Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Hypertens ; 41(2): 233-245, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36583351

ABSTRACT

BACKGROUND: Hypothalamic paraventricular nucleus (PVN) is an important central site for the control of the adipose afferent reflex (AAR) that increases sympathetic outflow and blood pressure in obesity-related hypertension (OH). METHOD: In this study, we investigated the effects of nitric oxide (NO) and cardiovascular bioactive polypeptide adrenomedullin (ADM) in the PVN on AAR and sympathetic nerve activity (SNA) in OH rats induced by a high-fat diet. RESULTS: The results showed that ADM, total neuronal NO synthase (nNOS) and phosphorylated-nNOS protein expression levels in the PVN of the OH rats were down-regulated compared to the control rats. The enhanced AAR in OH rats was attenuated by PVN acute application of NO donor sodium nitroprusside (SNP), but was strengthened by the nNOS inhibitor nNOS-I, guanylyl cyclase inhibitor (1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalin-1-one, ODQ) and gamma-aminobutyric acid A type receptor (GABAA) antagonist Bicuculline. Moreover, PVN ADM microinjection not only decreased basal SNA but also attenuated the enhanced AAR in OH rats, which were effectively inhibited by ADM receptor antagonist ADM22-52, nNOS-I, ODQ or Bicuculline pretreatment. Bilateral PVN acute microinjection of ADM also caused greater increases in NO and cyclic guanosine monophosphate (cGMP) levels, and nNOS phosphorylation. Adeno-associated virus vectors encoding ADM (AAV-ADM) transfection in the PVN of OH rats not only decreased the elevated AAR, basal SNA and blood pressure (BP), but also increased the expression and activation of nNOS. Furthermore, AAV-ADM transfection improved vascular remodeling in OH rats. CONCLUSION: Taken together, our data highlight the roles of ADM in improving sympathetic overactivation, enhanced AAR and hypertension, and its related mechanisms associated with receptors mediated NO-cGMP-GABAA pathway in OH condition.


Subject(s)
Hypertension , Paraventricular Hypothalamic Nucleus , Rats , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Adrenomedullin , Nitric Oxide/metabolism , Rats, Sprague-Dawley , Receptors, GABA/metabolism , Bicuculline/metabolism , Bicuculline/pharmacology , Obesity/complications , Reflex/physiology , Blood Pressure/physiology , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/pharmacology , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology , Sympathetic Nervous System
2.
J Vasc Res ; 59(5): 288-302, 2022.
Article in English | MEDLINE | ID: mdl-35947969

ABSTRACT

BACKGROUND: The mechanism of the perivascular adipose tissue (PVAT) anticontractile effect is well characterized in rodent visceral vascular beds; however, little is known about the mechanism of PVAT anticontractile function in subcutaneous vessels. In addition, we have previously shown that PVAT anticontractile function is nitric oxide synthase (NOS) dependent but have not investigated the roles of NOS isoforms. OBJECTIVE: Here, we examined PVAT anticontractile function in the mouse gracilis artery, a subcutaneous fat depot, in lean control and obese mice and investigated the mechanism in comparison to a visceral depot. METHOD: Using the wire myograph, we generated responses to noradrenaline and electrical field stimulation in the presence of pharmacological tools targeting components of the known PVAT anticontractile mechanism. In addition, we performed ex vivo "fat transplants" in the organ bath. RESULTS: The mechanism of PVAT anticontractile function is similar between subcutaneous and visceral PVAT depots. Both endothelial and neuronal NOS isoforms mediated the PVAT anticontractile effect. Loss of PVAT anticontractile function in obesity is independent of impaired vasoreactivity, and function can be restored in visceral PVAT by NOS activation. CONCLUSIONS: Targeting NOS isoforms may be useful in restoring PVAT anticontractile function in obesity, ameliorating increased vascular tone, and disease.


Subject(s)
Adipose Tissue , Obesity , Mice , Animals , Nitric Oxide Synthase Type I/pharmacology , Norepinephrine/pharmacology , Mice, Obese , Nitric Oxide Synthase , Protein Isoforms/pharmacology , Nitric Oxide , Vasoconstriction
3.
J Pain ; 23(10): 1765-1778, 2022 10.
Article in English | MEDLINE | ID: mdl-35705162

ABSTRACT

Dexamethasone supplementation to local anesthetics prolongs its action, yet the underlying mechanism is unclear. Previous studies have reported that increased p-p38 mitogen-activated protein kinase (MAPK) in the dorsal root ganglia (DRG) is associated with pain-associated behavior and that nitric oxide (NO), which is known to be a pronociceptive substance, directly inhibits sciatic nerve conduction. Here, we investigated the temporal changes in the hyperalgesic effect and p-p38 MAPK and NO synthase (NOS) expression levels in the DRG when dexamethasone was added to ropivacaine used for a sciatic nerve block (SNB) in postoperative pain model mice. Dexamethasone supplementation to ropivacaine significantly prolonged the analgesic effect of SNB via glucocorticoid receptor activation. Histological examination revealed that ropivacaine suppressed p-p38 MAPK expression in the DRG regardless of dexamethasone supplementation, suggesting that p-p38 MAPK was not involved in the prolonging effect of dexamethasone on nerve block. Contrastingly, plantar incision markedly increased the expression of neuronal NOS (nNOS) in DRG, and dexamethasone supplementation to ropivacaine significantly suppressed nNOS expression. Supplementation of L-NAME, an inhibitor of NOS, to ropivacaine markedly prolonged the effect of SNB, similar to dexamethasone. These results suggest that dexamethasone supplementation to local anesthetics prolongs the analgesic effect by inhibiting nNOS activity. PERSPECTIVE: The current study revealed that dexamethasone supplementation to local anesthetics prolongs the analgesic effect by inhibiting the activity of neuronal NOS and that p-p38 MAPK may not be involved in this phenomenon. Our findings offer a new target for the discovery of long-acting local anesthetics.


Subject(s)
Anesthetics, Local , Receptors, Glucocorticoid , Analgesics/pharmacology , Anesthetics, Local/pharmacology , Animals , Dexamethasone/pharmacology , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type I/pharmacology , Pain, Postoperative/drug therapy , Ropivacaine/pharmacology , Sciatic Nerve , p38 Mitogen-Activated Protein Kinases
4.
World Neurosurg ; 164: e214-e223, 2022 08.
Article in English | MEDLINE | ID: mdl-35472644

ABSTRACT

OBJECTIVE: This study aimed to determine the effects of sacral nerve electrical stimulation (SNS) on neuronal nitric oxide synthase (nNOS) in the colon and sacral cord of rats with defecation disorder after spinal cord injury (SCI). METHODS: Rats with severe SCI (T10) were used as models and randomly divided into an SCI group and an SNS group. After 14 days of treatment, the intestinal transport function was assessed. Finally, the differences in nNOS immunoreactive cells, protein levels, nNOS mRNA, and NO content in the colon and sacral cord tissues were estimated using immunohistochemistry, Western blot, real-time polymerase chain reaction, and nitrate reductase method. RESULTS: The intestinal transport function of the SNS group was superior to that of the SCI group (P < 0.05). The average optical density of nNOS immunoreactive positive cells in the SCI group were significantly increased compared with those in the sham group. The content of NO of the SCI group significant increased compared with those of the sham group and the SNS group (both P < 0.01). The nNOS mRNA and protein expression was higher in the SCI group than in the sham group (P < 0.01), while that in the SNS group was significantly lower than that in the SCI group. CONCLUSIONS: SNS could reduce nNOS expression in the colon and sacral cord of SCI rats. This reduction may be an important neuromodulation mechanism for SNS to improve defecation reflex and promote the recovery of intestinal transit function.


Subject(s)
Defecation , Spinal Cord Injuries , Animals , Colon , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/pharmacology , RNA, Messenger/metabolism , Rats , Spinal Cord
5.
J Plast Reconstr Aesthet Surg ; 74(1): 183-191, 2021 01.
Article in English | MEDLINE | ID: mdl-32893152

ABSTRACT

BACKGROUND: Diabetes mellitus increases the susceptibility of free tissue transplantations to ischemia-reperfusion injury. The aim of this study was to enhance nitric oxide (NO) bioavailability through exogenous NO synthase and the substrate L-arginine to attenuate ischemia reperfusion-induced alterations in a type 2 diabetes rodent model. MATERIAL AND METHODS: Sixty-four Wistar rats were divided into 8 experimental groups. Type 2 diabetes was established over 3 months with a combination of a high-fat diet and streptozotocin. A vascular pedicle isolated rat skin flap model that underwent 3 h of ischemia was used. At 30 min before ischemia, normal saline, endothelial NOSs (eNOSs), inducible NOSs, neuronal NOSs (1 and 2 IU), and L-arginine (50 mg/kg body weight) were administered by intravenous infusion alone or in combination. Ischemia-reperfusion-induced alterations were measured 5 days after the operation. RESULTS: The three isoforms of NOS significantly increased the flap vitality rate (VR) between 20% and 28% as compared to the control group (3%). Sole L-arginine administration increased the VR to 33%. The combination of L-arginine with NOS resulted in a further increase in flap VRs (39%-50%). Best results were achieved with the combination of eNOS and L-arginine (50%). An increase in enzyme dosage led to decreased VRs in all NOS isoforms alone and even in combination with L-arginine. CONCLUSION: Modulation of NO bioavailability through the exogenous application of NOSs and L-arginine significantly attenuated ischemia-reperfusion-induced alterations in a type 2 diabetic skin flap rat model. The combination of enzyme and substrate result in the highest VRs. Higher enzyme dosage seems to be less effective. This pharmacological preconditioning could be an easy and effective interventional strategy to support the conversion of L-arginine to NO in ischemic and in type 2 diabetic conditions.


Subject(s)
Arginine/pharmacology , Diabetes Mellitus, Type 2/metabolism , Nitric Oxide Synthase/pharmacology , Nitric Oxide/biosynthesis , Reperfusion Injury/metabolism , Surgical Flaps/physiology , Animals , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Male , Nitric Oxide Synthase Type I/pharmacology , Nitric Oxide Synthase Type II/pharmacology , Nitric Oxide Synthase Type III/pharmacology , Rats , Rats, Wistar , Skin/metabolism
6.
Mol Med ; 25(1): 31, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31266455

ABSTRACT

BACKGROUND: Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma leads to functional muscle ischemia. This contributes to the pathogenesis in cachexia, aging and muscular dystrophy. Mutations in the gene encoding dystrophin result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In many BMD patients and DMD patients that have been converted to BMD by gene therapy, sarcolemmal nNOS is missing due to the lack of dystrophin nNOS-binding domain. METHODS: Dystrophin spectrin-like repeats 16 and 17 (R16/17) is the sarcolemmal nNOS localization domain. Here we explored whether R16/17 protein therapy can restore nNOS to the sarcolemma and prevent functional ischemia in transgenic mice which expressed an R16/17-deleted human micro-dystrophin gene in the dystrophic muscle. The palmitoylated R16/17.GFP fusion protein was conjugated to various cell-penetrating peptides and produced in the baculovirus-insect cell system. The best fusion protein was delivered to the transgenic mice and functional muscle ischemia was quantified. RESULTS: Among five candidate cell-penetrating peptides, the mutant HIV trans-acting activator of transcription (TAT) protein transduction domain (mTAT) was the best in transferring the R16/17.GFP protein to the muscle. Systemic delivery of the mTAT.R16/17.GFP protein to micro-dystrophin transgenic mice successfully restored sarcolemmal nNOS without inducing T cell infiltration. More importantly, R16/17 protein therapy effectively prevented treadmill challenge-induced force loss and improved muscle perfusion during contraction. CONCLUSIONS: Our results suggest that R16/17 protein delivery is a highly promising therapy for muscle diseases involving sarcolemmal nNOS delocalizaton.


Subject(s)
Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type I/metabolism , Sarcolemma/metabolism , Utrophin/metabolism , Animals , Humans , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutation/genetics , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/pharmacology , Protein Binding/genetics , Sarcolemma/genetics , Sarcolemma/pathology , Utrophin/genetics
7.
BJU Int ; 120(6): 861-872, 2017 12.
Article in English | MEDLINE | ID: mdl-28782252

ABSTRACT

OBJECTIVES: To evaluate neuronal nitric oxide (NO) synthase (nNOS) phosphorylation, nNOS uncoupling, and oxidative stress in the penis and major pelvic ganglia (MPG), before and after the administration of the cAMP-dependent protein kinase A (PKA) agonist colforsin in a rat model of bilateral cavernous nerve injury (BCNI),which mimics nerve injury after prostatectomy. MATERIALS AND METHODS: Adult male Sprague-Dawley rats were divided into BCNI and sham-operated groups. Each group included two subgroups: vehicle and colforsin (0.1 mg/kg/day i.p.). After 3 days, erectile function (intracavernosal pressure) was measured and penis and MPG were collected for molecular analyses of phospho (P)-nNOS (Ser-1412 and Ser-847), total nNOS, nNOS uncoupling, binding of protein inhibitor of nNOS (PIN) to nNOS, gp91phox subunit of NADPH oxidase, active caspase 3, PKA catalytic subunit α (PKA-Cα; by Western blot) and oxidative stress (hydrogen peroxide [H2 O2 ] and superoxide by Western blot and microdialysis method). RESULTS: Erectile function was decreased 3 days after BCNI and normalized by colforsin. nNOS phosphorylation on both positive (Ser-1412) and negative (Ser-847) regulatory sites, and nNOS uncoupling, were increased after BCNI in the penis and MPG, and normalized by colforsin. H2 O2 and total reactive oxygen species production were increased in the penis after BCNI and normalized by colforsin. Protein expression of gp91phox was increased in the MPG after BCNI and was normalized by colforsin treatment. Binding of PIN to nNOS was increased in the penis after BCNI and was normalized by colforsin treatment. Protein expression of active Caspase 3 was increased in the MPG after BCNI and was normalized by colforsin treatment. Protein expression of PKA-Cα was decreased in the penis after BCNI and normalized by colforsin. CONCLUSION: Collectively, BCNI impairs nNOS function in the penis and MPG by mechanisms involving its phosphorylation and uncoupling in association with increased oxidative stress, resulting in erectile dysfunction. PKA activation by colforsin reverses these molecular changes and preserves penile erection in the face of BCNI.


Subject(s)
Erectile Dysfunction/physiopathology , Neuroprotective Agents , Nitric Oxide Synthase Type I , Penile Erection/drug effects , Protein Processing, Post-Translational , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Ganglia/drug effects , Male , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/pharmacology , Oxidative Stress , Pelvis/innervation , Phosphorylation , Rats , Rats, Sprague-Dawley
8.
Int. braz. j. urol ; 42(5): 1018-1027, Sept.-Oct. 2016. tab, graf
Article in English | LILACS | ID: lil-796875

ABSTRACT

ABSTRACT Objective: To evaluate the effect of neuronal nitric oxide synthase on the striated urethral sphincter and the urinary bladder. Materials and Methods: A coaxial catheter was implanted in the proximal urethra and another one in the bladder of female rats, which were anesthetized with subcutaneous injection of urethane. The urethral pressure with saline continuous infusion and bladder isovolumetric pressure were simultaneously recorded. Two groups of rats were formed. In group I, an intrathecal catheter was implanted on the day of the experiment at the L6-S1 level of the spinal cord; in group II, an intracerebroventricular cannula was placed 5-6 days before the experiment. Results: It was verified that the group treated with S-methyl-L-thio-citrulline, via intrathecal pathway, showed complete or partial inhibition of the urethral sphincter relaxation and total inhibition of the micturition reflexes. The urethral sphincter and the detrusor functions were recovered after L-Arginine administration. When S-methyl-L-thio-citrulline was administered via intracerebroventricular injection, there was a significant increase of urethral sphincter tonus while preserving the sphincter relaxation and the detrusor contractions, at similar levels as before the use of the drugs. Nevertheless there was normalization of the urethral tonus when L-Arginine was applied. Conclusions: The results indicate that, in female rats anaesthetized with urethane, the nNOS inhibitor administrated through the intrathecal route inhibits urethral sphincter relaxation, while intracerebroventricular injection increases the sphincter tonus, without changing bladder function. These changes were reverted by L-Arginine administration. These findings suggest that the urethral sphincter and detrusor muscle function is modulated by nitric oxide.


Subject(s)
Animals , Female , Thiourea/analogs & derivatives , Urethra/drug effects , Urination/drug effects , Urinary Bladder/drug effects , Citrulline/analogs & derivatives , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/pharmacology , Arginine/pharmacology , Pressure , Reference Values , Thiourea/pharmacology , Time Factors , Urethane/pharmacology , Urethra/physiology , Urination/physiology , Urinary Bladder/physiology , Injections, Spinal , Citrulline/pharmacology , Rats, Wistar , Anesthetics, Intravenous , Muscle Contraction/drug effects , Muscle Contraction/physiology
9.
An Acad Bras Cienc ; 88 Suppl 1: 609-22, 2016.
Article in English | MEDLINE | ID: mdl-27142540

ABSTRACT

The objective of this study was to investigate the effects of 2% L-glutamine supplementation on myenteric innervation in the ileum of diabetic rats, grouped as follows: normoglycemic (N); normoglycemic supplemented with L-glutamine (NG); diabetic (D); and diabetic supplemented with L-glutamine (DG). The ileums were subjected to immunohistochemical techniques to localize neurons immunoreactive to HuC/D protein (HuC/D-IR) and neuronal nitric oxide synthase enzyme (nNOS-IR) and to analyze varicosities immunoreactive to vasoactive intestinal polypeptide (VIP-IR) and calcitonin gene-related peptide (CGRP-IR). L-Glutamine in the DG group (i) prevented the increase in the cell body area of nNOS-IR neurons, (ii) prevented the increase in the area of VIP-IR varicosities, (iii) did not prevent the loss of HuC/D-IR and nNOS-IR neurons per ganglion, and (iv) reduced the size of CGRP-IR varicosities. L-Glutamine in the NG group reduced (i) the number of HuC/D-IR and nNOS-IR neurons per ganglion, (ii) the cell body area of nNOS-IR neurons, and (iii) the size of VIP-IR and CGRP-IR varicosities. 2% L-glutamine supplementation exerted differential neuroprotective effects in experimental diabetes neuropathy that depended on the type of neurotransmitter analyzed. However, the effects of this dose of L-glutamine on normoglycemic animals suggests there are additional actions of this beyond its antioxidant capacity.


Subject(s)
Diabetes Mellitus, Experimental , Glutamine/pharmacology , Ileum/innervation , Myenteric Plexus/drug effects , Animals , Calcitonin Gene-Related Peptide/pharmacology , Cell Body/drug effects , Glutamine/administration & dosage , Immunohistochemistry , Neurons/drug effects , Nitrergic Neurons , Nitric Oxide Synthase Type I/pharmacology , Rats , Rats, Wistar , Vasoactive Intestinal Peptide/pharmacology
10.
Int Braz J Urol ; 42(5): 1018-1027, 2016.
Article in English | MEDLINE | ID: mdl-24893916

ABSTRACT

OBJECTIVE: To evaluate the effect of neuronal nitric oxide synthase on the striated urethral sphincter and the urinary bladder. MATERIALS AND METHODS: A coaxial catheter was implanted in the proximal urethra and another one in the bladder of female rats, which were anesthetized with subcutaneous injection of urethane. The urethral pressure with saline continuous infusion and bladder isovolumetric pressure were simultaneously recorded. Two groups of rats were formed. In group I, an intrathecal catheter was implanted on the day of the experiment at the L6-S1 level of the spinal cord; in group II, an intracerebroventricular cannula was placed 5-6 days before the experiment. RESULTS: It was verified that the group treated with S-methyl-L-thio-citrulline, via intrathecal pathway, showed complete or partial inhibition of the urethral sphincter relaxation and total inhibition of the micturition reflexes. The urethral sphincter and the detrusor functions were recovered after L-Arginine administration. When S-methyl-Lthio-citrulline was administered via intracerebroventricular injection, there was a significant increase of urethral sphincter tonus while preserving the sphincter relaxation and the detrusor contractions, at similar levels as before the use of the drugs. Nevertheless there was normalization of the urethral tonus when L-Arginine was applied. CONCLUSIONS: The results indicate that, in female rats anaesthetized with urethane, the nNOS inhibitor administrated through the intrathecal route inhibits urethral sphincter relaxation, while intracerebroventricular injection increases the sphincter tonus, without changing bladder function. These changes were reverted by L-Arginine administration. These findings suggest that the urethral sphincter and detrusor muscle function is modulated by nitric oxide.


Subject(s)
Citrulline/analogs & derivatives , Enzyme Inhibitors/pharmacology , Nitric Oxide Synthase Type I/pharmacology , Thiourea/analogs & derivatives , Urethra/drug effects , Urinary Bladder/drug effects , Urination/drug effects , Anesthetics, Intravenous , Animals , Arginine/pharmacology , Citrulline/pharmacology , Female , Injections, Spinal , Muscle Contraction/drug effects , Muscle Contraction/physiology , Pressure , Rats, Wistar , Reference Values , Thiourea/pharmacology , Time Factors , Urethane/pharmacology , Urethra/physiology , Urinary Bladder/physiology , Urination/physiology
11.
J Neurosci ; 30(3): 973-84, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20089906

ABSTRACT

The molecular signaling that underpins synapse loss in neuropathological conditions remains unknown. Concomitant upregulation of the neuronal nitric oxide (NO) synthase (nNOS) in neurodegenerative processes places NO at the center of attention. We found that de novo nNOS expression was sufficient to induce synapse loss from motoneurons at adult and neonatal stages. In brainstem slices obtained from neonatal animals, this effect required prolonged activation of the soluble guanylyl cyclase (sGC)/protein kinase G (PKG) pathway and RhoA/Rho kinase (ROCK) signaling. Synapse elimination involved paracrine/retrograde action of NO. Furthermore, before bouton detachment, NO increased synapse myosin light chain phosphorylation (p-MLC), which is known to trigger actomyosin contraction and neurite retraction. NO-induced MLC phosphorylation was dependent on cGMP/PKG-ROCK signaling. In adulthood, motor nerve injury induced NO/cGMP-dependent synaptic stripping, strongly affecting ROCK-expressing synapses, and increased the percentage of p-MLC-expressing inputs before synapse destabilization. We propose that this molecular cascade could trigger synapse loss underlying early cognitive/motor deficits in several neuropathological states.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Motor Neurons/pathology , Myosin Light Chains/metabolism , Nitric Oxide Synthase Type I/metabolism , Synapses/pathology , rho-Associated Kinases/metabolism , Analysis of Variance , Animals , Animals, Newborn , Brain Stem/cytology , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Humans , Hypoglossal Nerve Diseases/pathology , In Vitro Techniques , Male , Microscopy, Immunoelectron/methods , Motor Neurons/drug effects , Motor Neurons/ultrastructure , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/pharmacology , Nuclear Proteins/genetics , Patch-Clamp Techniques , Phosphorylation/drug effects , Phosphorylation/physiology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Wistar , Synapses/drug effects , Synapses/ultrastructure , Synaptic Potentials/drug effects , Synaptic Potentials/genetics , Synaptophysin/metabolism , Transfection , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , rho-Associated Kinases/antagonists & inhibitors
12.
Intensive Care Med ; 33(11): 1993-2003, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17684724

ABSTRACT

OBJECTIVE: To investigate the role of neuronal nitric oxide synthase (NOS1) in murine polymicrobial peritonitis and sepsis. DESIGN: Randomized experimental trial. SETTING: Animal research facility. SUBJECTS: B6129S NOS1+/+ and B6;129S4 NOS-/- mice. INTERVENTIONS: NOS1+/+ and NOS1-/- animals underwent cecal ligation and puncture (CLP) or sham surgery and received the NOS1 inhibitor 7-nitroindazole (7-NI) or vehicle. MEASUREMENTS AND MAIN RESULTS: After CLP, genetic deficiency and pharmacologic inhibition of NOS1 significantly increased risk of mortality [8.69 (3.27, 23.1), p<0.0001 and 1.71 (1.00, 2.92) p=0.05, hazard ratio of death (95% confidence interval) for NOS1-/- and 7-NI-treated NOS1+/+ respectively] compared with NOS1+/+ animals. In 7-NI-treated NOS1+/+ animals, there were increases (6 h) and then decreases (24 h), whereas in NOS-/- animals persistent increases in blood bacteria counts (p=0.04 for differing effects of 7-NI and NOS1-/-) were seen compared with NOS1(+/+) animals. After CLP, NOS1(-/-) had upregulation of inducible NOS and proinflammatory cytokines and greater increases in serum tumor necrosis factor-alpha and interleukin-6 levels compared with NOS1+/+ mice (all p<0.05). Following CLP, there were similar significant decreases in circulating leukocytes and lung lavage cells (p

Subject(s)
Nitric Oxide Synthase Type I/deficiency , Peritonitis/physiopathology , Sepsis/physiopathology , Animals , Female , Gene Expression , Mice , Nitric Oxide Synthase Type I/adverse effects , Nitric Oxide Synthase Type I/analysis , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type I/pharmacology , Peritonitis/mortality , Random Allocation , Risk Assessment , Sepsis/mortality , Survival Analysis , United States
13.
Eur J Pharmacol ; 557(2-3): 212-20, 2007 Feb 28.
Article in English | MEDLINE | ID: mdl-17208221

ABSTRACT

Cholecystokinin and related peptides are involved in the control of intestinal motility and cholecystokinin receptor ligands might represent new pharmacological tools for the treatment of symptoms associated with functional bowel disorders. However, the respective roles played by cholecystokinin receptor subtypes and the mechanisms underlying these regulatory actions remain undetermined. This study was designed to examine the influence of cholecystokinin receptor subtypes on the motor activity of guinea-pig distal colon. The effects of drugs acting on CCK1 and CCK2 receptors were assessed in vitro on the contractile activity of longitudinal smooth muscle, both under basal conditions and in the presence of transmural electrical stimulation or KCl-induced contractions. The application of cholecystokinin octapeptide sulphate (cholecystokinin-8S) to colonic preparations induced concentration-dependent contractions which were prevented by devazepide (CCK1 receptor antagonist), enhanced by GV150013 (CCK2 receptor antagonist) or N(omega)-nitro-L-arginine methylester (L-NAME, nitric oxide synthase inhibitor), and unaffected by tetrodotoxin. The application of gastrin-17 to colonic preparations resulted in relaxant responses which were insensitive to devazepide, and prevented by GV150013, L-NAME or tetrodotoxin. L-NAME, N(omega)-propyl-L-arginine (NPA, neuronal nitric oxide synthase inhibitor) or GV150013 enhanced electrically evoked contractile responses, whereas devazepide did not. When tested in the presence of L-NAME or NPA the enhancing effect of GV150013 on electrically induced contractions no longer occurred. In the presence of KCl-induced pre-contractions, cholecystokinin-8S or gastrin-17 evoked concentration-dependent relaxations, which were unaffected by devazepide and were counteracted by GV150013, L-NAME, NPA or tetrodotoxin. In conclusion, the present results indicate that, at level of distal colon, CCK1 receptors mediate direct contractile effects on smooth muscle, whereas CCK2 receptors on enteric neurons mediate relaxant responses via nitric oxide release.


Subject(s)
Cholecystokinin/pharmacology , Colon/drug effects , Muscle Contraction/drug effects , Receptor, Cholecystokinin B/physiology , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Colon/physiology , Devazepide/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Electric Stimulation , Enzyme Inhibitors/pharmacology , Gastrins/pharmacology , Guinea Pigs , Hormone Antagonists/pharmacology , Male , Muscle, Smooth/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type I/pharmacology , Nootropic Agents/pharmacology , Perfusion , Phenylurea Compounds/pharmacology , Potassium Chloride/pharmacology , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/antagonists & inhibitors , Sincalide/analogs & derivatives , Sincalide/pharmacology
14.
Am J Physiol Regul Integr Comp Physiol ; 290(4): R1012-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16269576

ABSTRACT

The regulator of G protein signaling (RGS) 2, a GTPase-activating protein, is activated via the nitric oxide (NO)-cGMP pathway and thereby may influence blood pressure regulation. To test that notion, we measured mean arterial blood pressure (MAP) and heart rate (HR) with telemetry in N(omega)-nitro-l-arginine methyl ester (l-NAME, 5 mg l-NAME/10 ml tap water)-treated RGS2-deficient (RGS2(-/-)) and RGS2-sufficient (RGS2(+/+)) mice and assessed autonomic function. Without l-NAME, RGS2(-/-) mice showed during day and night a similar increase of MAP compared with controls. l-NAME treatment increased MAP in both strains. nNOS is involved in this l-NAME-dependent blood pressure increase, since 7-nitroindazole increased MAP by 8 and 9 mmHg (P < 0.05) in both strains. The l-NAME-induced MAP increase of 14-15 mmHg during night was similar in both strains. However, the l-NAME-induced MAP increase during the day was smaller in RGS2(-/-) than in RGS2(+/+) (11 +/- 1 vs. 17 +/- 2 mmHg; P < 0.05). Urinary norepinephrine and epinephrine excretion was higher in RGS2(-/-) than in RGS2(+/+) mice. The MAP decrease after prazosin was more pronounced in l-NAME-RGS2(-/-). HR variability parameters [root mean square of successive differences (RMSSD), low-frequency (LF) power, and high-frequency (HF) power] and baroreflex sensitivity were increased in RGS2(-/-). Atropine and atropine plus metoprolol markedly reduced RMSSD, LF, and HF. Our data suggest an interaction between RGS2 and the NO-cGMP pathway. The blunted l-NAME response in RGS2(-/-) during the day suggests impaired NO signaling. The MAP increases during the active phase in RGS2(-/-) mice may be related to central sympathetic activation and increased vascular adrenergic responsiveness.


Subject(s)
Arteries/physiology , Blood Pressure/drug effects , Heart Rate/drug effects , Nitric Oxide Synthase Type I/pharmacology , RGS Proteins/genetics , Animals , Atropine/pharmacology , Baroreflex/drug effects , Epinephrine/urine , Metoprolol/pharmacology , Mice , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Prazosin/pharmacology , Vasoconstriction
SELECTION OF CITATIONS
SEARCH DETAIL
...