Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R468-R479, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31868517

ABSTRACT

Cyclooxygenase (COX) is proposed to regulate cerebral blood flow (CBF); however, accurate regional contributions of COX are relatively unknown at baseline and particularly during hypoxia. We hypothesized that COX contributes to both basal and hypoxic cerebral vasodilation, but COX-mediated vasodilation is greater in the posterior versus anterior cerebral circulation. CBF was measured in 9 healthy adults (28 ± 4 yr) during normoxia and isocapnic hypoxia (fraction of inspired oxygen = 0.11), with COX inhibition (oral indomethacin, 100mg) or placebo. Four-dimensional flow magnetic resonance imaging measured cross-sectional area (CSA) and blood velocity to quantify CBF in 11 cerebral arteries. Cerebrovascular conductance (CVC) was calculated (CVC = CBF × 100/mean arterial blood pressure) and hypoxic reactivity was expressed as absolute and relative change in CVC [ΔCVC/Δ pulse oximetry oxygen saturation (SpO2)]. At normoxic baseline, indomethacin reduced CVC by 44 ± 5% (P < 0.001) and artery CSA (P < 0.001), which was similar across arteries. Hypoxia (SpO2 80%-83%) increased CVC (P < 0.01), reflected as a similar relative increase in reactivity (% ΔCVC/-ΔSpO2) across arteries (P < 0.05), in part because of increases in CSA (P < 0.05). Indomethacin did not alter ΔCVC or ΔCVC/ΔSpO2 to hypoxia. These findings indicate that 1) COX contributes, in a largely uniform fashion, to cerebrovascular tone during normoxia and 2) COX is not obligatory for hypoxic vasodilation in any regions supplied by large extracranial or intracranial arteries.


Subject(s)
Cerebral Arteries/enzymology , Cerebrovascular Circulation , Hypoxia/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Vasodilation , Adult , Biomarkers/blood , Blood Flow Velocity , Cerebral Arteries/diagnostic imaging , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Cerebrovascular Circulation/drug effects , Cyclooxygenase Inhibitors/administration & dosage , Double-Blind Method , Female , Humans , Hypoxia/blood , Hypoxia/diagnostic imaging , Hypoxia/physiopathology , Indomethacin/administration & dosage , Male , Oxygen/blood , Random Allocation , Vasodilation/drug effects , Young Adult
2.
Arterioscler Thromb Vasc Biol ; 38(3): 610-621, 2018 03.
Article in English | MEDLINE | ID: mdl-29348119

ABSTRACT

OBJECTIVE: Cigarette smoke exposure (CSE) is a risk factor for cerebral aneurysm (CA) formation, but the molecular mechanisms are unclear. Although CSE is known to contribute to excess reactive oxygen species generation, the role of oxidative stress on vascular smooth muscle cell (VSMC) phenotypic modulation and pathogenesis of CAs is unknown. The goal of this study was to investigate whether CSE activates a NOX (NADPH oxidase)-dependent pathway leading to VSMC phenotypic modulation and CA formation and rupture. APPROACH AND RESULTS: In cultured cerebral VSMCs, CSE increased expression of NOX1 and reactive oxygen species which preceded upregulation of proinflammatory/matrix remodeling genes (MCP-1, MMPs [matrix metalloproteinase], TNF-α, IL-1ß, NF-κB, KLF4 [Kruppel-like factor 4]) and downregulation of contractile genes (SM-α-actin [smooth muscle α actin], SM-22α [smooth muscle 22α], SM-MHC [smooth muscle myosin heavy chain]) and myocardin. Inhibition of reactive oxygen species production and knockdown of NOX1 with siRNA or antisense decreased CSE-induced upregulation of NOX1 and inflammatory genes and downregulation of VSMC contractile genes and myocardin. p47phox-/- NOX knockout mice, or pretreatment with the NOX inhibitor, apocynin, significantly decreased CA formation and rupture compared with controls. NOX1 protein and mRNA expression were similar in p47phox-/- mice and those pretreated with apocynin but were elevated in unruptured and ruptured CAs. CSE increased CA formation and rupture, which was diminished with apocynin pretreatment. Similarly, NOX1 protein and mRNA and reactive oxygen species were elevated by CSE, and in unruptured and ruptured CAs. CONCLUSIONS: CSE initiates oxidative stress-induced phenotypic modulation of VSMCs and CA formation and rupture. These molecular changes implicate oxidative stress in the pathogenesis of CAs and may provide a potential target for future therapeutic strategies.


Subject(s)
Aneurysm, Ruptured/enzymology , Cigarette Smoking/adverse effects , Intracranial Aneurysm/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , NADPH Oxidases/metabolism , Oxidative Stress , Smoke , Acetophenones/pharmacology , Aneurysm, Ruptured/genetics , Aneurysm, Ruptured/pathology , Aneurysm, Ruptured/prevention & control , Animals , Antioxidants/pharmacology , Cells, Cultured , Cerebral Arteries/enzymology , Cerebral Arteries/pathology , Dilatation, Pathologic , Disease Models, Animal , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Intracranial Aneurysm/prevention & control , Kruppel-Like Factor 4 , Male , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , NADPH Oxidase 1/genetics , NADPH Oxidase 1/metabolism , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , Phenotype , Rats, Sprague-Dawley , Signal Transduction , Vascular Remodeling
3.
Stroke ; 49(8): 1977-1980, 2018 08.
Article in English | MEDLINE | ID: mdl-29986930

ABSTRACT

Background and Purpose- Absent or diminished α-galactosidase A (GLA) and acid α-glucosidase (GAA) enzyme activity are core features of Fabry and Pompe disease, respectively. Patients with Fabry or Pompe disease may have dilated intracranial arteries but whether lower GLA or GAA enzyme activity relates to brain arterial dilatation in other populations is unknown. Methods- Participants included Parkinson disease patients and nonblood-related controls, whose GLA and GAA enzymatic activities were measured in dried blood spots. Independent readers measured the axial arterial diameter of the ascending portion of the cavernous internal carotid arteries and the most proximal segment of the basilar artery in T2 black voids. Linear regression models were built to investigate the relationship between brain arterial diameters and lysosomal enzymatic activities. Results- The cohort included 107 participants (mean age, 66.5±10.3; 67% men). In an adjusted linear regression model, lower GLA activity was associated with larger brain arterial diameters (B=0.50±0.23, P=0.03). The strength of association was the greatest for the basilar artery diameter (B=0.80±0.33, P=0.02). Similarly, lower GAA activity was associated with an increased basilar arterial diameter (B=0.73±0.35, P=0.04). Conclusions- Lower GLA and GAA enzymatic activities were associated with larger brain arterial diameters, particularly the basilar artery diameter. Lower lysosomal enzymatic function in patients without Fabry or Pompe disease may play a role in brain arterial dilatation.


Subject(s)
Cerebral Arteries/diagnostic imaging , Cerebral Arteries/enzymology , Glucan 1,4-alpha-Glucosidase/metabolism , Lysosomes/enzymology , alpha-Galactosidase/metabolism , Aged , Brain/blood supply , Brain/diagnostic imaging , Brain/enzymology , Cohort Studies , Dilatation, Pathologic/enzymology , Enzyme Activation/physiology , Female , Humans , Male , Middle Aged , Parkinsonian Disorders/diagnostic imaging , Parkinsonian Disorders/enzymology
4.
Cerebrovasc Dis ; 45(3-4): 180-186, 2018.
Article in English | MEDLINE | ID: mdl-29614486

ABSTRACT

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) from intracranial aneurysm rupture results in significant morbidity and mortality. In the present study, we examined the effect of most widely used antiplatelet drugs, aspirin and cilostazol, on aneurysm rupture prevention using a mouse intracranial aneurysm model. MATERIALS AND METHODS: Intracranial aneurysms were induced by a combination of deoxycorticosterone acetate-salt and a single injection of elastase into the cerebrospinal fluid in mice. Treatment with aspirin or cilostazol was started 1 day after aneurysm induction. Aneurysm rupture was detected by neurological symptoms and the presence of intracranial aneurysm with SAH was confirmed by post-mortem examination. RESULTS: Aspirin (10 mg/kg) significantly reduced aneurysm rupture (control:aspirin = 80%:31%, p < 0.05) without affecting the overall incidence of aneurysm formation (60%:62%). Cilostazol (3 mg/kg, 30 mg/kg) did not reduce both rupture rate (control:3 mg/kg:30 mg/kg = 81%:67%:77%) and the overall incidence of aneurysm formation (control:3 mg/kg:30 mg/kg = 72%:71%:76%). Tail vein bleeding time prolonged significantly in both aspirin and cilostazol groups (p < 0.01). CONCLUSION: Aspirin prevented aneurysm rupture in a mouse intracranial aneurysm model, while cilostazol did not. Aspirin, the most frequently used drug for patients with ischemic myocardial and cerebral diseases, is also effective in preventing cerebral aneurysmal rupture.


Subject(s)
Aneurysm, Ruptured/prevention & control , Aspirin/pharmacology , Cerebral Arteries/drug effects , Cilostazol/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Intracranial Aneurysm/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Subarachnoid Hemorrhage/prevention & control , Aneurysm, Ruptured/chemically induced , Aneurysm, Ruptured/enzymology , Aneurysm, Ruptured/pathology , Animals , Cerebral Arteries/enzymology , Cerebral Arteries/pathology , Cyclooxygenase 2/metabolism , Desoxycorticosterone Acetate , Disease Models, Animal , Intracranial Aneurysm/chemically induced , Intracranial Aneurysm/enzymology , Intracranial Aneurysm/pathology , Male , Mice, Inbred C57BL , Pancreatic Elastase , Subarachnoid Hemorrhage/chemically induced , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology
5.
Am J Physiol Cell Physiol ; 310(11): C885-93, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27076616

ABSTRACT

Plasma membrane-localized CaV1.2 channels are the primary calcium (Ca(2+)) influx pathway in arterial smooth muscle cells (myocytes). CaV1.2 channels regulate several cellular functions, including contractility and gene expression, but the trafficking pathways that control the surface expression of these proteins are unclear. Similarly, expression and physiological functions of small Rab GTPases, proteins that control vesicular trafficking in arterial myocytes, are poorly understood. Here, we investigated Rab proteins that control functional surface abundance of CaV1.2 channels in cerebral artery myocytes. Western blotting indicated that Rab25, a GTPase previously associated with apical recycling endosomes, is expressed in cerebral artery myocytes. Immunofluorescence Förster resonance energy transfer (immunoFRET) microscopy demonstrated that Rab25 locates in close spatial proximity to CaV1.2 channels in myocytes. Rab25 knockdown using siRNA reduced CaV1.2 surface and intracellular abundance in arteries, as determined using arterial biotinylation. In contrast, CaV1.2 was not located nearby Rab11A or Rab4 and CaV1.2 protein was unaltered by Rab11A or Rab4A knockdown. Rab25 knockdown resulted in CaV1.2 degradation by a mechanism involving both lysosomal and proteasomal pathways and reduced whole cell CaV1.2 current density but did not alter voltage dependence of current activation or inactivation in isolated myocytes. Rab25 knockdown also inhibited depolarization (20-60 mM K(+)) and pressure-induced vasoconstriction (myogenic tone) in cerebral arteries. These data indicate that Rab25 is expressed in arterial myocytes where it promotes surface expression of CaV1.2 channels to control pressure- and depolarization-induced vasoconstriction.


Subject(s)
Calcium Channels, L-Type/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , rab GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Cerebral Arteries/enzymology , Lysosomes/metabolism , Male , Membrane Potentials , Proteasome Endopeptidase Complex/metabolism , Protein Transport , Proteolysis , RNA Interference , Rats, Sprague-Dawley , Signal Transduction , Transfection , Vasoconstriction , rab GTP-Binding Proteins/genetics
6.
J Biol Chem ; 287(19): 15966-80, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22403397

ABSTRACT

The microvascular endothelial surface expresses multiple molecules whose sialylation state regulates multiple aspects of endothelial function. To better regulate these sialoproteins, we asked whether endothelial cells (ECs) might express one or more catalytically active sialidases. Human lung microvascular EC lysates contained heat-labile sialidase activity for a fluorogenic substrate, 2'-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (4-MU-NANA), that was dose-dependently inhibited by the competitive sialidase inhibitor, 2,3-dehydro-2-deoxy-N-acetylneuraminic acid but not its negative control. The EC lysates also contained sialidase activity for a ganglioside mixture. Using real time RT-PCR to detect mRNAs for the four known mammalian sialidases, NEU1, -2, -3, and -4, NEU1 mRNA was expressed at levels 2700-fold higher that those found for NEU2, -3, or -4. Western analyses indicated NEU1 and -3 protein expression. Using confocal microscopy and flow cytometry, NEU1 was immunolocalized to both the plasma membrane and the perinuclear region. NEU3 was detected both in the cytosol and nucleus. Prior siRNA-mediated knockdown of NEU1 and NEU3 each decreased EC sialidase activity for 4-MU-NANA by >65 and >17%, respectively, and for the ganglioside mixture by 0 and 40%, respectively. NEU1 overexpression in ECs reduced their migration into a wound by >40%, whereas NEU3 overexpression did not. Immunohistochemical studies of normal human tissues immunolocalized NEU1 and NEU3 proteins to both pulmonary and extrapulmonary vascular endothelia. These combined data indicate that human lung microvascular ECs as well as other endothelia express catalytically active NEU1 and NEU3. NEU1 restrains EC migration, whereas NEU3 does not.


Subject(s)
Cell Movement , Endothelial Cells/enzymology , Neuraminidase/metabolism , Aorta/enzymology , Carotid Arteries/enzymology , Cell Line , Cell Membrane/enzymology , Cell Nucleus/enzymology , Cerebral Arteries/enzymology , Cytosol/enzymology , Endothelial Cells/metabolism , Flow Cytometry , Gene Expression Regulation, Enzymologic , Humans , Hymecromone/analogs & derivatives , Hymecromone/pharmacology , Immunoblotting , Kidney/enzymology , Lung/enzymology , Microscopy, Confocal , N-Acetylneuraminic Acid/analogs & derivatives , N-Acetylneuraminic Acid/metabolism , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Substrate Specificity
7.
Clin Sci (Lond) ; 124(12): 695-700, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23438293

ABSTRACT

20-HETE (20-hydroxyeicosatetraenoic acid), a vasoconstrictor metabolite of arachidonic acid formed through the action of CYP4A (cytochrome P450-4A) in vascular smooth muscle cells, has been implicated in the development of hypertension and vascular dysfunction. There have been a number of reports in human subjects demonstrating an association between elevated urinary excretion of 20-HETE and hypertension, as well as increased 20-HETE production and vascular dysfunction. The Dahl SS (salt-sensitive) rat is a genetic model of salt-sensitive hypertension that exhibits vascular dysfunction, even when maintained on a normal-salt diet and before the development of hypertension. This mini-review highlights our current research on the role of CYP4A and 20-HETE in the vascular dysfunction of the Dahl SS rat. In our studies, the SS rat is compared with the consomic SS-5BN rat, having chromosome 5 from the salt-resistant Brown Norway rat (carrying all CYP4A genes) introgressed on to the SS genetic background. Our laboratory has demonstrated restoration of normal vascular function in the SS rat with inhibition of the CYP4A/20-HETE pathway, suggesting a direct role for this pathway in the vascular dysfunction in this animal model. Our studies have also shown that the SS rat has an up-regulated CYP4A/20-HETE pathway within their cerebral vasculature compared with the SS-5BN consomic rat, which causes endothelial dysfunction through the production of ROS (reactive oxygen species). Our data shows that ROS influences the expression of the CYP4A/20-HETE pathway in the SS rat in a feed-forward mechanism whereby elevated ROS stimulates production of 20-HETE. The presence of this vicious cycle offers a possible explanation for the spiralling effects of elevated 20-HETE on the development of vascular dysfunction in this animal model.


Subject(s)
Blood Pressure , Cytochrome P-450 CYP4A/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Hypertension/enzymology , Muscle, Smooth, Vascular/enzymology , Vasoconstriction , Animals , Cerebral Arteries/enzymology , Cerebral Arteries/physiopathology , Cytochrome P-450 CYP4A/genetics , Disease Models, Animal , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiopathology , Rats , Rats, Inbred BN , Rats, Inbred Dahl , Reactive Oxygen Species/metabolism , Signal Transduction , Sodium Chloride, Dietary , Up-Regulation
8.
Am J Respir Cell Mol Biol ; 46(3): 323-30, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21997485

ABSTRACT

We previously reported that hypoxia attenuates nitric oxide-cyclic guanosine monophosphate (NO-cGMP)-mediated fetal pulmonary vessel relaxation by inhibiting cGMP-dependent protein kinase 1 (PKG1) activity, but not all the mechanisms by which acute hypoxia inhibits PKG1 activity have been delineated. Here we demonstrate for the first time, to the best of our knowledge, that acute hypoxia induces an accumulation of ubiquitinated PKG1 in ovine fetal and newborn pulmonary artery smooth muscle cells. Such a modification was not evident in ovine fetal systemic (cerebral) artery smooth muscle cells. The accumulation of polyubiquitinated PKG1 observed after 4 hours of hypoxia was affected neither by the activation of PKG1 kinase activity with the cell-permeable cGMP analogue 8-bromo-cGMP, nor by its inhibition with DT-3 in fetal pulmonary artery smooth muscle cells. Ubiquitinated PKG1α was unable to bind the cGMP analogue 8-(2-aminoethyl)thioguanosine-3',5' (AET)-cGMP, a ligand for the unmodified protein. Inhibition of the proteasomal complex with MG132 led to the accumulation of polyubiquitinated PKG1 in normoxia, indicating the involvement of the ubiquitin-26S proteasomal system in degradation and clearance of this protein under normoxic conditions. The ubiquitinated PKG1 under hypoxic conditions, however, was not predominantly targeted for proteasomal degradation. Importantly, reoxygenation reversed the acute hypoxia-induced accumulation of ubiquitinated PKG1. Our results suggest that the PKG1 ubiquitination induced by acute hypoxia plays a unique role in the regulation of the pulmonary vascular smooth muscle cell vasoreactivity and relaxation mediated by the NO-cGMP-PKG1 pathway.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Oxygen/metabolism , Ubiquitination , Animals , Animals, Newborn , Cell Hypoxia , Cells, Cultured , Cerebral Arteries/enzymology , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activation , Enzyme Activators/pharmacology , Free Radical Scavengers/pharmacology , Ligands , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , Pulmonary Artery/enzymology , Reactive Oxygen Species/metabolism , Sheep , Signal Transduction , Time Factors , Up-Regulation
9.
Stroke ; 43(12): 3358-63, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160880

ABSTRACT

BACKGROUND AND PURPOSE: Angiotensin II produces oxidative stress and endothelial dysfunction in cerebral arteries, and angiotensin II type I receptors may play a role in longevity and vascular aging. Angiotensin-converting enzyme type 2 (ACE2) converts angiotensin II to angiotensin (1-7) and thus, may protect against effects of angiotensin II. We hypothesized that ACE2 deficiency increases oxidative stress and endothelial dysfunction in cerebral arteries and examined the role of ACE2 in age-related cerebrovascular dysfunction. METHODS: Endothelial function, expression of angiotensin system components, NADPH oxidase subunits, and proinflammatory cytokines were examined in cerebral arteries from adult (12 months old) and old (24 months old) ACE2 knockout (KO) and wild-type (WT) mice. The superoxide scavenger tempol was used to examine the role of oxidative stress on endothelial function. RESULTS: Vasodilatation to acetylcholine was impaired in adult ACE2 KO (24±6% [mean±SE]) compared with WT mice (52±7%; P<0.05). In old mice, vasodilatation to acetylcholine was impaired in WT mice (29±6%) and severely impaired in ACE2 KO mice (7±5%). Tempol improved endothelial function in adult and old ACE2 KO and WT mice. Aging increased mRNA for tumor necrosis factor-α in WT mice, and significantly increased mRNA levels of NAPDH oxidase 2, p47(phox), and Regulator of calcineurin 1 in both ACE2 KO and WT mice. mRNA levels of angiotensin system components did not change during aging. CONCLUSIONS: ACE2 deficiency impaired endothelial function in cerebral arteries from adult mice and augmented endothelial dysfunction during aging. Oxidative stress plays a critical role in cerebrovascular dysfunction induced by ACE2 deficiency and aging.


Subject(s)
Aging/metabolism , Cerebral Arteries/enzymology , Cerebrovascular Circulation/physiology , Oxidative Stress/physiology , Peptidyl-Dipeptidase A/genetics , Acetylcholine/pharmacology , Angiotensin I/biosynthesis , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Blood Pressure/physiology , Disease Models, Animal , Endothelium, Vascular/enzymology , Male , Mice , Mice, Knockout , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Peptide Fragments/biosynthesis , Peptidyl-Dipeptidase A/deficiency , RNA, Messenger/metabolism , Renin-Angiotensin System/physiology , Vasculitis/genetics , Vasculitis/metabolism , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology
10.
J Neuroinflammation ; 9: 274, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23259581

ABSTRACT

BACKGROUND: Subarachnoid hemorrhage (SAH) is associated with high morbidity and mortality. It is suggested that the associated inflammation is mediated through activation of the mitogen-activated protein kinase (MAPK) pathway which plays a crucial role in the pathogenesis of delayed cerebral ischemia after SAH. The aim of this study was first to investigate the timecourse of altered expression of proinflammatory cytokines and matrix metalloproteinase in the cerebral arteries walls following SAH. Secondly, we investigated whether administration of a specific mitogen-activated protein kinase kinase (MEK)1/2 inhibitor, U0126, given at 6 h after SAH prevents activation of the MEK/extracellular signal-regulated kinase 1/2 pathway and the upregulation of cerebrovascular inflammatory mediators and improves neurological function. METHODS: SAH was induced in rats by injection of 250 µl of autologous blood into basal cisterns. U0126 was given intracisternally using two treatment regimens: (A) treatments at 6, 12, 24 and 36 h after SAH and experiments terminated at 48 h after SAH, or (B) treatments at 6, 12, and 24 h after SAH and terminated at 72 h after SAH. Cerebral arteries were harvested and interleukin (IL)-6, IL-1ß, tumor necrosis factor α (TNF)α, matrix metalloproteinase (MMP)-9 and phosphorylated ERK1/2 (pERK1/2) levels investigated by immunohistochemistry. Early activation of pERK1/2 was measured by western blot. Functional neurological outcome after SAH was also analyzed. RESULTS: Expression levels of IL-1ß, IL-6, MMP-9 and pERK1/2 proteins were elevated over time with an early increase at around 6 h and a late peak at 48 to 72 h post-SAH in cerebral arteries. Enhanced expression of TNFα in cerebral arteries started at 24 h and increased until 96 h. In addition, SAH induced sensorimotor and spontaneous behavior deficits in the animals. Treatment with U0126 starting at 6 h after SAH prevented activation of MEK-ERK1/2 signaling. Further, U0126 significantly decreased the upregulation of inflammation proteins at 48 and 72 h following SAH and improved neurological function. We found no differences between treatment regimens A and B. CONCLUSIONS: These results show that SAH induces early activation of the MEK-ERK1/2 pathway in cerebral artery walls, which is associated with upregulation of proinflammatory cytokines and MMP-9. Inhibition of the MEK-ERK1/2 pathway by U0126 starting at 6 h post-SAH prevented upregulation of cytokines and MMP-9 in cerebral vessels, and improved neurological outcome.


Subject(s)
Cerebral Arteries/enzymology , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/physiology , Subarachnoid Hemorrhage/pathology , Animals , Astrocytes/drug effects , Astrocytes/enzymology , Blood Transfusion, Autologous/adverse effects , Butadienes/therapeutic use , Cerebral Arteries/pathology , Cytokines/genetics , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Exploratory Behavior/drug effects , Glial Fibrillary Acidic Protein/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Motor Activity/drug effects , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Nitriles/therapeutic use , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Statistics, Nonparametric , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
11.
Bull Exp Biol Med ; 154(1): 104-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23330102

ABSTRACT

The distribution of two enzymes involved in H(2)S synthesis, cystationine ß-synthase (CBS) and cystationine γ-liase (CSE), was studied in the walls of the internal carotid artery, order I-V branches of the middle cerebral artery basin, and intracerebral vessels of adult Wistar rats. Immunohistochemical staining showed the presence of CBS in the endothelium of small pial arteries (order IV-V branches) and intracerebral arterioles and in the capillary walls, neurons, and vascular nerves. As for CSE, in the internal carotid artery and large (order I-II) pial branches it was found mainly in the tunica media myocytes, in order III-IV vessels in myocytes and endothelium, and in smaller pial and intracerebral vessels in the endothelium. Along with enzyme-positive vessels, many pial and intracerebral arteries contained no these enzymes in the walls.


Subject(s)
Cerebral Arteries/enzymology , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Endothelium, Vascular/metabolism , Hydrogen Sulfide/metabolism , Animals , Carotid Artery, Internal/metabolism , Cerebral Arteries/innervation , Cerebral Arteries/metabolism , Immunohistochemistry , Male , Middle Cerebral Artery/metabolism , Rats , Rats, Wistar , Tunica Media/metabolism
12.
Pflugers Arch ; 462(3): 371-83, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21617900

ABSTRACT

We previously reported that in healthy mouse cerebral arteries, endothelial nitric oxide synthase (eNOS) produces H2O2, leading to endothelium-dependent dilation. In contrast, thromboxane A2 (TXA2), a potent pro-oxidant and pro-inflammatory endogenous vasoconstrictor, is associated with eNOS dysfunction. Our objectives were to elucidate whether (1) the cerebrovascular eNOS-H2O2 pathway was sensitive to oxidative stress associated with aging and dyslipidemia and (2) TXA2 contributed to cerebral eNOS dysfunction. Atherosclerotic (ATX = LDLR(-/-); hApoB(+/+)) and wild-type (WT) control mice were used at 3 and 12 months old (m/o). Three-m/o ATX mice were treated with the cardio-protective polyphenol catechin for 9 months. Dilations to ACh and the simultaneous eNOS-derived H2O2 production were recorded in isolated pressurized cerebral arteries. The age-associated decrease in cerebral eNOS-H2O2 pathway observed in WT was premature in ATX mice, decreasing at 3 m/o and abolished at 12 m/o. Thromboxane synthase inhibition by furegrelate increased dilations at 12 months in WT and at 3 and 12 months in ATX mice, suggesting an anti-dilatory role of TXA2 with age hastened by dyslipidemia. In addition, the non-selective NADP(H) oxidase inhibitor apocynin improved the eNOS-H2O2 pathway only in 12-m/o ATX mice. Catechin normalized the function of this pathway, which became sensitive to L-NNA and insensitive to furegrelate or apocynin; catechin also prevented the rise in TXA2 synthase expression. In conclusion, the age-dependent cerebral endothelial dysfunction is precocious in dyslipidemia and involves TXA2 production that limits eNOS activity. Preventive catechin treatment reduced the impact of endogenous TXA2 on the control of cerebral tone and maintained eNOS function.


Subject(s)
Aging/physiology , Atherosclerosis/enzymology , Cerebral Arteries/enzymology , Nitric Oxide Synthase Type III/metabolism , Thromboxane A2/metabolism , Up-Regulation , Acetophenones/metabolism , Animals , Antioxidants/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Benzofurans/metabolism , Catechin/therapeutic use , Cerebral Arteries/pathology , Enzyme Inhibitors/metabolism , Hemodynamics , Humans , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidants/metabolism , Oxidative Stress , Receptors, LDL/genetics , Receptors, LDL/metabolism , Thromboxane A2/genetics , Thromboxane-A Synthase/antagonists & inhibitors , Thromboxane-A Synthase/metabolism
13.
Am J Physiol Heart Circ Physiol ; 300(3): H1013-20, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21169403

ABSTRACT

Our goal was to examine whether exercise training (ExT) could normalize impaired nitric oxide synthase (NOS)-dependent dilation of cerebral (pial) arterioles during type 1 diabetes (T1D). We measured the in vivo diameter of pial arterioles in sedentary and exercised nondiabetic and diabetic rats in response to an endothelial NOS (eNOS)-dependent (ADP), an neuronal NOS (nNOS)-dependent [N-methyl-D-aspartate (NMDA)], and a NOS-independent (nitroglycerin) agonist. In addition, we measured superoxide anion levels in brain tissue under basal conditions in sedentary and exercised nondiabetic and diabetic rats. Furthermore, we used Western blot analysis to determine eNOS and nNOS protein levels in cerebral vessels/brain tissue in sedentary and exercised nondiabetic and diabetic rats. We found that ADP and NMDA produced a dilation of pial arterioles that was similar in sedentary and exercised nondiabetic rats. In contrast, ADP and NMDA produced only minimal vasodilation in sedentary diabetic rats. ExT restored impaired ADP- and NMDA-induced vasodilation observed in diabetic rats to that observed in nondiabetics. Nitroglycerin produced a dilation of pial arterioles that was similar in sedentary and exercised nondiabetic and diabetic rats. Superoxide levels in cortex tissue were similar in sedentary and exercised nondiabetic rats, were increased in sedentary diabetic rats, and were normalized by ExT in diabetic rats. Finally, we found that eNOS protein was increased in diabetic rats and further increased by ExT and that nNOS protein was not influenced by T1D but was increased by ExT. We conclude that ExT can alleviate impaired eNOS- and nNOS-dependent responses of pial arterioles during T1D.


Subject(s)
Arterioles/physiology , Cerebral Arteries/physiology , Cerebral Cortex/blood supply , Diabetes Mellitus, Type 1/physiopathology , Nitric Oxide Synthase/physiology , Physical Conditioning, Animal/physiology , Adenosine Diphosphate/pharmacology , Animals , Arterioles/drug effects , Arterioles/enzymology , Cerebral Arteries/drug effects , Cerebral Arteries/enzymology , Cerebral Cortex/chemistry , Cerebral Cortex/enzymology , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/enzymology , Male , N-Methylaspartate/pharmacology , Nitroglycerin/pharmacology , Rats , Rats, Sprague-Dawley , Superoxides/metabolism , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology
14.
Am J Physiol Heart Circ Physiol ; 301(5): H1882-90, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21856910

ABSTRACT

Dihydrotestosterone (DHT) attenuates cytokine-induced cyclooxygenase-2 (COX-2) in coronary vascular smooth muscle. Since hypoxia inducible factor-1α (HIF-1α) activation can lead to COX-2 production, this study determined the influence of DHT on HIF-1α and COX-2 following hypoxia or hypoxia with glucose deprivation (HGD) in the cerebral vasculature. COX-2 and HIF-1α levels were assessed via Western blot, and HIF-1α activation was indirectly measured via a DNA binding assay. Experiments were performed using cerebral arteries isolated from castrated male rats treated in vivo with placebo or DHT (18 days) followed by hypoxic exposure ex vivo (1% O(2)), cerebral arteries isolated from castrated male rats treated ex vivo with vehicle or DHT (10 or 100 nM; 18 h) and then exposed to hypoxia ex vivo (1% O(2)), or primary human brain vascular smooth muscle cells treated with DHT (10 nM; 6 h) or vehicle then exposed to hypoxia or HGD. Under normoxic conditions, DHT increased COX-2 (cells 51%; arteries ex vivo 31%; arteries in vivo 161%) but had no effect on HIF-1α. Following hypoxia or HGD, HIF-1α and COX-2 levels were increased; this response was blunted by DHT (cells HGD: -47% COX-2, -34% HIF-1α; cells hypoxia: -29% COX-2, -54% HIF-1α; arteries ex vivo: -37% COX-2; arteries in vivo: -35% COX-2) and not reversed by androgen receptor blockade. Hypoxia-induced HIF-1α DNA-binding was also attenuated by DHT (arteries ex vivo and in vivo: -55%). These results demonstrate that upregulation of COX-2 and HIF-1α in response to hypoxia is suppressed by DHT via an androgen receptor-independent mechanism.


Subject(s)
Cerebral Arteries/drug effects , Cyclooxygenase 2/metabolism , Dihydrotestosterone/administration & dosage , Glucose/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Androgen Antagonists/pharmacology , Animals , Cell Hypoxia , Cerebral Arteries/enzymology , Drug Implants , Humans , Inflammation Mediators/metabolism , Male , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Orchiectomy , Rats , Rats, Wistar , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Tissue Culture Techniques
15.
Open Heart ; 8(2)2021 11.
Article in English | MEDLINE | ID: mdl-34819349

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is associated with mental health disorders, in which neuroinflammation and cerebral microvascular dysfunction may play a role. Previously, we have shown that the proinflammatory factors Nε-(carboxymethyl)lysine (CML) and NADPH oxidase 2 (NOX2) are increased in the human infarcted heart microvasculature. The aim of this study was to analyse the presence of CML and NOX2 in the cerebral microvasculature of patients with MI. METHODS: Brain tissue was obtained at autopsy from 24 patients with MI and nine control patients. According to their infarct age, patients with MI were divided into three groups: 3-6 hours old (phase I), 6 hours-5 days old (phase II) and 5-14 days old (phase III). CML and NOX2 in the microvasculature were studied through immunohistochemical analysis. RESULTS: We observed a 2.5-fold increase in cerebral microvascular CML in patients with phase II and phase III MI (phase II: 21.39±7.91, p=0.004; phase III: 24.21±10.37, p=0.0007) compared with non-MI controls (8.55±2.98). NOX2 was increased in microvessels in patients with phase II MI (p=0.002) and phase III MI (p=0.04) compared with controls. No correlation was found between CML and NOX2 (r=0.58, p=0.13). CONCLUSIONS: MI coincides with an increased presence of CML and NOX2 in the brain microvasculature. These data point to proinflammatory alterations in the brain microvasculature that may underlie MI-associated mental health disorders.


Subject(s)
Cerebral Arteries/enzymology , Lysine/analogs & derivatives , Microvessels/enzymology , Myocardial Infarction/enzymology , NADPH Oxidase 2/biosynthesis , Neuroinflammatory Diseases/enzymology , Aged , Biomarkers/metabolism , Cerebral Arteries/pathology , Female , Humans , Immunohistochemistry , Lysine/biosynthesis , Male , Microvessels/pathology , Middle Aged , Myocardial Infarction/complications , Myocardial Infarction/pathology , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/pathology
16.
Neurobiol Dis ; 37(3): 656-60, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005954

ABSTRACT

Sandhoff disease is an autosomal recessive lysosomal disorder due to mutations in the beta-hexosaminidase beta-chain gene, resulting in beta-hexosaminidases A (alphabeta) and B (betabeta) deficiency and GM2 ganglioside accumulation in the brain. In this study, our aim was to demonstrate that transduction of cerebral endothelial cells cultured in two-chamber culture inserts with a lentiviral vector encoding the hexosaminidases alpha and beta chains could induce a vectorial secretion of hexosaminidases. Therefore, the human cerebral endothelial cell line hCMEC/D3 was infected with the bicistronic vector from the apical compartment, and beta-hexosaminidase activity was measured in transduced cells and in deficient fibroblasts co-cultured in the basal (i.e. brain) compartment. Induced beta-hexosaminidase secretion by transduced hCMEC/D3 cells was sufficient to allow for a 70-90% restoration of beta-hexosaminidase activity in deficient fibroblasts. On the basis of these in vitro data, we propose that brain endothelium be considered as a novel therapeutic target in Sandhoff disease.


Subject(s)
Endothelial Cells/enzymology , Genetic Therapy/methods , Sandhoff Disease/enzymology , Sandhoff Disease/therapy , Transduction, Genetic/methods , beta-N-Acetylhexosaminidases/metabolism , Cell Line, Transformed , Cerebral Arteries/cytology , Cerebral Arteries/enzymology , Cerebrum/blood supply , Cerebrum/enzymology , Cerebrum/physiopathology , Coculture Techniques , Diffusion Chambers, Culture , Endothelial Cells/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , G(M2) Ganglioside/metabolism , Genetic Vectors/pharmacology , Genetic Vectors/therapeutic use , Humans , Lentivirus/genetics , Sandhoff Disease/genetics , beta-N-Acetylhexosaminidases/genetics
17.
J Clin Invest ; 117(7): 1961-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17557122

ABSTRACT

NO plays critical roles in vascular function. We show that modulation of the eNOS serine 1179 (S1179) phosphorylation site affects vascular reactivity and determines stroke size in vivo. Transgenic mice expressing only a phosphomimetic (S1179D) form of eNOS show greater vascular reactivity, develop less severe strokes, and have improved cerebral blood flow in a middle cerebral artery occlusion model than mice expressing an unphosphorylatable (S1179A) form. These results provide a molecular mechanism by which multiple diverse cardiovascular risks, such as diabetes and obesity, may be centrally integrated by eNOS phosphorylation in vivo to influence blood flow and cardiovascular disease. They also demonstrate the in vivo relevance of posttranslational modification of eNOS in vascular function.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/pathology , Nitric Oxide Synthase Type II/metabolism , Alanine/genetics , Alanine/metabolism , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Brain Ischemia/genetics , Cerebral Arteries/enzymology , Cerebral Arteries/injuries , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Male , Mice , Mice, Transgenic , Mutation/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III , Phosphorylation , Regional Blood Flow , Serine/genetics , Serine/metabolism , Stroke/enzymology , Stroke/genetics , Stroke/pathology
18.
Am J Physiol Heart Circ Physiol ; 299(6): H2046-55, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20935146

ABSTRACT

The present study tested the hypotheses that 1) short-term dietary deficiency of magnesium (21 days) in rats would result in the upregulation of sphingomyelin synthase (SMS) and p53 in cardiac and vascular (aortic) smooth muscles, 2) low levels of Mg(2+) added to drinking water would either prevent or greatly reduce the upregulation of both SMS and p53, 3) exposure of primary cultured vascular smooth muscle cells (VSMCs) to low extracellular Mg(2+) concentration ([Mg(2)](o)) would lead to the de novo synthesis of ceramide, 4) inhibition of either SMS or p53 in primary culture VSMCs exposed to low [Mg(2+)](o) would lead to reductions in the levels of de novo ceramide synthesis, and 5) inhibition of sphingomyelin palmitoyl-CoA transferase (SPT) or ceramide synthase (CS) in primary cultured VSMCs exposed to low [Mg(2+)](o) would lead to a reduction in the levels of de novo ceramide synthesis. The data indicated that short-term magnesium deficiency (10% normal dietary intake) resulted in the upregulation of SMS and p53 in both ventricular and aortic smooth muscles; even very low levels of water-borne Mg(2+) (e.g., 15 mg·l(-1)·day(-1)) either prevented or ameliorated the upregulation in SMS and p53. Our experiments also showed that VSMCs exposed to low [Mg(2+)](o) resulted in the de novo synthesis of ceramide; the lower the [Mg(2+)](o), the greater the synthesis of ceramide. In addition, the data indicated that inhibition of either SMS, p53, SPT, or CS in VSMCs exposed to low [Mg(2+)](o) resulted in marked reductions in the de novo synthesis of ceramide.


Subject(s)
Ceramides/biosynthesis , Magnesium Deficiency/enzymology , Muscle, Smooth, Vascular/enzymology , Myocardium/enzymology , Myocytes, Smooth Muscle/enzymology , Transferases (Other Substituted Phosphate Groups)/metabolism , Tumor Suppressor Protein p53/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/metabolism , Animals , Aorta/enzymology , Cells, Cultured , Cerebral Arteries/enzymology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Male , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/metabolism , Rats , Rats, Wistar , Time Factors , Transferases (Other Substituted Phosphate Groups)/antagonists & inhibitors , Up-Regulation
19.
Am J Physiol Regul Integr Comp Physiol ; 299(5): R1377-86, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702800

ABSTRACT

In the developing fetus, cerebral arteries (CA) show striking differences in signal transduction mechanisms compared with the adult, and these differences are magnified in response to high-altitude long-term hypoxia (LTH). In addition, in the mature organism, cerebrovascular acclimatization to LTH may be associated with several clinical problems, the mechanisms of which are unknown. Because PKC plays a key role in regulating CA contractility, in fetal and adult cerebral arteries, we tested the hypothesis that LTH differentially regulates the PKC-mediated Ca(2+) sensitization pathways and contractility. In four groups of sheep [fetal normoxic (FN), fetal hypoxic (FH), adult normoxic (AN), and adult hypoxic (AH)], we examined, simultaneously, responses of CA tension and intracellular Ca(2+) concentration and measured CA levels of PKC, ERK1/2, RhoA, 20-kDa myosin light chain, and the 17-kDa PKC-potentiated myosin phosphatase inhibitor CPI-17. The PKC activator phorbol 12,13-dibutyrate (PDBu) produced robust contractions in all four groups. However, PDBu-induced contractions were significantly greater in AH CA than in the other groups. In all CA groups except AH, in the presence of MEK inhibitor (U-0126), the PDBu-induced contractions were increased a further 20-30%. Furthermore, in adult CA, PDBu led to increased phosphorylation of ERK1, but not ERK2; in fetal CA, the reverse was the case. PDBu-stimulated ERK2 phosphorylation also was significantly greater in FH than FN CA. Also, although RhoA/Rho kinase played a significant role in PDBu-mediated contractions of FN CA, this was not the case in FH or either adult group. Also, whereas CPI-17 had a significant role in adult CA contractility, this was not the case for the fetus. Overall, in ovine CA, the present study demonstrates several important maturational and LTH acclimatization changes in PKC-induced contractile responses and downstream pathways. The latter may play a key role in the pathophysiologic disorders associated with acclimatization to high altitude.


Subject(s)
Acclimatization , Aging , Calcium Signaling , Cerebral Arteries/enzymology , Fetal Hypoxia/enzymology , Hypoxia/enzymology , Protein Kinase C/metabolism , Vasoconstriction , Age Factors , Animals , Calcium Signaling/drug effects , Cerebral Arteries/drug effects , Cerebral Arteries/embryology , Cerebral Arteries/growth & development , Cerebral Arteries/physiopathology , Disease Models, Animal , Enzyme Activation , Enzyme Activators/pharmacology , Fetal Hypoxia/physiopathology , Gestational Age , Hypoxia/physiopathology , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myosin Light Chains/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Sheep , Time Factors , Vasoconstriction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
20.
J Vasc Res ; 47(2): 115-27, 2010.
Article in English | MEDLINE | ID: mdl-19729958

ABSTRACT

The interactive role of protein kinase C (PKC) isoforms and protein phosphatase 2A (PP2A) in the mechanisms underlying the gradual reduction in stretch-induced contraction through triphosphorylation of 20-kDa myosin light chain (MLC(20)) was investigated in the canine basilar artery. In the presence of 5 mM tetraethylammonium, stretching at a rate of 1 mm/s from the initial length (L(i)) to 1.5 L(i) produced a contraction. Maintaining the stretched state for 15 min (15-min stretch) produced triphosphorylation of MLC(20 )at Ser-19, Thr-18 and Thr-9, and a gradual reduction in the contraction, both of which were reversed by Gö6976 (1 microM), an inhibitor of conventional PKC. The 15-min stretch increased PKCalpha activity whereas it decreased PP2A activity, both of which were blocked by Y-27632, an inhibitor of rho kinase. Okadaic acid (OA; 1 microM), a PP2A inhibitor, also produced triphosphorylation of MLC(20) at the same amino acid residues and activated PKCalpha, which was inhibited by Gö6976. Stretching and OA increased phosphorylation of 17-kDa PKC-potentiated inhibitory phosphoprotein (CPI-17), and this phosphorylation was inhibited by Gö6976. The present results suggest that activation of PKCalpha mediated by an inhibitor of PP2A is involved in the stretch-induced triphosphorylation of MLC(20), and that this triphosphorylation counteracts the stretch-induced contraction.


Subject(s)
Basilar Artery/enzymology , Cerebral Arteries/enzymology , Myosin Light Chains/metabolism , Pressoreceptors/metabolism , Protein Kinase C-alpha/metabolism , Protein Phosphatase 2/metabolism , Vasoconstriction , Amides/pharmacology , Animals , Basilar Artery/drug effects , Carbazoles/pharmacology , Cerebral Arteries/drug effects , Dogs , Enzyme Activation , Female , In Vitro Techniques , Male , Okadaic Acid/pharmacology , Phosphorylation , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction , Time Factors , Vasoconstriction/drug effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL