Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Toxicol Appl Pharmacol ; 400: 115070, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32464219

ABSTRACT

Vascular dysfunction plays a critical role in the pathogenesis of sepsis. We elucidated the mechanisms underlying the amelioration of lipopolysaccharide (LPS)-induced vascular inflammation by oroxylin A (OroA) post-treatment in rats. The animals were intraperitoneally injected with LPS (10 mg/kg) to induce systemic inflammation and intravenously (iv) administered OroA (15 mg/kg) 6 h after the LPS treatment. The assessments included biochemical changes in peripheral blood, vascular reactivity which was evaluated by blood-vessel myography, morphological/histological assessment of inflammation, toll-like receptor (TLR)-4-mediated interleukin-1-receptor-associated-kinase (IRAK)-4 activation, changes in adhesion molecule expression, and endothelial junctional stability in the aorta. LPS significantly enhanced the proinflammatory cytokine release, increased vascular cell adhesion molecule (VCAM)-1 expression, disrupted endothelial tight junction, reduced vascular endothelial barrier stability, and increased macrophage infiltration and accumulation in the aorta. All observed pathological changes and vascular inflammation were significantly reversed by the OroA post-treatment. Importantly, OroA suppressed the increased adhesion molecule expression and the endothelial barrier disruption by inhibiting LPS-activated IRAK-4-targeted inhibitory nuclear factor kappa B kinase (IKK) α/ß complex phosphorylation, without directly affecting the interaction between LPS and TLR-4. Moreover, the iNOS activity induced by the LPS challenge was inhibited by the OroA pretreatment of the isolated aortic rings. These results suggest that OroA regulates the vascular tone by inhibiting vascular hyporeactivity caused by NO overproduction and reverses the endothelial barrier dysfunction and inflammation by inhibiting the IRAK-4-mediated IKKα/ß phosphorylation. Overall, these findings suggest OroA administration as a potentially useful therapeutic approach for clinical interventions in septic shock.


Subject(s)
Aorta/drug effects , Endothelium, Vascular/drug effects , Flavonoids/pharmacology , Sepsis/prevention & control , Vascular Cell Adhesion Molecule-1/genetics , Animals , Aorta/immunology , Aorta/pathology , Cells, Cultured , Chemokine CCL2/blood , Cytokines/blood , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Endotoxins/pharmacology , Flavonoids/therapeutic use , Gene Expression/drug effects , Male , Neutrophil Infiltration/drug effects , Rats, Sprague-Dawley , Sepsis/blood , Sepsis/pathology
2.
Eur J Pharmacol ; 868: 172851, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31836535

ABSTRACT

Nicotinic acetylcholine receptor activation on the perivascular sympathetic nerves via axo-axonal interaction mechanism causes norepinephrine release, which triggers the neurogenic nitrergic relaxation in basilar arteries to meet the need of a brain. Donepezil and huperzine A, which are the cholinesterase inhibitors used for Alzheimer's disease therapy, exert controversial effects on nicotinic acetylcholine receptors. Therefore, we investigated how donepezil and huperzine A via the axo-axonal interaction regulate the neurogenic vasodilation of isolated porcine basilar arteries and define their action on different subtypes of the nicotinic acetylcholine receptor by using blood vessel myography, calcium imaging, and electrophysiological techniques. Both nicotine (100 µM) and transmural nerve stimulation (TNS, 8 Hz) induce NO-mediated dilation in the arteries. Nicotine-induced vasodilations were concentration-dependently inhibited by huperzine A and donepezil, with the former being 30 fold less potent than the latter. Both cholinesterase inhibitors weakly and equally decreased TNS-elicited nitrergic vasodilations. Neither huperzine A nor donepezil affected isoproterenol (a ß adrenoceptor-agonist)- or sodium nitroprusside (a NO donor)-induced vasodilation. Further, huperzine A was less potent than donepezil in inhibiting nicotine-elicited calcium influxes in rodent superior cervical ganglionic neurons and inward currents in α7- and α3ß2-nicotinic acetylcholine receptor-expressing Xenopus oocytes. In conclusion, huperzine A may exert less harmful effect over donepezil on maintaining brainstem circulation and on the nicotinic acetylcholine receptor-associated cognition deficits during treatment for Alzheimer's disease.


Subject(s)
Basilar Artery/physiopathology , Cholinesterase Inhibitors/adverse effects , Cognitive Dysfunction/chemically induced , Nitrergic Neurons/drug effects , Vasodilation/drug effects , Alkaloids/administration & dosage , Alkaloids/adverse effects , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Basilar Artery/drug effects , Basilar Artery/innervation , Brain Stem/blood supply , Brain Stem/drug effects , Brain Stem/pathology , Brain Stem/physiopathology , Calcium/metabolism , Cholinesterase Inhibitors/administration & dosage , Cognitive Dysfunction/physiopathology , Donepezil/administration & dosage , Donepezil/adverse effects , Dose-Response Relationship, Drug , Humans , Models, Animal , Nicotine/metabolism , Nitrergic Neurons/metabolism , Nitrergic Neurons/physiology , Oocytes , Patch-Clamp Techniques , Rats , Receptors, Nicotinic/metabolism , Sesquiterpenes/administration & dosage , Sesquiterpenes/adverse effects , Swine , Synaptic Transmission/drug effects , Vasodilation/physiology , Xenopus laevis
3.
Vascul Pharmacol ; 87: 110-120, 2016 12.
Article in English | MEDLINE | ID: mdl-27614253

ABSTRACT

Nitric oxide (NO) overproduction via induction of inducible nitric oxide synthase (iNOS) is implicated in vasodilatory shock in sepsis, leading to septic encephalopathy and accelerating cerebral ischemic injury. An abbreviated urea-cycle (l-citrulline-l-arginine-NO cycle) has been demonstrated in cerebral perivascular nitrergic nerves and endothelial cells but not in normal cerebral vascular smooth muscle cell (CVSMC). This cycle indicates that argininosuccinate synthase (ASS) catalyzes l-citrulline (l-cit) conversion to form argininosuccinate (AS), and subsequent AS cleavage by argininosuccinate lyase (ASL) forms l-arginine (l-arg), the substrate for NO synthesis. The possibility that ASS enzyme in this cycle was induced in the CVSMC in sepsis was examined. Blood-vessel myography technique was used for measuring porcine isolated basilar arterial tone. NO in cultured CVSMC and in condition mediums were estimated by diaminofluorescein (DAF)-induced fluorescence and Griess reaction, respectively. Immunohistochemical and immunoblotting analyses were used to examine iNOS and ASS induction. l-cit and l-arg, which did not relax endothelium-denuded normal basilar arteries precontracted by U-46619, induced significant vasorelaxation with increased NO production in these arteries and the CVSMCs following 6-hour exposure to 20µg/ml lipopolysaccharide (LPS) or lipoteichoic acid (LTA). Pre-treatment with pyrrolidine dithiocarbamate (PDTC) and salicylate (SAL) (NFκB inhibitors), aminoguanidine (AG, an iNOS inhibitor), and nitro-l-arg (NLA, a non-specific NOS inhibitor) blocked NO synthesis in the CVSMC and attenuated l-cit- and l-arg-induced relaxation of LPS- and LTA-treated arteries. Furthermore, immunohistochemical and immunoblotting studies demonstrated that expression of basal iNOS and ASS in the smooth muscle cell of arterial segments denuded of endothelium and the cultured CVSMCs was significantly increased following 6-hour incubation with LPS or LTA. This increased iNOS- and ASS-proteins expression in both preparations was inhibited by SAL, but was further increased by AG. These results indicate that LPS and LTA induce the l-cit-l-arg-NO cycle via induction of iNOS and ASS in the CVSMCs, accounting for massively increased NO-production and cerebral vasodilation in septic shock. Simultaneous inhibition of both pathways and NFκB-activation may be necessary to efficiently decrease or normalize NO production in the CVSMCs in this disease condition, and/or prevention and treatment of cerebral vessel-related brain dysfunctions. Our results further suggest to avoid using iNOS inhibitors alone which may cause upregulation of iNOS and ASS resulted from feedback-inhibition of iNOS activity. Accordingly, combined treatments with specific iNOS-activity inhibitor and inhibitor for iNOS genomic expression may provide a strategy in optimally managing brain sepsis and related encephalopathy associated with enhanced iNOS expression and NO overproduction.


Subject(s)
Bacterial Toxins/pharmacology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Urea/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Arginine/metabolism , Argininosuccinate Synthase/metabolism , Cerebral Arteries/cytology , Cerebral Arteries/metabolism , Citrulline/metabolism , Female , Lipopolysaccharides/pharmacology , Male , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Synthase Type II/genetics , Swine , Teichoic Acids/pharmacology , Vasoconstrictor Agents/pharmacology
4.
Br J Pharmacol ; 173(7): 1179-95, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26694894

ABSTRACT

BACKGROUND AND PURPOSE: Effective management of hypotension refractory to vasoconstrictors in severe sepsis is limited. A new strategy to ameliorate endotoxemic hypotension by inducing endothelium-dependent constriction of large arteries was assessed. EXPERIMENTAL APPROACH: Endotoxemia in rats was induced by injection of LPS (10 mg·kg(-1), i.v.). Haemodynamics were measured in vivo, reactivity of isolated mesenteric arteries by myography and expression of proteins and enzyme activities by immunohistochemistry, biochemistry and molecular biology. KEY RESULTS: Six hours after LPS, the hypotension was promptly reversed following injection (i.v. or i.p.) of oroxylin-A (OroA) . In isolated LPS-treated but not normal mesenteric arteries, OroA (1-10 µM) induced endothelium-dependent, sustained constriction, blocked by endothelin-1 (ET-1) receptor antagonists. OroA further enhanced LPS-induced expression of endothelin-converting enzyme, ET-1 mRNA and proteins and ET-1 release, OroA also enhanced phosphorylation of Rho-associated protein kinase (ROCK) and reversed LPS-induced suppression of RhoA activities in smooth muscle of arteries with endothelium. Activated- phosphorylation of smooth muscle ROCK was blocked by ET-1-receptor antagonists and ROCK inhibitors. Moreover, OroA post-treatment suppressed, via inhibiting NF-κB activation, inducible NOS expression and circulating NO. CONCLUSIONS AND IMPLICATIONS: Reversal of endotoxemic hypotensive by OroA was due to release of endothelial ET-1, upregulated by LPS, from mesenteric arteries, inducing prompt and sustained vasoconstriction via activation of vascular smooth muscle RhoA/ROCK-pathway. In late endotoxemia, OroA-induced vasoconstriction was partly due to decreased circulating NO. Activation of endothelium-dependent constriction in large resistance arteries and suppression of systemic inflammation offer new strategies for acute management of endotoxemic hypotensive shock.


Subject(s)
Endothelin A Receptor Antagonists/therapeutic use , Endothelium, Vascular/drug effects , Endotoxemia/drug therapy , Flavonoids/therapeutic use , Hypotension/drug therapy , Mesenteric Arteries/drug effects , Shock, Septic/drug therapy , Animals , Endothelin-1/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Endotoxemia/physiopathology , Hypotension/physiopathology , Lipopolysaccharides/pharmacology , Male , Mesenteric Arteries/physiopathology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Rats, Sprague-Dawley , Shock, Septic/physiopathology , Vasoconstriction , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
5.
PLoS One ; 10(3): e0119843, 2015.
Article in English | MEDLINE | ID: mdl-25774877

ABSTRACT

Besides regulating calcium homeostasis, the effects of vitamin D on vascular tone and metabolic disturbances remain scarce in the literature despite an increase intake with high-fructose corn syrup worldwide. We investigated the effects of calcitriol, an active form of vitamin D, on vascular relaxation, glucose tolerance, and visceral fat pads in fructose-fed rats. Male Wistar-Kyoto rats were divided into 4 groups (n = 6 per group). Group Con: standard chow diet for 8 weeks; Group Fru: high-fructose diet (60% fructose) for 8 weeks; Group Fru-HVD: high-fructose diet as Group Fru, high-dose calcitriol treatment (20 ng / 100 g body weight per day) 4 weeks after the beginning of fructose feeding; and Group Fru-LVD: high-fructose diet as Group Fru, low-dose calcitriol treatment (10 ng / 100 g body weight per day) 4 weeks after the beginning of fructose feeding. Systolic blood pressure was measured twice a week by the tail-cuff method. Blood was examined for serum ionized calcium, phosphate, creatinine, glucose, triglycerides, and total cholesterol. Intra-peritoneal glucose intolerance test, aortic vascular reactivity, the weight of visceral fat pads, adipose size, and adipose angiotensin II levels were analyzed at the end of the study. The results showed that the fructose-fed rats significantly developed hypertension, impaired glucose tolerance, heavier weight and larger adipose size of visceral fat pads, and raised adipose angiotensin II expressions compared with the control rats. High- and low-dose calcitriol reduced modestly systolic blood pressure, increased endothelium-dependent aortic relaxation, ameliorated glucose intolerance, reduced the weight and adipose size of visceral fat pads, and lowered adipose angiotensin II expressions in the fructose-fed rats. However, high-dose calcitriol treatment mildly increased serum ionized calcium levels (1.44 ± 0.05 mmol/L). These results suggest a protective role of calcitriol treatment on endothelial function, glucose tolerance, and visceral adiposity in fructose-fed rats.


Subject(s)
Calcitriol/administration & dosage , Fructose/adverse effects , Glucose Intolerance/drug therapy , Hypertension/drug therapy , Obesity, Abdominal/drug therapy , Vitamins/administration & dosage , Adiposity/drug effects , Angiotensin II/metabolism , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Calcitriol/pharmacology , Dose-Response Relationship, Drug , Glucose Intolerance/chemically induced , Glucose Intolerance/pathology , Glucose Tolerance Test , Hypertension/chemically induced , Hypertension/pathology , Male , Obesity, Abdominal/chemically induced , Obesity, Abdominal/pathology , Rats , Rats, Inbred WKY , Vasodilation/drug effects , Vitamins/pharmacology
6.
Hypertens Res ; 36(2): 123-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22895064

ABSTRACT

Angiotensin-converting enzyme inhibitors and angiotensin II type 1 receptor blockers can improve insulin resistance and vascular dysfunction in insulin-resistant rats; however, there are few reports on the effects of direct renin inhibitors on these conditions. We investigated the effects of a direct renin inhibitor, aliskiren, on insulin resistance, aortic endothelial dysfunction and vascular remodeling in fructose-fed hypertensive rats. Male Wistar-Kyoto rats were divided into four groups (n=6 per group) and studied for 8 weeks: Group Con: standard chow diet; group Fru: high-fructose diet (60% fructose); Group FruA: high-fructose diet with concurrent aliskiren treatment (100 mg kg(-1) per day); and Group FruB: high-fructose diet with subsequent aliskiren treatment 4 weeks later. Blood was collected for biochemical assays, and isolated rings of the thoracic aorta were obtained for analysis of vascular reactivity, vascular structure and lipid peroxide. Rats fed with high-fructose diets developed significant systolic hypertension, decreased plasma nitrite (NO(2); nitric oxide metabolite) levels and increased plasma glucose, insulin, triglyceride, total cholesterol and aortic lipid peroxide levels, and aortic wall thickness compared with control rats. Aliskiren treatment, either concurrent or subsequent, elevated plasma NO(2) levels and reduced systolic hypertension, insulin resistance, dyslipidemia, aortic lipid peroxide levels and aortic wall hypertrophy in FHR. The peak endothelium-dependent aortic relaxations were significantly higher in rats that received aliskiren treatment than in those that did not. In conclusion, our findings suggest that aliskiren prevents and ameliorates insulin resistance, aortic endothelial dysfunction and oxidative vascular remodeling in fructose-fed hypertensive rats.


Subject(s)
Amides/pharmacology , Aorta, Thoracic/drug effects , Endothelium, Vascular/drug effects , Fructose/pharmacology , Fumarates/pharmacology , Hypertension/prevention & control , Insulin Resistance/physiology , Renin/antagonists & inhibitors , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Dietary Carbohydrates/pharmacology , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hypertension/blood , Hypertension/physiopathology , Lipid Peroxides/metabolism , Male , Nitrites/blood , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Inbred WKY , Vasodilation/drug effects , Vasodilation/physiology
7.
PLoS One ; 7(12): e50363, 2012.
Article in English | MEDLINE | ID: mdl-23236370

ABSTRACT

Microglia-mediated inflammation is implicated in pathogenesis of neurodegenerative diseases. Oroxylin A, a flavonoid isolated from Scutellariae baicalensis, has been shown to ameliorate microglia activation-mediated neurodegeneration in vivo. The molecular mechanism underlying the inhibitory effects of oroxylin A on microglia activation, however, remains unknown. In the present study, effects of oroxylin A co-treated with lipopolysaccharide (LPS, 100 ng/ml) on LPS-induced activation of cultured microglial BV-2 cells were examined. Nitric oxide (NO) production was determined by Greiss method. Expression of inducible nitric oxide synthase (iNOS), interleukin (IL)-1ß and IL-6 was assessed using real-time RT-PCR or Western blot analysis. Furthermore, activation of the nuclear factor κB (NFκB) and the signal transducer and activator of transcription 1 (STAT1) was examined by Western blot analysis and transcription factor DNA-binding activity assay. Our results indicated that oroxylin A (10-100 µM) in a concentration-dependent manner inhibited LPS-induced NO production via blocking iNOS expression at both mRNA and protein levels without affecting the degradation rate of iNOS mRNA. Moreover, oroxylin A significantly attenuated LPS-induced late expression (20 hours after LPS challenge) of IL-1ß and IL-6. Furthermore, oroxylin A significantly suppressed LPS-induced JAK2-mediated STAT1 phosphorylation without affecting LPS-induced NFκB-p65 nuclear translocation or NFκB-p65 DNA-binding activity. This is consistent with the finding that AG490, a specific JAK2 inhibitor, significantly inhibited LPS-induced STAT1 phosphorylation with almost completely diminished iNOS expression. These results suggest that oroxylin A, via suppressing STAT1 phosphorylation, inhibits LPS-induced expression of pro-inflammatory genes in BV-2 microglial cells.


Subject(s)
Flavonoids/pharmacology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Microglia/metabolism , Nitric Oxide Synthase Type II/metabolism , STAT1 Transcription Factor/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Interleukin-1beta/genetics , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Mice , Microglia/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Phosphorylation/drug effects , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects , Tyrphostins/pharmacology
8.
PLoS One ; 7(10): e47403, 2012.
Article in English | MEDLINE | ID: mdl-23071799

ABSTRACT

BACKGROUND AND PURPOSE: Successful drug treatment for sepsis-related acute lung injury (ALI) remains a major clinical problem. This study was designed to assess the beneficial effects of post-treatment of oroxylin A (OroA), a flavonoid, in ameliorating lipopolysaccharides (LPS)-induced lung inflammation and fatality. EXPERIMENTAL APPROACH: Rats were injected with LPS (10 mg/kg, iv) to induce ALI, and OroA was given (15 mg/kg, iv) 1 hr or 6 hrs after LPS challenge. Twenty four hrs after LPS challenge, biochemical changes in the blood and lung tissues, and morphological/histological alterations in the lung associated with inflammation and injury were examined. Therapeutic effect of OroA was assessed by measuring the survival rate in endotoxemic mice. KEY RESULTS: LPS (10 mg/kg, iv) significantly altered WBC counts, elevated plasma tumor necrosis factor (TNF)-α and nitric oxide (NO), increased pulmonary edema, thickened alveolar septa, and decreased survival rate. These changes were ameliorated by OroA (15 mg/kg, iv) administered 1 hr or 6 hrs after LPS challenge. This post-treatment also significantly attenuated LPS-induced activation of nuclear factor-κB (NF-κB) and the release of high mobility group box 1 (HMGB1) in lung tissues. Furthermore, post-treatment with OroA (60 mg/kg, ip) administered 1 hr or 6 hrs after LPS challenge in mice significantly increased survival rate. CONCLUSION AND IMPLICATION: OroA administered after induction of ALI by LPS significantly prevent and revere lung tissues injuries with increased survival rate. Positive post-treatment effects of OroA suggest that OroA is a potentially useful candidate for managing lung inflammation in LPS-induced endotoxemia and septic shock.


Subject(s)
Acute Lung Injury/drug therapy , Flavonoids/therapeutic use , Lipopolysaccharides/toxicity , NF-kappa B/metabolism , Signal Transduction/physiology , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Analysis of Variance , Animals , Fluorescent Antibody Technique , Immunoassay , Immunohistochemistry , Leukocyte Count , Male , Mice , Nitric Oxide/blood , Rats , Rats, Sprague-Dawley , Survival Analysis , Treatment Outcome , Tumor Necrosis Factor-alpha/blood
9.
Am J Physiol Heart Circ Physiol ; 302(5): H1123-30, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22210747

ABSTRACT

The close apposition between sympathetic and parasympathetic nerve terminals in the adventitia of cerebral arteries provides morphological evidence that sympathetic nerve activation causes parasympathetic nitrergic vasodilation via a sympathetic-parasympathetic interaction mechanism. The decreased parasympathetic nerve terminals in basilar arteries (BA) of spontaneously hypertensive rat (SHR) and renovascular hypertensive rats (RHR) compared with Wistar-Kyoto rats (WKY), therefore, would diminish this axo-axonal interaction-mediated neurogenic vasodilation in hypertension. Increased basilar arterial blood flow (BABF) via axo-axonal interaction during sympathetic activation was, therefore, examined in anesthetized rats by laser-Doppler flowmetry. Electrical stimulation (ES) of sympathetic nerves originating in superior cervical ganglion (SCG) and topical nicotine (10-30 µM) onto BA of WKY significantly increased BABF. Both increases were inhibited by tetrodotoxin, 7-nitroindazole (neuronal nitric oxide synthase inhibitor), and ICI-118,551 (ß(2)-adrenoceptor antagonist), but not by atenolol (ß(1)-adrenoceptor antagonist). Topical norepinephrine onto BA also increased BABF, which was abolished by atenolol combined with 7-nitroindazole or ICI-118,551. Similar results were found in prehypertensive SHR. However, in adult SHR and RHR, ES of sympathetic nerves or topical nicotine caused minimum or no increase of BABF. It is concluded that excitation of sympathetic nerves to BA in WKY causes parasympathetic nitrergic vasodilation with increased BABF. This finding indicates an endowed functional neurogenic mechanism for increasing the BABF or brain stem blood flow in coping with increased local sympathetic activities in acutely stressful situations such as the "fight-or-flight response." This increased blood flow in defensive mechanism diminishes in genetic and nongenetic hypertensive rats due most likely to decreased parasympathetic nitrergic nerve terminals.


Subject(s)
Basilar Artery/drug effects , Pons/blood supply , Pons/drug effects , Sympathetic Nervous System/drug effects , Animals , Atenolol/pharmacology , Blood Pressure/drug effects , Electric Stimulation , Enzyme Inhibitors/pharmacology , Ganglionic Stimulants/pharmacology , Indazoles/pharmacology , Laser-Doppler Flowmetry , Male , Nicotine/pharmacokinetics , Norepinephrine/pharmacology , Propanolamines/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/physiology , Sympathetic Nervous System/physiology , Tetrodotoxin/pharmacology
10.
Circulation ; 124(10): 1160-71, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21844078

ABSTRACT

BACKGROUND: Perivascular adipose tissue (PVAT)-derived relaxing factor (PVATRF) significantly regulates vascular tone. Its chemical nature remains unknown. We determined whether palmitic acid methyl ester (PAME) was the PVATRF and whether its release and/or vasorelaxing activity decreased in hypertension. METHODS AND RESULTS: Using superfusion bioassay cascade technique, tissue bath myography, and gas chromatography/mass spectrometry, we determined PVATRF and PAME release from aortic PVAT preparations of Wistar Kyoto rats and spontaneously hypertensive rats. The PVAT of Wistar Kyoto rats spontaneously and calcium dependently released PVATRF and PAME. Both induced aortic vasorelaxations, which were inhibited by 4-aminopyridine (2 mmol/L) and tetraethylammonium 5 and 10 mmol/L but were not affected by tetraethylammonium 1 or 3 mmol/L, glibenclamide (3 µmol/L), or iberiotoxin (100 nmol/L). Aortic vasorelaxations induced by PVATRF- and PAME-containing Krebs solutions were not affected after heating at 70°C but were equally attenuated after hexane extractions. Culture mediums of differentiated adipocytes, but not those of fibroblasts, contained significant PAME and caused aortic vasorelaxation. The PVAT of spontaneously hypertensive rats released significantly less PVATRF and PAME with an increased release of angiotensin II. In addition, PAME-induced relaxation of spontaneously hypertensive rats aortic smooth muscle diminished drastically, which was ameliorated significantly by losartan. CONCLUSIONS: We found that PAME is the PVATRF, causing vasorelaxation by opening voltage-dependent K+ channels on smooth muscle cells. Diminished PAME release and its vasorelaxing activity and increased release of angiotensin II in the PVAT suggest a noble role of PVAT in pathogenesis of hypertension. The antihypertensive effect of losartan is attributed partly to its reversing diminished PAME-induced vasorelaxation.


Subject(s)
Adipose Tissue/metabolism , Hypertension/metabolism , Palmitates/metabolism , Vasodilator Agents/metabolism , 4-Aminopyridine/pharmacology , Adipocytes/metabolism , Adipose Tissue/drug effects , Angiotensin II/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Calcium/metabolism , Cells, Cultured , Endothelium, Vascular/drug effects , Fibroblasts/metabolism , Glyburide/pharmacology , Hypertension/drug therapy , Losartan/pharmacology , Male , Peptides/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Tetraethylammonium/pharmacology , Vasodilation/drug effects
11.
Arch Med Sci ; 7(5): 882-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22291836

ABSTRACT

INTRODUCTION: The renin-angiotensin system plays a major role in the pathogenesis of metabolic syndrome. The objective of this study was to examine the effects of aliskiren, a direct renin inhibitor, on the metabolic syndrome of fructose-fed rats. MATERIAL AND METHODS: Male Sprague-Dawley rats were divided into 4 groups (n = 6 for each group). Group Con: rats were fed a standard chow diet for 8 weeks, group Fru: rats were fed a high fructose diet (60% fructose) for 8 weeks, group FruA: rats were fed a high fructose diet and were co-infused with aliskiren (100 mg/kg/day), and group FruB: rats were treated as group Fru, but aliskiren was administered 4 weeks later. Systolic blood pressure (SBP), homeostasis model assessment-insulin resistance (HOMA-IR), and blood profiles were measured. RESULTS: By the end of week 4 and 8 of a high fructose diet, SBP had increased significantly from 111 ±5 to 142 ±4 and 139 ±5 mmHg (p < 0.05), respectively. A high fructose diet significantly increased HOMA-IR from baseline (6.15 ±1.59) to 21.25 ±2.08 and 21.28 ±3.1 (p < 0.05) at week 4 and 8, respectively, and significantly induced metabolic syndrome. Concurrent aliskiren treatment prevented the development of hypertension and metabolic syndrome in fructose-fed rats. When fructose-induced hypertension was established, subsequent aliskiren treatment for 4 weeks reversed the elevated SBP and ameliorated metabolic syndrome. There were no significant differences in food, water intake, urine flow or body weight gain among groups. CONCLUSIONS: Aliskiren not only prevents but also ameliorates metabolic syndrome in fructose-fed rats.

12.
Invest Ophthalmol Vis Sci ; 51(9): 4746-53, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20357193

ABSTRACT

PURPOSE: To determine whether palmitic acid methyl ester (PAME) or methyl palmitate is the retina-derived relaxing factor (RRF). METHODS: A superfusion bioassay cascade technique was used with rat isolated retina as donor tissue and rat aortic ring as detector tissue. The superfusate was analyzed with gas chromatography/mass spectrometry (GC/MS). The biochemical and pharmacologic characteristics of RRF and PAME were compared. RESULTS: The authors demonstrated that the retina on superfusion with Krebs solution spontaneously released RRF (indicated by aortic ring relaxation) and PAME (measured by GC/MS). The release of RRF and PAME was calcium dependent because the release was abolished when the retinas were superfused with calcium-free Krebs solution. Furthermore, aortic relaxations induced by RRF and PAME were not affected after heating their solutions at 70 degrees C for 1 hour, suggesting that both are heat stable. Exogenous PAME concentration dependently induced aortic relaxation with EC50 of 0.82+/-0.75 pM. The aortic relaxations induced by RRF and exogenous PAME were inhibited by 4-aminopyridine (2 mM) and tetraethylammonium (TEA, 10 mM) but were not affected by TEA at 1 mM or 3 mM, glibenclamide (3 microM), or iberiotoxin (100 nM). The vasodilator activity of Krebs solution containing RRF or exogenous PAME was greatly attenuated after hexane extraction. CONCLUSIONS: RRF and PAME share similar biochemical properties and react similarly to all pharmacologic inhibitors examined. Both act primarily on the voltage-dependent K+ (Kv) channel of aortic smooth muscle cells, causing aortic relaxation. These results suggest that PAME is the hydrophobic RRF.


Subject(s)
Aorta/physiology , Palmitates/metabolism , Retina/metabolism , Retinal Vessels/physiology , Vasodilation/physiology , 4-Aminopyridine/pharmacology , Animals , Biological Assay , Calcium/metabolism , Calcium/pharmacology , Enzyme Inhibitors/pharmacology , Hexanes , Isotonic Solutions/metabolism , Isotonic Solutions/pharmacology , Male , Miconazole/pharmacology , Nitroarginine/pharmacology , Palmitates/isolation & purification , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Proadifen/pharmacology , Rats , Rats, Sprague-Dawley , Solvents , Tetraethylammonium/pharmacology , Vasodilation/drug effects
13.
Proc Natl Acad Sci U S A ; 105(49): 19526-31, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19057014

ABSTRACT

Nitric oxide (NO) is identified as the endothelium-derived relaxing factor and a neurotransmitter with a superfusion bioassay cascade technique. By using a similar technique with rat superior cervical ganglion (SCG) as donor tissue and rabbit endothelium-denuded aortic ring as detector tissue, we report here that a vasodilator, which is more potent than NO, is released in the SCG upon field electrical stimulation (FES) or addition of nicotine. Release of this vasodilator was enhanced by arginine analogs, including N(omega)-nitro-l-arginine (a NO synthase inhibitor), suggesting that it is not NO. Analysis by gas chromatography/mass spectrometry identified 2 saturated fatty acids, palmitic acid methyl ester (PAME) and stearic acid methyl ester (SAME), being released from the SCG upon FES in the presence of arginine analogs. Exogenous PAME but not SAME induced significant aortic dilation (EC(50) = 0.19 nM), indicating that PAME is the potent vasodilator. Release of PAME and SAME was significantly diminished in chronically decentralized SCG but not denervated SCG, suggesting the preganglionic origin. Furthermore, release of both fatty acids was calcium- and myosin light chain kinase-dependent, suggesting that both were released from axoplasmic vesicular stores. Electrophysiological studies further demonstrated that PAME but not SAME inhibited nicotine-induced inward currents in cultured SCG and the alpha7-nicotinic acetylcholine receptor-expressing Xenopus oocytes. Endogenous PAME appears to play a role in modulation of the autonomic ganglionic transmission and to complement the vasodilator effect of NO.


Subject(s)
Palmitates/metabolism , Receptors, Nicotinic/metabolism , Superior Cervical Ganglion/metabolism , Synaptic Transmission/physiology , Animals , Cells, Cultured , Electric Stimulation , Enzyme Inhibitors/pharmacology , Male , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nitric Oxide/metabolism , Nitroarginine/pharmacology , Oocytes/physiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Stearic Acids/metabolism , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/drug effects , Sympathectomy , Synaptic Transmission/drug effects , Vasodilation/physiology , Xenopus , alpha7 Nicotinic Acetylcholine Receptor
14.
Nanotechnology ; 19(20): 205102, 2008 May 21.
Article in English | MEDLINE | ID: mdl-21825732

ABSTRACT

Biological molecules conjugating with nanoparticles are valuable for applications including bio-imaging, bio-detection, and bio-sensing. Nanometer-sized diamond particles have excellent electronic and chemical properties for bio-conjugation. In this study, we manipulated the carboxyl group produced on the surface of nanodiamond (carboxylated nanodiamond, cND) for conjugating with alpha-bungarotoxin (α-BTX), a neurotoxin derived from Bungarus multicinctus with specific blockade of alpha7-nicotinic acetylcholine receptor (α7-nAChR). The electrostatic binding of cND-α-BTX was mediated by the negative charge of the cND and the positive charge of the α-BTX in physiological pH conditions. Sodium dodecyl sulfate-polyacrylamide gel analysis and matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI/TOF-MS) spectra displayed that α-BTX proteins were conjugated with cND particles via non-covalent bindings. The green fluorescence of the cND particles combining with the red fluorescence of tetramethylrhodamine-labeled α-BTX presented a yellow color at the same location, which indicated that α-BTX proteins were conjugated with cND particles. Xenopus laevis's oocytes expressed the human α7-nAChR proteins by microinjection with α7-nAChR mRNA. The cND-α-BTX complexes were bound to α7-nAChR locating on the cell membrane of oocytes and human lung A549 cancer cells analyzed by laser scanning confocal microscopy. The choline-evoked α7-nAChR-mediated inward currents of the oocytes were blocked by cND-α-BTX complexes in a concentration-dependent manner using two-electrode voltage-clamp recording. Furthermore, the fluorescence intensity of cND-α-BTX binding on A549 cells could be quantified by flow cytometry. These results indicate that cND-conjugated α-BTX still preserves its biological activity in blocking the function of α7-nAChR, and provide a visual system showing the binding of α-BTX to α7-nAChR.

15.
Am J Physiol Heart Circ Physiol ; 293(3): H1737-44, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17557921

ABSTRACT

Statins are reported to be beneficial in treating a multitude of disorders including dementia due to Alzheimer disease (AD) and vascular dementia (VaD) with varying, yet-to-be determined mechanisms of actions. Although cholinesterase inhibitors (ChEIs) are still recommended as the primary drug of choice for AD and related diseases, their efficacy is frequently questioned. We recently reported that alpha7-neuronal acetylcholine nicotinic receptor (alpha7-nAChR)-mediated neurogenic vasodilation of porcine cerebral arteries was blocked by ChEIs, and this blockade was prevented by statin pretreatment. The exact mechanism of interaction between ChEIs and statins remains unclear. Activation of alpha7-nAChRs located on perivascular postganglionic sympathetic nerve terminals releases norepinephrine, which then acts on presynaptic beta(2)-adrenoceptors located on neighboring nitrergic nerve terminals, resulting in nitric oxide release and vasodilation. The present study, therefore, was designed to determine whether interaction of ChEIs and statins occurs at the alpha7-nAChR level. We examined effects of concurrent application of ChEIs and statins on alpha7-nAChR-mediated inward currents in primary neuronal cultures of rat superior cervical ganglion cells, the origin of the perivascular sympathetic innervation to the cerebral arteries. The results indicated that physostigmine, neostigmine, and galantamine inhibited choline- and nicotine-induced whole cell currents in a concentration-dependent manner. This inhibition, which was noncompetitive in nature, was prevented by concurrent application of mevastatin and lovastatin in a concentration-dependent manner. These results suggest that statins protect alpha7-nAChR function directly at the receptor level. Since alpha7-nAChR is neuroprotective, having beneficial effects on memory and cerebral vascular function, its functional inhibition by ChEIs may explain in part the limitation of its effectiveness in AD and VaD therapy. Protection of alpha7-nAChR function from ChEI inhibition by concurrent administration of statins may provide an alternative strategy in improving the efficacy of AD and VaD therapy.


Subject(s)
Cholinesterase Inhibitors/pharmacology , Evoked Potentials/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurons/physiology , Receptors, Nicotinic/physiology , Superior Cervical Ganglion/physiology , Animals , Cells, Cultured , Choline/pharmacology , Dose-Response Relationship, Drug , Evoked Potentials/physiology , Male , Neurons/drug effects , Nicotine/pharmacology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Superior Cervical Ganglion/drug effects , alpha7 Nicotinic Acetylcholine Receptor
16.
Am J Physiol Heart Circ Physiol ; 291(1): H202-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16772524

ABSTRACT

We have proposed that activation of cerebral perivascular sympathetic alpha7-nicotinic acetylcholine receptors (alpha7-nAChRs) by nicotinic agonists releases norepinephrine, which then acts on parasympathetic nitrergic nerves, resulting in release of nitric oxide and vasodilation. Using patch-clamp electrophysiology, immunohistochemistry, and in vitro tissue bath myography, we tested this axo-axonal interaction hypothesis further by examining whether blocking norepinephrine reuptake enhanced alpha7-nAChR-mediated cerebral nitrergic neurogenic vasodilation. The results indicated that choline- and nicotine-induced alpha7-nAChR-mediated nitrergic neurogenic relaxation in endothelium-denuded isolated porcine basilar artery rings was enhanced by desipramine and imipramine at lower concentrations (0.03-0.1 microM) but inhibited at higher concentrations (0.3-10 microM). In cultured superior cervical ganglion (SCG) neurons of the pig and rat, choline (0.1-30 mM)-evoked inward currents were reversibly blocked by 1-30 microM mecamylamine, 1-30 microM methyllycaconitine, 10-300 nM alpha-bungarotoxin, and 0.1-10 microM desipramine and imipramine, providing electrophysiological evidence for the presence of similar functional alpha7-nAChRs in cerebral perivascular sympathetic neurons of pigs and rats. In alpha7-nAChR-expressing Xenopus oocytes, choline-elicited inward currents were attenuated by alpha-bungarotoxin, imipramine, and desipramine. These monoamine uptake inhibitors appeared to directly block the alpha7-nAChR, resulting in diminished nicotinic agonist-induced cerebral nitrergic vasodilation. The enhanced nitrergic vasodilation by lower concentrations of monoamine uptake inhibitors is likely due to a greater effect on monoamine uptake than on alpha7-nAChR blockade. These results further support the hypothesis of axo-axonal interaction in nitrergic regulation of cerebral vascular tone.


Subject(s)
Antidepressive Agents, Tricyclic/administration & dosage , Basilar Artery/physiology , Biogenic Monoamines/metabolism , Cerebrovascular Circulation/physiology , Neurons/physiology , Receptors, Nicotinic/metabolism , Vasodilation/physiology , Animals , Basilar Artery/drug effects , Cells, Cultured , Cerebrovascular Circulation/drug effects , Desipramine/administration & dosage , Dose-Response Relationship, Drug , Female , Imipramine/administration & dosage , Male , Metabolic Clearance Rate/drug effects , Nitric Oxide/metabolism , Oocytes/drug effects , Oocytes/physiology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Superior Cervical Ganglion/drug effects , Superior Cervical Ganglion/physiology , Swine , Vasodilation/drug effects , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
17.
J Cereb Blood Flow Metab ; 26(12): 1562-76, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16639428

ABSTRACT

Cholinesterase inhibitors (ChEIs) have been used to treat Alzheimer's disease (AD). The efficacy of these drugs, however, is less than satisfactory. The possibility that ChEIs may have effects unrelated to ChE activity, such as negatively modulate neuronal nicotinic acetylcholine receptors (nAChRs) was evaluated. Since alpha7-nAChRs on cerebral perivascular sympathetic neurons mediate cerebral parasympathetic-nitrergic vasodilation, effects of physostigmine, neostigmine, and galantamine on alpha7-nAChR-mediated dilation in isolated porcine basilar arterial rings denuded of endothelium was examined using in vitro tissue bath technique. The results indicated that these ChEIs blocked vasodilation induced by choline (0.3 mmol/L), nicotine (0.1 mmol/L), and transmural nerve stimulation (TNS). The ChEI inhibition of dilation induced by TNS but not by choline or nicotine was prevented by atropine (0.1 micromol/L) pretreatment. Furthermore, using confocal microscopy, significant calcium influx induced by choline and nicotine in cultured porcine superior cervical ganglion (SCG) cells was attenuated by ChEIs. In alpha7-nAChR-expressed Xenopus oocytes, nicotine-induced inward currents were attenuated by alpha-bungarotoxin and ChEIs. Moreover, ChEI inhibition of nicotine- and choline-induced dilation was prevented by pretreatment with mevastatin and lovastatin (10 micromol/L), which did not affect ChEI inhibition of TNS-induced relaxation. These findings suggest that ChEIs inhibit the alpha7-nAChRs located on postganglionic sympathetic nerve terminals of SCG origin, causing a decreased release of nitric oxide in the neighboring nitrergic nerves and cerebral vasodilation. Inhibition of alpha7-nAChRs leading to a potential cerebral hypoperfusion may contribute to the limitation of ChEIs and question the validity of using a ChEI alone in treating AD. The efficacy of ChEIs may be improved by concurrent use of statins.


Subject(s)
Basilar Artery/metabolism , Cholinesterase Inhibitors/pharmacology , Nitrergic Neurons/metabolism , Receptors, Nicotinic/metabolism , Superior Cervical Ganglion/metabolism , Vasodilation , Animals , Anticholesteremic Agents/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cells, Cultured , Female , Gene Expression , Male , Microscopy, Confocal/methods , Nitric Oxide/biosynthesis , Oocytes/cytology , Oocytes/metabolism , Organ Culture Techniques , Parasympathetic Nervous System/metabolism , Receptors, Nicotinic/genetics , Superior Cervical Ganglion/cytology , Swine , Vasodilation/drug effects , Vasodilation/genetics , Xenopus , alpha7 Nicotinic Acetylcholine Receptor
18.
Mol Cancer Ther ; 4(2): 281-90, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15713899

ABSTRACT

Colorectal carcinoma is a leading cause of human mortality due to its high metastatic ability. Because the activation of matrix metalloproteinases (MMP) is a key factor in the metastatic process, agents with the ability to inhibit MMP activity have potential in the treatment of colorectal carcinoma. In the present study, among 36 flavonoids examined, myricetin was found to be the most potent inhibitor of MMP-2 enzyme activity in COLO 205 cells (IC50 = 7.82 micromol/L). Myricetin inhibition of MMP-2 enzyme activity was also found in the human colorectal carcinoma cell lines COLO 320HSR, COLO 320DM, HT 29, and COLO 205-X (IC50 = 11.18, 11.56, 13.25, and 23.51 micromol/L, respectively). In contrast, no inhibitory effect of MMP-2 protein expression or enzyme activity was observed in myricitrin (myricetin-3-rhamnoside)-treated cells. In 12-O-tetradecanoylphorbol-13-acetate (TPA)-stimulated COLO 205 cells, an increase in MMP-2 protein expression and enzyme activity, as well as of protein kinase C (PKC) alpha protein translocation, extracellular signal-regulated kinase (ERK) 1/2 protein phosphorylation, and c-Jun protein expression was observed. ERK inhibitor (PD98059) and PKC inhibitors (GF-109203X and H-7), but not p38 inhibitor (SB203580) or c-jun-NH2-kinase inhibitor (SP600125), significantly inhibited TPA-induced MMP-2 protein expression, with reduced ERK phosphorylation and c-Jun protein expression. Addition of myricetin but not myricitrin suppressed TPA-induced MMP-2 protein expression in COLO 205 cells by blocking the TPA-induced events, including translocation of PKCalpha from cytosol to membrane, phosphorylation of ERK1/2 protein, and induction of c-Jun protein expression. Addition of PD98059 or GF-109203X significantly enhanced the inhibitory effect of myricetin on MMP-2 enzyme activity induced by TPA. Furthermore, myricetin, but not myricitrin, suppressed TPA-induced invasion of COLO 205 cells in an in vitro invasion assay using Engelbreth-Holm-Swarm sarcoma tumor extract Matrigel-coated Transwells. Results of the present study indicate that myricetin significantly blocked both endogenous and TPA-induced MMP-2 enzyme activity by inhibiting its protein expression and enzyme activity. The blockade involved suppression of PKC translocation, ERK phosphorylation, and c-Jun protein expression.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma/enzymology , Colorectal Neoplasms/enzymology , Flavonoids/pharmacology , Matrix Metalloproteinase Inhibitors , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein Transport/drug effects , Tetradecanoylphorbol Acetate/pharmacology
19.
Toxicol Lett ; 153(2): 191-200, 2004 Nov 02.
Article in English | MEDLINE | ID: mdl-15451549

ABSTRACT

Nicotine has been shown to induce relaxation via nitric oxide (NO) production with activation of endothelium nitric oxide synthase (eNOS), however the effect of nicotine on lipopolysaccharide/interferon-gamma (LPS/IFN-gamma)-induced NO production and inducible NOS (iNOS) gene expression is still undefined. Here, nicotine alone did not affect the NO and PGE2 production in RAW264.7 and primary peritoneal macrophages. Interestingly, nicotine showed the dose-dependent stimulatory effect on LPS (20 ng/ml)/IFN-gamma (10 ng/ml)-induced NO but not PGE2 production in both cells. Although nicotine stimulates NO production in the presence of LPS/IFN-gamma, LPS at the dose of 20 ng/ml, nicotine showed no obvious inductive effect on the expression of iNOS protein by Western blotting in both cells. However, nicotine significantly stimulates LPS (2.5, 5 ng/ml)/IFN-gamma (10 ng/ml)-induced iNOS expression and NO production in RAW264.7 cells. Cytotoxicity assay showed that nicotine enhanced LPS (20 ng/ml) and IFN-gamma (10 ng/ml)-induced cytotoxicity, which was inhibited by an NOS inhibitor N-nitro-L-arginine (NLA) in RAW264.7 cells. Direct and indirect NOS activity assays indicated that nicotine did not affect NOS activity. And, iNOS protein stability was not changed by nicotine after LPS/IFN-gamma treatment. These data indicates that nicotine may potentiate LPS/IFN-gamma-induced cytotoxic effects by enhancing NO production; enhancing iNOS gene expression induced by LPS/IFN-gamma is involved. A cross-talk between inflammation and smoking was proposed in the present study.


Subject(s)
Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Nicotine/pharmacology , Nitric Oxide/biosynthesis , Animals , Cell Line , Cell Survival/drug effects , Dinoprostone/biosynthesis , Mice , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitroarginine/pharmacology
20.
J Cell Biochem ; 86(2): 331-9, 2002.
Article in English | MEDLINE | ID: mdl-12112002

ABSTRACT

Induction of heme oxygenase (HO)-1 during inflammation has been demonstrated in many cell types, but the contribution of inflammatory molecules nitric oxide (NO) and prostaglandin E(2) (PGE(2)) has remained unresolved. Here we show that NO donors including sodium nitroprusside (SNP) and spermine nonoate (SP-NO), and PGE(2) significantly stimulate HO-1 expression in RAW264.7 macrophages, associated with alternative induction on NO and PGE(2) in medium, respectively. NO donors also show the inductive effect on cyclo-oxygenase 2 protein and PGE(2) production. In the presence of lipopolysaccharide and interferon-gamma (LPS/IFN-gamma), HO-1 protein was induced slightly but significantly, and SNP, SP-NO, and PGE(2) enhanced HO-1 protein induced by LPS/IFN-gamma. L-Arginine analogs N-nitro-L-arginine methyl ester (L-NAME) and N-nitro-L-arginine (NLA) significantly block HO-1 protein induced by LPS/IFN-gamma associated with a decrease in NO (not PGE(2)) production. And, NSAIDs aspirin and diclofenase dose dependently inhibited LPS/IFN-gamma-induced HO-1 protein accompanied by suppression of PGE(2) (not NO) production. PD98059 (a specific inhibitor of MEKK), but not SB203580 (a specific inhibitor of p38 kinase), attenuated PGE(2) (not SP-NO) induced HO-1 protein. Under UVC (100 J/m(2)) and UVB (50 J/m(2)) irradiation, PGE(2) or SP-NO treatment prevents cells from UVC or UVB-induced cell death, and HO-1 inhibitor tin protoporphyrin (SnPP) reverses the preventive effects of PGE(2) and SP-NO. The protective activity induced by PGE(2) on UVC or UVB irradiation-induced cell death was blocked by MAPK inhibitor PD98059 (not SB203580). These results demonstrated that inflammatory molecules NO and PGE(2) were potent inducers of HO-1 gene, and protected cells from UV-irradiation-induced cell death through HO-1 induction.


Subject(s)
Cell Death/radiation effects , Dinoprostone/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/radiation effects , Nitric Oxide/metabolism , Animals , Cell Line , Cell Survival/drug effects , Drug Synergism , Enzyme Induction/drug effects , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase-1 , Macrophages/enzymology , Macrophages/metabolism , Membrane Proteins , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Donors/pharmacology , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL