Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Acta Physiol (Oxf) ; 230(1): e13484, 2020 09.
Article in English | MEDLINE | ID: mdl-32336027

ABSTRACT

For a long time, the vascular tone was considered to be regulated exclusively by tonic innervation of vasoconstrictor adrenergic nerves. However, accumulating experimental evidence has revealed the existence of nerves mediating vasodilatation, including perivascular nitrergic nerves (PNN), in a wide variety of mammalian species. Functioning of nitrergic vasodilator nerves is evidenced in several territories, including cerebral, mesenteric, pulmonary, renal, penile, uterine and cutaneous arteries. Nitric oxide (NO) is the main neurogenic vasodilator in cerebral arteries and acts as a counter-regulatory mechanism for adrenergic vasoconstriction in other vascular territories. In the penis, NO relaxes the vascular and cavernous smooth muscles leading to penile erection. Furthermore, when interacting with other perivascular nerves, NO can act as a neuromodulator. PNN dysfunction is involved in the genesis and maintenance of vascular disorders associated with arterial and portal hypertension, diabetes, ageing, obesity, cirrhosis and hormonal changes. For example defective nitrergic function contributes to enhanced sympathetic neurotransmission, vasoconstriction and blood pressure in some animal models of hypertension. In diabetic animals and humans, dysfunctional nitrergic neurotransmission in the corpus cavernosum is associated with erectile dysfunction. However, in some vascular beds of hypertensive and diabetic animals, an increased PNN function has been described as a compensatory mechanism to the increased vascular resistance. The present review summarizes current understanding on the role of PNN in control of vascular tone, its alterations under different conditions and the associated mechanisms. The knowledge of these changes can serve to better understand the mechanisms involved in these disorders and help in planning new treatments.


Subject(s)
Arteries/innervation , Vasoconstriction , Vasodilation , Animals , Humans , Male , Nitric Oxide , Penis/innervation
2.
Curr Pharm Des ; 26(30): 3633-3651, 2020.
Article in English | MEDLINE | ID: mdl-32242780

ABSTRACT

Cardiovascular diseases (CVD) are considered a major health problem worldwide, being the main cause of mortality in developing and developed countries. Endothelial dysfunction, characterized by a decline in nitric oxide production and/or bioavailability, increased oxidative stress, decreased prostacyclin levels, and a reduction of endothelium-derived hyperpolarizing factor is considered an important prognostic indicator of various CVD. Changes in cyclic nucleotides production and/ or signalling, such as guanosine 3', 5'-monophosphate (cGMP) and adenosine 3', 5'-monophosphate (cAMP), also accompany many vascular disorders that course with altered endothelial function. Phosphodiesterases (PDE) are metallophosphohydrolases that catalyse cAMP and cGMP hydrolysis, thereby terminating the cyclic nucleotide-dependent signalling. The development of drugs that selectively block the activity of specific PDE families remains of great interest to the research, clinical and pharmaceutical industries. In the present review, we will discuss the effects of PDE inhibitors on CVD related to altered endothelial function, such as atherosclerosis, diabetes mellitus, arterial hypertension, stroke, aging and cirrhosis. Multiple evidences suggest that PDEs inhibition represents an attractive medical approach for the treatment of endothelial dysfunction-related diseases. Selective PDE inhibitors, especially PDE3 and PDE5 inhibitors are proposed to increase vascular NO levels by increasing antioxidant status or endothelial nitric oxide synthase expression and activation and to improve the morphological architecture of the endothelial surface. Thereby, selective PDE inhibitors can improve the endothelial function in various CVD, increasing the evidence that these drugs are potential treatment strategies for vascular dysfunction and reinforcing their potential role as an adjuvant in the pharmacotherapy of CVD.


Subject(s)
Cyclic GMP , Phosphoric Diester Hydrolases , Biological Factors , Humans , Nitric Oxide , Nucleotides, Cyclic
3.
Vascul Pharmacol ; 115: 33-45, 2019 04.
Article in English | MEDLINE | ID: mdl-30790705

ABSTRACT

Perinatal sodium overload induces endothelial dysfunction in adult offspring, but the underlying mechanisms are not fully known. The involvement of tissue renin-angiotensin system on high sodium-programmed endothelial dysfunction was examined. Acetylcholine and angiotensin I and II responses were analyzed in aorta and mesenteric resistance arteries from 24-week-old male offspring of normal-salt (O-NS, 1.3% NaCl) and high-salt (O-HS, 8% NaCl) fed dams. COX-2 expression, O2- production and angiotensin converting enzyme (ACE) activity were determined. A separated O-HS was treated with losartan (15 mg kg-1/day) for eight weeks. Compared to O-NS, O-HS were normotensive. Acetylcholine-induced relaxation was impaired in O-HS arteries, which was improved by tempol, apocynin or indomethacin. The angiotensin I-induced contraction was greater in O-HS arteries, whereas the angiotensin II responses were unchanged. ACE activity, O2- production and COX-2 expression were increased in O-HS arteries. In this group, the increased O2- production was inhibited by apocynin or losartan. Chronic losartan decreased COX-2 expression and restored the endothelium-dependent vasodilation in O-HS. Our findings reiterate that perinatal sodium overload programs endothelial dysfunction in adult offspring through a blood pressure-independent mechanism. Our results also suggest that vascular angiotensin II is the main mediator of high sodium-programmed endothelial dysfunction, promoting COX-2 expression and oxidative stress.


Subject(s)
Animal Nutritional Physiological Phenomena , Aorta, Thoracic/physiopathology , Endothelium, Vascular/physiopathology , Maternal Nutritional Physiological Phenomena , Mesenteric Arteries/physiopathology , Prenatal Exposure Delayed Effects , Renin-Angiotensin System , Sodium Chloride, Dietary/adverse effects , Age Factors , Angiotensin I/pharmacology , Angiotensin II/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Cyclooxygenase 2/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Oxidative Stress/drug effects , Peptidyl-Dipeptidase A/metabolism , Pregnancy , Rats, Wistar , Renin-Angiotensin System/drug effects , Superoxides/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
4.
Biochim Biophys Acta Mol Basis Dis ; 1864(12): 3577-3587, 2018 12.
Article in English | MEDLINE | ID: mdl-30254014

ABSTRACT

We investigated whether hypertension induced by maternal lipopolysaccharide (LPS) administration during gestation is linked to peripheral vascular and renal hemodynamic regulation, through angiotensin II → NADPH-oxidase signalling, and whether these changes are directly linked to intrauterine oxidative stress. Female Wistar rats were submitted to LPS, in the absence or presence of α-tocopherol during pregnancy. Malondialdehyde in placenta and in livers from dams and foetuses was enhanced by LPS. Tail-cuff systolic blood pressure (tcSBP) was elevated in the 16-week-old LPS offspring. Renal malondialdeyde and protein expression of NADPH oxidase isoform 2 were elevated in these animals at 20 weeks of age. Maternal α-tocopherol treatment prevented the elevation in malondialdehyde induced by LPS on placenta and livers from dams and foetuses, as well as prevented the elevation in tcSBP and the elevation in renal malondialdehyde in adult life. LPS offspring presented impairment of endothelium-dependent relaxation in aorta and mesenteric rings, which was blunted by angiotensin type 1 receptor (AT1R) blockade and NADPH oxidase inhibition. At age of 32 weeks, renal hemodynamic parameters were unchanged in anaesthetised LPS offspring, but angiotensin II infusion led to an increased glomerular filtration rate paralleled by filtration fraction elevation. The renal haemodynamic changes provoked by angiotensin II was prevented by early treatment with α-tocopherol and by late treatment with NADPH oxidase inhibitor. These results point to oxidative stress as a mediator of offspring hypertension programmed by maternal inflammation and to the angiotensin II → NADPH oxidase signalling pathway as accountable for vascular and renal dysfunctions that starts and maintains hypertension.


Subject(s)
Antioxidants/therapeutic use , Hemodynamics/drug effects , Hypertension/prevention & control , Lipopolysaccharides/metabolism , Oxidative Stress/drug effects , Pregnancy Complications, Cardiovascular/prevention & control , alpha-Tocopherol/therapeutic use , Acetophenones/therapeutic use , Angiotensin II/metabolism , Animals , Blood Pressure/drug effects , Female , Hypertension/metabolism , Kidney/blood supply , Kidney/drug effects , Kidney/metabolism , Kidney/physiopathology , Malondialdehyde/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Pregnancy , Pregnancy Complications, Cardiovascular/metabolism , Rats, Wistar
5.
Eur J Pharmacol ; 822: 59-68, 2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29355555

ABSTRACT

Ageing impairs endothelial function, which is considered a hallmark of the development of cardiovascular diseases in elderly. Cilostazol, a phosphodiesterase-3 inhibitor, has antiplatelet, antithrombotic and protective effects on endothelial cells. Here, we hypothesized that cilostazol could improve endothelial function in mesenteric resistance arteries (MRA) from old rats. Using eight-week cilostazol-treated (100mg/kg/day) or untreated 72-week-old Wistar rats, we evaluate the relaxation to acetylcholine, sodium nitroprusside (SNP), forskolin and isoproterenol and the noradrenaline-induced contraction in MRA. Superoxide anion and nitric oxide (NO) was measured by dihydroethidium- and diaminofluorescein-2-emitted fluorescence, respectively. Normotensive old rats had impaired acetylcholine-induced NO- and EDHF-mediated relaxation and increased noradrenaline vasoconstriction than young rats. This age-associated endothelial dysfunction was restored by cilostazol treatment. Relaxation to SNP, forskolin or isoproterenol remained unmodified by cilostazol. Diaminofluorescein-2-emitted fluorescence was increased while dihydroethidium-emitted was decreased by cilostazol, indicating increased NO and reduced superoxide generation, respectively. Cilostazol improves endothelial function in old MRA without affecting blood pressure. This protective effect of cilostazol could be attributed to reduced oxidative stress, increased NO bioavailability and EDHF-type relaxation. Although these results are preliminary, we believe that should stimulate further interest in cilostazol as an alternative for the treatment of age-related vascular disorders.


Subject(s)
Aging/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Endothelium, Vascular/drug effects , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Phosphodiesterase 3 Inhibitors/pharmacology , Tetrazoles/pharmacology , Animals , Arterial Pressure/drug effects , Cilostazol , Endothelium, Vascular/metabolism , Male , Mesenteric Arteries/cytology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Superoxides/metabolism , Vasoconstriction/drug effects , Vasodilation/drug effects
6.
PLoS One ; 11(6): e0156793, 2016.
Article in English | MEDLINE | ID: mdl-27272874

ABSTRACT

INTRODUCTION: We analysed possible time-dependent changes in nitrergic perivascular innervation function from diabetic rats and mechanisms implicated. MATERIALS AND METHODS: In endothelium-denuded mesenteric arteries from control and four- (4W) and eight-week (8W) streptozotocin-induced diabetic rats the vasoconstriction to EFS (electrical field stimulation) was analysed before and after preincubation with L-NAME. Neuronal NO release was analysed in the absence and presence of L-arginine, tetrahydrobiopterine (BH4) and L-arginine plus BH4. Superoxide anion (O2-), peroxynitrite (ONOO-) and superoxide dismutase (SOD) activity were measured. Expressions of Cu-Zn SOD, nNOS, p-nNOS Ser1417, p-nNOS Ser847, and Arginase (Arg) I and II were analysed. RESULTS: EFS response was enhanced at 4W, and to a lesser extent at 8W. L-NAME increased EFS response in control rats and at 8W, but not at 4W. NO release was decreased at 4W and restored at 8W. L-arginine or BH4 increased NO release at 4W, but not 8W. SOD activity and O2- generation were increased at both 4W and 8W. ONOO- decreased at 4W while increased at 8W. Cu-Zn SOD, nNOS and p-NOS Ser1417 expressions remained unmodified at 4W and 8W, whereas p-nNOS Ser847 was increased at 4W. ArgI was overexpressed at 4W, remaining unmodified at 8W. ArgII expression was similar in all groups. CONCLUSIONS: Our results show a time-dependent effect of diabetes on neuronal NO release. At 4W, diabetes induced increased O2- generation, nNOS uncoupling and overexpression of ArgI and p-nNOS Ser847, resulting in decreased NO release. At 8W, NO release was restored, involving normalisation of ArgI and p-nNOS Ser847 expressions.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Mesenteric Arteries/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Vasoconstriction/drug effects , Animals , Electric Stimulation , Male , Mesenteric Arteries/metabolism , Peroxynitrous Acid/metabolism , Rats , Streptozocin , Superoxide Dismutase/metabolism , Superoxides/metabolism , Time Factors
7.
PLoS One ; 9(7): e100356, 2014.
Article in English | MEDLINE | ID: mdl-24992476

ABSTRACT

BACKGROUND AND PURPOSE: Tranilast, in addition to its capacity to inhibit mast cell degranulation, has other biological effects, including inhibition of reactive oxygen species, cytokines, leukotrienes and prostaglandin release. In the current study, we analyzed whether tranilast could alter endothelial function in rat mesenteric resistance arteries (MRA). EXPERIMENTAL APPROACH: Acetylcholine-induced relaxation was analyzed in MRA (untreated and 1-hour tranilast treatment) from 6 month-old Wistar rats. To assess the possible participation of endothelial nitric oxide or prostanoids, acetylcholine-induced relaxation was analyzed in the presence of L-NAME or indomethacin. The participation of endothelium-derived hyperpolarizing factor (EDHF) in acetylcholine-induced response was analyzed by preincubation with TRAM-34 plus apamin or by precontraction with a high K+ solution. Nitric oxide (NO) and superoxide anion levels were measured, as well as vasomotor responses to NO donor DEA-NO and to large conductance calcium-activated potassium channel opener NS1619. KEY RESULTS: Acetylcholine-induced relaxation was greater in tranilast-incubated MRA. Acetylcholine-induced vasodilation was decreased by L-NAME in a similar manner in both experimental groups. Indomethacin did not modify vasodilation. Preincubation with a high K+ solution or TRAM-34 plus apamin reduced the vasodilation to ACh more markedly in tranilast-incubated segments. NO and superoxide anion production, and vasodilator responses to DEA-NO or NS1619 remained unmodified in the presence of tranilast. CONCLUSIONS AND IMPLICATIONS: Tranilast increased the endothelium-dependent relaxation to acetylcholine in rat MRA. This effect is independent of the nitric oxide and cyclooxygenase pathways but involves EDHF, and is mediated by an increased role of small conductance calcium-activated K+ channels.


Subject(s)
Acetylcholine/pharmacology , Biological Factors/physiology , Mesenteric Arteries/drug effects , Vasodilator Agents/pharmacology , ortho-Aminobenzoates/pharmacology , Animals , Benzimidazoles/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Hydrazines/pharmacology , Indomethacin/pharmacology , Male , Mast Cells/drug effects , Mast Cells/pathology , Mesenteric Arteries/pathology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Rats , Rats, Wistar , Superoxides/metabolism
8.
PLoS One ; 8(8): e73232, 2013.
Article in English | MEDLINE | ID: mdl-23977380

ABSTRACT

OBJECTIVES: We analyzed whether mast cell stabilization by either ketotifen or tranilast could alter either sympathetic or nitrergic innervation function in rat mesenteric arteries. METHODS: Electrical field stimulation (EFS)-induced contraction was analyzed in mesenteric segments from 6-month-old Wistar rats in three experimental groups: control, 3-hour ketotifen incubated (0.1 αmol/L), and 3-hour tranilast incubated (0.1 mmol/L). To assess the possible participation of nitrergic or sympathetic innervation, EFS contraction was analyzed in the presence of non-selective nitric oxide synthase (NOS) inhibitor L-NAME (0.1 mmol/L), α-adrenergic receptor antagonist phentolamine (0.1 µmol/L), or the neurotoxin 6-hydroxydopamine (6-OHDA, 1.46 mmol/L). Nitric oxide (NO) and superoxide anion (O2.(-) levels were measured, as were vasomotor responses to noradrenaline (NA) and to NO donor DEA-NO, in the presence and absence of 0.1 mmol/L tempol. Phosphorylated neuronal NOS (P-nNOS) expression was also analyzed. RESULTS: EFS-induced contraction was increased by ketotifen and decreased by tranilast. L-NAME increased the vasoconstrictor response to EFS only in control segments. The vasodilator response to DEA-NO was higher in ketotifen- and tranilast-incubated segments, while tempol increased vasodilator response to DEA-NO only in control segments. Both NO and O2(-) release, and P-nNOS expression were diminished by ketotifen and by tranilast treatment. The decrease in EFS-induced contraction produced by phentolamine was lower in tranilast-incubated segments. NA vasomotor response was decreased only by tranilast. The remnant vasoconstriction observed in control and ketotifen-incubated segments was abolished by 6-OHDA. CONCLUSION: While both ketotifen and tranilast diminish nitrergic innervation function, only tranilast diminishes sympathetic innnervation function, thus they alter the vasoconstrictor response to EFS in opposing manners.


Subject(s)
Ketotifen/pharmacology , Mast Cells/physiology , Mesenteric Arteries/physiology , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , ortho-Aminobenzoates/pharmacology , Acetylcholine/pharmacology , Animals , Electric Stimulation , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Histamine Release/drug effects , In Vitro Techniques , Male , Mast Cells/drug effects , Mesenteric Arteries/drug effects , Neurons/drug effects , Neurons/metabolism , Nitrergic Neurons/drug effects , Nitrergic Neurons/physiology , Potassium Chloride/pharmacology , Rats, Wistar , Staining and Labeling , Superoxides/metabolism , Tolonium Chloride/metabolism , Vasodilator Agents/pharmacology , Vasomotor System/drug effects
9.
PLoS One ; 7(11): e50593, 2012.
Article in English | MEDLINE | ID: mdl-23209788

ABSTRACT

This study analyzed the effect of in utero exposure to maternal diabetes on contraction to noradrenaline in mesenteric resistance arteries (MRA) from adult offspring, focusing on the role of cyclooxygenase (COX)-derived prostanoids. Diabetes in the maternal rat was induced by a single injection of streptozotocin (50 mg/kg body weight) on day 7 of pregnancy. Contraction to noradrenaline was analyzed in isolated MRA from offspring of diabetic (O-DR) and non-diabetic (O-CR) rats at 3, 6 and 12 months of age. Release of thromboxane A(2) (TxA(2)) and prostaglandins E(2) (PGE(2)) and F(2α) (PGF(2α)), was measured by specific enzyme immunoassay kits. O-DR developed hypertension from 6 months of age compared with O-CR. Arteries from O-DR were hyperactive to noradrenaline only at 6 and 12 months of age. Endothelial removal abolished this hyperreactivity to noradrenaline between O-CR and O-DR. Preincubation with either the COX-1/2 (indomethacin) or COX-2 inhibitor (NS-398) decreased noradrenaline contraction only in 6- and 12-month-old O-DR, while it remained unmodified by COX-1 inhibitor SC-560. In vessels from 6-month-old O-DR, a similar reduction in the contraction to noradrenaline produced by NS-398 was observed when TP and EP receptors were blocked (SQ29548+AH6809). In 12-month-old O-DR, this effect was only achieved when TP, EP and FP were blocked (SQ29548+AH6809+AL8810). Noradrenaline-stimulated TxB(2) and PGE(2) release was higher in 6- and 12-month-old O-DR, whereas PGF(2α) was increased only in 12-month-old O-DR. Our results demonstrated that in utero exposure to maternal hyperglycaemia in rats increases the participation of COX-2-derived prostanoids on contraction to noradrenaline, which might help to explain the greater response to this agonist in MRA from 6- and 12-month-old offspring. As increased contractile response in resistance vessels may contribute to hypertension, our results suggest a role for these COX-2-derived prostanoids in elevating vascular resistance and blood pressure in offspring of diabetic rats.


Subject(s)
Cyclooxygenase 2/metabolism , Diabetes Mellitus, Experimental/metabolism , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Norepinephrine/pharmacology , Prostaglandins/metabolism , Animals , Cells, Cultured , Dinoprost/analogs & derivatives , Dinoprost/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Immunoenzyme Techniques , Indomethacin/pharmacology , Male , Nitrobenzenes/pharmacology , Pregnancy , Prostaglandins E/metabolism , Prostaglandins F/metabolism , Rats , Sulfonamides/pharmacology , Thromboxane A2/metabolism , Xanthones/pharmacology
10.
Eur J Pharmacol ; 654(3): 280-8, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21262224

ABSTRACT

This study analyzed the effect of aldosterone (0.05mg/kg per day, 3 weeks) on vasoconstriction induced by noradrenaline in mesenteric resistance arteries from WKY rats and SHR. Contraction to noradrenaline was measured in mesenteric resistance arteries from untreated and aldosterone-treatedrats from both strains. Participation of nitric oxide (NO), superoxide anions, thromboxane A(2) (TxA(2)) and prostacyclin in this response was determined. 6-keto-prostaglandin (PG)F1alpha and thromboxane B(2) (TxB(2)) releases were determined by enzyme immunoassay. NO and superoxide anion release were also determined by fluorescence and chemiluminiscence, respectively. Aldosterone did not modify noradrenaline-induced contraction in either strain. In mesenteric resistance arteries from both aldosterone-treated groups, endothelium removal or preincubation with NO synthesis inhibitor L-NAME increased the noradrenaline-induced contraction, while incubation with the superoxide anion scavenger tempol decreased it. Preincubation with either the COX-1/2 or COX-2 inhibitor (indomethacin and NS-398, respectively) decreased the noradrenaline contraction in aldosterone-treated animals, while this response was not modified by COX-1 inhibitor SC-560. TxA(2) synthesis inhibitor (furegrelate), or TxA2 receptor antagonist (SQ 29 548) also decreased the noradrenaline contraction in aldosterone-treated animals. In untreated SHR, but not WKY rats, this response was increased by L-NAME, and reduced by tempol, indomethacin, NS-398 or SQ 29 548. Aldosterone treatment did not modify NO or TxB(2) release, but it did increase superoxide anion and 6-keto-PGF(1alpha) release in mesenteric resistance arteries from both strains. In conclusion, chronic aldosterone treatment reduces smooth muscle contraction to alpha-adrenergic stimuli, producing a new balance in the release of endothelium-derived prostanoids and NO.


Subject(s)
Aldosterone/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Hypertension/physiopathology , Mesenteric Arteries/drug effects , Norepinephrine/pharmacology , Vasoconstriction/drug effects , Aldosterone/blood , Animals , Blood Pressure/drug effects , Collagen Type I/metabolism , Gene Expression Regulation/drug effects , Hydrocortisone/blood , Hypertension/blood , Hypertension/metabolism , Hypertension/pathology , In Vitro Techniques , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiology , Mesenteric Arteries/physiopathology , Nitric Oxide/metabolism , Norepinephrine/blood , Potassium Chloride/pharmacology , Prostaglandins/biosynthesis , Rats , Rats, Inbred WKY , Superoxides/metabolism
11.
J. physiol. biochem ; 66(4): 301-309, dic. 2010.
Article in English | IBECS | ID: ibc-122817

ABSTRACT

No disponible


Spontaneously hypertensive rat (SHR) offspring from L-arginine- and antioxidant-supplemented SHR dams had persistent lower blood pressure in adulthood. We investigated the influence of vascular mechanism in this effect. We analyzed response to acetylcholine and phenylephrine in aorta and superior mesenteric arteries from Wistar–Kyoto (WKY), SHR, and SHR perinatally supplemented with L-arginine and 4-hydroxy-2,2,6,6-tetramethylpiperidinoxyl (TEMPOL; SHR-suppl). Supplements reduced blood pressure persistently in SHR. Relaxation to acetylcholine was greater in WKY than SHR and remained unmodified in SHR-suppl compared with SHR. Acute TEMPOL did not alter relaxation to acetylcholine in WKY but increased it similarly in SHR and SHR-suppl. Phenylephrine contraction was increased in SHR compared to WKY. In SHR-suppl, this response was similar to SHR. Endothelium removal or N-nitro-L-arginine methyl ester (L-NAME) increased contraction to phenylephrine more in WKY than SHR. In SHR-suppl, this was similar to SHR. In both SHR and SHR-suppl, TEMPOL similarly reduced phenylephrine response. This effect was prevented by L-NAME. Results exposed reinforce the concept that oxidative stress during perinatal period is a contributing factor to the development of hypertension in SHR. Results also reveal that the beneficial effect of this supplementation does not appear to be related to improved endothelial function, suggesting that other regulatory mechanisms of blood pressure may be involved (AU)


Subject(s)
Animals , Rats , Hypertension/drug therapy , Peripheral Vascular Diseases/physiopathology , Arginine/pharmacokinetics , Antioxidants/pharmacokinetics , Oxidative Stress , Rats, Inbred SHR , Endothelial Cells , Protective Agents/pharmacokinetics , Disease Models, Animal
12.
J Physiol Biochem ; 66(4): 301-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20865358

ABSTRACT

Spontaneously hypertensive rat (SHR) offspring from L-arginine- and antioxidant-supplemented SHR dams had persistent lower blood pressure in adulthood. We investigated the influence of vascular mechanism in this effect. We analyzed response to acetylcholine and phenylephrine in aorta and superior mesenteric arteries from Wistar-Kyoto (WKY), SHR, and SHR perinatally supplemented with L-arginine and 4-hydroxy-2,2,6,6-tetramethylpiperidinoxyl (TEMPOL; SHR-suppl). Supplements reduced blood pressure persistently in SHR. Relaxation to acetylcholine was greater in WKY than SHR and remained unmodified in SHR-suppl compared with SHR. Acute TEMPOL did not alter relaxation to acetylcholine in WKY but increased it similarly in SHR and SHR-suppl. Phenylephrine contraction was increased in SHR compared to WKY. In SHR-suppl, this response was similar to SHR. Endothelium removal or N-nitro-L-arginine methyl ester (L-NAME) increased contraction to phenylephrine more in WKY than SHR. In SHR-suppl, this was similar to SHR. In both SHR and SHR-suppl, TEMPOL similarly reduced phenylephrine response. This effect was prevented by L-NAME. Results exposed reinforce the concept that oxidative stress during perinatal period is a contributing factor to the development of hypertension in SHR. Results also reveal that the beneficial effect of this supplementation does not appear to be related to improved endothelial function, suggesting that other regulatory mechanisms of blood pressure may be involved.


Subject(s)
Antioxidants/metabolism , Arginine/metabolism , Acetylcholine/chemistry , Animals , Animals, Newborn , Antioxidants/chemistry , Blood Pressure , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Drug , Hypertension/drug therapy , Hypertension/pathology , NG-Nitroarginine Methyl Ester/metabolism , Nitric Oxide/chemistry , Oxidative Stress , Phenylephrine/chemistry , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Spin Labels
13.
Life Sci ; 87(9-10): 302-8, 2010 Aug 28.
Article in English | MEDLINE | ID: mdl-20637778

ABSTRACT

AIMS: Na(+), K(+)-ATPase activity contributes to the regulation of vascular contractility and it has been suggested that vascular Na(+), K(+)-ATPase activity may be altered during the progression of diabetes; however the mechanisms involved in the altered Na(+), K(+)-ATPase activity changes remain unclear. Thus, the aim of the present study was to evaluate ouabain-sensitive Na(+), K(+)-ATPase activity and the mechanism(s) responsible for any alterations on this activity in aortas from 1- and 4-week streptozotocin-pretreated (50 mg kg(-1), i.v.) rats. MAIN METHODS: Aortic rings were used to evaluate the relaxation induced by KCl (1-10mM) in the presence and absence of ouabain (0.1 mmol/L) as an index of ouabain-sensitive Na(+), K(+)-ATPase activity. Protein expression of COX-2 and p-PKC-betaII in aortas were also investigated. KEY FINDINGS: Ouabain-sensitive Na(+), K(+)-ATPase activity was unaltered following 1-week of streptozotocin administration, but was increased in the 4-week diabetic aorta (27%). Endothelium removal or nitric oxide synthase inhibition with l-NAME decreased ouabain-sensitive Na(+), K(+)-ATPase activity only in control aortas. In denuded aortic rings, indomethacin, NS-398, ridogrel or Gö-6976 normalized ouabain-sensitive Na(+), K(+)-ATPase activity in 4-week diabetic rats. In addition, COX-2 (51%) and p-PKC-betaII (59%) protein expression were increased in 4-week diabetic aortas compared to controls. SIGNIFICANCE: In conclusion, diabetes led to a time-dependent increase in ouabain-sensitive Na(+), K(+)-ATPase activity. The main mechanism involved in this activation is the release of TxA(2)/PGH(2) by COX-2 in smooth muscle cells, linked to activation of the PKC pathway.


Subject(s)
Aorta, Thoracic/physiopathology , Diabetes Mellitus, Experimental/physiopathology , Ouabain/pharmacology , Prostaglandins/metabolism , Protein Kinase C/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/enzymology , Blotting, Western , Cyclooxygenase 2/biosynthesis , Diabetes Mellitus, Experimental/enzymology , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Male , Prostaglandin H2/metabolism , Rats , Rats, Wistar , Thromboxane A2/metabolism , Time Factors , Vasoconstriction/drug effects , Vasodilation/drug effects
14.
Clin Sci (Lond) ; 119(7): 283-92, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20459396

ABSTRACT

Our present study examines, in mesenteric resistance arteries, possible vasodilation alterations, and the role of NO and COX (cyclo-oxygenase) derivatives, in cirrhosis. The vasodilator response to acetylcholine was analysed in segments from control and cirrhotic rats. The effects of the non-specific COX inhibitor indomethacin, the specific COX-1 inhibitor SC-560 and the specific COX-2 inhibitor NS-398 were analysed in segments from both groups of rats. NO release was measured, and eNOS [endothelial NOS (NO synthase)], phospho-eNOS, iNOS (inducible NOS), COX-1 and COX-2 protein expression was also analysed. The effects of the TP receptor [TXA2 (thromboxane A(2)) receptor] antagonist SQ 29548, the TXA(2) synthesis inhibitor furegrelate, the PGI(2) (prostaglandin I(2)) synthesis inhibitor TCP (tranylcypromine) or TCP+furegrelate were only determined in segments from cirrhotic rats. The vasodilator response to acetylcholine was higher in segments from cirrhotic rats. Indomethacin, SC-560 and NS-398 did not modify the vasodilator response in control rats; however, indomethacin, NS-398 and TCP+furegrelate increased, whereas SC-560 did not modify and SQ 29548, furegrelate or TCP decreased, the vasodilator response to acetylcholine in cirrhotic rats. NO release was higher in cirrhotic rats. Furegrelate decreased, whereas TCP+furegrelate increased, the NO release in segments from cirrhotic rats. eNOS and COX-1 protein expression was not modified, whereas phosho-eNOS, iNOS and COX-2 protein expression was higher in cirrhotic rats. Therefore the increase in iNOS expression and eNOS activity may mediate increases in endothelial NO release. The COX-2 derivatives TXA(2) and PGI(2) may act simultaneously, producing a compensatory effect that reduces NO release and may limit the hyperdynamic circulation.


Subject(s)
Liver Cirrhosis, Experimental/metabolism , Mesenteric Arteries/metabolism , Nitric Oxide/metabolism , Prostaglandins A/physiology , Thromboxane A2/physiology , Animals , Blood Pressure/physiology , Body Weight/physiology , C-Reactive Protein/metabolism , Lipid Metabolism/physiology , Liver/pathology , Liver Cirrhosis, Experimental/physiopathology , Male , Organ Size/physiology , Prostaglandins/metabolism , Prostaglandins A/biosynthesis , Rats , Rats, Sprague-Dawley , Spleen/pathology , Superoxides/metabolism , Thromboxane A2/biosynthesis , Vasodilation/physiology
15.
J Hypertens ; 27(6): 1233-42, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19337130

ABSTRACT

OBJECTIVE: Hypertensive rats are more sensitive to the pressor effects of acute ouabain than normotensive rats. We analyzed the effect of chronic ouabain (approximately 8.0 microg/day, 5 weeks) treatment on the blood pressure of spontaneously hypertensive rats (SHRs) and Wistar-Kyoto rats and the contribution of vascular mechanisms. METHODS: Responses to acetylcholine and phenylephrine were analyzed in isolated tail arteries. Protein expression of endothelial nitric oxide synthase and cyclooxygenase-2 (COX-2) were also investigated. RESULTS: Ouabain treatment enhanced blood pressure only in SHRs. The pD2 for acetylcholine was decreased in arteries from SHRs compared with Wistar-Kyoto rats, and ouabain did not change this parameter. However, ouabain was able to increase the pD2 to phenylephrine in SHRs. Nitric oxide synthase inhibition with NG-nitro-L-arginine methyl ester or potassium channel blockade by tetraetylamonium increased the response to phenylephrine in SHRs, with a smaller increase in response observed in ouabain-treated SHRs. In addition, indomethacin (a COX inhibitor) and ridogrel (a thromboxane A2 synthase inhibitor and prostaglandin H2/thromboxane A2 receptor antagonist) decreased contraction to phenylephrine in tail rings from ouabain-treated SHRs. Protein expression of endothelial nitric oxide synthase was unaltered following ouabain treatment in SHRs, whereas COX-2 expression was increased. CONCLUSION: Chronic ouabain treatment further increases the raised blood pressure of SHRs. This appears to involve a vascular mechanism, related to a reduced vasodilator influence of nitric oxide and endothelium-derived hyperpolarizing factor and increased production of vasoconstrictor prostanoids by COX-2. These data suggest that the increased plasma levels of ouabain could play an important role in the maintenance of hypertension and the impairment of endothelial function.


Subject(s)
Blood Pressure/drug effects , Blood Pressure/physiology , Hypertension/etiology , Hypertension/physiopathology , Ouabain/toxicity , Acetylcholine/pharmacology , Animals , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , In Vitro Techniques , Indomethacin/pharmacology , Male , Ouabain/administration & dosage , Pentanoic Acids/pharmacology , Phenylephrine/pharmacology , Pyridines/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Thromboxane-A Synthase/antagonists & inhibitors , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology
16.
Clin Sci (Lond) ; 117(8): 305-12, 2009 Aug 24.
Article in English | MEDLINE | ID: mdl-19267693

ABSTRACT

Neuronal NO plays a functional role in many vascular tissues, including MAs (mesenteric arteries). Glucocorticoids alter NO release from endothelium and the CNS (central nervous system), but no results from peripheral innervation have been reported. In the present study we investigated the effects of dexamethasone on EFS (electrical field stimulation)-induced NO release in MAs from WKY (Wistar-Kyoto) rats and SHRs (spontaneously hypertensive rats) and the role of PKC (protein kinase C) in this response. In endothelium-denuded MAs, L-NAME (NG-nitro-L-arginine methyl ester) increased the contractile response to EFS only in segments from SHRs. EFS-induced contraction was reduced by 1 micromol/l dexamethasone in segments from SHRs, but not WKY rats, and this effect was abolished in the presence of dexamethasone. EFS induced a tetrodotoxin-resistant NO release in WKY rat MAs, which remained unchanged by 1 micromol/l dexamethasone. In SHR MAs, dexamethasone decreased basal and EFS-induced neuronal NO release, and this decrease was prevented by the glucocorticoid receptor antagonist mifepristone. Dexamethasone did not affect nNOS [neuronal NOS (NO synthase)] expression in either strain. In SHR MAs, incubation with calphostin C (a non-selective PKC inhibitor), Gö6983 (a classic PKC delta and zeta inhibitor), LY379196 (a PKCbeta inhibitor) or PKCzeta-PI (PKCzeta pseudosubstrate inhibitor) decreased both basal and EFS-induced neuronal NO release. Additionally, PKC activity was reduced by dexamethasone. The PKC inhibitor-induced reduction in NO release was unaffected by dexamethasone. In conclusion, results obtained in the present study indicate that PKC activity positively modulates the neuronal NO release in MAs from SHRs. They also reveal that by PKC inhibition, through activation of glucocorticoid receptors, dexamethasone reduces neuronal NO release in these arteries.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Hypertension/metabolism , Mesenteric Arteries/drug effects , Nitric Oxide/metabolism , Protein Kinase C/physiology , Animals , Electric Stimulation/methods , Enzyme Activation/physiology , Male , Mesenteric Arteries/metabolism , Nitric Oxide Synthase Type I/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Tissue Culture Techniques , Vasoconstriction/drug effects
17.
Regul Pept ; 146(1-3): 125-30, 2008 Feb 07.
Article in English | MEDLINE | ID: mdl-17904662

ABSTRACT

OBJECTIVE: To analyze the effect of aldosterone on the expression of calcitonin gene-related peptide (CGRP) receptor components, calcitonin-like receptor (CL receptor) and receptor activity modifying protein 1 (RAMP1), as well as the effect of this mineralocorticoid on CGRP-mediated vasodilation in middle cerebral arteries from Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). RESULTS: CGRP 0.1 nM-0.1 microM induced a concentration-dependent relaxation that was nitric oxide independent and higher in SHR middle cerebral arteries. CL receptor and RAMP1 expression were similar in both strains. The relaxation to CGRP was not modified by aldosterone 1 microM in either strain, although aldosterone 1 microM increased expression of CL receptor without modifying RAMP1 in segments from SHR rats. CONCLUSIONS: CGRP elicits greater vasodilation in middle cerebral arteries from SHR than WKY rats, that is nitric oxide independent, and by mechanism independent of CGRP receptor components expression. Although aldosterone increases the expression of CL receptor in SHR, it does not alter vasodilation to CGRP, since RAMP1 expression is not increased. These results indicate that the increase in CL receptor, without an increase in RAMP1, does not correlate with changes in functional role of the CGRP receptor.


Subject(s)
Aldosterone/pharmacology , Cerebral Arteries/drug effects , Receptors, Calcitonin Gene-Related Peptide/physiology , Receptors, Calcitonin/metabolism , Animals , Calcitonin Receptor-Like Protein , Cerebral Arteries/metabolism , Hypertension , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptor Activity-Modifying Proteins , Receptors, Calcitonin/drug effects , Up-Regulation
18.
J Pharmacol Exp Ther ; 322(3): 1129-36, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17562850

ABSTRACT

Glucocorticoids play a role in the control of vascular smooth muscle tone through the alteration of vasoconstrictor and vasodilator factor production. We studied the effect of dexamethasone on vasoconstriction induced by electrical field stimulation (EFS) in rat mesenteric arteries (MAs) and the role of hypertension in this effect. Endothelium-denuded MAs were obtained from Wistar-Kyoto rats and spontaneously hypertensive rats (SHRs). EFS response was analyzed by isometric tension recordings and cyclooxygenase (COX-1 and COX-2) expression by Western blot. Noradrenaline (NA) release was evaluated in segments incubated with [(3)H]NA. Dexamethasone (0.1 and 1 microM; 2-8 h) reduced vasoconstriction to EFS (200 mA, 0.3 ms, 1-16 Hz), in a dose- and time-dependent manner only in SHRs. However, the EFS-induced release of [(3)H]NA was increased in SHR arteries preincubated with dexamethasone (1 microM; 6 h). The thromboxane A(2) (TxA(2)) synthase inhibitor furegrelate (10 microM), the selective COX-2 inhibitor NS-398 (N-[2-cyclohexyloxy-4-nitrophenyl] methanesulfonamide; 10 microM), or the TxA(2) receptor antagonist SQ 29548 (1 microM), reduced EFS and NA induced vasoconstrictor responses. However, the effect of these drugs was abolished in arteries preincubated with dexamethasone. Both dexamethasone and phentolamine (1 microM) inhibited the increased thromboxane B(2) levels observed after EFS. COX-2 protein expression was reduced by dexamethasone in SHR arteries. Results suggest that dexamethasone reduces vasoconstriction to EFS in MAs from SHRs by decreasing COX-2 expression, thereby decreasing the smooth muscle TXA(2) release induced by alpha-adrenoceptor activation. The undetectable COX-2 expression in MAs from normotensive animals explains the noneffect of dexamethasone in their arteries.


Subject(s)
Cyclooxygenase 2/drug effects , Dexamethasone/pharmacology , Hypertension/physiopathology , Mesenteric Arteries/drug effects , Thromboxane A2/metabolism , Vasoconstriction/drug effects , Animals , Dose-Response Relationship, Drug , Electric Stimulation , Mesenteric Arteries/physiopathology , Muscle, Smooth, Vascular/metabolism , Phentolamine/pharmacology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Thromboxane B2
19.
Cardiovasc Res ; 75(2): 398-407, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17412316

ABSTRACT

OBJECTIVE: Experimental studies and opinion articles emphasize that cardiovascular alterations associated with ageing can be improved by the long-term use of fenofibrates. We analyzed the effect of fenofibrate treatment on the acetylcholine-induced relaxation in rat aorta and the participation of nitric oxide (NO) and cyclooxygenase (COX)-derived factors in this effect. METHODS: Acetylcholine relaxation in untreated and 6-week fenofibrate-treated Wistar rats was analyzed in the absence and presence of the NO synthase (NOS) inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME), the specific inducible NO (iNOS) synthase inhibitor 1400W, the nonspecific COX inhibitor indomethacin, the specific COX-2 inhibitor NS-398, the specific thromboxane receptor antagonist SQ-29548, the thromboxane synthesis inhibitor furegrelate, the prostacyclin synthesis inhibitor tranylcypromine, or the 20-HETES synthesis inhibitor formamidine. eNOS, iNOS, COX-1, and COX-2 expression was studied by Western blotting. In addition, production of prostaglandin F(2alpha) (PGF(2alpha)), thromboxane A(2) (TxA(2)), prostaglandin E(2) (PGE(2)), isoprostanes, and prostacyclin (PGI(2)) was also measured. RESULTS: Fenofibrate treatment reduced acetylcholine relaxation. Indomethacin, NS-398, and tranylcypromine decreased acetylcholine relaxation in untreated rats but enhanced relaxation in treated rats. SQ-29548 increased acetylcholine responses in segments from treated rats but not in segments from untreated rats. L-NAME decreased vasodilator response to acetylcholine in both groups while furegrelate, NS-398, 1400W, and formamidine did not affect acetylcholine responses in either group. eNOS and COX-2 expression was higher in aorta from treated rats while COX-1 and iNOS remained unmodified. Basal and acetylcholine-stimulated NO and PGE(2) release were increased, and that of PGI(2) decreased in treated rats. TxA(2) release was similar, but PGF(2alpha) release was undetectable in both groups. CONCLUSIONS: Although it increases NO production through increases in eNOS expression, fenofibrate treatment induces endothelial dysfunction. This effect seems to be mediated by decreased PGI(2) and increased PGE(2) release, and it may help to explain the rise in thromboembolic events observed after long-term fenofibrate treatment in humans.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/metabolism , Fenofibrate/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Acetylcholine/pharmacology , Animals , Blotting, Western/methods , Cyclooxygenase 1/analysis , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/analysis , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/analysis , Dinoprostone/metabolism , Endothelium, Vascular/drug effects , In Vitro Techniques , Indomethacin/pharmacology , Male , Nitric Oxide/analysis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type III/analysis , Nitric Oxide Synthase Type III/metabolism , Nitrobenzenes/pharmacology , Prostaglandins F/analysis , Prostaglandins F/metabolism , Rats , Rats, Wistar , Sulfonamides/pharmacology , Time Factors , Vasodilation/drug effects , Vasodilator Agents/pharmacology
20.
Life Sci ; 79(16): 1537-45, 2006 Sep 13.
Article in English | MEDLINE | ID: mdl-16716361

ABSTRACT

Chronic ouabain treatment produces hypertension acting on the central nervous system and at vascular levels. However, cardiac effects in this model of hypertension are still poorly understood. Hence, the effects of hypertension induced by chronic ouabain administration ( approximately 8 microg day(-1), s.c.) for 5 weeks on the cardiac function were studied in Wistar rats. Ouabain induces hypertension but not myocardial hypertrophy. Awake ouabain-treated rats present an increment of the left ventricular systolic pressure and of the maximum positive and negative dP/dt. Isolated papillary muscles from ouabain-treated rats present an increment in isometric force, and this effect was present even when inotropic interventions (external Ca(2+) increment and increased heart rate) were performed. However, the sarcoplasmic reticulum activity and the SERCA-2 protein expression did not change. On the other hand, the activity of myosin ATPase increased without changes in myosin heavy chain protein expression. In addition, the expression of alpha(1) and alpha(2) isoforms of Na(+), K(+)-ATPase also increased in the left ventricle from ouabain-hypertensive rats. The present results showed positive inotropic and lusitropic effects in hearts from awake ouabain-treated rats, which are associated with an increment of the isometric force development and of the activity of myosin ATPase and expression of catalytic subunits of the Na(+), K(+)-ATPase.


Subject(s)
Cardiotonic Agents/toxicity , Hypertension/chemically induced , Myocardial Contraction/drug effects , Ouabain/toxicity , Ventricular Function, Left/drug effects , Adaptation, Physiological , Animals , Blood Pressure/drug effects , Calcium-Transporting ATPases/metabolism , Heart Ventricles/metabolism , Hypertension/enzymology , In Vitro Techniques , Male , Myosin Heavy Chains/metabolism , Myosins/metabolism , Papillary Muscles/drug effects , Papillary Muscles/physiology , Protein Isoforms/metabolism , Rats , Rats, Wistar , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Potassium-Exchanging ATPase/metabolism , Ventricular Function
SELECTION OF CITATIONS
SEARCH DETAIL
...