Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Stroke ; 52(7): 2404-2413, 2021 07.
Article in English | MEDLINE | ID: mdl-34107734

ABSTRACT

Background and Purpose: Hypertension is a leading risk factor for cerebrovascular disease and loss of brain health. While the brain renin-angiotensin system (RAS) contributes to hypertension, its potential impact on the local vasculature is unclear. We tested the hypothesis that activation of the brain RAS would alter the local vasculature using a modified deoxycorticosterone acetate (DOCA) model. Methods: C57BL/6 mice treated with DOCA (50 mg SQ; or shams) were given tap H2O and H2O with 0.9% NaCl for 1 to 3 weeks. Results: In isolated cerebral arteries and parenchymal arterioles from DOCA-treated male mice, endothelium- and nitric oxide-dependent dilation was progressively impaired, while mesenteric arteries were unaffected. In contrast, cerebral endothelial function was not significantly affected in female mice treated with DOCA. In males, mRNA expression of renal Ren1 was markedly reduced while RAS components (eg, Agt and Ace) were increased in both brain and cerebral arteries with central RAS activation. In NZ44 reporter mice expressing GFP (green fluorescent protein) driven by the angiotensin II type 1A receptor (Agtr1a) promoter, DOCA increased GFP expression ≈3-fold in cerebral arteries. Impaired endothelial responses were restored to normal by losartan, an AT1R (angiotensin II type 1 receptor) antagonist. Last, DOCA treatment produced inward remodeling of parenchymal arterioles. Conclusions: These findings suggest activation of the central and cerebrovascular RAS impairs endothelial (nitric oxide dependent) signaling in brain through expression and activation of AT1R and sex-dependent effects. The central RAS may be a key contributor to vascular dysfunction in brain in a preclinical (low renin) model of hypertension. Because the brain RAS is also activated during aging and other diseases, a common mechanism may promote loss of endothelial and brain health despite diverse cause.


Subject(s)
Cerebrovascular Disorders/metabolism , Endothelium, Vascular/metabolism , Hypertension/metabolism , Nitric Oxide Synthase Type III/biosynthesis , Receptor, Angiotensin, Type 1/biosynthesis , Renin-Angiotensin System/physiology , Animals , Cerebrovascular Disorders/chemically induced , Cerebrovascular Disorders/genetics , Desoxycorticosterone Acetate/toxicity , Female , Hypertension/chemically induced , Hypertension/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type III/genetics , Receptor, Angiotensin, Type 1/genetics , Renin-Angiotensin System/drug effects
2.
Hypertension ; 71(5): 921-927, 2018 05.
Article in English | MEDLINE | ID: mdl-29531174

ABSTRACT

Vascular aging fundamentally contributes to large and small vessel disease. Despite the importance of such changes for brain function, mechanisms that mediate such changes are poorly defined. We explored mechanisms that underlie changes with age, testing the hypothesis that ROCK (Rho kinase) plays an important role. In C57BL/6 mice, baseline diameters of isolated pressurized parenchymal arterioles were similar in adult (4-5 month) and old mice (22±1 month; ≈15±1 µm). Endothelium-dependent dilation was impaired in old mice compared with adults in a pathway-specific manner. Vasodilation to NS-309 (which activates small- and intermediate-conductance Ca2+ activated K+ channels in endothelial cells) was intact while endothelial nitric oxide synthase-mediated vasodilation was reduced by ≥60%, depending on the concentration (P<0.05). A similar reduction was present in basilar arteries. Inhibiting both ROCK isoforms with Y-27632 restored the majority of endothelial function in old mice. Because genetic background is a determinant of vascular disease, we performed similar studies using FVB/N mice. Endothelial dysfunction was seen with aging in both FVB/N and C57BL/6 mice although the magnitude was increased ≈2-fold in the latter strain (P<0.05). In both strains of mice, age-induced endothelial dysfunction was reversed by inhibition of ROCK2 with SLX-2119. Thus, aging impairs endothelial function in both cerebral arteries and parenchymal arterioles, predominantly via effects on endothelial nitric oxide synthase-dependent regulation of vascular tone. The magnitude of these changes was influenced by genetic background and mediated by ROCK2.


Subject(s)
Aging/genetics , Genetic Background , Vascular Diseases/genetics , rho-Associated Kinases/genetics , Analysis of Variance , Animals , Arterioles/metabolism , Cerebral Arteries/metabolism , Endothelium, Vascular/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Sensitivity and Specificity , Vascular Diseases/physiopathology
3.
Hypertension ; 70(3): 559-565, 2017 09.
Article in English | MEDLINE | ID: mdl-28674038

ABSTRACT

Pharmacological activation of PPAR-γ (peroxisome proliferator-activated receptor-γ) protects the vasculature. Much less is known on the cell-specific impact of PPAR-γ when driven by endogenous ligands. Recently, we found that endothelial PPAR-γ protects against angiotensin II-induced endothelial dysfunction. Here, we explored that concept further examining whether effects were sex dependent along with underlying mechanisms. We studied mice expressing a human dominant-negative mutation in PPAR-γ driven by the endothelial-specific vascular cadherin promoter (E-V290M), using nontransgenic littermates as controls. Acetylcholine (an endothelium-dependent agonist) produced similar relaxation of carotid arteries from nontransgenic and E-V290M mice. Incubation of isolated arteries with angiotensin II (1 nmol/L) overnight had no effect in nontransgenic, but reduced responses to acetylcholine by about 50% in male and female E-V290M mice (P<0.05). Endothelial function in E-V290M mice was restored to normal by inhibitors of superoxide (tempol), NADPH oxidase (VAS-2870), Rho kinase (Y-27632), ROCK2 (SLX-2119), NF-κB (nuclear factor-kappa B essential modulator-binding domain peptide), or interleukin-6 (neutralizing antibody). In addition, we hypothesized that PPAR-γ may influence the angiotensin 1-7 arm of the renin-angiotensin system. In the basilar artery, dilation to angiotensin 1-7 was selectively reduced in E-V290M mice by >50% (P<0.05), an effect reversed by Y-27632. Thus, effects of angiotensin II are augmented by interference with endothelial PPAR-γ through sex-independent mechanisms, involving oxidant-inflammatory signaling and ROCK2 (Rho kinase). The study also provides the first evidence that endothelial PPAR-γ interacts with angiotensin 1-7 responses. These critical roles for endothelial PPAR-γ have implications for pathophysiology and therapeutic approaches for vascular disease.


Subject(s)
Angiotensin II , Angiotensin I , PPAR gamma/metabolism , Peptide Fragments , Vascular Diseases , Vasodilation , Amides , Angiotensin I/metabolism , Angiotensin I/pharmacology , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Animals, Genetically Modified , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Female , Interleukin-6/metabolism , Male , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Pyridines , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Vascular Diseases/metabolism , Vascular Diseases/physiopathology , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
4.
Hypertension ; 68(3): 809-17, 2016 09.
Article in English | MEDLINE | ID: mdl-27432870

ABSTRACT

Rho kinase (ROCK) has been implicated in physiological and pathophysiological processes, including regulation of vascular function. ROCK signaling is thought to be a critical contributor to cardiovascular disease, including hypertension and effects of angiotensin II (Ang II). Two isoforms of ROCK (1 and 2) have been identified and are expressed in vascular cells. In this study, we examined the importance of ROCK2 in relation to vessel function using several models and a novel inhibitor of ROCK2. First, incubation of carotid arteries with the direct RhoA activator CN-03 or Ang II impaired endothelium-dependent relaxation by ≈40% to 50% (P<0.05) without altering endothelium-independent relaxation. Both CN-03- and Ang II-induced endothelial dysfunction was prevented by Y-27632 (an inhibitor of both ROCK isoforms) or the selective ROCK2 inhibitor SLX-2119. In contrast, SLX-2119 had little effect on contraction of carotid arteries to receptor-mediated agonists (serotonin, phenylephrine, vasopressin, or U46619). Second, in basilar arteries, SLX-2119 inhibited constriction to Ang II by ≈90% without significantly affecting responses to serotonin or KCl. Third, in isolated pressurized brain parenchymal arterioles, SLX-2119 inhibited myogenic tone in a concentration-dependent manner (eg, 1 µmol/L SLX-2119 dilated by 79±4%). Finally, SLX-2119 dilated small pial arterioles in vivo, an effect that was augmented by inhibition of nitric oxide synthase. These findings suggest that ROCK2 has major, but heterogeneous, effects on function of endothelium and vascular muscle. The data support the concept that aberrant ROCK2 signaling may be a key contributor to select aspects of large and small vessel disease, including Ang II-induced endothelial dysfunction.


Subject(s)
Cerebral Arteries/metabolism , Endothelium, Vascular/metabolism , rho-Associated Kinases/metabolism , Acetylcholine/pharmacology , Analysis of Variance , Angiotensin II/pharmacology , Animals , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Cerebral Arteries/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice , Mice, Inbred C57BL , Nitroprusside/pharmacology , Random Allocation , Reference Values , Vasoconstriction/drug effects , rho-Associated Kinases/genetics
5.
Am J Physiol Heart Circ Physiol ; 311(1): H146-56, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27106041

ABSTRACT

Carotid artery disease is a major contributor to stroke and cognitive deficits. Angiotensin II (Ang II) promotes vascular dysfunction and disease through mechanisms that include the IL-6/STAT3 pathway. Here, we investigated the importance of suppressor of cytokine signaling 3 (SOCS3) in models of Ang II-induced vascular dysfunction. We examined direct effects of Ang II on carotid arteries from SOCS3-deficient (SOCS3(+/-)) mice and wild-type (WT) littermates using organ culture and then tested endothelial function with acetylcholine (ACh). A low concentration of Ang II (1 nmol/l) did not affect ACh-induced vasodilation in WT but reduced that of SOCS3(+/-) mice by ∼50% (P < 0.05). In relation to mechanisms, effects of Ang II in SOCS3(+/-) mice were prevented by inhibitors of STAT3, IL-6, NF-κB, or superoxide. Systemic Ang II (1.4 mg/kg per day for 14 days) also reduced vasodilation to ACh in WT. Surprisingly, SOCS3 deficiency prevented most of the endothelial dysfunction. To examine potential underlying mechanisms, we performed bone marrow transplantation. WT mice reconstituted with SOCS3(+/-) bone marrow were protected from Ang II-induced endothelial dysfunction, whereas reconstitution of SOCS3(+/-) mice with WT bone marrow exacerbated Ang II-induced effects. The SOCS3 genotype of bone marrow-derived cells did not influence direct effects of Ang II on vascular function. These data provide new mechanistic insight into the influence of SOCS3 on the vasculature, including divergent effects depending on the source of Ang II. Bone marrow-derived cells deficient in SOCS3 protect against systemic Ang II-induced vascular dysfunction.


Subject(s)
Angiotensin II , Aorta/metabolism , Basilar Artery/metabolism , Bone Marrow Cells/metabolism , Carotid Arteries/metabolism , Hypertension/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Vasodilation , Animals , Aorta/drug effects , Aorta/physiopathology , Basilar Artery/drug effects , Basilar Artery/physiopathology , Bone Marrow Transplantation , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Genotype , Hypertension/chemically induced , Hypertension/physiopathology , Hypertension/prevention & control , Interleukin-6/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Organ Culture Techniques , Phenotype , STAT3 Transcription Factor/metabolism , Signal Transduction , Superoxides/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Vasodilation/drug effects , Vasodilator Agents/pharmacology
6.
Hypertension ; 64(5): 1088-93, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25185134

ABSTRACT

Although peroxisome proliferator-activated receptor-γ (PPARγ) is thought to play a protective role in the vasculature, its cell-specific effect, particularly in resistance vessels, is poorly defined. Nitric oxide (NO) plays a major role in vascular biology in the brain. We examined the hypothesis that selective interference with PPARγ in vascular muscle would impair NO-dependent responses and augment vasoconstrictor responses in the cerebral circulation. We studied mice expressing a dominant negative mutation in human PPARγ (P467L) under the control of the smooth muscle myosin heavy chain promoter (S-P467L). In S-P467L mice, dilator responses to exogenously applied or endogenously produced NO were greatly impaired in cerebral arteries in vitro and in small cerebral arterioles in vivo. Select NO-independent responses, including vasodilation to low concentrations of potassium, were also impaired in S-P467L mice. In contrast, increased expression of wild-type PPARγ in smooth muscle had little effect on vasomotor responses. Mechanisms underlying impairment of both NO-dependent and NO-independent vasodilator responses after interference with PPARγ involved Rho kinase with no apparent contribution by oxidative stress-related mechanisms. These findings support the concept that via effects on Rho kinase-dependent signaling, PPARγ in vascular muscle is a major determinant of vascular tone in resistance vessels and, in particular, NO-mediated signaling in cerebral arteries and brain microvessels. Considering the importance of NO and Rho kinase, these findings have implications for regulation of cerebral blood flow and the pathogenesis of large and small vessel disease in brain.


Subject(s)
Cerebrovascular Circulation/physiology , Muscle, Smooth, Vascular/physiology , PPAR gamma/physiology , Animals , Arterioles/metabolism , Female , Humans , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , Nitric Oxide/metabolism , PPAR gamma/genetics , Signal Transduction/physiology , Vasodilation/physiology , rho-Associated Kinases/physiology
7.
Hypertension ; 61(2): 437-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23266544

ABSTRACT

Angiotensin II (Ang II) is known to promote vascular disease and hypertension in part by formation of cytokines, such as interleukin-6. However, the role of signal transducer and activator of transcription 3 (STAT3) in these processes and Ang II/interleukin-6 signaling is unclear. Using 2 models, we tested the hypothesis that STAT3 is essential for Ang II-induced vascular dysfunction and hypertension. Incubation of isolated carotid arteries from C57BL/6J mice with Ang II overnight increased superoxide ≈2-fold and reduced vasodilator responses to the endothelium-dependent agonist acetylcholine by ≈50% versus controls (P<0.05). These effects were prevented by the addition of small-molecular inhibitors of STAT3 activation (S3I-201 or STATTIC). In vivo, administration of Ang II (1.4 mg kg(-1) day(-1)) using osmotic minipumps increased arterial pressure by ≈40 mm Hg at day 14 compared with vehicle-treated mice, and this effect was prevented by S3I-201 treatment (5 mg/kg IP, QOD). After systemic treatment with Ang II, dilator responses to acetylcholine were reduced by ≈30% to 50% in carotid artery and basilar arteries, whereas S3I-201 treatment prevented most of this impairment (P<0.05). In contrast to effects on vascular function and blood pressure, S31-201 did not prevent Ang II-induced hypertrophy in the carotid artery. These findings provide the first evidence that inhibitors of STAT3 activation protect against Ang II-induced oxidative stress, endothelial dysfunction, and hypertension. Because Ang II promotes vascular disease in the presence of multiple cardiovascular risk factors, these results suggest that selective targeting of STAT3 may have substantial therapeutic potential.


Subject(s)
Angiotensin II/toxicity , Endothelium, Vascular/drug effects , Hypertension/prevention & control , Oxidative Stress/physiology , STAT3 Transcription Factor/antagonists & inhibitors , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Aminosalicylic Acids/pharmacology , Animals , Antihypertensive Agents/pharmacology , Aorta/drug effects , Aorta/metabolism , Aorta/physiopathology , Benzenesulfonates/pharmacology , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Carotid Arteries/physiopathology , Cyclic S-Oxides/pharmacology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Male , Mice , Nitroprusside/pharmacology , Oxidative Stress/drug effects , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism , Superoxides/metabolism , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
8.
Am J Physiol Regul Integr Comp Physiol ; 302(10): R1184-90, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22461176

ABSTRACT

Vascular disease occurs commonly during aging. Carotid artery and cerebrovascular disease are major causes of stroke and contributors to dementia. Recent evidence suggests that peroxisome proliferator-activated receptor-γ (PPARγ) may play a protective role in the vasculature, but the potential importance of PPARγ in vascular aging is unknown. To examine the hypothesis that PPARγ normally protects against vascular aging, we studied heterozygous knockin mice expressing a human dominant-negative mutation in PPARγ (P465L, designated L/+). Endothelial dysfunction, a major contributor to vascular disease, was studied using carotid arteries from adult (8 ± 1 mo) and old (24 ± 1 mo) L/+ mice and wild-type littermates. In arteries from wild-type mice, responses to the endothelium-dependent agonist ACh were similar in adult and old wild-type mice but were reduced by ∼50% in old L/+ mice (n = 7-10, P < 0.05). Impaired responses in arteries from old L/+ mice were restored to normal by a scavenger of superoxide. Relaxation of arteries to nitroprusside (an NO donor) was similar in all groups. Contraction of arteries to U46619 was not affected by age or genotype, while maximal responses to endothelin-1 were reduced with age in both wild-type and L/+ mice. Vascular expression (mRNA) of the catalytic component of NADPH oxidase (Nox2) was not altered in wild-type mice but was increased significantly in old L/+ mice. These findings provide the first evidence that interference with PPARγ function accelerates vascular aging, suggesting a novel role for PPARγ in protecting against age-induced oxidative stress and endothelial dysfunction.


Subject(s)
Aging/physiology , Aorta/physiopathology , Carotid Arteries/physiopathology , Endothelium, Vascular/physiopathology , PPAR gamma/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Aorta/drug effects , Aorta/metabolism , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Animal , Nitroprusside/pharmacology , Oxidative Stress/physiology , PPAR gamma/genetics , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
9.
J Cereb Blood Flow Metab ; 29(6): 1130-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19352401

ABSTRACT

Although arachidonic acid (AA) has diverse vascular effects, the mechanisms that mediate these effects are incompletely defined. The goal of our study was to use genetic approaches to examine the role of hydrogen peroxide (H2O2), glutathione peroxidase (Gpx1, which degrades H2O2), and CuZn-superoxide dismutase (SOD1, which produces H2O2 from superoxide) in mediating and in determining vascular responses to AA. In basilar arteries in vitro, AA produced dilation in nontransgenic mice, and this response was reduced markedly in transgenic mice overexpressing Gpx1 (Gpx1 Tg) or in those genetically deficient in SOD1. For example, AA (1 nmol/L to 1 mumol/L) dilated the basilar artery and this response was reduced by approximately 90% in Gpx1 Tg mice (P<0.01), although responses to acetylcholine were not altered. Dilation of cerebral arterioles in vivo in response to AA was inhibited by approximately 50% by treatment with catalase (300 U/mL) (P<0.05) and reduced by as much as 90% in Gpx1 Tg mice compared with that in controls (P<0.05). These results provide the first evidence that Gpx1 has functional effects in the cerebral circulation, and that AA-induced vascular effects are mediated by H2O2 produced by SOD1. In contrast, cerebral vascular responses to the endothelium-dependent agonist acetylcholine are not mediated by H2O2.


Subject(s)
Brain/blood supply , Brain/metabolism , Glutathione Peroxidase/metabolism , Hydrogen Peroxide/metabolism , Animals , Arachidonic Acid/metabolism , Brain/drug effects , Catalase/metabolism , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Glutathione Peroxidase/genetics , Indomethacin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Vasodilation/drug effects , Glutathione Peroxidase GPX1
10.
Am J Physiol Heart Circ Physiol ; 296(6): H1914-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19395552

ABSTRACT

Vascular dysfunction occurs with aging. We hypothesized that oxidative stress and ANG II [acting via ANG II type 1 (AT(1)) receptors] promotes cerebral vascular dysfunction with aging. We studied young (5-6 mo), old (17-19 mo), and very old (23 +/- 1 mo) mice. In basilar arteries in vitro, acetylcholine (an endothelium-dependent agonist) produced dilation in young wild-type mice that was reduced by approximately 60 and 90% (P < 0.05) in old and very old mice, respectively. Similar effects were seen using A23187, a second endothelium-dependent agonist. The vascular response to acetylcholine in very old mice was almost completely restored with tempol (a scavenger of superoxide) and partly restored by PJ34, an inhibitor of poly(ADP-ribose) polymerase (PARP). We used mice deficient in Mn-SOD (Mn-SOD(+/-)) to test whether this form of SOD protected during aging but found that age-induced endothelial dysfunction was not altered by Mn-SOD deficiency. Cerebral vascular responses were similar in young mice lacking AT(1) receptors (AT(1)(-/-)) and wild-type mice. Vascular responses to acetylcholine and A23187 were reduced by approximately 50% in old wild-type mice (P < 0.05) but were normal in old AT(1)-deficient mice. Thus, aging produces marked endothelial dysfunction in the cerebral artery that is mediated by ROS, may involve the activation of PARP, but was not enhanced by Mn-SOD deficiency. Our findings suggest a novel and fundamental role for ANG II and AT(1) receptors in age-induced vascular dysfunction.


Subject(s)
Aging/physiology , Cerebrovascular Circulation/physiology , Oxidative Stress/physiology , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism , Acetylcholine/pharmacology , Angiotensin II/metabolism , Animals , Basilar Artery/drug effects , Basilar Artery/physiology , Calcimycin/pharmacology , Cerebrovascular Circulation/drug effects , Ionophores/pharmacology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Vasodilation/physiology , Vasodilator Agents/pharmacology
11.
Circ Res ; 103(6): 654-61, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18676352

ABSTRACT

The ligand-activated transcription factor peroxisome proliferator activated receptor gamma (PPARgamma) is expressed in vascular endothelium where it exerts anti-inflammatory and antioxidant effects. However, its role in regulating vascular function remains undefined. We examined endothelial function in transgenic mice expressing dominant-negative mutants of PPARgamma under the control of an endothelial-specific promoter to test the hypothesis that endothelial PPARgamma plays a protective role in the vasculature. Under baseline conditions, responses to the endothelium-dependent agonist acetylcholine were not affected in either aorta or the basilar artery in vitro. In response to feeding a high-fat diet for 12 weeks, acetylcholine produced dilation that was markedly impaired in the basilar artery of mice expressing dominant-negative mutants, but not in mice expressing wild-type PPARgamma controlled by the same promoter. Unlike basilar artery, 12 weeks of a high-fat diet was not sufficient to cause endothelial dysfunction in the aorta of mice expressing dominant-negative PPARgamma, although aortic dysfunction became evident after 25 weeks. The responses to acetylcholine in basilar artery were restored to normal after treatment with a scavenger of superoxide. Baseline blood pressure was only slightly elevated in the transgenic mice, but the pressor response to angiotensin II was augmented. Thus, interference with PPARgamma in the endothelium produces endothelial dysfunction in the cerebral circulation through a mechanism involving oxidative stress. Consistent with its role as a fatty acid sensor, these findings provide genetic evidence that endothelial PPARgamma plays a critical role in protecting blood vessels in response to a high-fat diet.


Subject(s)
Cerebrovascular Circulation/genetics , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/metabolism , Dietary Fats/administration & dosage , Endothelium, Vascular/physiopathology , PPAR gamma/physiology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Cells, Cultured , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/genetics , PPAR gamma/genetics
12.
Hypertension ; 51(4): 867-71, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18285614

ABSTRACT

The transcription factor PPARgamma is expressed in endothelium and vascular muscle where it may exert antiinflammatory and antioxidant effects. We tested the hypothesis that PPARgamma plays a protective role in the vasculature by examining vascular structure and function in heterozygous knockin mice expressing the P465L dominant negative mutation in PPARgamma (L/+). In L/+ aorta, responses to the endothelium-dependent agonist acetylcholine (ACh) were not affected, but there was an increase in contraction to serotonin, PGF(2alpha), and endothelin-1. In cerebral blood vessels both in vitro and in vivo, ACh produced dilation that was markedly impaired in L/+ mice. Superoxide levels were elevated in cerebral arterioles from L/+ mice and responses to ACh were restored to normal with a scavenger of superoxide. Diameter of maximally dilated cerebral arterioles was less, whereas wall thickness and cross-sectional area was greater in L/+ mice, indicating cerebral arterioles underwent hypertrophy and remodeling. Thus, interference with PPARgamma signaling produces endothelial dysfunction via a mechanism involving oxidative stress and causes vascular hypertrophy and inward remodeling. These findings indicate that PPARgamma has vascular effects which are particularly profound in the cerebral circulation and provide genetic evidence that PPARgamma plays a critical role in protecting blood vessels.


Subject(s)
Cerebrovascular Circulation/physiology , Hypertension/physiopathology , PPAR gamma/genetics , PPAR gamma/metabolism , Signal Transduction/physiology , Acetylcholine/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Arterioles/pathology , Arterioles/physiopathology , Dinoprost/pharmacology , Endothelin-1/pharmacology , Female , Gene Expression Profiling , Genes, Dominant , Hypertension/pathology , Hypertrophy , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Serotonin/pharmacology , Serotonin Agents/pharmacology , Signal Transduction/drug effects , Vasodilator Agents/pharmacology
13.
Am J Physiol Heart Circ Physiol ; 291(5): H2301-7, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16782846

ABSTRACT

20-Hydroxyeicosatetraenoic acid (20-HETE), an arachidonic acid (AA) metabolite synthesized by cytochrome P-450 omega-oxidases, is reported to produce vasoconstriction in the cerebral circulation. However, we find that like 14,15-epoxyeicosatrienoic acid (14,15-EET), 20-HETE produces dilation of mouse basilar artery preconstricted with U-46619 in vitro. Indomethacin inhibited the vasodilation produced by 20-HETE but not by 14,15-EET, suggesting a cyclooxygenase (COX)-dependent mechanism. Metabolic studies indicated several mechanisms that may play a role in this process. Mouse brain endothelial cells (MBEC) converted 20-HETE to 20-OH-PGE(2), which was as potent as PGE(2) in dilating the basilar artery. 20-HETE also stimulated AA release and PGE(2) and 6-keto-PGF(1alpha) production in MBEC. Furthermore, the basilar artery converted 20-HETE to 20-COOH-AA, which also produced COX-dependent dilation of the basilar artery. 20-COOH-AA increased AA release and PGE(2) and 6-keto-PGF(1alpha) production by the MBEC, but to a lesser extent than 20-HETE. Whereas the conversion of 20-HETE to 20-OH-PGE(2) and production of endogenous prostaglandins probably are primarily responsible for vasodilation, the production of 20-COOH-AA also may contribute to this process.


Subject(s)
Basilar Artery/drug effects , Hydroxyeicosatetraenoic Acids/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , Vasodilation/drug effects , Vasodilation/physiology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Cell Culture Techniques , Cells, Cultured , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Indomethacin/pharmacology , Mice , Mice, Inbred C57BL , Telencephalon/blood supply , Vasoconstrictor Agents/pharmacology
14.
J Cereb Blood Flow Metab ; 26(4): 449-55, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16094317

ABSTRACT

Very little is known regarding the mechanisms of action of angiotensin II (Ang II) or the consequences of Ang II-dependent hypertension in the cerebral circulation. We tested the hypothesis that Ang II produces constriction of cerebral arteries that is mediated by activation of AT1A receptors and Rho-kinase. Basilar arteries (baseline diameter approximately 130 microm) from mice were isolated, cannulated and pressurized to measure the vessel diameter. Angiotensin II was a potent constrictor in arteries from male, but not female, mice. Vasoconstriction in response to Ang II was prevented by an inhibitor of Rho-kinase (Y-27632) in control mice, and was reduced by approximately 85% in mice deficient in expression of AT1A receptors. We also examined the chronic effects of Ang II using a model of Ang II-dependent hypertension, mice which overexpress human renin (R+) and angiotensinogen (A+). Responses to the endothelium-dependent agonist acetylcholine were markedly impaired in R+A+ mice (P<0.01) compared with controls, but were restored to normal by a superoxide scavenger (PEG-SOD). A-23187 (another endothelium-dependent agonist) produced vasodilation in control mice, but no response or vasoconstriction in R+A+ mice. In contrast, dilation of the basilar artery in response to a NO donor (NONOate) was similar in R+A+ mice and controls. Thus, Ang II produces potent constriction of cerebral arteries via activation of AT1A receptors and Rho-kinase. There are marked gender differences in cerebral vascular responses to Ang II. Endothelial function is greatly impaired in a genetic model of Ang II-dependent hypertension via a mechanism that involves superoxide.


Subject(s)
Angiotensin II/pharmacology , Cerebrovascular Circulation/drug effects , Hypertension/physiopathology , Angiotensinogen/genetics , Animals , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Endothelium, Vascular/pathology , Female , Humans , Hypertension/etiology , Hypertension/pathology , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Serine-Threonine Kinases/physiology , Receptor, Angiotensin, Type 1/physiology , Renin/genetics , Sex Factors , Superoxides , Vasoconstriction/drug effects , rho-Associated Kinases
15.
Biol Reprod ; 66(2): 468-74, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11804964

ABSTRACT

During ovine pregnancy, when both estrogen and progesterone are elevated, prostacyclin (PGI2) production by uterine arteries and the key enzymes for PGI2 production, phospholipase A2 (cPLA2), cyclooxygenase 1 (COX-1), and prostacyclin synthetase (PGIS), are increased. This study was conducted to determine whether exogenous estradiol-17beta (E2beta) with or without progesterone (P4) treatment would increase cPLA2, COX-1, and PGIS protein expression in ovine uterine, mammary, and systemic (renal, mental, and coronary) arteries. Nonpregnant ovariectomized sheep received vehicle (n = 10), P(4) (0.9-g controlled internal drug release vaginal implants; n = 13), E2beta (5 microg/kg bolus followed by 6 microg x kg(-1) x day(-1); n = 10), or P4 + E2beta (n = 12). Arteries were procured on Day 10, and cPLA2, COX-1, and PGIS protein were measured by Western immunoblot analysis in endothelial isolated proteins and vascular smooth muscle (VSM). The levels of cPLA2 was increased in uterine artery endothelium in ewes treated with P4 + E2beta but was not altered by any steroid treatment in renal, coronary, mammary, or omental artery endothelium or in VSM of any evaluated artery. Similarly, COX-1 was increased in uterine artery endothelium with P4 + E2beta but was not significantly altered by treatment in other endothelium or VSM. E2beta treatment increased PGIS protein in uterine and renal artery endothelium but did not alter PGIS in other endothelial tissue. P4 increased PGIS expression in the uterine, mammary, omental, and renal artery VSM, and E2beta increased PGIS expression in the uterine and omental artery VSM. Both E2beta and P4 treatments differentially alter protein expression of the key enzymes involved in PGI2 production in different artery types and may play an important role in the control of blood flow redistribution during hormone replacement therapy.


Subject(s)
Arteries/metabolism , Cytochrome P-450 Enzyme System/biosynthesis , Endothelium, Vascular/metabolism , Estrogens/pharmacology , Intramolecular Oxidoreductases/biosynthesis , Isoenzymes/biosynthesis , Phospholipases A/biosynthesis , Progesterone/pharmacology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Uterus/metabolism , Vasodilation/physiology , Animals , Blotting, Western , Cyclooxygenase 1 , Endothelium, Vascular/physiology , Female , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Ovariectomy , Phospholipases A2 , Pregnancy , Regional Blood Flow/physiology , Sheep , Uterus/blood supply
16.
Am J Physiol Heart Circ Physiol ; 282(1): H342-8, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11748080

ABSTRACT

The follicular phase (FOL) and pregnancy exhibit increases in uterine blood flow (UBF), estrogen levels, and uterine artery (UA) endothelial nitric oxide synthase (eNOS) expression. UA branching within the mesometrium increases the total vascular cross-sectional area, which reduces the vascular perfusion pressure gradient, thus locally decreasing the blood flow velocity. Shear stress (SS) activates eNOS and may be associated with UBF elevations during FOL and pregnancy. We hypothesized that regional differences in eNOS responses are observed with both decreases in vessel diameter and during the ovarian cycle and pregnancy. Endothelial isolated proteins were collected from renal (RA) and internal iliac arteries (II) as well as from primary (UA 1 degrees ), secondary (UA 2 degrees), and tertiary (UA 3 degrees) UA branches of nonpregnant luteal phase (LUT; n = 6) and FOL (n = 6) as well as midpregnant (MP; 82 +/- 1 days gestation, n = 6) and late pregnant (LP; 127 +/- 3 days gestation, n = 6) ewes (term = 145 +/- 3 days gestation) for Western blot analysis. LUT RA, II, and UA 1 degrees eNOS levels were similar. There was a 60.7 +/- 9.8% reduction in eNOS expression in UA 2 degrees and UA 3 degrees. A similar decreasing eNOS regional expression gradient was observed in LP ewes. No eNOS regional expression gradient was observed in FOL or MP ewes because eNOS increased in UA 2 degrees and UA 3 degrees. In UA 2 degrees and UA 3 degrees, MP > LP = FOL > LUT. Thus, with increasing UBF, FOL and pregnancy rises in SS may regulate eNOS protein expression in smaller diameter UAs. A decrease in LUT and LP UA 2 degrees and UA 3 degrees endothelial eNOS suggest a possible negative feedback mechanism due to downregulation of eNOS if SS is normalized.


Subject(s)
Arteries/physiology , Estrus/physiology , Iliac Artery/physiology , Nitric Oxide Synthase/metabolism , Pregnancy, Animal/physiology , Uterus/blood supply , Animals , Dinoprost/pharmacology , Endothelium, Vascular/enzymology , Female , Nitric Oxide Synthase Type III , Pregnancy , Regional Blood Flow , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...