminopropene) and beta-aminopropionitrile (betaAPN) combined treatment (AA + betaAPN) results in myocardial protection from AA-induced subendocardial necrosis and a rapid and extensive aortic medial smooth muscle injury in rats. To determine the mechanisms of AA + betaAPN-induced vascular toxicity, cardiovascular parameters were monitored during a 10-day exposure by gavage in male Sprague-Dawley rats (180-200 g). Water intake and urine output were measured in rats treated with water, AA (100 mg kg(-1) body weight), betaAPN (1 g kg(-1) body weight), and AA + betaAPN for 10 days in metabolic cages. Plasma and urine samples were analyzed for blood urea nitrogen, CO2, creatinine, hematocrit, electrolytes (Na+, K+, Cl-), and osmolality. Heart and plasma semicarbazide-sensitive amine oxidase metabolic capacity (SSAO)was also measured following 1, 3 and 10 days of treatment. Following 10 day exposure to control or AA + betaAPN treatment, thoracic aortic rings (approximately 3 mm) were removed, and aortic reactivity to contractile and relaxant agonists was tested in vitro. In addition, cultured rat aorta vascular smooth muscle cells or rat heart beating myocytes were exposed to various concentrations of AA and betaAPN or AA metabolites and betaAPN to test for synergism in vitro. Several of the changes in in vivo cardiovascular parameters were shared, both in direction and magnitude, between the AA + betaAPN and the AA alone or the betaAPN alone treatments. This suggests that these effects (e.g. increased water intake and urine flow, decreased hematocrit, decreased heart and plasma SSAO metabolic capacity) were dependent on an AA alone or a betaAPN alone effect and were not AA + betaAPN specific effects. Significant inhibition of plasma and heart SSAO metabolic capacity occurred in the betaAPN alone and the AA + betaAPN treatments, but not in the AA alone treatment. Aortic rings from AA + betaAPN treated rats were contracted significantly less than anatomically-matched control rat aortic rings by 100 mM potassium chloride or by 10 microM norepinephrine. BetaAPN offered substantial protection against AA cytotoxicity in cultured vascular smooth muscle cells and beating myocytes, but did not alter the cytotoxicity of AA metabolites (i.e. acrolein, H2O2, or ammonia) in vascular smooth muscle cells as determined by the MTT viability assay. Overall, these data suggest that myocardial protection from AA injury that occurs in the combined AA + betaAPN treatment is likely due to inhibition of plasma SSAO. This may result in an increase in the AA dose accumulation and metabolism in the aorta leading to the severe aortic medial injury.


Subject(s)
Allylamine/antagonists & inhibitors , Amine Oxidase (Copper-Containing)/blood , Aminopropionitrile/pharmacology , Cardiovascular Diseases/prevention & control , Myocardium/pathology , Allylamine/toxicity , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Animals , Aorta/cytology , Aorta/drug effects , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/physiopathology , Cell Survival/drug effects , Cells, Cultured , Drinking/drug effects , Enzyme Inhibitors/pharmacology , Hemodynamics/drug effects , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Rats , Rats, Sprague-Dawley
16.
Gen Comp Endocrinol ; 114(1): 36-46, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10094857

ABSTRACT

Arginine vasotocin (AVT) is present in the neurohypophysis of all nonmammalian vertebrates and it appears to be the antecedent of the neurohypophysial nonapeptide hormones. Relatively little is known about AVT receptors in lower vertebrates, especially fish, and the present study was designed to examine AVT receptor interactions in trout vascular and nonvascular smooth muscle in vitro. AVT produced dose-dependent contraction of isolated rings from celiacomesenteric, coronary, and efferent branchial arteries, ventral aorta, anterior cardinal vein, and strips of ductus Cuvier. The greatest efficacy (magnitude of contraction per unit tissue weight) and sensitivity (effective concentration for half-maximal response, EC50) to AVT was found in the efferent bronchial artery (EBA) and its receptors were characterized further. Other neurohypophysial peptides, including arginine vasopressin (AVP), lysine vasopressin (LVP), isotocin (IST), and oxytocin (OXY), contracted EBA with an efficacy order of (most to least) AVT = AVP = OXY > LVP > IST and a sensitivity order of AVT > OXY >/= AVP > IST > LVP. Neither Desmopressin, an AVP V2-receptor agonist, nor the AVP ring fragment, AVP4-9, contracted EBA nor did they inhibit AVT contraction. Pretreatment of EBA rings with the selective AVP V1-receptor antagonists (deamino-Pen1, O-Me-Tyr2, Arg8-vasopressin and deamino-Pen1, Val4, Arg8-vasopressin), the selective V2-receptor antagonist (adamantaneacetyl1, O-Et-D-Tyr0, Val4, aminobutyryl6, Arg8,9-vasopressin), or the combined V1-oxytocin receptor antagonist (d(CH2)5[Tyr(Me)2, Orn8-AVT]) competitively inhibited AVT contractions without affecting AVT efficacy. Receptor affinity constants (pA2) determined by Schild analysis were in the range of 6.8-7.3, with slightly higher constants for the AVP V1-/oxytocin receptor antagonists than for the selective V2-receptor antagonist. Endothelium removal had no effect on EBA sensitivity to AVT. EBA rings were an order of magnitude more sensitive to AVT than nonvascular gastrointestinal and urinary bladder smooth muscle rings or strips. However, AVT (10(-7) M) was as efficacious as acetylcholine (10(-5) M) in gastrointestinal, gallbladder, and urinary bladder smooth muscle. It is concluded that trout EBA possess an AVT smooth muscle receptor that shares a similar pharmacological profile with the mammalian vascular AVP V1a-receptor and the OXY-receptor, but it is distinct from the previously reported gill epithelial cell receptor.


Subject(s)
Glycoproteins/chemistry , Muscle, Smooth, Vascular/chemistry , Oncorhynchus mykiss/metabolism , Pituitary Hormones/chemistry , Vasotocin/chemistry , Acetylcholine/pharmacology , Animals , Colforsin/pharmacology , Deamino Arginine Vasopressin/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Glycoproteins/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Pituitary Hormones/metabolism , Vasodilation/drug effects , Vasotocin/drug effects , Vasotocin/metabolism
17.
Toxicol Appl Pharmacol ; 148(2): 245-51, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9473532

ABSTRACT

Allylamine (AA, 3-aminopropene) is a specific cardiovascular toxin used experimentally to model myocardial necrosis and atherosclerosis. In these physiologic experiments, 10-day AA exposure (100 mg . kg-1 . day-1 by gavage) produced severe myocardial necrosis and increased heart rate but did not affect systolic blood pressure in rats. Mid-thoracic aortic ring segments were removed, and reactivity to contractile and relaxant agonists was tested. Aortic rings (approximately 3 mm) from AA-treated rats were contracted significantly more by high potassium (100 mM) and slightly more by norepinephrine (NE, 10 microM) than anatomically matched control aortic rings. No difference in aortic ring NE sensitivity or percentage relaxation in response to acetylcholine (1 microM) or sodium nitroprusside (100 microM) was detected between control and AA-treated rat aortic rings. Allylamine (1 microM-1 mM) induced modest, concentration-dependent contractions and tension oscillations in aortic rings from both control and AA-treated rats. Aortic rings from AA-treated rats, however, were more sensitive to AA. Vascular smooth muscle cells derived from control and AA-treated rat aortas had similar toxic sensitivity to AA in vitro using the MTT viability assay. The mechanisms by which AA exposure increased heart rate in vivo and contractility of aortic rings are unknown. These experiments support the previously proposed concept that AA-induced acute myocardial necrosis is due to coronary vasospasm and myocardial ischemia and cell injury.


Subject(s)
Allylamine/toxicity , Blood Pressure/drug effects , Heart Rate/drug effects , Muscle, Smooth, Vascular/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Male , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/physiology , Myocardium/pathology , Nitroprusside/pharmacology , Norepinephrine/pharmacology , Potassium/pharmacology , Rats , Vascular Resistance/drug effects
18.
Toxicol Sci ; 46(2): 386-92, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10048142

ABSTRACT

Semicarbazide-sensitive amine oxidase (SSAO) plays a role in the in vivo and in vitro toxicity of several environmental and endogenous amines. We investigated the role of SSAO as a component of cell culture medium (through addition of fetal calf serum (FCS)) compared to intracellular SSAO in the in vitro cytotoxicity of three amines and metabolites. Smooth muscle cells and beating cardiac myocytes were grown in 96-well plates and exposed to various concentrations and combinations of FCS in medium, amines (allylamine, AA; benzylamine, BZA; and methylamine, MA), and amine metabolites (aldehydes: acrolein, benzaldehyde, and formaldehyde; hydrogen peroxide, H2O2; ammonia, NH3). Amine and amine metabolite cytotoxicity was quantified by monitoring cell viability. SSAO activity was measured in FCS, cardiovascular cells, or rat plasma by a radioenzymatic assay using [14C]BZA. Our data show that AA and its aldehyde metabolite, acrolein, were the most toxic compounds to both cell types. However, AA toxicity was FCS-dependent in both cell types, while BZA, MA, and amine metabolite (i.e., aldehydes, H2O2, and NH3) cytotoxicity showed little FCS dependence. In these experiments, medium containing 10% FCS had a calculated amine metabolic capacity that was 30- to 50-fold that of the cultured smooth muscle cellular content in a single well of a 96-well plate. Our study demonstrates that SSAO in FCS contributes to amine metabolism and cytotoxicity to rat cardiovascular cells in vitro and how critical it is to evaluate serum for its role in mechanisms of amine toxicity in vitro and in vivo.


Subject(s)
Amine Oxidase (Copper-Containing)/physiology , Amines/metabolism , Amines/toxicity , Cell Survival/drug effects , Muscle, Smooth, Vascular/metabolism , Acrolein/toxicity , Allylamine/toxicity , Amine Oxidase (Copper-Containing)/blood , Animals , Benzylamines/toxicity , Cattle , Cells, Cultured , Drug Interactions , Fetus/chemistry , Male , Methylamines/toxicity , Rats
19.
Am J Physiol ; 273(2 Pt 2): R527-39, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9277535

ABSTRACT

Active venous regulation of cardiovascular function is well known in mammals but has not been demonstrated in fish. In the present studies, the natriuretic peptides (NP) rat atrial natriuretic peptide (ANP) and trout ventricular natriuretic peptide (VNP), clearance receptor inhibitor SC-46542, and sodium nitroprusside (SNP) were infused into unanesthetized trout fitted with pressure cannulas in the ventral aorta, dorsal aorta, and ductus Cuvier, and a ventral aorta (VA) flow probe was used to measure cardiac output (CO). In another group, in vivo vascular (venous) capacitance curves were obtained during ANP or SNP infusion. The in vitro effects of NP on vessels and the heart were also examined. ANP, VNP, and SC-46542 decreased central venous pressure (PVen), CO, stroke volume (SV), and gill resistance (RG), whereas systemic resistance (RS) and heart rate (HR) increased. Dorsal aortic pressure (PDA) transiently increased and then fell even though RS remained elevated. ANP decreased mean circulatory filling pressure (MCFP), increased vascular compliance at all blood volumes, and increased unstressed volume in hypovolemic fish. ANP had no direct effect on the heart. ANP responses in vivo were not altered in trout made hypotensive by prior treatment with the angiotensin-converting enzyme inhibitor lisinopril. SNP reduced ventral aortic pressure (PVA), PDA, and RS, increased CO and HR, but did not affect PVen, SV, or RG. SNP slightly decreased MCFP but did not affect compliance or unstressed volume. In vitro, large systemic arteries were more responsive than veins to NP, whereas SNP relaxed both. These results show that, in vivo, NP decrease venous compliance, thereby decreasing venous return, CO, and arterial pressure. Conversely, SNP hypotension is due to decreased RS. This is the first evidence for active regulation of venous capacitance in fish, which probably occurs in small veins or venules. The presence of venous baroreceptors is also suggested.


Subject(s)
Antihypertensive Agents/pharmacology , Atrial Natriuretic Factor/pharmacology , Nitroprusside/pharmacology , Oncorhynchus mykiss/physiology , Veins/drug effects , Veins/physiology , Animals , Cardiac Output/drug effects , Cardiovascular System/drug effects , Female , Heart Rate/drug effects , In Vitro Techniques , Male , Rats , Vascular Capacitance/drug effects
20.
Am J Physiol ; 272(4 Pt 2): R1112-20, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9140009

ABSTRACT

Bradykinins have only recently been identified in fish, and a detailed analysis of their cardiovascular actions is lacking. The present study examines the cardiovascular effects of trout bradykinin ([Arg0,Trp5,Leu8]bradykinin; tBK) in conscious trout, Oncorhynchus mykiss. tBK (1-10 nmol/kg body wt bolus) produced triphasic pressor-depressor-pressor responses. In phase 1, cardiac output (CO), ventral aortic (P(VA)), dorsal aortic (P(DA)), and central venous pressure increased, whereas systemic (R(S)) and gill resistance (R(G)) were unchanged. In phase 2, R(G) increased, whereas R(S), CO, and heart rate decreased, reducing P(VA) and P(DA). Plasma prostaglandin E2 and the prostacyclin metabolite, 6-ketoprostaglandin F1alpha, were significantly elevated during phase 2, whereas leukotrienes C4 and B4 and thromboxane B2 were unaffected. Phase 3 was produced by an increased CO and R(S) and the return of R(G) to control. Phase 1 pressor response was not blocked by inhibitors of cyclooxygenase, angiotensin-converting enzyme (ACE) or alpha-adrenoceptors (alpha-AD), whereas phase 2 depressor and plasma prostaglandin responses were prevented by cyclooxygenase inhibition. Phase 3 was partially blocked by ACE and alpha-AD inhibitors and is a response to the preceding hypotension. In vitro, tBK only decreased vascular resistance in the perfused splanchnic or skeletal muscle-kidney preparations. These results show that although tBK has multiple effects on the trout cardiovascular system, none of the effects are due to direct tBK stimulation of vascular smooth muscle. Phase 2 vasodilation has features consistent with release of vasodilator prostaglandins while the mechanism of phase 1 constriction is unknown.


Subject(s)
Arachidonic Acids/blood , Blood Pressure/drug effects , Bradykinin/pharmacology , Hemodynamics/drug effects , 6-Ketoprostaglandin F1 alpha/blood , Activity Cycles , Animals , Bradykinin/administration & dosage , Cardiac Output/drug effects , Gills/blood supply , Heart Rate/drug effects , Indomethacin/pharmacology , Injections, Intra-Arterial , Leukotrienes/blood , Oncorhynchus mykiss , Stroke Volume/drug effects , Thromboxane B2/blood , Time Factors , Vascular Resistance/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...