1
|
Xavier FE. Nitrergic perivascular innervation in health and diseases: Focus on vascular tone regulation. Acta Physiol (Oxf) 2020; 230:e13484. [PMID: 32336027 DOI: 10.1111/apha.13484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
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.
Collapse
Affiliation(s)
- Fabiano E. Xavier
- Departamento de Fisiologia e Farmacologia Centro de Biociências Universidade Federal de Pernambuco Recife Brazil
| |
Collapse
|
2
|
Role of Nitric Oxide in the Cardiovascular and Renal Systems. Int J Mol Sci 2018; 19:ijms19092605. [PMID: 30177600 PMCID: PMC6164974 DOI: 10.3390/ijms19092605] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
The gasotransmitters are a family of gaseous signaling molecules which are produced endogenously and act at specific receptors to play imperative roles in physiologic and pathophysiologic processes. As a well-known gasotransmitter along with hydrogen sulfide and carbon monoxide, nitric oxide (NO) has earned repute as a potent vasodilator also known as endothelium-derived vasorelaxant factor (EDRF). NO has been studied in greater detail, from its synthesis and mechanism of action to its physiologic, pathologic, and pharmacologic roles in different disease states. Different animal models have been applied to investigate the beneficial effects of NO as an antihypertensive, renoprotective, and antihypertrophic agent. NO and its interaction with different systems like the renin–angiotensin system, sympathetic nervous system, and other gaseous transmitters like hydrogen sulfide are also well studied. However, links that appear to exist between the endocannabinoid (EC) and NO systems remain to be fully explored. Experimental approaches using modulators of its synthesis including substrate, donors, and inhibitors of the synthesis of NO will be useful for establishing the relationship between the NO and EC systems in the cardiovascular and renal systems. Being a potent vasodilator, NO may be unique among therapeutic options for management of hypertension and resulting renal disease and left ventricular hypertrophy. Inclusion of NO modulators in clinical practice may be useful not only as curatives for particular diseases but also for arresting disease prognoses through its interactions with other systems.
Collapse
|
3
|
Effects of endogenous nitric oxide on adrenergic nerve-mediated vasoconstriction and calcitonin gene-related peptide-containing nerve-mediated vasodilation in pithed rats. Eur J Pharmacol 2017; 802:69-75. [DOI: 10.1016/j.ejphar.2017.02.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 01/20/2023]
|
4
|
Fujiwara H, Hashikawa-Hobara N, Wake Y, Takatori S, Goda M, Higuchi H, Zamami Y, Tangsucharit P, Kawasaki H. Neurogenic vascular responses in male mouse mesenteric vascular beds. J Pharmacol Sci 2012; 119:260-70. [PMID: 22785022 DOI: 10.1254/jphs.12014fp] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rat mesenteric arteries were maintained by both adrenergic vasoconstrictor nerves and calcitonin gene-related peptide (CGRP) vasodilator nerves. However, functions of these nerves in a pathophysiological state have not fully been analyzed. The use of disease models developed genetically in mice is expected to clarify neural function of perivascular nerves. Thus, we investigated basic mouse vascular responses. Mesenteric vascular beds isolated from male C57BL/6 mouse were perfused with Krebs solution and perfusion pressure was measured. Periarterial nerve stimulation (PNS, 8 - 24 Hz) induced frequency-dependent vasoconstriction, which increased flow rate-dependently. PNS-induced vasoconstriction was abolished by tetrodotoxin (neurotoxin) and guanethidine (adrenergic neuron blocker) and blunted by prazosin (α(1)-adrenoceptor antagonist). Injection of norepinephrine caused vasoconstriction, which was abolished by prazosin. In preparations with active tone, PNS (1 - 8 Hz) induced frequency-dependent vasodilation, which was inhibited by tetrodotoxin, capsaicin (CGRP depletor), and CGRP8-37 (CGRP-receptor antagonist). Injections of CGRP, acetylcholine, and sodium nitroprusside induced vasodilations. Vasodilator response to CGRP was inhibited by CGRP8-37. Immunohistochemical study showed innervation of tyrosine hydroxylase- and CGRP-immunopositive fibers in mesenteric arteries and veins. These results suggest that male mouse mesenteric vascular beds are useful for studying neural regulation of mesenteric arteries, which are innervated by adrenergic and CGRPergic nerves regulating vascular tone.
Collapse
Affiliation(s)
- Hiroki Fujiwara
- Department of Clinical Pharmaceutical Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Okayama 700-8530, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Chan NYK, Seyedi N, Takano K, Levi R. An unsuspected property of natriuretic peptides: promotion of calcium-dependent catecholamine release via protein kinase G-mediated phosphodiesterase type 3 inhibition. Circulation 2011; 125:298-307. [PMID: 22158783 DOI: 10.1161/circulationaha.111.059097] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Although natriuretic peptides are considered cardioprotective, clinical heart failure trials with recombinant brain natriuretic peptide (nesiritide) failed to prove it. Unsuspected proadrenergic effects might oppose the anticipated benefits of natriuretic peptides. METHODS AND RESULTS We investigated whether natriuretic peptides induce catecholamine release in isolated hearts, sympathetic nerve endings (cardiac synaptosomes), and PC12 cells bearing a sympathetic neuron phenotype. Perfusion of isolated guinea pig hearts with brain natriuretic peptide elicited a 3-fold increase in norepinephrine release, which doubled in ischemia/reperfusion conditions. Brain natriuretic peptide and atrial natriuretic peptide also released norepinephrine from cardiac synaptosomes and dopamine from nerve growth factor-differentiated PC12 cells in a concentration-dependent manner. These catecholamine-releasing effects were associated with an increase in intracellular calcium and abolished by blockade of calcium channels and calcium transients, demonstrating a calcium-dependent exocytotic process. Activation of the guanylyl cyclase-cyclic GMP-protein-kinase-G system with nitroprusside or membrane-permeant cyclic GMP analogs mimicked the proexocytotic effect of natriuretic peptides, an action associated with an increase in intracellular cyclic AMP (cAMP) and protein-kinase-A activity. Cyclic AMP enhancement resulted from an inhibition of phosphodiesterase type 3-induced cAMP hydrolysis. Collectively, these findings indicate that, by inhibiting phosphodiesterase type 3, natriuretic peptides sequentially enhance intracellular cAMP levels, protein kinase A activity, intracellular calcium, and catecholamine exocytosis. CONCLUSIONS Our results show that natriuretic peptides, at concentrations likely to be reached at cardiac sympathetic nerve endings in advanced congestive heart failure, promote norepinephrine release via a protein kinase G-induced inhibition of phosphodiesterase type 3-mediated cAMP hydrolysis. We propose that this proadrenergic action may counteract the beneficial cardiac and hemodynamic effects of natriuretic peptides and thus explain the ineffectiveness of nesiritide as a cardiac failure medication.
Collapse
Affiliation(s)
- Noel Yan-Ki Chan
- Dept of Pharmacology, Weill Cornell Medical College, 1300 York Ave, New York, NY 10065-4896, USA
| | | | | | | |
Collapse
|
6
|
Macarthur H, Wilken GH, Westfall TC, Kolo LL. Neuronal and non-neuronal modulation of sympathetic neurovascular transmission. Acta Physiol (Oxf) 2011; 203:37-45. [PMID: 21362154 PMCID: PMC3139802 DOI: 10.1111/j.1748-1716.2010.02242.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Noradrenaline, neuropeptide Y and adenosine triphosphate are co-stored in, and co-released from, sympathetic nerves. Each transmitter modulates its own release as well as the release of one another; thus, anything affecting the release of one of these transmitters has consequences for all. Neurotransmission at the sympathetic neurovascular junction is also modulated by non-sympathetic mediators such as angiotensin II, serotonin, histamine, endothelin and prostaglandins through the activation of specific pre-junctional receptors. In addition, nitric oxide (NO) has been identified as a modulator of sympathetic neuronal activity, both as a physiological antagonist against the vasoconstrictor actions of the sympathetic neurotransmitters, and also by directly affecting transmitter release. Here, we review the modulation of sympathetic neurovascular transmission by neuronal and non-neuronal mediators with an emphasis on the actions of NO. The consequences for co-transmission are also discussed, particularly in light of hypertensive states where NO availability is diminished.
Collapse
Affiliation(s)
- H Macarthur
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, MO 63104, USA.
| | | | | | | |
Collapse
|
7
|
Altered function of nitrergic nerves inhibiting sympathetic neurotransmission in mesenteric vascular beds of renovascular hypertensive rats. Hypertens Res 2010; 33:485-91. [PMID: 20379183 DOI: 10.1038/hr.2010.48] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neuronal nitric oxide (NO) has been shown to modulate perivascular adrenergic neurotransmission by inhibiting noradrenaline release from terminals in rat mesenteric arteries. This study was conducted to investigate changes in the inhibitory function of NO-containing nerves (nitrergic nerves) in mesenteric vascular beds of 2-kidney, 1-clip renovascular hypertensive rats (2K1C-RHR). Rat mesenteric vascular beds without endothelium were perfused with Krebs solution and the perfusion pressure was measured. In preparations from sham-operated rats (control) and 2K1C-RHRs, vasoconstriction induced by periarterial nerve stimulation (PNS; 2-8 Hz), but not vasoconstriction induced by exogenously injected noradrenaline (0.5, 1.0 nmol), was markedly facilitated in the presence of a nonselective NO synthase (NOS) inhibitor, N-omega-nitro-L-arginine methyl ester (L-NAME) (100 microM). The facilitatory effect of L-NAME in preparations from 2K1C-RHR was smaller than that in control preparations. L-NAME augmented PNS-evoked noradrenaline release, which was smaller in 2K1C-RHRs than in controls. The expression of neuronal NO synthase (nNOS) measured by western blotting in mesenteric arteries from 2K1C-RHRs was significantly decreased compared with control arteries. Immunohistochemical staining of mesenteric arteries showed dense innervation of nNOS-immunopositive nerves that was significantly smaller in arteries from 2K1C-RHR than that in control arteries. Mesenteric arteries were densely innervated by tyrosine hydroxylase-immunopositive nerves, which coalesced with nNOS-immunopositive nerves. These results suggest that the inhibitory function of nitrergic nerves in adrenergic neurotransmission is significantly decreased in 2K1C-RHRs. This functional alteration based on the decrease in nNOS expression and nitrergic innervation leads to enhanced adrenergic neurotransmission and contributes to the initiation and development of renovascular hypertension.
Collapse
|
8
|
Macarthur H, Westfall TC, Wilken GH. Oxidative stress attenuates NO-induced modulation of sympathetic neurotransmission in the mesenteric arterial bed of spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 2008; 294:H183-9. [DOI: 10.1152/ajpheart.01040.2007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Current evidence suggests that hyperactivity of the sympathetic nervous system and endothelial dysfunction are important factors in the development and maintenance of hypertension. Under normal conditions the endothelial mediator nitric oxide (NO) negatively modulates the activity of the norepinephrine portion of sympathetic neurotransmission, thereby placing a “brake” on the vasoconstrictor ability of this transmitter. This property of NO is diminished in the isolated, perfused mesenteric arterial bed taken from the spontaneously hypertensive rat (SHR), resulting in greater nerve-stimulated norepinephrine and lower neuropeptide Y (NPY) overflow from this mesenteric preparation compared with that of the normotensive Wistar-Kyoto rat (WKY). We hypothesized that increased oxidative stress in the SHR contributes to the dysfunction in the NO modulation of sympathetic neurotransmission. Here we demonstrate that the antioxidant N-acetylcysteine reduced nerve-stimulated norepinephrine and increased NPY overflow in the mesenteric arterial bed taken from the SHR. Furthermore, this property of N-acetylcysteine was prevented by inhibiting nitric oxide synthase with Nω-nitro-l-arginine methyl ester, demonstrating that the effect of N-acetylcysteine was due to the preservation of NO from oxidation. Despite a reduction in norepinephrine overflow, the nerve-stimulated perfusion pressure response in the SHR mesenteric bed was not altered by the inclusion of N-acetylcysteine. Studies including the Y1 antagonist BIBO 3304 with N-acetylcysteine demonstrated that this preservation of the perfusion pressure response was due to elevated NPY overflow. These results demonstrate that the reduction in the bioavailability of NO as a result of elevated oxidative stress contributes to the increase in norepinephrine overflow from the SHR mesenteric sympathetic neuroeffector junction.
Collapse
|
9
|
Whalen EJ, Saurer TB, Johnson AK, Lewis SJ. Intracellular cGMP may promote Ca2+-dependent and Ca2+-independent release of catecholamines from sympathetic nerve terminals. Vascul Pharmacol 2006; 45:102-11. [PMID: 16697265 DOI: 10.1016/j.vph.2006.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2005] [Revised: 03/06/2006] [Accepted: 03/20/2006] [Indexed: 12/31/2022]
Abstract
OBJECTIVE This study examined the hypothesis that intracellular cGMP stimulates the release of catecholamines from sympathetic nerve terminals (SNTs) in conscious rats. METHODS Conscious rats were prepared to determine the effects of intravenously-administered agents on heart rate (HR) and mean arterial blood pressure (MAP). RESULTS Bolus intravenous injections of the membrane-permeable cGMP analogue, 8-(4-chlorophenylthio)-cGMP (8-CPT-cGMP), elicited immediate and pronounced increases in HR before any changes in MAP were observed. In contrast, injections of cGMP did not elicit changes in HR or MAP. The 8-CPT-cGMP-induced tachycardia was markedly diminished by (1) the beta(1,2)-adrenoceptor antagonist, propranolol, (2) the ganglion blocking agent, chlorisondamine, and (3) bretylium, which blocks Ca2+-dependent mobilization of vesicular stores of catecholamines from SNTs. 8-CPT-cGMP also elicited minor falls in MAP in propranolol-treated rats but elicited pronounced falls in MAP in rats treated with chlorisondamine, bretylium, or combined administration of bretylium and the muscarinic receptor antagonist, methyl-atropine. CONCLUSIONS These findings suggest that (1) intracellular cGMP elicits the release of Ca2+-sensitive and Ca2+-insensitive stores of catecholamines from SNTs in conscious rats, and (2) cGMP-mediated release of catecholamines from SNTs antagonizes cGMP-mediated relaxation of vascular smooth muscle in resistance arteries. Taken together, these findings support the concept that increases in intracellular cGMP levels by atrial natriuretic peptide and endothelium- and cardiac-derived nitric oxide regulate sympathetic control of the heart and the microvasculature of conscious rats via cGMP-dependent release of catecholamines.
Collapse
Affiliation(s)
- Erin J Whalen
- Department of Psychology, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | |
Collapse
|
10
|
Bagnall NM, Dent PC, Walkowska A, Sadowski J, Johns EJ. Nitric oxide inhibition and the impact on renal nerve-mediated antinatriuresis and antidiuresis in the anaesthetized rat. J Physiol 2005; 569:849-56. [PMID: 16239274 PMCID: PMC1464264 DOI: 10.1113/jphysiol.2005.097709] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/29/2005] [Accepted: 10/18/2005] [Indexed: 12/26/2022] Open
Abstract
The contribution of nitric oxide (NO) to the antinatriuresis and antidiuresis caused by low-level electrical stimulation of the renal sympathetic nerves (RNS) was investigated in rats anaesthetized with chloralose-urethane. Groups of rats, n= 6, were given i.v. infusions of vehicle, l-NAME (10 microg kg(-1) min(-1)), 1400W (20 microg kg(-1) min(-1)), or S-methyl-thiocitrulline (SMTC) (20 microg kg(-1) min(-1)) to inhibit NO synthesis non-selectively or selectively to block the inducible or neuronal NOS isoforms (iNOS and nNOS, respectively). Following baseline measurements of blood pressure (BP), renal blood flow (RBF), glomerular filtration rate (GFR), urine flow (UV) and sodium excretion (U(Na)V), RNS was performed at 15 V, 2 ms duration with a frequency between 0.5 and 1.0 Hz. RNS did not cause measurable changes in BP, RBF or GFR in any of the groups. In untreated rats, RNS decreased UV and U(Na)V by 40-50% (both P < 0.01), but these excretory responses were prevented in l-NAME-treated rats. In the presence of 1400W i.v., RNS caused reversible reductions in both UV and U(Na)V of 40-50% (both P < 0.01), while in SMTC-treated rats, RNS caused an inconsistent fall in UV, but a significant reduction (P < 0.05) in U(Na)V of 21%. These data demonstrated that the renal nerve-mediated antinatriuresis and antidiuresis was dependent on the presence of NO, generated in part by nNOS. The findings suggest that NO importantly modulates the neural control of fluid reabsorption; the control may be facilitatory at a presynaptic level but inhibitory on tubular reabsorptive processes.
Collapse
Affiliation(s)
- N M Bagnall
- Department of Physiology, Aras Windle, University College Cork, Cork, Republic of Ireland
| | | | | | | | | |
Collapse
|
11
|
Lindqvist M, Melcher A, Hjemdahl P. Hemodynamic and sympathoadrenal responses to mental stress during nitric oxide synthesis inhibition. Am J Physiol Heart Circ Physiol 2004; 287:H2309-15. [PMID: 15256378 DOI: 10.1152/ajpheart.01216.2003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiovascular and sympathoadrenal responses to a reproducible mental stress test were investigated in eight healthy young men before and during intravenous infusion of the nitric oxide (NO) synthesis inhibitor N-monomethyl-L-arginine (L-NMMA). Before L-NMMA, stress responses included significant increases in heart rate, mean arterial pressure, and cardiac output (CO) and decreases in systemic and forearm vascular resistance. Arterial plasma norepinephrine (NE) increased. At rest after 30 min of infusion of L-NMMA (0.3 mg.kg(-1).min(-1) iv), mean arterial pressure increased from 98 +/- 4 to 108 +/- 3 mmHg (P <0.001) because of an increase in systemic vascular resistance from 12.9 +/- 0.5 to 18.5 +/- 0.9 units (P <0.001). CO decreased from 7.7 +/- 0.4 to 5.9 +/- 0.3 l/min (P <0.01). Arterial plasma NE decreased from 2.08 +/- 0.16 to 1.47 +/- 0.14 nmol/l. Repeated mental stress during continued infusion of L-NMMA (0.15 mg.kg(-1).min(-1)) induced qualitatively similar cardiovascular responses, but there was a marked attenuation of the increase in mean arterial blood pressure, resulting in similar "steady-state" blood pressures during mental stress without and with NO blockade. Increases in heart rate and CO were attenuated, but stress-induced decreases in systemic and forearm vascular resistance were essentially unchanged. Arterial plasma NE increased less than during the first stress test. Thus the increased arterial tone at rest during L-NMMA infusion is compensated for by attenuated increases in blood pressure during mental stress, mainly through a markedly attenuated CO response and suppressed sympathetic nerve activity.
Collapse
Affiliation(s)
- Madeleine Lindqvist
- Dept. of Clinical Physiology, Danderyd Hospital, SE-182 88 Stockholm, Sweden.
| | | | | |
Collapse
|
12
|
Kolo LL, Westfall TC, Macarthur H. Nitric oxide decreases the biological activity of norepinephrine resulting in altered vascular tone in the rat mesenteric arterial bed. Am J Physiol Heart Circ Physiol 2004; 286:H296-303. [PMID: 14684362 DOI: 10.1152/ajpheart.00668.2003] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) reacts with catecholamines resulting in their deactivation. In this study, we demonstrated that coincubation of NO donors with sympathetic neurotransmitters decreased the amount of norepinephrine detected but not ATP or neuropeptide Y (NPY). Furthermore, we found that the ability of norepinephrine to increase perfusion pressure in the isolated perfused mesenteric arterial bed of the rat was attenuated by the incubation of norepinephrine with the NO donor diethylamine NONOate. Conversely, the vasoconstrictive ability of NPY and ATP was unaffected by incubation with NONOate. Periarterial nerve stimulation in the presence of the NO synthase (NOS) inhibitor Nω-nitro-l-arginine methyl ester (l-NAME) resulted in an increase in both perfusion pressure response and norepinephrine levels. This was prevented by l-arginine, demonstrating that the effects of l-NAME were indeed specific to the inhibition of NOS. To confirm that NO was not altering the release of norepinephrine from the sympathetic nerve via presynaptic activation of guanylate cyclase, we repeated the experiments in the presence of the guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]-quinoxaloine-one (ODQ). Unlike l-NAME, ODQ infusion did not increase norepinephrine overflow, demonstrating that modulation of norepinephrine by NO at the vascular neuroeffector junction of the rat mesenteric vascular bed is not the result of presynaptic guanylate cyclase activation. These results demonstrate that, in addition to being a direct vasodilatator, NO can also alter vascular reactivity at the sympathetic neuroeffector junction in the rat mesenteric bed by deactivating the vasoconstrictor norepinephrine.
Collapse
Affiliation(s)
- Lacy L Kolo
- Department of Pharmacological and Physiological Science, Saint Louis University School of Medicine, 1402 South Grand Blvd., St. Louis, MO 63104, USA
| | | | | |
Collapse
|
13
|
Peredo HA. Opposite effects of endogenous nitric oxide and prostaglandin F2alpha in the rat mesenteric bed. ACTA ACUST UNITED AC 2003; 23:167-72. [PMID: 14690491 DOI: 10.1046/j.1474-8673.2003.00292.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
1. The relationship between the effects of endogenous nitric oxide (NO) and prostanoids on the noradrenaline (NA)-induced contractions and the mechanisms involved were investigated in the rat perfused mesenteric bed, using NG-nitro-L-arginine methyl ester (l-NAME), a NO synthase inhibitor and sodium nitroprusside (SNP), a NO donor. 2. The constrictor responses to NA were reduced to 50% by the cyclooxygenase inhibitor 10 microm indomethacin as well as by 1 microM SNP. When indomethacin and SNP were perfused simultaneously the contractions were further reduced. 3. The NA-induced contractions were increased by the addition of 400 microM L-NAME and the addition of either indomethacin or SNP abolished such increases. The simultaneous perfusion of both agents further reduced the contractions. 4. Removal of the endothelium increased NA-induced contractions to a similar extent as L-NAME and this increase was abolished by indomethacin as well as by SNP. 5. The perfusion of 10 microM NA augmented the release of prostaglandin (PG) F2 alpha by the mesenteric bed without modifications in any other prostanoid. In the presence of L-NAME, this effect was further increased. However, SNP abolished the NA-induced stimulation of PGF2 alpha release. 6. In de-endothelialized preparations NA also stimulated PGF2 alpha production as observed in intact preparations. This effect was more marked in the presence of L-NAME; in contrast, SNP abolished the stimulation. 7. In conclusion, the present results suggest an opposite action between NO and PGF2 alpha on the NA-induced contractions in the rat mesenteric bed.
Collapse
Affiliation(s)
- H A Peredo
- Instituto de Investigaciones Farmacológicas, CONICET, Junín 956-number 5 piso, 1113 Buenos Aires, Argentina
| |
Collapse
|
14
|
Tanioka H, Nakamura K, Fujimura S, Yoshida M, Suzuki-Kusaba M, Hisa H, Satoh S. Facilitatory role of NO in neural norepinephrine release in the rat kidney. Am J Physiol Regul Integr Comp Physiol 2002; 282:R1436-42. [PMID: 11959687 DOI: 10.1152/ajpregu.00697.2001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We examined modulation by nitric oxide (NO) of sympathetic neurotransmitter release and vasoconstriction in the isolated pump-perfused rat kidney. Electrical renal nerve stimulation (RNS; 1 and 2 Hz) increased renal perfusion pressure and renal norepinephrine (NE) efflux. Nonselective NO synthase (NOS) inhibitors [N(omega)-nitro-L-arginine methyl ester (L-NAME) or N(omega)-nitro-L-arginine], but not a selective neuronal NO synthase inhibitor (7-nitroindazole sodium salt), suppressed the NE efflux response and enhanced the perfusion pressure response. Pretreatment with L-arginine prevented the effects of L-NAME on the RNS-induced responses. 2-(4-Carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (carboxy-PTIO), which eliminates NO by oxidizing it to NO(2), suppressed the NE efflux response, whereas the perfusion pressure response was less susceptible to carboxy-PTIO. 8-Bromoguanosine cGMP suppressed and a guanylate cyclase inhibitor [4H-8-bromo-1,2,4-oxadiazolo(3,4-d)benz(b)(1,4)oxazin-1-one] enhanced the RNS-induced perfusion pressure response, but neither of these drugs affected the NE efflux response. These results suggest that endogenous NO facilitates the NE release through cGMP-independent mechanisms, NO metabolites formed after NO(2) rather than NO itself counteract the vasoconstriction, and neuronal NOS does not contribute to these modulatory mechanisms in the sympathetic nervous system of the rat kidney.
Collapse
Affiliation(s)
- Hideki Tanioka
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Rabelo FA, Russo EM, Salgado MC, Coelho EB. Nonendothelial NO blunts sympathetic response of normotensive rats but not of SHR. Hypertension 2001; 38:565-8. [PMID: 11566932 DOI: 10.1161/01.hyp.38.3.565] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The inhibitory role of NO on sympathetic-induced contraction of resistance vessels of spontaneously hypertensive rats (SHR) has not been defined. Accordingly, we investigated the effect of endothelial removal or NO synthase inhibition on vasoconstrictor responses to sympathetic stimulation or phenylephrine in perfused mesenteric beds isolated from normotensive rats (NR) and SHR. Electrical stimulation (10 to 64 Hz) of perivascular nerves elicited a frequency-dependent increase in perfusion pressure that was greater in preparations from SHR (maximal effect: 223.4+/-8.4 versus 117.6+/-10.3 mm Hg in NR, n=6, P<0.001), and endothelium removal did not affect these responses in arteries from NR but caused a significant shift to the left of the frequency-response curve in arteries from SHR. In arteries with endothelium, inhibition of NO synthase with N(G)-nitro-L-arginine (L-NNA, 50 micromol/L) augmented the vasoconstrictor responses to sympathetic stimulation in both NR and SHR preparations. In preparations that had the endothelium removed, however, L-NNA potentiated only the responses to sympathetic stimulation of NR arteries. Vasoconstrictor responses to phenylephrine was potentiated by endothelium removal and in the presence of L-NNA only when the endothelium was intact in both NR and SHR arteries. The number of NADPH-diaphorase-positive cells in the superior mesenteric sympathetic ganglion of SHR was significantly less compared with that of NR. In conclusion, these data suggest a prejunctional inhibitory action of non-endothelial-derived NO, most probably neuronal-derived NO, on sympathetic-mediated vasoconstriction in NR arteries. In contrast, enhancement of the sympathetic-mediated vasoconstriction in SHR arteries elicited by L-NNA can be attributed to inhibition of endothelial-derived NO.
Collapse
Affiliation(s)
- F A Rabelo
- Department of Internal Medicine, Renal Division, Faculty of Medicine, Ribeirão Preto, University of São Paulo, Brazil
| | | | | | | |
Collapse
|
16
|
Ohlmann P, Martínez MC, Bucher B, Andriantsitohaina R, Muller B, Schneider F, Stoclet JC. Effect of lipopolysaccharide treatment on neurogenic contraction and noradrenaline release in rat arteries. Fundam Clin Pharmacol 2000; 14:593-600. [PMID: 11206710 DOI: 10.1111/j.1472-8206.2000.tb00445.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In the present study, contractile responses and [3H]-noradrenaline overflow evoked by electrical field stimulation were assessed, respectively, in the small mesenteric artery and in tail artery removed from rats pre-treated with either saline or lipopolysaccharide (LPS). In small mesenteric arteries, LPS treatment did not significantly modify the contractile responses elicited by electrical stimulation, in the absence or in the presence of L-arginine. However, in arteries removed from rats treated with LPS, L-arginine addition produced relaxation of vessels pre-contracted with noradrenaline. The amplification of neurogenic contraction by the nitric oxide (NO) synthase inhibitor Nomega-nitro-L-arginine methyl ester (L-NAME) was similar in arteries removed from saline and LPS-infused rats. In mesenteric arteries, LPS treatment suppressed the potentiation of the neurogenic responses by the alpha2-adrenoceptor antagonist, yohimbine and by the inhibitor of neuronal uptake of noradrenaline, cocaine. In rat tail artery exposed to L-arginine, LPS treatment produced an increase in [3H]-noradrenaline overflow evoked by electrical stimulation. Altogether, these data suggest that an enhanced noradrenaline release from sympathetic nerves, probably resulting from inhibition of the modulatory effect of both prejunctional alpha2-adrenoceptors and neuronal uptake mechanism, may play a role in the preservation of neurogenic response after LPS treatment despite evidence of the induction of NO synthase.
Collapse
Affiliation(s)
- P Ohlmann
- Pharmacologie et physico-chimie des interactions cellulaires et moléculaires, UMR CNRS 7034, Faculté de pharmacie, Université Louis Pasteur, Illkirch, France
| | | | | | | | | | | | | |
Collapse
|
17
|
Skarzynski DJ, Okuda K. Different actions of noradrenaline and nitric oxide on the output of prostaglandins and progesterone in cultured bovine luteal cells. Prostaglandins Other Lipid Mediat 2000; 60:35-47. [PMID: 10680774 DOI: 10.1016/s0090-6980(99)00046-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The effects of noradrenaline (NA) and nitric oxide (NO) on prostaglandins (PGs) and progesterone (P4) secretion during the development of the bovine corpus luteum (CL) were investigated. Bovine luteal cells of early and mid-cycle CL were cultured for 20 to 24 h in medium containing 10% calf serum, washed, and treated with NA or nitrergic agents for an additional 16 h in a serum-free medium. NA (10(-5) M) stimulated P4 from early and mid-cycle CL by 238% and 154% (P < 0.01), respectively. Moreover, although NA induced a twofold increase in PGE2 secretion (P < 0.01) in both examined periods, the effect of NA on PGF2alpha secretion was approximately 1.5 times higher (P < 0.05) in early than in mid-cycle CL. Two NO synthase inhibitors, L-NAME and L-NOARG (both 10(-4) M), stimulated P4 secretion only in mid-luteal cells (P < 0.01), although they did not affect the cells from early CL. Although a NO donor, S-NAP (10(-4) M) inhibited P4 secretion from mid-cycle luteal cells (P < 0.05), it strongly stimulated PGE2 in both examined phases (P < 0.001). On the other hand, the output of PGF2alpha was stimulated by S-NAP only in the cells of the mid-cycle CL (P < 0.01). The overall results suggest that adrenergic and nitrergic agents play opposite roles in the regulation of bovine CL functions. Whereas NA may play a supporting role in luteal development, NO may participate in the functional regression of the bovine CL by inhibiting steroidogenesis.
Collapse
Affiliation(s)
- D J Skarzynski
- Laboratory of Reproductive Endocrinology, Faculty of Agriculture, Okayama University, Japan
| | | |
Collapse
|
18
|
Qu XW, Rozenfeld RA, Huang W, Sun X, Tan XD, Hsueh W. Roles of nitric oxide synthases in platelet-activating factor-induced intestinal necrosis in rats. Crit Care Med 1999; 27:356-64. [PMID: 10075061 DOI: 10.1097/00003246-199902000-00043] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To examine the role of constitutive and inducible nitric oxide synthases (cNOS and iNOS) in platelet-activating factor (PAF)-induced shock and intestinal injury. DESIGN Prospective, randomized, controlled experimental study. SETTING Hospital research laboratory. SUBJECTS Young adult male Sprague-Dawley rats were anesthetized and studied. INTERVENTIONS Rats were injected with PAF, either alone or after the following pretreatments: a) selective iNOS inhibitors aminoguanidine or S-methylisothiourea; b) 3-morpholinosydnonimine, a NO donor; c) S-methylisothiourea + 3-morpholinosydnonimine; and d) antineutrophil antibody (to deplete neutrophils). MEASUREMENTS AND MAIN RESULTS Blood pressure, hematocrit, white blood cell counts, intestinal injury, and intestinal cNOS and iNOS activities were assessed. We found that: a) cNOS is the predominant NOS in the intestine and its activity is inversely correlated to the level of tissue injury; b) there is a time-dependent increase in cNOS activity in sham-operated animals, which was abolished by PAF; c) Western blotting and immunohistochemistry showed iNOS present in the normal intestine, localizing mainly in crypt cells; d) iNOS inhibitors attenuated PAF-induced injury in animals with high cNOS activity, but had no protective effect in animals with low cNOS activity; e) 3-morpholinosydnonimine, alone or together with S-methylisothiourea, alleviated PAF-induced injury; and f) neutrophil depletion blocked the suppressive effect of PAF on cNOS and prevented injury. CONCLUSIONS We conclude that cNOS and iNOS play different roles in PAF-induced intestinal injury. Caution should be exerted concerning potential therapeutic uses of iNOS inhibitors.
Collapse
Affiliation(s)
- X W Qu
- Department of Pathology, Children's Memorial Hospital, Northwestern University Medical School, Chicago, IL 60614, USA
| | | | | | | | | | | |
Collapse
|
19
|
Lindqvist M, Melcher A, Hjemdahl P. Flow dependence of forearm noradrenaline overflow, as assessed during mental stress and sodium nitroprusside infusion. J Hypertens 1999; 17:91-7. [PMID: 10100099 DOI: 10.1097/00004872-199917010-00014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the influence of blood flow on measurements of regional sympathetic nerve activity by radiotracer methodology ([3H]noradrenaline). DESIGN Ten healthy men were studied under two conditions of elevated forearm blood flow: mental stress (Stroop colour word conflict test) and an intra-arterial infusion of sodium nitroprusside. METHODS Arterial blood pressure was measured invasively and forearm blood flow with strain-gauge plethysmography. Arterial and venous plasma adrenaline and noradrenaline were measured with high-performance liquid chromatography, and regional and total noradrenaline spillover were calculated. RESULTS During mental stress, mean arterial pressure increased by 17%, heart rate by 16 beats/min, forearm blood flow by 117%, while forearm vascular resistance decreased by 44% (P < 0.001 for all). Sodium nitroprusside increased forearm blood flow dose-dependently, but elicited only minor effects on systemic haemodynamics. Mental stress increased arterial plasma noradrenaline by 52% (P < 0.001), and total body noradrenaline spillover by 75% (P < 0.001). During sodium nitroprusside infusion, arterial plasma noradrenaline increased only slightly and total body noradrenaline spillover was unaffected Forearm noradrenaline overflow increased from 5.4 +/- 0.9 to 16.9 +/- 2.6 pmol/min per I (P < 0.001) during mental stress and from 6.6 +/- 0.8 to 16.9 +/- 3.7 pmol/min per I (P < 0.001) during the second dose-step of sodium nitroprusside infusion. By intra-individual comparisons of forearm noradrenaline overflow increases during mental stress and during sodium nitroprusside infusion, with similar forearm blood flow increases, the flow dependence of forearm noradrenaline overflow was estimated. During mental stress, about 60% (median value, range 29-112%) of the increase in forearm noradrenaline overflow was attributed to the increase in forearm blood flow, whereas 40% was considered to reflect increased sympathetic nerve activity. CONCLUSIONS There seems to be a considerable flow dependence of the regional overflow of noradrenaline, that is, a component of simple wash-out of noradrenaline from the forearm tissues during vasodilation. However, the present results still indicate that sympathetic nerve activity in the forearm is increased during mental stress, justifying the radiotracer technique for semiquantitative measurements, also during vasodilation.
Collapse
Affiliation(s)
- M Lindqvist
- Division of Clinical Physiology, Karolinska Institutet, Danderyd Hospital, Sweden
| | | | | |
Collapse
|
20
|
Diéguez G, Fernández N, Sánchez MA, García-Villalón AL, Monge L, Gómez B. Adrenergic reactivity after inhibition of nitric oxide synthesis in the cerebral circulation of awake goats. Brain Res 1998; 813:381-9. [PMID: 9838199 DOI: 10.1016/s0006-8993(98)01064-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The interaction between nitric oxide (NO) and adrenergic reactivity in the cerebral circulation was studied using in vivo and in vitro preparations. Blood flow to one brain hemisphere (cerebral blood flow) was electromagnetically measured in conscious goats, and the effects of norepinephrine, tyramine and cervical sympathetic nerve stimulation were recorded before (control) and after inhibition of NO formation with Nw-nitro-l-arginine methyl ester (l-NAME). The responses to norepinephrine, tyramine and electrical field stimulation were also recorded in segments, 4 mm in length, from the goat's middle cerebral artery under control conditions and after l-NAME. In vivo, l-NAME (10 goats, 47 mg kg-1 administered i.v.) reduced resting cerebral blood flow by 37+/-2%, increased mean systemic arterial pressure by 24+/-3%, reduced heart rate by 35+/-2%, and decreased cerebrovascular conductance by 52+/-2% (all P<0.01). Norepinephrine (0.3-9 microgram), tyramine (50-500 microgram), and supramaximal electrical sympathetic cervical nerve stimulation (1. 5-6 Hz) decreased cerebrovascular conductance, and these decreases were significantly higher after l-NAME than under control conditions, remaining higher for about 48 h after this treatment. Norepinephrine (10-8-10-3 M), tyramine (10-6-10-3 M) and electrical field stimulation (1.5-6 Hz) contracted isolated cerebral arteries, and the maximal contraction, but not the sensitivity, was significantly higher in the arteries treated than in non-treated with l-NAME (10-4 M). Therefore, the reactivity of cerebral vasculature to exogenous and endogenous norepinephrine may be increased after inhibition of NO synthesis. This increase might be related, at least in part, to changes at postjunctional level in the adrenergic innervation of the vessel wall, and it might contribute to the observed decreases in resting cerebral blood flow after inhibition of NO synthesis.
Collapse
Affiliation(s)
- G Diéguez
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma, Arzobispo Morcillo 4, 28029, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
21
|
Fan WQ, Smolich JJ, Wild J, Yu VY, Walker AM. Major vasodilator role for nitric oxide in the gastrointestinal circulation of the mid-gestation fetal lamb. Pediatr Res 1998; 44:344-50. [PMID: 9727711 DOI: 10.1203/00006450-199809000-00013] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
As nitric oxide (NO) may be a particularly important vasodilator in early life, we investigated its role in the regulation of the gastrointestinal (GI) circulation at mid-gestation. Cardiac output and GI blood flow were measured by the radioactive microsphere technique in eight chronically instrumented and unanesthetized mid-gestation fetal sheep. Mean arterial pressure (MAP), heart rate, blood flow, oxygen delivery, and vascular resistance were determined before and after infusion of the specific NO synthase inhibitor, Nomega-nitro-L-arginine (L-NNA) at doses of 10 and 25 mg/kg. In response to L-NNA infusion, MAP increased (p < 0.01) and combined ventricular output decreased (p < 0.001). GI blood flow and oxygen delivery decreased and vascular resistance increased in the stomach and all segments of the small and large intestine (all p < 0.001). The greatest reduction in blood flow was in the small intestine (p < 0.01) and the basal differential pattern of small intestinal blood flow exceeding large intestinal flow was completely abolished. These changes were much greater than those previously described in late-gestation fetuses. Our results suggest that, at mid-gestation, NO plays a major role in the regulation of blood flow and vascular tone across all segments of the fetal GI tract, with its effects being more pronounced than later in development.
Collapse
Affiliation(s)
- W Q Fan
- Institute of Reproduction and Development, Monash University, Clayton, Victoria, Australia
| | | | | | | | | |
Collapse
|
22
|
Gottesfeld Z. Sympathetic neural response to immune signals involves nitric oxide: effects of exposure to alcohol in utero. Alcohol 1998; 16:177-81. [PMID: 9665320 DOI: 10.1016/s0741-8329(97)00188-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In response to infection, inflammation, or injury, the neural-immune-endocrine networks are activated to restore or maintain stability in the internal environment. Disruption of any one of the functional components may impair the effectiveness of the immune response to challenges, and may consequently jeopardize the wellness of the host. Studies in the author's laboratory have shown that the normal activation of splenic sympathetic neurons in response to the endotoxin lipopolysaccharide, a tool frequently used to mimic infection or inflammation, does not occur in fetal alcohol-exposed (FAE) rats. The sympathetic innervation of lymphoid organs is considered an important immune modulator. Thus, the anomalous splenic sympathetic response may partly account for the impaired immunity associated with FAE. Although the underlying mechanism is far from clear, studies described in this report suggest that nitric oxide (NO), a gaseous free radical, is involved in the altered splenic sympathetic neural response to immune signals. The suggestion is supported by the following findings: (1) blockade of NO synthesis prevented the blunted sympathetic response to lipopolysaccharide or interleukin-1 in FAE rats, and (2) there was a further increase in NO formation in response to lipopolysaccharide in the FAE rats compared to their control cohorts. This was demonstrated by an augmented increase in the inducible NO synthase immunoreactivity in the spleen as well as in circulating levels of NO metabolites. It is suggested, therefore, that the altered splenic sympathetic response to immune signals involves excessive formation of NO that may account, at least in part, for the impaired immunity associated with FAE.
Collapse
Affiliation(s)
- Z Gottesfeld
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston 77225, USA
| |
Collapse
|
23
|
Gottesfeld Z, Maier M, Mailman D, Lai M, Weisbrodt NW. Splenic sympathetic response to endotoxin is blunted in the fetal alcohol-exposed rat: role of nitric oxide. Alcohol 1998; 16:19-24. [PMID: 9650632 DOI: 10.1016/s0741-8329(98)00009-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This study was designed to test the hypothesis that nitric oxide (NO) mediates the blunted splenic sympathetic response to lipopolysaccharide (endotoxin) that occurs in young rats exposed to alcohol in utero (FAE). The subjects, 26-29-day-old rats, were progeny of pregnant dams fed an alcohol diet (35% of the calories were derived from ethanol) or their control and pair-fed (PFC) cohorts. We examined the effects of lipopolysaccharide (LPS) (0.5 mg/kg, i.p.) on splenic norepinephrine (NE) turnover, an index of sympathetic neural activity, splenic inducible NO synthase (iNOS) protein immunoreactivity, and NO metabolites nitrite/nitrate concentrations in plasma. In response to LPS, splenic NE turnover was increased by more than twofold in the PFC groups, but the increase did not occur in their FAE cohorts. The blockade of NOS with L-NAME (30 mg/kg, i.p.) reversed this difference. In both the PFC and FAE rats, basal levels of splenic iNOS protein immunoreactivity were equally barely detected and plasma NO metabolite levels were relatively low (25 microM in both groups). In response to LPS, however, iNOS protein displayed a marked increase in the PFC group and an even greater increase (by close to threefold) in the FAE rats. LPS also substantially increased plasma NO metabolite levels by close to eightfold in the control groups, but by 15-fold in their FAE cohorts compared to the basal levels. These findings support the hypothesis that in the FAE rat, an augmented NO formation accounts for the blunted sympathetic response to endotoxin.
Collapse
Affiliation(s)
- Z Gottesfeld
- Department of Neurobiology and Anatomy, University of Texas Medical School, Houston 77225, USA.
| | | | | | | | | |
Collapse
|
24
|
Dötsch J, Hänze J, Beste O, Behrendt J, Weber WM, Dittrich K, Rascher W. Suppression of neuropeptide Y1 receptor function in SK-N-MC cells by nitric oxide. THE AMERICAN JOURNAL OF PHYSIOLOGY 1997; 273:C618-21. [PMID: 9277359 DOI: 10.1152/ajpcell.1997.273.2.c618] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The neuropeptide Y1 receptor (NPY1) predominantly mediates the vasoconstrictor effects of NPY in smooth muscle cells. The present experiments were planned to study the direct influence of the vasodilator nitric oxide (NO) on NPY1-receptor function. SK-N-MC and CHP-234 cells expressing Y1 and Y2 receptor, respectively, were incubated with the NO donors sodium nitroprusside (SNP), 3-morpholinosydnonimine (SIN-1), and S-nitroso-N-acetyl-penicillamine (SNAP). Receptor binding, Y1-receptor mRNA expression by Northern blot, and adenosine 3',5'-cyclic monophosphate (cAMP) and intracellular Ca2+ concentration ([Ca2+]i) responses were studied. SNP, SIN-1, and SNAP decreased normal binding of NPY to the NPY1 receptor in SK-N-MC cells in a concentration-dependent manner. SNP (500 microM), SIN-1 (1,000 microM), and SNAP (500 microM) significantly decreased binding to approximately 50%. The cell viability was not reduced. None of the NO donors affected Y2 receptor binding. Pretreatment with SNP significantly attenuated NPY-induced inhibition of cAMP formation in SK-N-MC cells but had no effect on CHP cells. The NPY-induced [Ca2+]i response was reduced to 50% by SNP pretreatment. NPY1 mRNA expression was reduced to one-third after SNAP treatment of SK-N-MC cells. In vitro, NPY1 receptor function of SK-N-MC cells is inhibited by NO-donor incubation on an mRNA level.
Collapse
Affiliation(s)
- J Dötsch
- Department of Pediatrics, Justus-Liebig-Universität, Giessen, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Kvetnanský R, Pacák K, Tokarev D, Jeloková J, Jezová D, Rusnák M. Chronic blockade of nitric oxide synthesis elevates plasma levels of catecholamines and their metabolites at rest and during stress in rats. Neurochem Res 1997; 22:995-1001. [PMID: 9239755 DOI: 10.1023/a:1022426910111] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Formation of nitric oxide, and endothelium-derived relaxing factor, can be inhibited by administration of N-nitro-L-arginine methylesther (L-NAME). In the present study, the activity of the sympathoadrenal system in rats with blood pressure (BP) elevation induced by L-NAME was investigated. L-NAME was administered in a dose of 50 mg/kg, i.p. every 12 h for 4 days. Blood samples were collected via chronically inserted arterial catheters in conscious, freely moving rats at rest and during immobilization stress. Plasma epinephrine (EPI), norepinephrine (NE), and dopamine (DA), as well as catecholamine metabolites dihydroxyphenylglycol (DHPG) and dihydroxyphenylacetic acid (DOPAC) were measured by HPLC method. In L-NAME treated animals, which slowed a significant increase in BP, plasma EPI levels were markedly elevated both before and during stress. Plasma NE levels were not significantly increased, however, DHPG levels, which indicate NE turnover and reuptake, were highly elevated. Plasma DA levels were not changed after L-NAME administration but DA metabolite DOPAC showed a significant elevation both under basal conditions and during stress. Thus, the present results indicate that the prolonged blockade of nitric oxide synthesis that causes arterial hypertension is associated with an activation of the sympathoadrenal system.
Collapse
Affiliation(s)
- R Kvetnanský
- Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
26
|
Dötsch J, Hänze J, Dittrich K, Demirakça S, Haberberger R, Rascher W. Stimulation of neuropeptide Y release in rat pheochromocytoma cells by nitric oxide. Eur J Pharmacol 1997; 331:313-7. [PMID: 9274994 DOI: 10.1016/s0014-2999(97)01050-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Neuropeptide Y and nitric oxide (NO) synthase are colocalized in nervous tissues. We tested the hypothesis whether or not NO might be involved in the release of neuropeptide Y. Neuropeptide Y concentration in the supernatant of PC12 rat pheochromocytoma cells, shown to express NO synthase I by immunohistochemistry, rose threefold in a time- and dose-dependent manner following sodiumnitroprusside and 3-morpholinosydnonimine (SIN-1) incubation. Neuropeptide Y mRNA expression was induced by NO-donors as a function of incubation-time. Neuropeptide Y production rose fivefold with zaprinast, an inhibitor of the phosphodiesterase V and threefold with nerve growth factor (NGF). Combined application of zaprinast and NGF did not further increase neuropeptide Y production while combination of zaprinast and sodiumnitroprusside potentiated the NO effect on neuropeptide Y release. The data suggest that NO regulates neuropeptide Y secretion of PC12 pheochromocytoma cells on the mRNA level.
Collapse
Affiliation(s)
- J Dötsch
- Department of Pediatrics, Justus-Liebig-Universität Giessen, Germany
| | | | | | | | | | | |
Collapse
|
27
|
Gironacci MM, Lorenzo PS, Adler-Graschinsky E. Possible participation of nitric oxide in the increase of norepinephrine release caused by angiotensin peptides in rat atria. Hypertension 1997; 29:1344-50. [PMID: 9180639 DOI: 10.1161/01.hyp.29.6.1344] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In rat atria isolated with their cardioaccelerans nerves and labeled with [3H]norepinephrine, exposure to 1 x 10(-7) mol/L angiotensin II (Ang II) and 1 x 10(-7) mol/L Ang-(1-7) increased the release of radioactivity elicited by nerve stimulation (0.5-millisecond-long square-wave pulses at 2 Hz during 2 minutes) by 90% and 60%, respectively. The facilitatory effect on noradrenergic neurotransmission caused by both peptides was stereospecifically prevented by N omega-nitro-L-arginine methyl ester (1 x 10(-4) mol/L), an inhibitor of nitric oxide synthase that catalyzes the conversion of L-arginine to nitric oxide, as well as by 1 x 10(-5) mol/L methylene blue, a substance that inhibits the guanylate cyclase considered as the final target of nitric oxide action. On the other hand, the precursor of nitric oxide synthesis. L-arginine (1 x 10(-3) mol/L), reversed the prevention produced by N omega-nitro-L-arginine methyl ester on the increased release of norepinephrine caused by Ang II and Ang-(1-7). The present results suggest that nitric oxide could be involved in the neuromodulatory function elicited by both Ang II and Ang-(1-7) in rat atria. The physiological role of this observation is still under study.
Collapse
Affiliation(s)
- M M Gironacci
- Instituto de Química y Fisicoquímica Biológicas (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | | | | |
Collapse
|
28
|
Tatchum-Talom R, Atkinson J. Disruption of the rat mesenteric arterial bed endothelial function by air perfusion. Life Sci 1997; 60:2407-16. [PMID: 9199485 DOI: 10.1016/s0024-3205(97)00301-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The impact of air perfusion on the endothelial function of the rat mesenteric arterial bed (MAB; perfused with Krebs' bicarbonate plus indomethacin) was compared to that of the NO synthase inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME). Air shifted the dose-response curve for the alpha-adrenoceptor agonist, norepinephrine (NE) to the left (ED50%: 2.9+/-0.7 to 0.9+/-0.7 microg, P < 0.05); maximal vasoconstriction did not change. L-NAME produced a similar increase in midrange sensitivity (ED50% 1.4+/-0.7 microg, P < 0.05) and a 20% increase in maximum (152+/-6 to 183+/-7 mmHg, P < 0.05). Electromechanical stimulation with potassium chloride (KCl) was not modified by reserpine. Neither air nor L-NAME modified midrange sensitivity to KCl. L-NAME produced a 17% increase in maximum (91+/-4 to 107+/-5 mmHg, P < 0.05); reserpine abolished the latter effect. Air and L-NAME diminished endothelium-dependent vasodilation elicited by carbachol. Air did not modify endothelium-dependent vasodilation elicited by sodium nitroprusside; this response was potentiated by L-NAME. In summary, air and L-NAME produced similar effects on receptor-dependent activation of the endothelial L-arginine nitric oxide (NO) pathway. Potentiation by L-NAME of the maximal electromechanical response suggests the existence of a tone-dependent NO system. Abolition of the latter response by reserpine suggests that this system is of sympathetic origin.
Collapse
Affiliation(s)
- R Tatchum-Talom
- Laboratoire de Pharmacologie Cardio-vasculaire, Faculté de Pharmacie, Université Henri Poincaré Nancy 1, France.
| | | |
Collapse
|
29
|
|
30
|
Yuhi T, Wada A, Kobayashi H, Yamamoto R, Yanagita T, Niina H. Up-regulation of functional voltage-dependent sodium channels by cyclic AMP-dependent protein kinase in adrenal medulla. Brain Res 1996; 709:37-43. [PMID: 8869554 DOI: 10.1016/0006-8993(95)01252-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Treatment of cultured bovine adrenal chromaffin cells with dbcAMP increased [3H]STX binding with an EC50 of 126 microM and a half-effective time of 12 h; dbcAMP (1 mM x 18 h) raised the Bmax approximately 1.5-fold without altering the Kd value. Forskolin (0.1 mM) or IBMX (0.3 mM) also increased [3H]STX binding, while dbcGMP had no effect. Effects of dbcAMP and forskolin were abolished by H-89, an inhibitor of cAMP-dependent protein kinase. Cycloheximide (10 microgram/ml) and actinomycin D (10 microgram/ml), inhibitors of protein synthesis, nullified the stimulatory effect of dbcAMP, whereas tunicamycin, an inhibitor of protein glycosylation, had no effect. Treatment with dbcAMP augmented veratridine-induced 22Na influx, 45Ca influx via voltage-dependent Ca channels and catecholamine secretion, while the same treatment did not alter 45Ca influx and catecholamine secretion caused by high K (a direct activation of voltage-dependent Ca channels) [25]. Na influx via single Na channel calculated from 22Na influx and [3H]STX binding was quantitatively similar between non-treated and dbcAMP-treated cells. Brevetoxin allosterically enhanced veratridine-induced 22Na influx approximately 3-fold in dbcAMP-treated cells as in non-treated cells. These results suggest that cAMP-dependent protein kinase is involved in the modulation of Na channel expression in adrenal medulla.
Collapse
Affiliation(s)
- T Yuhi
- Department of Pharmacology, Miyazaki Medical College, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Schwarz P, Diem R, Dun NJ, Förstermann U. Endogenous and exogenous nitric oxide inhibits norepinephrine release from rat heart sympathetic nerves. Circ Res 1995; 77:841-8. [PMID: 7554131 DOI: 10.1161/01.res.77.4.841] [Citation(s) in RCA: 174] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
This study was designed to elucidate whether nitric oxide (NO) controls norepinephrine (NE) release from sympathetic nerves of the rat heart. Hearts were perfused in the Langendorff mode with Tyrode's solution. The right sympathetic nerve was stimulated with trains of 1 or 3 Hz and NE release was measured. The NO synthase (NOS) inhibitor NG-nitro-L-arginine (L-NNA) enhanced the evoked NE release in a concentration-dependent manner. This facilitation was independent of the increase in perfusion pressure and was stereospecifically reversed by L-arginine but not D-arginine. Another NOS inhibitor, NG-methyl-L-arginine, produced a similar increase in NE release. The NO-donor compound S-nitroso-N-acetyl-D,L-penicillamine, added in the presence of L-NNA, restored the suppression of NE release in a concentration-dependent fashion. A similar suppression was achieved with 3-morpholinosydnonimine. These results demonstrated that NE release is under the inhibitory control of endogenous NO. Western blots demonstrated the presence of neuronal NOS I and endothelial NOS III in the hearts. Perfusion of the hearts with a low concentration of the detergent CHAPS produced functional damage of the endothelium, as evidenced by an increase in perfusion pressure and a conversion of the acetylcholine-induced coronary vasodilation to a constriction. However, CHAPS treatment did not produce a facilitation of NE release (as did the NOS inhibitors), and L-NNA still increased NE release in CHAPS-treated hearts. Double-labeling immunofluorescence histochemistry showed NOS I immunoreactivity in stellate ganglion cells and in neurons of the heart, some of which also stained positive for tyrosine hydroxylase.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- P Schwarz
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | | | | | | |
Collapse
|
32
|
Levi R, Park KH, Imamura M, Seyedi N, Lander HM. Nitric oxide and peripheral adrenergic neuromodulation. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 1995; 34:399-413. [PMID: 8562448 DOI: 10.1016/s1054-3589(08)61100-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- R Levi
- Department of Pharmacology, Cornell University Medical College, New York, New York 10021, USA
| | | | | | | | | |
Collapse
|