Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 11(7): e0159130, 2016.
Article in English | MEDLINE | ID: mdl-27415822

ABSTRACT

There is emerging evidence identifying microRNAs (miRNAs) as mediators of statin-induced cholesterol efflux, notably through the ATP-binding cassette transporter A1 (ABCA1) in macrophages. The objective of this study was to assess the impact of an HMG-CoA reductase inhibitor, pitavastatin, on macrophage miRNAs in the presence and absence of oxidized-LDL, a hallmark of a pro-atherogenic milieu. Treatment of human THP-1 cells with pitavastatin prevented the oxLDL-mediated suppression of miR-33a, -33b and -758 mRNA in these cells, an effect which was not uniquely attributable to induction of SREBP2. Induction of ABCA1 mRNA and protein by oxLDL was inhibited (30%) by pitavastatin, while oxLDL or pitavastatin alone significantly induced and repressed ABCA1 expression, respectively. These findings are consistent with previous reports in macrophages. miRNA profiling was also performed using a miRNA array. We identified specific miRNAs which were up-regulated (122) and down-regulated (107) in THP-1 cells treated with oxLDL plus pitavastatin versus oxLDL alone, indicating distinct regulatory networks in these cells. Moreover, several of the differentially expressed miRNAs identified are functionally associated with cholesterol trafficking (six miRNAs in cells treated with oxLDL versus oxLDL plus pitavastatin). Our findings indicate that pitavastatin can differentially modulate miRNA in the presence of oxLDL; and, our results provide evidence that the net effect on cholesterol homeostasis is mediated by a network of miRNAs.


Subject(s)
Cholesterol/metabolism , Macrophages/drug effects , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Quinolines/pharmacology , ATP Binding Cassette Transporter 1/genetics , Biological Transport, Active/drug effects , Cell Line , Gene Expression Regulation/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipoproteins, LDL/metabolism , Sterol Regulatory Element Binding Protein 2/genetics
2.
Am J Physiol Heart Circ Physiol ; 305(5): H687-98, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23792683

ABSTRACT

Protein 3-nitrotyrosine (3-NT) formation is frequently regarded as a simple biomarker of disease, an irreversible posttranslational modification that can disrupt protein structure and function. Nevertheless, evidence that protein 3-NT modifications may be site selective and reversible, thus allowing for physiological regulation of protein activity, has begun to emerge. We have previously reported that cyclooxygenase (COX)-1 undergoes heme-dependent nitration of Tyr(385), an internal and catalytically essential residue. In the present study, we demonstrate that nitrated COX-1 undergoes a rapid reversal of nitration by substrate-selective and biologically regulated denitrase activity. Using nitrated COX-1 as a substrate, denitrase activity was validated and quantified by analytic HPLC with electrochemical detection and determined to be constitutively active in murine and human endothelial cells, macrophages, and a variety of tissue samples. Smooth muscle cells, however, contained little denitrase activity. Further characterizing this denitrase activity, we found that it was inhibited by free 3-NT and may be enhanced by endogenous nitric oxide and exogenously administered carbon monoxide. Finally, we describe a purification protocol that results in significant enrichment of a discrete denitrase-containing fraction, which maintains activity throughout the purification process. These findings reveal that nitrated COX-1 is a substrate for a denitrase in cells and tissues, implying that the reciprocal processes of nitration and denitration may modulate bioactive lipid synthesis in the setting of inflammation. In addition, our data reveal that denitration is a controlled process that may have broad importance for regulating cell signaling events in nitric oxide-generating systems during oxidative/nitrosative stress.


Subject(s)
Cyclooxygenase 1/metabolism , Endothelium, Vascular/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Nitrates/metabolism , Oxidoreductases/metabolism , Adaptation, Physiological/physiology , Animals , Cell Line , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Macrophages/cytology , Mice , Mice, Inbred C57BL , Models, Animal , Muscle, Smooth, Vascular/cytology , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Oxidative Stress/physiology , Rats , Tyrosine/analogs & derivatives , Tyrosine/metabolism
3.
Antioxid Redox Signal ; 18(5): 471-80, 2013 Feb 10.
Article in English | MEDLINE | ID: mdl-22867102

ABSTRACT

AIMS: Hydrogen peroxide (H(2)O(2)), a nonradical oxidant, is employed to ascertain the role of redox mechanisms in regulation of vascular tone. Where both dilation and constriction have been reported, we examined the hypothesis that the ability of H(2)O(2) to effect vasoconstriction or dilation is conditioned by redox mechanisms and may be modulated by antioxidants. RESULTS: Exogenous H(2)O(2) (0.1-10.0 µM), dose-dependently reduced the internal diameter of rat renal interlobular and 3rd-order mesenteric arteries (p<0.05). This response was obliterated in arteries pretreated with antioxidants, including tempol, pegylated superoxide dismutase (PEG-SOD), butylated hydroxytoluene (BHT), and biliverdin (BV). However, as opposed to tempol or PEG-SOD, BHT & BV, antioxidants targeting radicals downstream of H(2)O(2), also uncovered vasodilation. INNOVATIONS: Redox-dependent vasoconstriction to H(2)O(2) was blocked by inhibitors of cyclooxygenase (COX) (indomethacin-10 µM), thromboxane (TP) synthase (CGS13080-10 µM), and TP receptor antagonist (SQ29548-1 µM). However, H(2)O(2) did not increase vascular thromboxane B(2) release; instead, it sensitized the vasculature to a TP agonist, U46619, an effect reversed by PEG-SOD. Antioxidant-conditioned dilatory response to H(2)O(2) was accompanied by enhanced vascular heme oxygenase (HO)-dependent carbon monoxide generation and was abolished by HO inhibitors or by HO-1 & 2 antisense oligodeoxynucleotides treatment of SD rats. CONCLUSION: These results demonstrate that H(2)O(2) has antioxidant-modifiable pleiotropic vascular effects, where constriction and dilation are brought about in the same vascular segment. H(2)O(2)-induced oxidative stress increases vascular TP sensitivity and predisposes these arterial segments to constrictor prostanoids. Conversely, vasodilation is reliant upon HO-derived products whose synthesis is stimulated only in the presence of antioxidants targeting radicals downstream of H(2)O(2).


Subject(s)
Antioxidants/pharmacology , Blood Vessels/drug effects , Heme Oxygenase (Decyclizing)/metabolism , Hydrogen Peroxide/pharmacology , Receptors, Thromboxane/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Base Sequence , Blood Vessels/enzymology , Blood Vessels/metabolism , Bridged Bicyclo Compounds, Heterocyclic , DNA Primers , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Hydrazines/pharmacology , Imidazoles/pharmacology , Male , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Thromboxane/physiology
4.
Am J Physiol Heart Circ Physiol ; 301(2): H617-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21602468

ABSTRACT

Nitric oxide (NO) is an important vasoactive molecule produced by three NO synthase (NOS) enzymes: neuronal (nNOS), inducible (iNOS), and endothelial NOS (eNOS). While eNOS contributes to blood vessel dilation that protects against the development of hypertension, iNOS has been primarily implicated as a disease-promoting isoform during atherogenesis. Despite this, iNOS may play a physiological role via the modulation of cyclooxygenase and thromboregulatory eicosanoid production. Herein, we examined the role of iNOS in a murine model of thrombosis. Blood flow was measured in carotid arteries of male and female wild-type (WT) and iNOS-deficient mice following ferric chloride-induced thrombosis. Female WT mice were more resistant to thrombotic occlusion than male counterparts but became more susceptible upon iNOS deletion. In contrast, male mice (with and without iNOS deletion) were equally susceptible to thrombosis. Deletion of iNOS was not associated with a change in the balance of thromboxane A(2) (TxA(2)) or antithrombotic prostacyclin (PGI(2)). Compared with male counterparts, female WT mice exhibited increased urinary nitrite and nitrate levels and enhanced ex vivo induction of iNOS in hearts and aortas. Our findings suggest that iNOS-derived NO in female WT mice may attenuate the effects of vascular injury. Thus, although iNOS is detrimental during atherogenesis, physiological iNOS levels may contribute to providing protection against thrombotic occlusion, a phenomenon that may be enhanced in female mice.


Subject(s)
Carotid Arteries/enzymology , Carotid Artery Injuries/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Thrombosis/prevention & control , Analysis of Variance , Animals , Aorta/drug effects , Aorta/enzymology , Blood Coagulation , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Carotid Arteries/physiopathology , Carotid Artery Injuries/chemically induced , Carotid Artery Injuries/complications , Carotid Artery Injuries/physiopathology , Chlorides , Disease Models, Animal , Estradiol/urine , Female , Ferric Compounds , Interferon-gamma/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/urine , Nitric Oxide Synthase Type II/deficiency , Nitric Oxide Synthase Type II/genetics , Regional Blood Flow , Sex Factors , Thrombosis/enzymology , Thrombosis/etiology , Thrombosis/physiopathology , Time Factors
5.
Am J Physiol Heart Circ Physiol ; 299(3): H613-23, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20543082

ABSTRACT

Cyclooxygenase (COX)-2 and inducible nitric oxide (NO) synthase (iNOS) are responsive to a wide array of inflammatory stimuli, have been localized to vascular smooth muscle cells (SMCs), and are intimately linked to the progression of vascular disease, including atherosclerotic lesion formation. We and others have shown that the production and subsequent impact of COX products appear to be correlative with the status of NO synthesis. This study examined the impact of inflammation-driven NO production on COX-2 expression in SMCs. Concurrent stimulation of quiescent rat aortic SMCs with lipopolysaccharide (LPS) and interferon (IFN)-gamma increased COX-2, iNOS, and nitrite production. Pharmacological inhibition of NO synthase (N(G)-monomethyl-l-arginine) concentration- and time-dependently magnified LPS + IFN-gamma-mediated COX-2 mRNA and protein induction in a cGMP-independent manner. COX-2 induction was associated with activation of the ERK, p38, and JNK mitogen-activated protein kinase (MAPK) pathways. Interestingly, NO synthase inhibition enhanced ERK, p38, and to a lesser extent JNK phosphorylation but suppressed MAPK phosphatase (MKP)-1 induction in response to LPS + IFN-gamma. Similarly, the exposure of SMCs from iNOS(-/-) mice to LPS + IFN-gamma produced an enhancement of COX-2 induction, p38, and JNK phosphorylation and an attenuated upregulation of MKP-1 versus their wild-type counterparts. Taken together, our data indicate that NO, in part derived from iNOS, negatively regulates the immediate early induction of COX-2 in response to inflammatory stimuli.


Subject(s)
Aorta/metabolism , Cyclooxygenase 2/metabolism , Inflammation/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Synthase Type II/genetics , Analysis of Variance , Animals , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Interferon-gamma/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
6.
J Am Chem Soc ; 132(11): 3914-22, 2010 Mar 24.
Article in English | MEDLINE | ID: mdl-20196542

ABSTRACT

Prostaglandin biosynthesis is catalyzed by two spatially and functionally distinct active sites in cyclooxygenase (COX) enzymes. Despite the crucial role of COXs in biology, molecular details regarding the function and regulation of these enzymes are incompletely defined. Reactive nitrogen species, formed during oxidative stress, produce modifications that alter COX functionalities and prostaglandin biosynthesis. We previously established that COX-1 undergoes selective nitration on Tyr385 via a mechanism that requires the presence of bound heme cofactor. As this is a critical residue for COX-1 catalysis, nitration at this site results in enzyme inactivation. We now show that occupancy of the COX-1 active site with substrate protects against Tyr385 nitration and redirects nitration to alternative Tyr residues on COX-1, preserving catalytic activity. This study reveals a novel role for the substrate in protecting COX-1 from inactivation by nitration in pathophysiological settings.


Subject(s)
Peroxynitrous Acid/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Stress, Physiological/drug effects , Amino Acid Sequence , Animals , Arachidonic Acid/metabolism , Catalytic Domain , Chromatography, High Pressure Liquid , Chromatography, Liquid , Cyclooxygenase 1/chemistry , Cyclooxygenase 1/metabolism , Cyclooxygenase Inhibitors/pharmacology , Electrochemistry , Enzyme Activation/drug effects , Heme/metabolism , Humans , Mice , Molecular Sequence Data , Prostaglandin-Endoperoxide Synthases/chemistry , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Rats , Substrate Specificity , Tandem Mass Spectrometry , Tyrosine
7.
Circ Res ; 105(8): 775-83, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19745167

ABSTRACT

RATIONALE: Vascular tissues produce carbon monoxide (CO) via HO-dependent and HO-independent mechanisms; the former in tandem with biliverdin and iron and the latter as a lone product. CO has been shown to function as both a vasoconstrictor and vasodilator; however, factors that dictate the vasoregulatory phenotype of this gas are unknown. OBJECTIVE: We investigated whether CO-mediated vasoconstriction is mechanistically linked to enhanced reactive oxygen species production that masks vasodilatory pathways. METHODS AND RESULTS: Sprague-Dawley rat interlobar and interlobular arteries were examined in terms of superoxide (O2*-) generation and vascular reactivity in the absence and presence of antioxidants. Both authentic CO and the CO-releasing molecule (CORM)-3 constricted renal arteries and increased O2*- production in a dose-dependent manner. The antioxidants tempol, ebselen, and deferoxamine inhibited CO-induced O2*- production and converted CO from constrictor to dilator. CO-induced O2*- generation was found to involve the activity of multiple oxidases including nitric oxide synthase, NADPH oxidase, xanthine oxidase, and complex IV of the mitochondrial electron chain. Furthermore, inhibition of these enzymes converted CO from constrictor to dilator. Similarly, biliverdin and bilirubin inhibited CO-induced O2*- production and vasoconstriction, allowing for a vasodilatory response to CO to be expressed. CO-induced vasoconstriction was dependent on a non-thromboxane agonist of the thromboxane receptor, whereas vasodilatory mechanisms of CO relied on the activation of soluble guanylate cyclase and calcium-gated potassium channels. CONCLUSIONS: CO-induced vasoconstriction involves the generation of reactive oxygen species, which, when negated, allows for the expression of vasodilatory pathways which are masked by the primary oxidative stress response to this gas.


Subject(s)
Antimetabolites/pharmacology , Arteries/metabolism , Carbon Monoxide/pharmacology , Kidney/metabolism , Oxidative Stress/drug effects , Vasoconstriction/drug effects , Animals , Antimetabolites/metabolism , Carbon Monoxide/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Guanylate Cyclase/metabolism , Male , Organometallic Compounds/pharmacology , Oxidoreductases/metabolism , Potassium Channels, Calcium-Activated , Rats , Rats, Sprague-Dawley , Superoxides/metabolism
8.
Eur J Pharmacol ; 606(1-3): 72-6, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19374880

ABSTRACT

Statins have been demonstrated to elicit a broad range of cellular events resulting in an attenuation of the inflammatory response and enhanced protection to the components of the vessel wall. The present study was designed to examine the effect of pitavastatin on pathways associated with the proinflammatory gene, early growth response (Egr)-1, in human vascular smooth muscle cells. Pretreatment with pitavastatin resulted in a dose-dependent reduction in Egr-1 protein and suppressed Egr-1 mRNA expression in response to phorbol 12-myristate 13-acetate (PMA). A reduction in Egr-1 expression reduced the activation of NGFI-A binding protein (NAB)-2, an Egr-1-dependent gene. Furthermore, these events appeared to be dependent on the ability of pitavastatin to attenuate signaling cascades associated with extracellular regulated kinase (ERK) 1/2, but not p38 and c-Jun N-terminal kinase (JNK).


Subject(s)
Early Growth Response Protein 1/genetics , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Quinolines/pharmacology , Transcriptional Activation/drug effects , Animals , Early Growth Response Protein 1/deficiency , Early Growth Response Protein 1/metabolism , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism
9.
Curr Hypertens Rev ; 5(4): 273-282, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-20368751

ABSTRACT

The endothelium generates powerful mediators that regulate blood flow, temper inflammation and maintain a homeostatic environment to prevent both the initiation and progression of vascular disease. Nitric oxide (NO) is arguably the single most influential molecule in terms of dictating blood vessel homeostasis. In addition to direct effects associated with altered NO production (e.g. vasoconstriction, excessive inflammation, endothelial dysfunction), NO is a critical modulator of vaso-relevant pathways including cyclooxygenase (COX)-derived prostaglandin production and angiotensin II generation by the renin-angiotensin system. Furthermore, NO may influence the selectivity of COX-2 inhibitors and ultimately contribute to controversies associated with the use of these drugs. Consistent with a central role for NO in vascular disease, disruptions in the production and bioavailability of NO have been linked to hypertension, diabetes, hypercholesterolemia, obesity, aging, and smoking. The ability of the vessel wall to control disease-associated oxidative stress may be the most critical determinant in maintaining homeostatic levels of NO and subsequently the prospect of stroke, myocardial infarction and other CV abnormalities. To this end, investigation of mechanisms that alter the balance of protective mediators, including pathways that are indirectly modified by NO, is critical to the development of effective therapy in the treatment of CV disease.

10.
Am J Pathol ; 173(5): 1253-64, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18948435

ABSTRACT

Despite the multifactorial nature of atherosclerosis, substantial evidence has established inflammation as an often surreptitious, yet critical and unifying driving force which promotes disease progression. To this end, research has defined molecular networks initiated by cytokines, growth factors and other pro-inflammatory molecules which promote hallmarks of atherosclerosis such as endothelial dysfunction, macrophage infiltration, LDL oxidation, cell proliferation and thrombosis. Although commonly associated with risk factors such as dyslipidemia, diabetes and hypertension, the global etiology of atherosclerosis may be alternatively attributed to underlying anthropological pressures. The agricultural, industrial and technological revolutions produced alterations in dietary, social and economic factors which have collectively exaggerated the exposure of the human genome to environmental stimuli. Furthermore, advances in sanitation, nutrition, and medicine have increased the lifespan of humans, effectively prolonging blood vessel exposure to these factors. As a result, the vasculature has become conditioned to respond to injury with what is arguably an overzealous immunological response; thus setting the stage for the prevalence of cardiovascular disease, including atherosclerotic plaque development in Western populations. Evidence suggests that each of these alterations can be linked to specific mediators in the inflammatory process. Integration of these factors with an inflammation-based hypothesis of atherosclerosis has yet to be extrapolated to observations in the realms of basic and clinical sciences and is the focus of this review.


Subject(s)
Anthropology , Atherosclerosis/pathology , Inflammation/pathology , Atherosclerosis/genetics , Atherosclerosis/therapy , Diet , Genomics , Humans , Inflammation/genetics , Motor Activity
12.
Free Radic Biol Med ; 40(12): 2198-205, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16785033

ABSTRACT

Hyperglycemia has been linked to increased oxidative stress, a resultant endothelial cell dysfunction, and, ultimately, apoptosis. Heme oxygenases (HO-1/HO-2) and the products of their activity, biliverdin/bilirubin and carbon monoxide (CO), play a physiological role in the vascular system. The effects of heme-mediated HO-1 induction, CO, and biliverdin on urinary 8-epi-isoprostane PGF(2alpha) and endothelial cell sloughing were examined in an animal model of streptozotocin (STZ)-induced diabetes. Hyperglycemia itself did not affect HO-1 and HO-2 protein levels, but caused a net decrease in HO activity. Weekly heme administration induced HO-1 protein, as demonstrated by immunohistochemistry and Western blot analyses. Administration of biliverdin or the CO donor, CORM-3, decreased urinary 8-epi-isoprostane PGF(2alpha), P < 0.5 compared to diabetes. Hyperglycemia increased endothelial cell sloughing; 8.2 +/- 0.8 cells/ml blood in control rats vs. 48 +/- 4.8 cells/ml blood in diabetic rats (P < 0.05). Heme administration significantly increased endothelial cell sloughing in diabetic rats (98 +/- 8.1 cells/ml blood, P < 0.0007) whereas biliverdin modestly decreased endothelial cell sloughing (26 +/- 3.5 cells/ml blood, P < 0.003). Administration of CORM-3 to diabetic rats resulted in a significant decrease in endothelial cell sloughing to 21.3 +/- 2.3 (P < 0.001). Administration of SnMP to CORM-3 diabetic rats only partially reversed the protective effects of CORM-3 on endothelial cell sloughing from 21.3 +/- 2.3 to 29 +/- 2.1 cells/ml, thus confirming a direct protective of CO, in addition to the ability of CORM-3 to induce HO-1 protein. These results demonstrate that exogenously administered CO or bilirubin can prevent endothelial cell sloughing in diabetic rats, likely via a decrease in oxidative stress, and thus represents a novel approach to prophylactic vascular protection in diabetes.


Subject(s)
Biliverdine/pharmacology , Carbon Monoxide/pharmacology , Diabetes Mellitus, Type 1/pathology , Diabetic Angiopathies/prevention & control , Endothelium, Vascular/drug effects , Protective Agents/pharmacology , Animals , Cell Aggregation/drug effects , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/enzymology , Diabetic Angiopathies/enzymology , Diabetic Angiopathies/pathology , Dinoprost/analogs & derivatives , Dinoprost/urine , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/enzymology , Endothelium, Vascular/pathology , Heme/pharmacology , Heme Oxygenase (Decyclizing)/analysis , Heme Oxygenase-1/analysis , Organometallic Compounds/pharmacology , Rats
13.
Hypertension ; 43(2): 347-51, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14698998

ABSTRACT

We tested the hypothesis that the status of NO synthesis influences the renal heme-heme oxygenase system. Studies were conducted in untreated rats and rats treated with the NO synthesis inhibitor N(G)-nitro-L-arginine methyl ester for 2 days. Treated and untreated rats were contrasted in terms of renal expression of heme oxygenase-1 and -2, renal carbon monoxide (CO)-generating activity, and urinary CO concentration and excretion rate. Heme oxygenase-1 and -2 proteins were similarly expressed in the kidneys of untreated and treated rats. In contrast, the NADPH-dependent component of the CO-generating activity of renal homogenates incubated with heme (a measure of heme oxygenase activity) was higher (P<0.05) in kidneys from rats treated with the NO synthesis inhibitor relative to corresponding data in untreated rats (1015+/-95 versus 379+/-111 pmol CO/mg per hour). Similarly, relative to corresponding data in untreated rats, rats treated with the NO synthesis inhibitor displayed increased (P<0.05) urinary CO concentration (920+/-174 versus 2286+/-472 pmol/mL) and urinary CO excretion (4.7+/-0.4 versus 14.3+/-2.7 pmol/min). This study demonstrates that NO synthesis inhibition upregulates the urinary concentration and excretion rate of CO, and the HO-dependent generation of CO by renal homogenates, without affecting the expression of renal heme oxygenase isoforms. Our findings imply that endogenous NO is an inhibitory regulator of renal CO generation by HO.


Subject(s)
Carbon Monoxide/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Kidney/enzymology , Nitric Oxide Synthase/antagonists & inhibitors , Animals , Carbon Monoxide/urine , Enzyme Inhibitors/pharmacology , Heme Oxygenase-1 , Kidney/drug effects , Kidney/metabolism , Kinetics , Male , NG-Nitroarginine Methyl Ester/pharmacology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...