Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Pharmacol Res ; 171: 105788, 2021 09.
Article in English | MEDLINE | ID: mdl-34311071

ABSTRACT

Uterine perivascular adipose tissue (PVAT) contributes to uterine blood flow regulation in pregnancy, at least in part, due to its effects on uterine artery reactivity. We tested the hypothesis that uterine PVAT modulates the balance between the contribution of nitric oxide synthase (NOS)- and cyclooxygenase (COX)-dependent pathways to acetylcholine (ACh)-induced relaxation in isolated uterine arteries. Concentration-response curves to ACh (1 nM - 30 µM) were performed on uterine arteries from pregnant and non-pregnant rats. Arteries were exposed to Krebs-Henseleit solution (control) or PVAT-conditioned media (PVATmedia) in the presence of the following inhibitors: L-NAME (NOS inhibitor), indomethacin (COX inhibitor), SC560 (COX-1 inhibitor), NS398 (COX-2 inhibitor), SQ 29,548 (thromboxane receptor (TP) inhibitor). In arteries incubated with PVATmedia, the presence of indomethacin increased ACh-induced relaxation, reversing the anti-dilatory effect of PVATmedia. NOS inhibition reduced ACh-induced relaxation in uterine arteries from pregnant rats, and exposure to PVATmedia did not change this effect. Selective inhibition of COX-1 but not COX-2 suppressed relaxation responses to ACh in control arteries. The presence of PVATmedia abolished the effect of COX-1 inhibition. Incubation of uterine arteries from pregnant rats with PVATmedia increased production of thromboxane B2 (TxB2, p = 0.01) but thromboxane receptor (TP) inhibition did not affect the anti-dilatory properties of PVATmedia. In conclusion, inhibition of COX signaling suppressed the anti-dilatory effects of PVATmedia, while PVATmedia had no effect on the contribution of the NOS/NO pathway to ACh-induced relaxation in uterine arteries from pregnant rats, indicating that the anti-dilatory effects of uterine PVAT are mediated in part by COX-dependent mechanisms.


Subject(s)
Adipose Tissue/physiology , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Membrane Proteins/physiology , Uterine Artery/physiology , Acetylcholine/pharmacology , Adipose Tissue/drug effects , Animals , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Female , Indomethacin/pharmacology , Male , Membrane Proteins/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/physiology , Nitrobenzenes/pharmacology , Pregnancy , Pyrazoles/pharmacology , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Uterine Artery/drug effects , Uterine Artery/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
2.
FASEB J ; 34(8): 10027-10040, 2020 08.
Article in English | MEDLINE | ID: mdl-32592197

ABSTRACT

Aspirin prevents thrombosis by inhibiting platelet cyclooxygenase (COX)-1 activity and the production of thromboxane (Tx)A2 , a pro-thrombotic eicosanoid. However, the non-platelet actions of aspirin limit its antithrombotic effects. Here, we used platelet-COX-1-ko mice to define the platelet and non-platelet eicosanoids affected by aspirin. Mass-spectrometry analysis demonstrated blood from platelet-COX-1-ko and global-COX-1-ko mice produced similar eicosanoid profiles in vitro: for example, formation of TxA2 , prostaglandin (PG) F2α , 11-hydroxyeicosatraenoic acid (HETE), and 15-HETE was absent in both platelet- and global-COX-1-ko mice. Conversely, in vivo, platelet-COX-1-ko mice had a distinctly different profile from global-COX-1-ko or aspirin-treated control mice, notably significantly higher levels of PGI2 metabolite. Ingenuity Pathway Analysis (IPA) predicted that platelet-COX-1-ko mice would be protected from thrombosis, forming less pro-thrombotic TxA2 and PGE2 . Conversely, aspirin or lack of systemic COX-1 activity decreased the synthesis of anti-aggregatory PGI2 and PGD2 at non-platelet sites leading to predicted thrombosis increase. In vitro and in vivo thrombosis studies proved these predictions. Overall, we have established the eicosanoid profiles linked to inhibition of COX-1 in platelets and in the remainder of the cardiovascular system and linked them to anti- and pro-thrombotic effects of aspirin. These results explain why increasing aspirin dosage or aspirin addition to other drugs may lessen antithrombotic protection.


Subject(s)
Aspirin/pharmacology , Blood Platelets/metabolism , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/pharmacology , Eicosanoids/metabolism , Membrane Proteins/physiology , Thrombosis/metabolism , Animals , Arachidonic Acid/administration & dosage , Blood Platelets/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thrombosis/drug therapy , Thrombosis/pathology
3.
Ann Thorac Surg ; 110(1): 40-49, 2020 07.
Article in English | MEDLINE | ID: mdl-31760051

ABSTRACT

BACKGROUND: The molecular mechanisms underlying aortic valve calcification are poorly understood. Here, we aimed to identify the master regulators of calcification by comparison of genes in valve interstitial cells (VICs) with calcified and noncalcified aortic valves. METHODS: Calcified aortic valves were surgically excised from patients with aortic valve stenosis who required aortic valve replacements. Noncalcified and calcified sections were obtained from aortic valve leaflets. Collagenase-digested tissues were seeded into dishes, and VICs adhering to the dishes were cultured for 3 weeks, followed by comprehensive gene expression analysis. Functional analyses of identified proteins were performed by in vitro calcification assays. Tissue localization was determined by immunohistochemical staining for normal (n = 11) and stenotic valves (n = 30). RESULTS: We found 87 genes showing greater than a twofold change in calcified tissues. Among these genes, 68 were downregulated and 19 were upregulated. Cyclooxygenase-1 (COX1) messenger RNA and protein levels were upregulated in VICs from calcified tissues. The COX1 messenger RNA and protein levels in VICs were also strongly increased by stimulation with osteoblast differentiation medium. These were VIC-specific phenotypes and were not observed in other cell types. Immunohistochemical staining revealed that COX1-positive VICs were specifically localized in the calcified area of aortic valve tissues. CONCLUSIONS: The VIC-specific COX1 overexpression played a crucial role in calcification by promoting osteoblast differentiation in aortic valve tissues.


Subject(s)
Aortic Valve Stenosis/enzymology , Aortic Valve/enzymology , Aortic Valve/pathology , Calcinosis/enzymology , Cyclooxygenase 1/physiology , Fibroblasts/enzymology , Aged , Aged, 80 and over , Aortic Valve/cytology , Aortic Valve/metabolism , Aortic Valve/surgery , Aortic Valve Stenosis/surgery , Calcinosis/surgery , Calcium/metabolism , Cells, Cultured , Culture Media/pharmacology , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 1/genetics , Female , Gene Expression Profiling , Heart Valve Prosthesis Implantation , Humans , Male , Middle Aged , Osteoblasts/pathology , Osteogenesis , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , Vimentin/analysis
4.
Life Sci ; 239: 117039, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31704447

ABSTRACT

AIMS: Obesity is a risk factor for endothelial dysfunction, the severity of which is likely to vary depending on extent and impact of adiposity on the vasculature. This study investigates the roles of cyclooxygenase isoforms and thromboxane receptor activities in the differential endothelial dilatory capacities of arteries derived from omental and subcutaneous adipose tissues in obesity. MAIN METHODS: Small arteries were isolated from omental and subcutaneous adipose tissues obtained from consented morbidly obese patients (n = 65, BMI 45 ±â€¯6 kg m-2 [Mean ±â€¯SD]) undergoing bariatric surgery. Relaxation to acetylcholine was studied by wire myography in the absence or presence of indomethacin (10 µM, cyclooxygenase inhibitor), FR122047 (1 µM, cyclooxygenase-1 inhibitor), Celecoxib (4 µM, cyclooxygenase-2 inhibitor), Nω-Nitro-L-arginine methyl ester (L-NAME, 100 µM, nitric oxide synthase inhibitor) or combination of apamin (0.5 µM) and charybdotoxin (0.1 µM) that together inhibit endothelium-derived hyperpolarizing factor (EDHF). Contractions to U46619 (thromboxane A2 mimetic) were also studied. KEY FINDINGS: Acetylcholine relaxation was significantly attenuated in omental compared with subcutaneous arteries from same patients (p < 0.01). Indomethacin (p < 0.01) and FR122047 (p < 0.001) but not Celecoxib significantly improved the omental arteriolar relaxation. Cyclooxygenase-1 mRNA and U46619 contractions were both increased in omental compared with subcutaneous arteries (p < 0.05). L-NAME comparably inhibited acetylcholine relaxation in both arteries, while apamin+charybdotoxin were less effective in omental compared with subcutaneous arteries. SIGNIFICANCE: The results show that the depot-specific reduction in endothelial dilatory capacity of omental compared with subcutaneous arteries in obesity is in large part due to altered cyclooxygenase-1 and enhanced thromboxane receptor activities, which cause EDHF deficiency.


Subject(s)
Cyclooxygenase 1/metabolism , Gastroepiploic Artery/drug effects , Receptors, Thromboxane/metabolism , Adipose Tissue/blood supply , Adipose Tissue/metabolism , Adult , Apamin/pharmacology , Arteries/drug effects , Celecoxib/pharmacology , Charybdotoxin/pharmacology , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/pharmacology , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/drug effects , Female , Gastroepiploic Artery/metabolism , Humans , Indomethacin/pharmacology , Male , Middle Aged , Muscle Relaxation/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Obesity, Morbid/metabolism , Omentum/blood supply , Omentum/metabolism , Receptors, Thromboxane/physiology , Vasodilation/drug effects
5.
Naunyn Schmiedebergs Arch Pharmacol ; 392(4): 437-450, 2019 04.
Article in English | MEDLINE | ID: mdl-30552456

ABSTRACT

Various studies have confirmed that prostaglandins (PG) alter the bladder motor activity and micturition reflex in both human and animals. However, no sufficient data is reported about the effect of cyclooxygenase (COX) inhibitors neither in normal bladder physiology nor in pathological conditions. This study aims to compare the potential effects of some COX inhibitors with varying COX-1/COX-2 selectivities (indomethacin, ketoprofen, and diclofenac) with that of the selective COX-2 inhibitor (DFU) on bladder function. The role played by some PGs and their receptors in controlling detrusor muscle function in normal condition and in cystitis is also studied. Organ bath experiments were performed using isolated rat detrusor muscle. Direct and neurogenic contractions were induced using ACh and electric stimulation (EFS), respectively. A model of hemorrhagic cystitis was induced by single injection of cyclophosphamide (300 mg/kg) in rats, and confirmed by histophathological examination. Results are expressed as mean ± SEM of 5-9 rats. Alprostadil and iloprost (1 nM- 10 µM) concentration-dependently potentiated ACh (100 µM)- and EFS (4 Hz)-induced contraction, with maximum potentiation of 40.01 ± 5.29 and 27.59 ± 6.64%, respectively, in case of ACh contractions. In contrast, ONO-AE1-259 (selective EP2 agonist, 1 nM-10 µM) inhibited muscle contraction. SC51322 (EP1-antagonist, 10 µM) and RO1138452 (IP antagonist, 10 µM) inhibited both direct and neurogenic responses. Hemorrhagic cystitis reduced both ACh and EFS responses as well as the potentiatory effect of iloprost and the inhibitory effect of RO1138452 on ACh contractions. ONO-AE3-237 (DP1 antagonist, 1 µM) significantly potentiated contractions in cystitis but showed no effect in normal bladder. A significant inhibition of contractile response was observed in presence of indomethacin, ketoprofen, and diclofenac at all tested concentrations (20, 50, and 100 µM). Highest effect was induced by diclofenac. The effect of these COX inhibitors on EFS contractions was intensified in case of cystitis, indomethacin being the most potent. Atropine (1 nM) significantly reduced indomethacin effect on ACh contraction only in normal rats. On the other hand, DFU (10-6 M) significantly potentiated the contractile effect of ACh in case of cystitis although it showed no effect in normal rats. EP1 receptors seem to play an important role in rat bladder contractility. DP1 receptors as COX-2, on the other hand, gain an important role only in case of cystitis. The use of non-selective COX inhibitors in cystitis may be associated with bladder hypoactivity; selective COX-2 inhibitors may be a safer option.


Subject(s)
Cyclooxygenase Inhibitors/adverse effects , Cystitis/pathology , Muscle, Smooth/drug effects , Receptors, Prostaglandin/antagonists & inhibitors , Urinary Bladder/drug effects , Animals , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Cyclophosphamide , Cystitis/chemically induced , Cystitis/physiopathology , Disease Models, Animal , Male , Muscle Contraction/drug effects , Muscle, Smooth/pathology , Muscle, Smooth/physiology , Rats, Wistar , Receptors, Prostaglandin/physiology , Urinary Bladder/pathology , Urinary Bladder/physiology
6.
J Leukoc Biol ; 104(6): 1173-1186, 2018 12.
Article in English | MEDLINE | ID: mdl-30145840

ABSTRACT

Inflammation-limiting nonsteroidal pain relievers magnify myocardial infarction (MI) incidences and increase re-admission events in heart failure (HF) patients. However, the molecular and cellular mechanism of this provocative adverse effect is unclear. Our goal was to determine whether carprofen (CAP) impedes splenic leukocyte-directed acute inflammation-resolving response in cardiac injury. After subacute CAP treatment, mice were subjected to permanent coronary ligation maintaining MI- and naïve-controls. Spleen and left ventricle (LV) leukocytes were quantitated using flow cytometry pre- and 24 h post-MI. The inflammation resolution mediators were quantified using mass spectrometry while splenocardiac apoptosis and leukocyte phagocytosis were measured by immunofluorescence and ImageStream, respectively. Subacute CAP treatment promoted strain and cardiac dysfunction before MI and coronary occlusion showed signs of acute HF in CAP and MI-controls. Subacute CAP-injected mice had pre-activated splenic neutrophils, an over activated "don't eat me" signal (CD47) with reduced total Mϕs (F4/80+ ) and reparative Mϕs (F4/80/Ly6Clo /CD206) compared with control in LV and spleen. Post-MI, CAP pre-activated neutrophils (Ly6G+ ) were intensified and reduced reparative neutrophils (Ly6G+ /CD206+ ) and Mϕs (F4/80/Ly6Clo ) in LV was indicative of non-resolving inflammation compared with MI-control. Subacute CAP treatment deferred neutrophil phagocytosis functions in the spleen and LV and was more evident post-MI compared with MI-control. CAP pre-activated splenic neutrophils that tailored the Mϕ phagocytosis thereby increased splenocardiac leukocyte death. CAP over amplified COX-1 and COX-2 compared with MI-control and failed to limit prostaglandins and thromboxane in post-MI setting. Further, CAP reduced cardiac-protective epoxyeicosatrienoic acids and over amplified pyrogenic inflammatory cytokines and reduced reparative cytokines, thereby non-resolving inflammation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Carbazoles/toxicity , Heart Ventricles/drug effects , Inflammation/chemically induced , Leukocytes/drug effects , Myocardial Infarction/physiopathology , Spleen/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Carbazoles/pharmacology , Cyclooxygenase 1/physiology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/physiology , Eicosanoids/metabolism , Heart Failure/etiology , Heart Ventricles/immunology , Heart Ventricles/physiopathology , Inflammation/etiology , Inflammation Mediators/metabolism , Leukocytes/immunology , Macrophage Activation/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Neutrophil Activation/drug effects , Phagocytosis/drug effects , Prostaglandins/metabolism , Spleen/immunology , Spleen/physiopathology
7.
J Chin Med Assoc ; 80(8): 467-475, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28676445

ABSTRACT

BACKGROUND: Liver fibrosis causes portal hypertension which dilates collateral vasculature and enhances extra-hepatic angiogenesis including intrapulmonary shunts, which subsequently complicates with hepatopulmonary syndrome. Metformin is an anti-diabetic agent which possesses anti-inflammation and anti-angiogenesis properties. This study evaluated the effect of metformin treatment on liver and lung in a non-diabetic rat model with biliary cirrhosis induced via common bile duct ligation (CBDL). METHODS: CBDL rats were fed with metformin 150 mg/kg/day during the 8th-28th day post operation. The hemodynamic and biochemistry parameters were tested, and blood gas analysis was performed. The liver and lung were dissected for protein analysis and immuno-histochemical stains. Intrapulmonary shunting degree was determined using color microsphere method. RESULTS: Metformin treatment neither induced obvious hypoglycemic event nor altered hemodynamics in cirrhotic rats. The plasma levels of alanine aminotransferase were significantly reduced by metformin (control vs. metformin: 269 ± 56 vs. 199 ± 21 IU/L, P = 0.02). Sirius Red stains and CD-68 stains showed that metformin reduced intrahepatic fibrosis and CD-68-positive macrophages. Metformin did not influence hypoxia and intrapulmonary angiogenesis; however, it significantly reduced intrapulmonary shunts (31.7 ± 10.1 vs. 15.0 ± 6.6%, P = 0.006.). Furthermore, metformin reduced the protein expressions of COX-2 and PI3K in liver and COX-1 in lung. CONCLUSION: Metformin reduced liver injury and improved hepatic fibrosis in cirrhotic rats. It also attenuated the intrapulmonary shunts. However, the effects of metformin on pulmonary angiogenesis and hypoxia were insignificant.


Subject(s)
Hepatopulmonary Syndrome/prevention & control , Liver Cirrhosis, Biliary/drug therapy , Liver Cirrhosis/prevention & control , Metformin/therapeutic use , AMP-Activated Protein Kinases/physiology , Animals , Cyclooxygenase 1/physiology , Male , Rats , Rats, Sprague-Dawley
8.
J Cereb Blood Flow Metab ; 37(3): 1060-1068, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27178425

ABSTRACT

Cortical spreading depression (CSD) is considered a significant phenomenon for human neurological conditions and one of its key signatures is the development of persistent cortical oligemia. The factors underlying this reduction in cerebral blood flow (CBF) remain incompletely understood but may involve locally elaborated vasoconstricting eicosanoids. We employed laser Doppler flowmetry in urethane-anesthetized rats, together with a local pharmacological blockade approach, to test the relative contribution of cyclooxygenase (COX)-derived prostanoids to the oligemic response following CSD. Administration of the non-selective COX inhibitor naproxen completely inhibited the oligemic response. Selective inhibition of COX-1 with SC-560 preferentially reduced the early reduction in CBF while selective COX-2 inhibition with NS-398 affected only the later response. Blocking the action of thromboxane A2 (TXA2), using the selective thromboxane synthase inhibitor ozagrel, reduced only the initial CBF decrease, while inhibition of prostaglandin F2alpha action, using the selective FP receptor antagonist AL-8810, blocked the later phase of the oligemia. Our results suggest that the long-lasting oligemia following CSD consists of at least two distinct temporal phases, mediated by preferential actions of COX-1- and COX-2-derived prostanoids: an initial phase mediated by COX-1 that involves TXA2 followed by a later phase, mediated by COX-2 and PGF2alpha.


Subject(s)
Cerebrovascular Circulation/drug effects , Cortical Spreading Depression/drug effects , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Prostaglandins/physiology , Animals , Cyclooxygenase Inhibitors/pharmacology , Dinoprost/physiology , Laser-Doppler Flowmetry , Rats , Thromboxane A2/physiology , Time Factors
9.
J Chin Med Assoc ; 79(11): 583-588, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27580512

ABSTRACT

BACKGROUND: Hepatic encephalopathy (HE) is a complex neuropsychiatric syndrome secondary to acute or chronic liver failure. However, its pathophysiology remains obscure. Recently, we found that the inhibition of cyclooxygenase by indomethacin aggravated HE in rats with thioacetamide-induced acute hepatic failure, suggesting a pivotal role of cyclooxygenase in HE. This study was aimed at surveying the roles of cyclooxygenase isoforms responsible for prostaglandins synthesis, cyclooxygenase-1 (COX1) and COX2, in cirrhotic rats with HE. METHODS: Liver cirrhosis was induced (using formalin) in male Sprague-Dawley rats with bile duct ligation (FBDL). Sham-operated rats served as the surgical controls. The severity of HE was assessed by motor activity counts. Plasma 6-keto-prostaglandin-F1α [6-keto-PGF1α; a relatively stable metabolite of prostacyclin (PGI2)], alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALK-P), and total bilirubin were measured. Hepatic mRNA expressions of COX1 and COX2 were tested. RESULTS: The FBDL group showed lower motor activity counts than the sham group in total (1472 ± 156 vs. 2174 ± 262 counts/30 min, p = 0.034), ambulatory (824 ± 99 vs. 1443 ± 206 counts/30 min, p = 0.014), and vertical movement (431 ± 69 vs. 849 ± 145 counts/30 min, p = 0.018). The mRNA expression of hepatic COX2 was significantly higher in the FBDL group. Plasma ALK-P and bilirubin levels were negatively correlated with total movements, respectively (both p < 0.05). In addition, hepatic COX2 mRNA expression was positively correlated with AST, ALK-P, total bilirubin, and 6-keto-PGF1α (all p < 0.05), but not correlated with total movements. CONCLUSION: Hepatic COX2 expression and PGI2 production are enhanced in cirrhotic rats, but the correlation with HE is not remarkable. Cyclooxygenase and PGI2 may not play important roles in HE in the setting of chronic liver failure.


Subject(s)
Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Hepatic Encephalopathy/etiology , Liver Cirrhosis/complications , 6-Ketoprostaglandin F1 alpha/blood , Animals , Epoprostenol/biosynthesis , Liver/enzymology , Male , Motor Activity , Rats , Rats, Sprague-Dawley
10.
J Neurosci ; 35(39): 13463-74, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424891

ABSTRACT

According to the current model of neurovascular coupling, blood flow is controlled regionally through phasic changes in the activity of neurons and astrocytes that signal to alter arteriole diameter. Absent in this model, however, is how brain blood flow is tonically regulated independent of regional changes in activity. This is important because a large fraction of brain blood flow is required to maintain basal metabolic needs. Using two-photon fluorescence imaging combined with patch-clamp in acute rat brain slices of sensory-motor cortex, we demonstrate that reducing resting Ca(2+) in astrocytes with intracellular BAPTA causes vasoconstriction in adjacent arterioles. BAPTA-induced vasoconstriction was eliminated by a general COX blocker and the effect is mimicked by a COX-1, but not COX-2, antagonist, suggesting that astrocytes provide tonic, steady-state vasodilation by releasing prostaglandin messengers. Tonic vasodilation was insensitive to TTX, as well as a variety of synaptic and extrasynaptic receptor antagonists, indicating that the phenomenon operates largely independent of neural activity. Using in vivo two-photon fluorescence imaging of the barrel cortex in fully awake mice, we reveal that acute COX-1 inhibition reduces resting arteriole diameter but fails to affect vasodilation in response to vibrissae stimulation. Our findings demonstrate that astrocytes provide tonic regulation of arterioles using resting intracellular Ca(2+) in a manner that is independent of phasic, neuronal-evoked vasodilation. Significance statement: The brain requires both phasic and tonic regulation of its blood supply to service energy needs over various temporal windows. While many mechanisms have been described for phasic blood flow regulation, how the brain accomplishes tonic control is largely unknown. Here we describe a way in which astrocytes contribute to the management of basal brain blood flow by providing steady-state vasodilation to arterioles via resting astrocyte Ca(2+) and the continuous release of prostaglandin messengers. This phenomenon may be important for understanding the declines in basal brain blood flow that occur in aging and dementia, as well as for the interpretation of fMRI data.


Subject(s)
Astrocytes/physiology , Cerebrovascular Circulation/physiology , Neurovascular Coupling/physiology , Animals , Arterioles/physiology , Chelating Agents/pharmacology , Cyclooxygenase 1/drug effects , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , In Vitro Techniques , Male , Mice , Neurons/physiology , Patch-Clamp Techniques , Physical Stimulation , Prostaglandins/metabolism , Rats , Rats, Sprague-Dawley , Somatosensory Cortex/physiology , Tetrodotoxin/pharmacology , Vasoconstriction/physiology , Vibrissae/innervation
11.
PLoS One ; 10(8): e0136738, 2015.
Article in English | MEDLINE | ID: mdl-26308616

ABSTRACT

This study tested the hypothesis that in hypertensive arteries cyclooxygenase-1 (COX-1) remains as a major form, mediating prostacyclin (prostaglandin I2; PGI2) synthesis that may evoke a vasoconstrictor response in the presence of functional vasodilator PGI2 (IP) receptors. Two-kidney-one-clip (2K1C) hypertension was induced in wild-type (WT) mice and/or those with COX-1 deficiency (COX-1-/-). Carotid arteries were isolated for analyses 4 weeks after. Results showed that as in normotensive mice, the muscarinic receptor agonist ACh evoked a production of the PGI2 metabolite 6-keto-PGF1α and an endothelium-dependent vasoconstrictor response; both of them were abolished by COX-1 inhibition. At the same time, PGI2, which evokes contraction of hypertensive vessels, caused relaxation after thromboxane-prostanoid (TP) receptor antagonism that abolished the contraction evoked by ACh. Antagonizing IP receptors enhanced the contraction to the COX substrate arachidonic acid (AA). Also, COX-1-/- mice was noted to develop hypertension; however, their increase of blood pressure and/or heart mass was not to a level achieved with WT mice. In addition, we found that either the contraction in response to ACh or that evoked by AA was abolished in COX-1-/- hypertensive mice. These results demonstrate that as in normotensive conditions, COX-1 is a major contributor of PGI2 synthesis in 2K1C hypertensive carotid arteries, which leads to a vasoconstrictor response resulting from opposing dilator and vasoconstrictor activities of IP and TP receptors, respectively. Also, our data suggest that COX-1-/- attenuates the development of 2K1C hypertension in mice, reflecting a net adverse role yielded from all COX-1-mediated activities under the pathological condition.


Subject(s)
Carotid Arteries/metabolism , Cyclooxygenase 1/physiology , Epoprostenol/metabolism , Hypertension/physiopathology , Renal Artery/surgery , Vasoconstriction , Vasomotor System/physiology , Animals , Carotid Arteries/pathology , Hypertension/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, Thromboxane/metabolism
12.
Eur J Pharmacol ; 764: 249-255, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26151307

ABSTRACT

The soluble guanylyl cyclase/cGMP system plays an important role in the vasodilator response to nitric oxide (NO) in various vascular beds. However, in rat retinal arterioles, the cyclooxygenase-1/cAMP-mediated pathway contributes to the vasodilator effects of NO, although the specific prostanoid involved remains to be elucidated. In the present study, we investigated the role of prostaglandin I2 and its receptor (prostanoid IP receptor) system in NO-induced vasodilation of rat retinal arterioles in vivo. Fundus images were captured using a digital camera that was equipped with a special objective lens. Changes in diameter of retinal arterioles were assessed. The NO donor (±)-(E)-4-ethyl-2-[(E)-hydroxyimino]-5-nitro-3-hexenamide (NOR3) increased the diameter of retinal arterioles but decreased systemic blood pressure in a dose-dependent manner. Treatment of rats with indomethacin, a non-selective cyclooxygenase inhibitor, markedly attenuated the retinal vasodilator, but not depressor responses to NOR3. The prostanoid IP receptor antagonist 4,5-dihydro-N-[4-[[4-(1-methylethoxy)phenyl]methyl]phenyl]-1H-imadazol-2-amine (CAY10441), and the prostaglandin I2 synthase inhibitor 9α,11α-azoprosta-5Z,13E-dien-1-oic acid (U-51605), both showed similar preventive effects against the NOR3-induced retinal vasodilator response. Neither CAY10441 nor U-51605 showed any significant effects on the depressor response to NOR3. NOR3 enhanced the release of prostaglandin I2 from cultured human retinal microvascular endothelial cells and the NOR3-induced prostaglandin I2 release was almost completely abolished by the cyclooxygenase-1 inhibitor SC-560, but not by the cyclooxygenase-2 inhibitor NS-398. However, NOR3 did not increase the release of prostaglandin I2 from human intestinal microvascular endothelial cells. These results suggest that NO exerts its dilatory effect via cyclooxygenase-1/prostaglandin I2/prostanoid IP receptor signaling mechanisms in the retinal vasculature.


Subject(s)
Arterioles/physiology , Epoprostenol/physiology , Retinal Vessels/physiology , Animals , Arterioles/drug effects , Benzyl Compounds/pharmacology , Cells, Cultured , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/pharmacology , Endothelial Cells/metabolism , Humans , Hydroxylamines/pharmacology , Imidazoles/pharmacology , Male , Nitric Oxide/physiology , Nitric Oxide Donors/pharmacology , Nitro Compounds , Nitrobenzenes/pharmacology , Prostaglandins H/pharmacology , Pyrazoles/pharmacology , Rats, Wistar , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/physiology , Retinal Vessels/drug effects , Sulfonamides/pharmacology , Vasodilation/drug effects
13.
J Gastroenterol Hepatol ; 29 Suppl 4: 3-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25521725

ABSTRACT

BACKGROUND AND AIM: We investigated the roles of cyclooxygenase (COX) isozymes and prostaglandins (PGs) and their receptors in mucosal defense against cold-restraint stress (CRS)-induced gastric lesions. METHODS: Male C57BL/6 wild-type (WT) mice and those lacking COX-1 or COX-2 as well as those lacking EP1, EP3, or IP receptors were used after 18 h fasting. Animals were restrained in Bollman cages and kept in a cold room at 10°C for 90 min. RESULTS: CRS induced multiple hemorrhagic lesions in WT mouse stomachs. The severity of these lesions was significantly worsened by pretreatment with the nonselective COX inhibitors (indomethacin, loxoprofen) or selective COX-1 inhibitor (SC-560), while neither of the selective COX-2 inhibitors (rofecoxib and celecoxib) had any effect. These lesions were also aggravated in animals lacking COX-1, but not COX-2. The expression of COX-2 mRNA was not detected in the stomach after CRS, while COX-1 expression was observed under normal and stressed conditions. The gastric ulcerogenic response to CRS was similar between EP1 or EP3 knockout mice and WT mice, but was markedly worsened in animals lacking IP receptors. Pretreating WT mice with iloprost (the PGI2 analog) significantly prevented CRS-induced gastric lesions in the presence of indomethacin. PGE2 also reduced the severity of these lesions, and the effect was mimicked by the EP4 agonist, AE1-329. CONCLUSIONS: These results suggest that endogenous PGs derived from COX-1 play a crucial role in gastric mucosal defense during CRS, and this action is mainly mediated by PGI2 /IP receptors and partly by PGE2 /EP4 receptors.


Subject(s)
Cold Temperature/adverse effects , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/adverse effects , Gastric Mucosa/pathology , Gastrointestinal Hemorrhage/etiology , Gastrointestinal Hemorrhage/pathology , Prostaglandins I/physiology , Stress, Physiological/physiology , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Dinoprostone/physiology , Epoprostenol/physiology , Gene Expression , Indomethacin/adverse effects , Male , Mice, Inbred C57BL , Phenylpropionates/adverse effects , Pyrazoles/adverse effects , RNA, Messenger/metabolism , Receptors, Epoprostenol/physiology , Receptors, Prostaglandin E/physiology , Receptors, Prostaglandin E, EP4 Subtype/physiology
14.
Braz. j. med. biol. res ; 47(12): 1050-1056, 12/2014. graf
Article in English | LILACS | ID: lil-727667

ABSTRACT

People who suffer from traumatic brain injury (TBI) often experience cognitive deficits in spatial reference and working memory. The possible roles of cyclooxygenase-1 (COX-1) in learning and memory impairment in mice with TBI are far from well known. Adult mice subjected to TBI were treated with the COX-1 selective inhibitor SC560. Performance in the open field and on the beam walk was then used to assess motor and behavioral function 1, 3, 7, 14, and 21 days following injury. Acquisition of spatial learning and memory retention was assessed using the Morris water maze on day 15 post-TBI. The expressions of COX-1, prostaglandin E2 (PGE2), interleukin (IL)-6, brain-derived neurotrophic factor (BDNF), platelet-derived growth factor BB (PDGF-BB), synapsin-I, and synaptophysin were detected in TBI mice. Administration of SC560 improved performance of beam walk tasks as well as spatial learning and memory after TBI. SC560 also reduced expressions of inflammatory markers IL-6 and PGE2, and reversed the expressions of COX-1, BDNF, PDGF-BB, synapsin-I, and synaptophysin in TBI mice. The present findings demonstrated that COX-1 might play an important role in cognitive deficits after TBI and that selective COX-1 inhibition should be further investigated as a potential therapeutic approach for TBI.


Subject(s)
Animals , Brain Injuries/complications , Cerebral Cortex/injuries , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/therapeutic use , Learning/drug effects , Memory Disorders/drug therapy , Pyrazoles/therapeutic use , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Decortication , Cyclooxygenase 1/metabolism , Disease Models, Animal , Dinoprostone/analysis , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Hippocampus/metabolism , /blood , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Recovery of Function/drug effects , Synaptophysin/analysis , Synaptophysin/metabolism
15.
Braz J Med Biol Res ; 47(12): 1050-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25387671

ABSTRACT

People who suffer from traumatic brain injury (TBI) often experience cognitive deficits in spatial reference and working memory. The possible roles of cyclooxygenase-1 (COX-1) in learning and memory impairment in mice with TBI are far from well known. Adult mice subjected to TBI were treated with the COX-1 selective inhibitor SC560. Performance in the open field and on the beam walk was then used to assess motor and behavioral function 1, 3, 7, 14, and 21 days following injury. Acquisition of spatial learning and memory retention was assessed using the Morris water maze on day 15 post-TBI. The expressions of COX-1, prostaglandin E2 (PGE2), interleukin (IL)-6, brain-derived neurotrophic factor (BDNF), platelet-derived growth factor BB (PDGF-BB), synapsin-I, and synaptophysin were detected in TBI mice. Administration of SC560 improved performance of beam walk tasks as well as spatial learning and memory after TBI. SC560 also reduced expressions of inflammatory markers IL-6 and PGE2, and reversed the expressions of COX-1, BDNF, PDGF-BB, synapsin-I, and synaptophysin in TBI mice. The present findings demonstrated that COX-1 might play an important role in cognitive deficits after TBI and that selective COX-1 inhibition should be further investigated as a potential therapeutic approach for TBI.


Subject(s)
Brain Injuries/complications , Cerebral Cortex/injuries , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/therapeutic use , Learning/drug effects , Memory Disorders/drug therapy , Pyrazoles/therapeutic use , Animals , Becaplermin , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Decortication , Cyclooxygenase 1/metabolism , Dinoprostone/analysis , Dinoprostone/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hippocampus/metabolism , Interleukin-6/blood , Maze Learning/drug effects , Memory Disorders/etiology , Memory Disorders/metabolism , Mice, Inbred C57BL , Proto-Oncogene Proteins c-sis/metabolism , Recovery of Function/drug effects , Synaptophysin/analysis , Synaptophysin/metabolism
16.
PLoS One ; 9(9): e107503, 2014.
Article in English | MEDLINE | ID: mdl-25238071

ABSTRACT

We sought to determine a role for platelets in in vivo angiogenesis, quantified by changes in the capillary to fibre ratio (C:F) of mouse skeletal muscle, utilising two distinct forms of capillary growth to identify differential effects. Capillary sprouting was induced by muscle overload, and longitudinal splitting by chronic hyperaemia. Platelet depletion was achieved by anti-GPIbα antibody treatment. Sprouting induced a significant increase in C:F (1.42±0.02 vs. contralateral 1.29±0.02, P<0.001) that was abolished by platelet depletion, while the significant C:F increase caused by splitting (1.40±0.03 vs. control 1.28±0.03, P<0.01) was unaffected. Granulocyte/monocyte depletion showed this response was not immune-regulated. VEGF overexpression failed to rescue angiogenesis following platelet depletion, suggesting the mechanism is not simply reliant on growth factor release. Sprouting occurred normally following antibody-induced GPVI shedding, suggesting platelet activation via collagen is not involved. BrdU pulse-labelling showed no change in the proliferative potential of cells associated with capillaries after platelet depletion. Inhibition of platelet activation by acetylsalicylic acid abolished sprouting, but not splitting angiogenesis, paralleling the response to platelet depletion. We conclude that platelets differentially regulate mechanisms of angiogenesis in vivo, likely via COX signalling. Since endothelial proliferation is not impaired, we propose a link between COX1 and induction of endothelial migration.


Subject(s)
Blood Platelets/physiology , Cyclooxygenase 1/physiology , Membrane Proteins/physiology , Neovascularization, Physiologic/physiology , Animals , Capillaries/growth & development , Capillaries/physiology , Cell Proliferation , Cyclooxygenase 1/metabolism , Membrane Proteins/metabolism , Mice , Vascular Endothelial Growth Factor A/metabolism
17.
Mol Hum Reprod ; 20(11): 1144-57, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25103627

ABSTRACT

The interplay between inflammation, cervical cancer and HIV acquisition in women is poorly understood. We have previously shown that seminal plasma (SP) can promote cervical tumour cell growth in vitro and in vivo via the activation of potent inflammatory pathways. In this study, we investigated whether SP could regulate expression of chemokine receptors with known roles in HIV infection, in the cervix and in cervical cancer. The expression of CD4 and CCR5 was investigated by RT-PCR analysis and immunohistochemistry. CD4 and CCR5 expression was elevated in cervical cancer tissue compared with normal cervix. Ex vivo studies conducted on cervical tissues and HeLa cells showed that SP significantly increases the expression of CD4 and CCR5 transcripts. Furthermore, it was found that SP also up-regulates CCR5 protein in HeLa cells. The regulation of CCR5 expression was investigated following treatment of HeLa cells with SP in the presence/absence of chemical inhibitors of intracellular signalling, EP2 and EP4 antagonists, prostaglandin (PG) E2 and a cyclooxygenase (COX)-1 doxycycline-inducible expression system. These experiments demonstrated that the regulation of CCR5 expression by SP occurs via the epidermal growth factor receptor (EGFR)-COX-1-PGE2 pathway. This study provides a link between activation of inflammatory pathways and regulation of HIV receptor expression in cervical cancer cells.


Subject(s)
Receptors, CCR5/metabolism , Semen/metabolism , Up-Regulation , Uterine Cervical Neoplasms/genetics , Adult , CD4 Antigens/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 1/physiology , Female , HeLa Cells , Humans , Immunohistochemistry , Male , RNA, Messenger/metabolism , Receptors, CCR5/genetics , Uterine Cervical Neoplasms/metabolism
18.
J Am Heart Assoc ; 3(3): e000875, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24815497

ABSTRACT

BACKGROUND: Obstructive sleep apnea (OSA) is associated with increased risk of cardiovascular and cerebrovascular disease resulting from intermittent hypoxia (IH)-induced inflammation. Cyclooxygenase (COX)-formed prostanoids mediate the inflammatory response, and regulate blood pressure and cerebral blood flow (CBF), but their role in blood pressure and CBF responses to IH is unknown. Therefore, this study's objective was to determine the role of prostanoids in cardiovascular and cerebrovascular responses to IH. METHODS AND RESULTS: Twelve healthy, male participants underwent three, 6-hour IH exposures. For 4 days before each IH exposure, participants ingested a placebo, indomethacin (nonselective COX inhibitor), or Celebrex(®) (selective COX-2 inhibitor) in a double-blind, randomized, crossover study design. Pre- and post-IH blood pressure, CBF, and urinary prostanoids were assessed. Additionally, blood pressure and urinary prostanoids were assessed in newly diagnosed, untreated OSA patients (n=33). Nonselective COX inhibition increased pre-IH blood pressure (P ≤ 0.04) and decreased pre-IH CBF (P=0.04) while neither physiological variable was affected by COX-2 inhibition (P ≥ 0.90). Post-IH, MAP was elevated (P ≤ 0.05) and CBF was unchanged with placebo and nonselective COX inhibition. Selective COX-2 inhibition abrogated the IH-induced MAP increase (P=0.19), but resulted in lower post-IH CBF (P=0.01). Prostanoids were unaffected by IH, except prostaglandin E2 was elevated with the placebo (P=0.02). Finally, OSA patients had elevated blood pressure (P ≤ 0.4) and COX-1 formed thromboxane A2 concentrations (P=0.02). CONCLUSIONS: COX-2 and COX-1 have divergent roles in modulating vascular responses to acute and chronic IH. Moreover, COX-1 inhibition may mitigate cardiovascular and cerebrovascular morbidity in OSA. CLINICAL TRIAL REGISTRATION URL: www.clinicaltrials.gov. Unique identifier: NCT01280006.


Subject(s)
Blood Pressure/physiology , Cerebrovascular Circulation/physiology , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Hypoxia/physiopathology , Sleep Apnea, Obstructive/physiopathology , Adult , Blood Pressure/drug effects , Celecoxib , Cerebrovascular Circulation/drug effects , Cross-Over Studies , Cyclooxygenase 1/drug effects , Cyclooxygenase 2/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dinoprost/urine , Dinoprostone/urine , Double-Blind Method , Epoprostenol/urine , Female , Heart Rate/drug effects , Heart Rate/physiology , Humans , Indomethacin/pharmacology , Male , Middle Aged , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Thromboxane A2/urine
19.
Br J Pharmacol ; 171(15): 3728-40, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24758697

ABSTRACT

BACKGROUND AND PURPOSE: The COX isoforms (COX-1, COX-2) regulate human gut motility, although their role under pathological conditions remains unclear. This study examines the effects of COX inhibitors on excitatory motility in colonic tissue from patients with diverticular disease (DD). EXPERIMENTAL APPROACH: Longitudinal muscle preparations, from patients with DD or uncomplicated cancer (controls), were set up in organ baths and connected to isotonic transducers. Indomethacin (COX-1/COX-2 inhibitor), SC-560 (COX-1 inhibitor) or DFU (COX-2 inhibitor) were assayed on electrically evoked, neurogenic, cholinergic and tachykininergic contractions, or carbachol- and substance P (SP)-induced myogenic contractions. Distribution and expression of COX isoforms in the neuromuscular compartment were assessed by RT-PCR, Western blot and immunohistochemical analysis. KEY RESULTS: In control preparations, neurogenic cholinergic contractions were enhanced by COX inhibitors, whereas tachykininergic responses were blunted. Carbachol-evoked contractions were increased by indomethacin or SC-560, but not DFU, whereas all inhibitors reduced SP-induced motor responses. In preparations from DD patients, COX inhibitors did not affect electrically evoked cholinergic contractions. Both indomethacin and DFU, but not SC-560, decreased tachykininergic responses. COX inhibitors did not modify carbachol-evoked motor responses, whereas they counteracted SP-induced contractions. COX-1 expression was decreased in myenteric neurons, whereas COX-2 was enhanced in glial cells and smooth muscle. CONCLUSIONS AND IMPLICATIONS: In control colon, COX-1 and COX-2 down-regulate cholinergic motility, whereas both isoforms enhance tachykininergic motor activity. In the presence of DD, there is a loss of modulation by both COX isoforms on the cholinergic system, whereas COX-2 displays an enhanced facilitatory control on tachykininergic contractile activity.


Subject(s)
Colon/physiology , Cyclooxygenase 1/physiology , Cyclooxygenase 2/physiology , Diverticulitis, Colonic/physiopathology , Adult , Aged , Aged, 80 and over , Colon/drug effects , Cyclooxygenase Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Indomethacin/pharmacology , Male , Middle Aged , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Pyrazoles/pharmacology
20.
Pharmacol Res ; 81: 74-82, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24548820

ABSTRACT

OBJECTIVE: The vasorelaxant effect of 2-arachidonoylglycerol (2-AG) has been well characterised in animals. 2-AG is present in human vascular cells and is up-regulated in cardiovascular pathophysiology. However, the acute vascular actions of 2-AG have not been explored in humans. APPROACH: Mesenteric arteries were obtained from patients receiving colorectal surgery and mounted on a myograph. Arteries were contracted and 2-AG concentration-response curves were carried out. Mechanisms of action were characterised pharmacologically. Post hoc analysis was carried out to assess the effects of cardiovascular disease/risk factors on 2-AG responses. RESULTS: 2-AG caused vasorelaxation of human mesenteric arteries, independent of cannabinoid receptor or transient receptor potential vanilloid-1 activation, the endothelium, nitric oxide or metabolism via monoacyglycerol lipase or fatty acid amide hydrolase. 2-AG-induced vasorelaxation was reduced in the presence of indomethacin and flurbiprofen, suggesting a role for cyclooxygenase metabolism 2-AG. Responses to 2-AG were also reduced in the presence of Cay10441, L-161982 and potentiated in the presence of AH6809, suggesting that metabolism of 2-AG produces both vasorelaxant and vasoconstrictor prostanoids. Finally, 2-AG-induced vasorelaxation was dependent on potassium efflux and the presence of extracellular calcium. CONCLUSIONS: We have shown for the first time that 2-AG causes vasorelaxation of human mesenteric arteries. Vasorelaxation is dependent on COX metabolism, activation of prostanoid receptors (EP4 & IP) and ion channel modulation. 2-AG responses are blunted in patients with cardiovascular risk factors.


Subject(s)
Arachidonic Acids/pharmacology , Cyclooxygenase 1/physiology , Endocannabinoids/pharmacology , Glycerides/pharmacology , Mesenteric Arteries/drug effects , Vasodilation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Flurbiprofen/pharmacology , Humans , In Vitro Techniques , Indomethacin/pharmacology , Mesenteric Arteries/physiology , Vasodilation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...