Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Neurodegener ; 11: 9, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26801396

ABSTRACT

BACKGROUND: Substantial evidence suggests that amyloid-ß (Aß) species induce oxidative stress and cerebrovascular (CV) dysfunction in Alzheimer's disease (AD), potentially contributing to the progressive dementia of this disease. The upstream molecular pathways governing this process, however, are poorly understood. In this report, we examine the role of heparan sulfate proteoglycans (HSPG) in Aß-induced vascular smooth muscle cell (VSMC) dysfunction in vitro. RESULTS: Our results demonstrate that pharmacological depletion of HSPG (by enzymatic degradation with active, but not heat-inactivated, heparinase) in primary human cerebral and transformed rat VSMC mitigates Aß(1-40⁻) and Aß(1-42⁻)induced oxidative stress. This inhibitory effect is specific for HSPG depletion and does not occur with pharmacological depletion of other glycosaminoglycan (GAG) family members. We also found that Aß(1-40) (but not Aß(1-42)) causes a hypercontractile phenotype in transformed rat cerebral VSMC that likely results from a HSPG-mediated augmentation in intracellular Ca(2+) activity, as both Aß(1-40⁻)induced VSMC hypercontractility and increased Ca(2+) influx are inhibited by pharmacological HSPG depletion. Moreover, chelation of extracellular Ca(2+) with ethylene glycol tetraacetic acid (EGTA) does not prevent the production of Aß(1-40⁻) or Aß(1-42⁻)mediated reactive oxygen species (ROS), suggesting that Aß-induced ROS and VSMC hypercontractility occur through different molecular pathways. CONCLUSIONS: Taken together, our data indicate that HSPG are critical mediators of Aß-induced oxidative stress and Aß(1-40⁻)induced VSMC dysfunction.


Subject(s)
Amyloid beta-Peptides/metabolism , Heparan Sulfate Proteoglycans/metabolism , Muscle, Smooth, Vascular/metabolism , Oxidative Stress/physiology , Peptide Fragments/metabolism , Reactive Oxygen Species/metabolism , Alzheimer Disease/metabolism , Cell Line , Cells, Cultured , Humans
2.
Neurosci Res ; 107: 57-62, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26712324

ABSTRACT

We investigated in cerebral penetrating arterioles the signaling mechanisms and dose-dependency of extracellular magnesium-induced vasodilation and also its vasodilatory effects in vessels preconstricted with agonists associated with delayed cerebral vasospasm following SAH. Male rat penetrating arterioles were cannulated. Their internal diameters were monitored. To investigate mechanisms of magnesium-induced vasodilation, inhibitors of endothelial function, potassium channels and endothelial impairment were tested. To simulate cerebral vasospasm we applied several spasmogenic agonists. Increased extracellular magnesium concentration produced concentration-dependent vasodilation, which was partially attenuated by non-specific calcium-sensitive potassium channel inhibitor tetraethylammonium, but not by other potassium channel inhibitors. Neither the nitric oxide synthase inhibitor L-NNA nor endothelial impairment induced by air embolism reduced the dilation. Although the magnesium-induced vasodilation was slightly attenuated by the spasmogen ET-1, neither application of PF2α nor TXA2 analog effect the vasodilation. Magnesium induced a concentration- and smooth muscle cell-dependent dilation in cerebral penetrating arterioles. Calcium-sensitive potassium channels of smooth muscle cells may play a key role in magnesium-induced vasodilation. Magnesium also dilated endothelium-impaired vessels as well as vessels preconstricted with spasmogenic agonists. These results provide a fundamental background for the clinical use of magnesium, especially in treatment against delayed cerebral ischemia or vasospasm following SAH.


Subject(s)
Cerebral Cortex/drug effects , Magnesium Compounds/pharmacology , Vasodilation/drug effects , Animals , Arterioles/drug effects , Arterioles/physiopathology , Cations, Divalent , Cerebral Cortex/blood supply , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Extracellular Space/chemistry , Magnesium Compounds/analysis , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/physiopathology
3.
J Korean Neurosurg Soc ; 57(1): 1-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25674336

ABSTRACT

OBJECTIVE: Cerebral vessels, such as intracerebral perforating arterioles isolated from rat brain, have been widely used as an ex vivo model to study the cerebrovascular function associated with cerebrovascular disorders and the therapeutic effects of various pharmacological agents. These perforating arterioles, however, have demonstrated differences in the vascular architecture and reactivity compared with a larger leptomeningeal artery which has been commonly implicated in cerebrovascular disease. In this study, therefore, we developed the method for studying cerebrovascular function utilizing the olfactory artery isolated from the mouse brain. METHODS: The olfactory artery (OA) was isolated from the C57/BL6 wild-type mouse brain. After removing connective tissues, one side of the isolated vessel segment (approximately -500 µm in length) was cannulated and the opposite end of the vessel was completely sealed while being viewed with an inverted microscope. After verifying the absence of pressure leakage, we examined the vascular reactivity to various vasoactive agents under the fixed intravascular pressure (60 mm Hg). RESULTS: We found that the isolated mouse OAs were able to constrict in response to vasoconstrictors, including KCl, phenylephrine, endothelin-1, and prostaglandin PGH2. Moreover, this isolated vessel demonstrated vasodilation in a dose-dependent manner when vasodilatory agents, acetylcholine and bradykinin, were applied. CONCLUSION: Our findings suggest that the isolated olfactory artery would provide as a useful ex vivo model to study the molecular and cellular mechanisms of vascular function underlying cerebrovascular disorders and the direct effects of such disease-modifying pathways on cerebrovascular function utilizing pharmacological agents and genetically modified mouse models.

4.
Proc Natl Acad Sci U S A ; 112(8): E881-90, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675483

ABSTRACT

Cerebral amyloid angiopathy (CAA) is characterized by deposition of amyloid ß peptide (Aß) within walls of cerebral arteries and is an important cause of intracerebral hemorrhage, ischemic stroke, and cognitive dysfunction in elderly patients with and without Alzheimer's Disease (AD). NADPH oxidase-derived oxidative stress plays a key role in soluble Aß-induced vessel dysfunction, but the mechanisms by which insoluble Aß in the form of CAA causes cerebrovascular (CV) dysfunction are not clear. Here, we demonstrate evidence that reactive oxygen species (ROS) and, in particular, NADPH oxidase-derived ROS are a key mediator of CAA-induced CV deficits. First, the NADPH oxidase inhibitor, apocynin, and the nonspecific ROS scavenger, tempol, are shown to reduce oxidative stress and improve CV reactivity in aged Tg2576 mice. Second, the observed improvement in CV function is attributed both to a reduction in CAA formation and a decrease in CAA-induced vasomotor impairment. Third, anti-ROS therapy attenuates CAA-related microhemorrhage. A potential mechanism by which ROS contribute to CAA pathogenesis is also identified because apocynin substantially reduces expression levels of ApoE-a factor known to promote CAA formation. In total, these data indicate that ROS are a key contributor to CAA formation, CAA-induced vessel dysfunction, and CAA-related microhemorrhage. Thus, ROS and, in particular, NADPH oxidase-derived ROS are a promising therapeutic target for patients with CAA and AD.


Subject(s)
Aging/pathology , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy/physiopathology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/physiopathology , Reactive Oxygen Species/metabolism , Vasomotor System/physiopathology , Acetophenones/pharmacology , Animals , Apolipoproteins E/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cerebral Amyloid Angiopathy/complications , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cerebral Hemorrhage/complications , Cricetinae , Cyclic N-Oxides/pharmacology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Oxidative Stress/drug effects , Spin Labels , Vasomotor System/drug effects , Vasomotor System/pathology
5.
Am J Physiol Heart Circ Physiol ; 306(5): H654-66, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24414067

ABSTRACT

Elastin (Eln) insufficiency in mice and humans is associated with hypertension and altered structure and mechanical properties of large arteries. However, it is not known to what extent functional or structural changes in resistance arteries contribute to the elevated blood pressure that is characteristic of Eln insufficiency. Here, we investigated how Eln insufficiency affects the structure and function of the resistance vasculature. A functional profile of resistance vasculature in Eln(+/-) mice was generated by assessing small mesenteric artery (MA) contractile and vasodilatory responses to vasoactive agents. We found that Eln haploinsufficiency had a modest effect on phenylephrine-induced vasoconstriction, whereas ANG II-evoked vasoconstriction was markedly increased. Blockade of ANG II type 2 receptors with PD-123319 or modulation of Rho kinase activity with the inhibitor Y-27632 attenuated the augmented vasoconstriction, whereas acute Y-27632 administration normalized blood pressure in Eln(+/-) mice. Sodium nitroprusside- and isoproterenol-induced vasodilatation were normal, whereas ACh-induced vasodilatation was severely impaired in Eln(+/-) MAs. Histologically, the number of smooth muscle layers did not change in Eln(+/-) MAs; however, an additional discontinuous layer of Eln appeared between the smooth muscle layers that was absent in wild-type arteries. We conclude that high blood pressure arising from Eln insufficiency is due partly to permanent changes in vascular tone as a result of increased sensitivity of the resistance vasculature to circulating ANG II and to impaired vasodilatory mechanisms arising from endothelial dysfunction characterized by impaired endothelium-dependent vasodilatation. Eln insufficiency causes augmented ANG II-induced vasoconstriction in part through a novel mechanism that facilitates contraction evoked by ANG II type 2 receptors and altered G protein signaling.


Subject(s)
Arterial Pressure , Elastin/deficiency , Hypertension/metabolism , Mesenteric Arteries/metabolism , Vascular Resistance/drug effects , Vasoconstriction , Vasodilation , Angiotensin II/metabolism , Animals , Arterial Pressure/drug effects , Calcium/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Elastin/genetics , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Genetic Predisposition to Disease , Haploinsufficiency , Hemizygote , Hypertension/drug therapy , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/pathology , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Kinase Inhibitors/pharmacology , Receptor, Angiotensin, Type 2/drug effects , Receptor, Angiotensin, Type 2/metabolism , Signal Transduction/drug effects , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
6.
J Am Heart Assoc ; 2(4): e000365, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23974906

ABSTRACT

BACKGROUND: KATP channels, assembled from pore-forming (Kir6.1 or Kir6.2) and regulatory (SUR1 or SUR2) subunits, link metabolism to excitability. Loss of Kir6.2 results in hypoglycemia and hyperinsulinemia, whereas loss of Kir6.1 causes Prinzmetal angina-like symptoms in mice. Conversely, overactivity of Kir6.2 induces neonatal diabetes in mice and humans, but consequences of Kir6.1 overactivity are unknown. METHODS AND RESULTS: We generated transgenic mice expressing wild-type (WT), ATP-insensitive Kir6.1 [Gly343Asp] (GD), and ATP-insensitive Kir6.1 [Gly343Asp,Gln53Arg] (GD-QR) subunits, under Cre-recombinase control. Expression was induced in smooth muscle cells by crossing with smooth muscle myosin heavy chain promoter-driven tamoxifen-inducible Cre-recombinase (SMMHC-Cre-ER) mice. Three weeks after tamoxifen induction, we assessed blood pressure in anesthetized and conscious animals, as well as contractility of mesenteric artery smooth muscle and KATP currents in isolated mesenteric artery myocytes. Both systolic and diastolic blood pressures were significantly reduced in GD and GD-QR mice but normal in mice expressing the WT transgene and elevated in Kir6.1 knockout mice as well as in mice expressing dominant-negative Kir6.1 [AAA] in smooth muscle. Contractile response of isolated GD-QR mesenteric arteries was blunted relative to WT controls, but nitroprusside relaxation was unaffected. Basal KATP conductance and pinacidil-activated conductance were elevated in GD but not in WT myocytes. CONCLUSIONS: KATP overactivity in vascular muscle can lead directly to reduced vascular contractility and lower blood pressure. We predict that gain of vascular KATP function in humans would lead to a chronic vasodilatory phenotype, as indeed has recently been demonstrated in Cantu syndrome.


Subject(s)
Blood Pressure , Hypotension/metabolism , KATP Channels/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/genetics , Dose-Response Relationship, Drug , Genetic Predisposition to Disease , Hypotension/genetics , Hypotension/physiopathology , KATP Channels/genetics , Membrane Potentials , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Mutation , Phenotype , Potassium/metabolism , Vasoconstriction , Vasodilation , Vasodilator Agents/pharmacology
7.
Stroke ; 44(3): 779-85, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23362079

ABSTRACT

BACKGROUND AND PURPOSE: Reduced risk and severity of stroke in adult females are thought to depend on normal levels of endogenous estrogen, which is a known neuro- and vasoprotective agent in experimental cerebral ischemia. Recently, a novel G protein-coupled estrogen receptor (GPER, formerly GPR30) has been identified and may mediate the vasomotor and -protective effects of estrogen. However, the signaling mechanisms associated with GPER in the cerebral microcirculation remain unclear. We investigated the mechanism of GPER-mediated vasoreactivity and also its vasoprotective effect after hypoxia/reoxygenation (H/RO) injury. METHODS: Rat cerebral penetrating arterioles from both sexes were isolated, cannulated, and pressurized. Vessel diameters were recorded by computer-aided videomicroscopy. To investigate vasomotor mechanism of the GPER agonist (G-1), several inhibitors with or without endothelial impairment were tested. Ischemia/reperfusion injury was simulated using H/RO. Vasomotor responses to adenosine triphophate after H/RO were measured with or without G-1 and compared with controls. RESULTS: G-1 produced a vasodilatory response, which was partially dependent on endothelium-derived nitric oxide (NO) but not arachidonic acid cascades and endothelial hyperpolarization factor. Attenuation of G-1-vasodilation by the NO synthase inhibitor and endothelium-impairment were greater in vessels from female than male animals. G-1 treatment after H/RO injury fully restored arteriolar dilation to adenosine triphophate compared with controls. CONCLUSIONS: GPER agonist elicited dilation, which was partially caused by endothelial NO pathway and induced by direct relaxation of smooth muscle cells. Further, GPER agonist restored vessel function of arterioles after H/RO injury and may play an important role in the ability of estrogen to protect the cerebrovasculature against ischemia/reperfusion injury.


Subject(s)
Brain/blood supply , Cerebrovascular Circulation/drug effects , Cyclopentanes/pharmacology , Microvessels/drug effects , Quinolines/pharmacology , Receptors, G-Protein-Coupled/agonists , Reperfusion Injury/physiopathology , Animals , Benzodioxoles/pharmacology , Brain/drug effects , Cerebrovascular Circulation/physiology , Estrogens/physiology , Female , Male , Microvessels/physiopathology , Models, Animal , Rats , Rats, Inbred Lew , Receptors, G-Protein-Coupled/antagonists & inhibitors , Sex Factors , Signal Transduction/drug effects , Signal Transduction/physiology , Vasodilation/drug effects , Vasodilation/physiology
8.
Microcirculation ; 19(5): 461-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22372561

ABSTRACT

OBJECTIVE: VaD is the second-most common form of dementia, second only to that caused by AD. As the name indicates, VaD is predominantly considered a disease caused by vascular phenomena. METHODS: In this invited review, we introduce the reader to recent developments in defining VaD as a unique form of dementia by reviewing the current pertinent literature. We discuss the clinical and experimental evidence that supports the notion that the microcirculation, specifically cell-to-cell communication, likely contributes to the development of VaD. Through exploration of the concept of the NVU, we elucidate the extensive cerebrovascular communication that exists and highlight models that may help test the contribution(s) of cell-to-cell communication at the microvascular level to the development and progression of VaD. Lastly, we explore the possibility that some dementia, generally considered to be purely neurodegenerative, may actually have a vascular component at the neurovascular level. CONCLUSION: This latter recognition potentially broadens the critical involvement of microvascular events that contribute to the numerous dementias affecting an increasingly larger sector of the adult population.


Subject(s)
Cell Communication , Dementia, Vascular/physiopathology , Models, Cardiovascular , Adult , Animals , Humans
9.
J Biol Chem ; 287(15): 12541-9, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22354966

ABSTRACT

Regulator of G protein signaling 2 (RGS2) is a GTPase-activating protein for G(q/11)α and G(i/o)α subunits. RGS2 deficiency is linked to hypertension in mice and humans, although causative mechanisms are not understood. Because endothelial dysfunction and increased peripheral resistance are hallmarks of hypertension, determining whether RGS2 regulates microvascular reactivity may reveal mechanisms relevant to cardiovascular disease. Here we have determined the effects of systemic versus endothelium- or vascular smooth muscle-specific deletion of RGS2 on microvascular contraction and relaxation. Contraction and relaxation of mesenteric resistance arteries were analyzed in response to phenylephrine, sodium nitroprusside, or acetylcholine with or without inhibitors of nitric oxide (NO) synthase or K(+) channels that mediate endothelium-derived hyperpolarizing factor (EDHF)-dependent relaxation. The results showed that deleting RGS2 in vascular smooth muscle had minor effects. Systemic or endothelium-specific deletion of RGS2 strikingly inhibited acetylcholine-evoked relaxation. Endothelium-specific deletion of RGS2 had little effect on NO-dependent relaxation but markedly impaired EDHF-dependent relaxation. Acute, inducible deletion of RGS2 in endothelium did not affect blood pressure significantly. Impaired EDHF-mediated vasodilatation was rescued by blocking G(i/o)α activation with pertussis toxin. These findings indicated that systemic or endothelium-specific RGS2 deficiency causes endothelial dysfunction resulting in impaired EDHF-dependent vasodilatation. RGS2 deficiency enables endothelial G(i/o) activity to inhibit EDHF-dependent relaxation, whereas RGS2 sufficiency facilitates EDHF-evoked relaxation by squelching endothelial G(i/o) activity. Mutation or down-regulation of RGS2 in hypertension patients therefore may contribute to endothelial dysfunction and defective EDHF-dependent relaxation. Blunting G(i/o) signaling might improve endothelial function in such patients.


Subject(s)
Biological Factors/physiology , Endothelial Cells/metabolism , Endothelium, Vascular/physiopathology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , RGS Proteins/deficiency , Vasodilation , Acetylcholine/pharmacology , Animals , Biological Factors/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Endothelin Receptor Antagonists , Endothelium, Vascular/pathology , Gene Knockout Techniques , Hemodynamics , Hypertension/metabolism , In Vitro Techniques , Mesenteric Arteries/drug effects , Mesenteric Arteries/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Pertussis Toxin/pharmacology , RGS Proteins/genetics , Signal Transduction , Vasodilator Agents/pharmacology
10.
Am J Physiol Heart Circ Physiol ; 298(6): H2208-20, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20382858

ABSTRACT

Activation of phospholipases leads to the release of arachidonic acid and lysophospholipids that play prominent roles in regulating vasomotor tone. To identify the role of calcium-independent phospholipase A(2)beta (iPLA(2)beta) in vasomotor function, we measured vascular responses to phenylephrine (PE) and ACh in mesenteric arterioles from wild-type (WT; iPLA(2)beta(+/+)) mice and those lacking the beta-isoform (iPLA(2)beta(-/-)) both ex vivo and in vivo. Vessels isolated from iPLA(2)beta(-/-) mice demonstrated increased constriction to PE, despite lower basal smooth muscle calcium levels, and decreased vasodilation to ACh compared with iPLA(2)beta(+/+) mice. PE constriction resulted in initial intracellular calcium release with subsequent steady-state constriction that depended on extracellular calcium influx. Endothelial denudation had no effect on vessel tone or PE-induced constriction although the dilation to ACh was significantly reduced in iPLA(2)beta(+/+) vessels. In contrast, vessels from iPLA(2)beta(-/-) constricted by 54% after denudation, indicating smooth muscle hypercontractility. In vivo, blood pressure, resting vessel diameter, and constriction of mesenteric vessels to PE were not different in iPLA(2)beta(-/-) vessels compared with WT mouse vessels. However, relaxation after ACh administration in situ was attenuated, indicating an endothelial inability to induce dilation in response to ACh. In cultured endothelial cells, inhibition of iPLA(2)beta with (S)-(E)-6-(bromomethylene)tetrahydro-3-(1-naphthalenyl)-2H-pyran-2-one (BEL) decreased endothelial nitric oxide synthase phosphorylation and reduced endothelial agonist-induced intracellular calcium release as well as extracellular calcium influx. We conclude that iPLA(2)beta is an important mediator of vascular relaxation and intracellular calcium homeostasis in both smooth muscle and endothelial cells and that ablation of iPLA(2)beta causes agonist-induced smooth muscle hypercontractility and reduced agonist-induced endothelial dilation.


Subject(s)
Acetylcholine/pharmacology , Endothelium, Vascular/physiology , Muscle, Smooth, Vascular/physiology , Phospholipases A2, Calcium-Independent/genetics , Phospholipases A2, Calcium-Independent/physiology , Vasoconstriction/physiology , Vasodilation/physiology , Animals , Calcium/metabolism , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Group VI Phospholipases A2/genetics , Group VI Phospholipases A2/physiology , Homeostasis/physiology , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nitric Oxide Synthase Type III/metabolism , Phenylephrine/pharmacology , Phosphorylation , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
11.
Mol Neurodegener ; 5: 15, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20388225

ABSTRACT

BACKGROUND: Evidence indicates that soluble forms of amyloid-beta (Abeta) are vasoactive, which may contribute to cerebrovascular dysfunction noted in patients with Alzheimer's Disease and cerebral amyloid angiopathy. The effects of soluble Abeta on penetrating cerebral arterioles - the vessels most responsible for controlling cerebrovascular resistance - have not been studied. RESULTS: Freshly dissolved Abeta1-40 and Abeta1-42, but not the reverse peptide Abeta40-1 constricted isolated rat penetrating arterioles and diminished dilation to adenosine tri-phosphate (ATP). Abeta1-42 also enhanced ATP-induced vessel constriction. Abeta1-40 diminished arteriolar myogenic response, and an anti-Abeta antibody reduced Abeta1-40 induced arteriolar constriction. Prolonged Abeta exposure in vessels of Tg2576 mice resulted in a marked age-dependent effect on ATP-induced vascular responses. Vessels from 6 month old Tg2576 mice had reduced vascular responses whereas these were absent from 12 month old animals. Abeta1-40 and Abeta1-42 acutely increased production of reactive oxygen species (ROS) in cultured rat cerebro-microvascular cells. The radical scavenger MnTBAP attenuated this Abeta-induced oxidative stress and Abeta1-40-induced constriction in rat arterioles. CONCLUSIONS: Our results suggest that soluble Abeta1-40 and Abeta1-42 directly affect the vasomotor regulation of isolated rodent penetrating arterioles, and that ROS partially mediate these effects. Once insoluble Abeta deposits are present, arteriolar reactivity is greatly diminished.

12.
Physiology (Bethesda) ; 24: 107-16, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19364913

ABSTRACT

Through oxygen-dependent release of the vasodilator ATP, the mobile erythrocyte plays a fundamental role in matching microvascular oxygen supply with local tissue oxygen demand. Signal transduction within the erythrocyte and microvessels as well as feedback mechanisms controlling ATP release have been described. Our understanding of the impact of this novel control mechanism will rely on the integration of in vivo experiments and computational models.


Subject(s)
Erythrocytes/physiology , Muscle, Smooth, Vascular/physiology , Oxygen Consumption/physiology , Adenosine Triphosphate/blood , Animals , Blood Vessels/physiology , Erythrocytes/metabolism , Humans , Muscle Tonus/physiology , Signal Transduction/physiology
13.
J Vasc Res ; 46(3): 253-64, 2009.
Article in English | MEDLINE | ID: mdl-18984964

ABSTRACT

BACKGROUND: Adenosine triphosphate (ATP), a potent vascular regulator in the cerebral circulation, initiates conducted vasomotor responses which may be impaired after pathological insults. We analyzed the mechanism of ATP-induced local vasomotor responses and their effect on conducted vasomotor responses in rat cerebral penetrating arterioles. METHODS: Arterioles were cannulated and their internal diameter monitored. Vasomotor responses to ATP were observed in the presence or absence of inhibitors, or after endothelial impairment. Smooth muscle membrane potentials were measured in some vessels. RESULTS: Microapplication of ATP produced a biphasic response (constriction followed by dilation), which resulted in conducted dilation preceded by a membrane hyperpolarization. alpha,beta-methylene-ATP or pyridoxal phosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) blunted the ATP-mediated constriction and enhanced local and conducted dilation. N(omega)-monomethyl-L-arginine, endothelial impairment and N-methylsulfonyl-6-(2-propargyloxyphenyl) hexanamide (MS-PPOH) reduced the local dilation caused by ATP. The conducted dilation was attenuated by MS-PPOH and endothelial impairment, but not N(omega)-monomethyl-L-arginine or indomethacin. CONCLUSION: ATP-induced conducted dilation is preceded by membrane hyperpolarization. Local ATP induces initial local constriction via smooth-muscle P(2X1) and subsequent dilation via endothelial P(2Y) receptors. Nitric oxide, cytochrome P450 metabolites, and intermediate and large conductance K(Ca) channels mediate dilation caused by ATP. ATP-induced conducted dilation is dependent upon both the endothelium and cytochrome P450 metabolites.


Subject(s)
Adenosine Triphosphate/pharmacology , Arterioles/drug effects , Cerebral Arteries/drug effects , Adenosine Triphosphate/analogs & derivatives , Amides/pharmacology , Animals , Arterioles/physiology , Biological Factors/physiology , Cerebral Arteries/physiology , Cytochrome P-450 Enzyme System/physiology , Endothelium, Vascular/physiology , Male , Membrane Potentials/drug effects , Nitric Oxide/physiology , Potassium Channels, Calcium-Activated/physiology , Prostaglandins/physiology , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/physiology , Vasodilation/drug effects
14.
J Neurosci ; 28(50): 13542-50, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19074028

ABSTRACT

The contributing effect of cerebrovascular pathology in Alzheimer's disease (AD) has become increasingly appreciated. Recent evidence suggests that amyloid-beta peptide (Abeta), the same peptide found in neuritic plaques of AD, may play a role via its vasoactive properties. Several studies have examined young Tg2576 mice expressing mutant amyloid precursor protein (APP) and having elevated levels of soluble Abeta but no cerebral amyloid angiopathy (CAA). These studies suggest but do not prove that soluble Abeta can significantly impair the cerebral circulation. Other studies examining older Tg2576 mice having extensive CAA found even greater cerebrovascular dysfunction, suggesting that CAA is likely to further impair vascular function. Herein, we examined vasodilatory responses in young and older Tg2576 mice to further assess the roles of soluble and insoluble Abeta on vessel function. We found that (1) vascular impairment was present in both young and older Tg2576 mice; (2) a strong correlation between CAA severity and vessel reactivity exists; (3) a surprisingly small amount of CAA led to marked reduction or complete loss of vessel function; 4) CAA-induced vasomotor impairment resulted from dysfunction rather than loss or disruption of vascular smooth muscle cells; and 5) acute depletion of Abeta improved vessel function in young and to a lesser degree older Tg2576 mice. These results strongly suggest that both soluble and insoluble Abeta cause cerebrovascular dysfunction, that mechanisms other than Abeta-induced alteration in vessel integrity are responsible, and that anti-Abeta therapy may have beneficial vascular effects in addition to positive effects on parenchymal amyloid.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Brain/blood supply , Cerebrovascular Circulation/physiology , Muscle, Smooth, Vascular/physiopathology , Alzheimer Disease/physiopathology , Animals , Brain/pathology , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Amyloid Angiopathy/physiopathology , Mice , Mice, Transgenic
16.
Stroke ; 34(6): 1473-8, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12730558

ABSTRACT

BACKGROUND AND PURPOSE: P2 receptors are important regulators of cerebrovascular tone. However, there is functional heterogeneity of P2Y receptors along the vascular tree, and the functionality of P2Y receptors in small arterioles has not been studied in detail. We investigated the effects of activating P2Y1 and P2Y2 receptors and their underlying dilator mechanisms in rat intracerebral arterioles. METHODS: We used computer-aided videomicroscopy to measure diameter responses from isolated and pressurized rat penetrating arterioles (39.9+/-1.2 microm) to the natural P2 receptor agonist ATP in addition to ADP-beta-S (P2Y1-selective) and ATP-gamma-S (P2Y2-selective) and inhibitors of signaling pathways. RESULTS: Extraluminal application of ATP-gamma-S and ADP-beta-S initiated a biphasic response (initial constriction followed by the secondary dilation) similar to ATP-induced responses. Pyridoxal phosphate-6-azophenyl-2',4'-disulphonic acid (0.1 mmol/L; a P2Y1 receptor antagonist) blocked ADP-beta-S- but not ATP-gamma-S-induced dilation and affected the ATP-mediated dilation at low concentrations. Nomega-Monomethyl-l-arginine partially inhibited the dilation of ATP and ADP-beta-S but not ATP-gamma-S. High K+ saline suppressed the dilation of all agonists. Indomethacin had no effect. CONCLUSIONS: Both P2Y1 and P2Y2 receptors are functionally present in cerebral arterioles. ATP stimulates P2Y1 receptors at low concentrations, while high concentrations of ATP activate P2Y2 in addition to P2Y1 receptors. Nitric oxide is involved in P2Y1 but not P2Y2 receptor activation. Potassium channels play an important role in the regulation of P2Y receptor-mediated dilation.


Subject(s)
Adenosine Diphosphate/analogs & derivatives , Adenosine Triphosphate/analogs & derivatives , Arterioles/physiology , Brain/blood supply , Pyridoxal Phosphate/analogs & derivatives , Receptors, Purinergic P2/metabolism , Vasodilation/physiology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Arterioles/drug effects , Cerebrovascular Circulation/physiology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , In Vitro Techniques , Indomethacin/pharmacology , Male , Microscopy, Video , Nitric Oxide/metabolism , Potassium Chloride/pharmacology , Purinergic P2 Receptor Agonists , Purinergic P2 Receptor Antagonists , Pyridoxal Phosphate/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y2 , Thionucleotides/pharmacology , Vascular Patency/drug effects , Vasodilation/drug effects , omega-N-Methylarginine/pharmacology
17.
Stroke ; 33(11): 2692-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12411663

ABSTRACT

BACKGROUND AND PURPOSE: Extracellular concentration of potassium ion ([K+]o) may have a significant influence on the cerebral circulation in health and disease. Mechanisms of [K+]o-induced conducted vasomotor responses in cerebral arterioles, possibly linking microvascular regulation to neuronal activity, have not been examined. METHODS: We analyzed vascular responses to small increases of [K+]o (up to 5 mmol/L) in isolated, cannulated, and pressurized rat cerebral arterioles (36.5+/-1.4 micro m). [K+]o was elevated globally through extraluminal application or locally through micropipette, while arteriolar diameter was measured online. RESULTS: Elevation of [K+]o (5 mmol/L) produced dilation that was inhibited by ouabain but not BaCl2. Locally applied [K+]o (3 to 5 mmol/L) produced a biphasic response (initial constriction followed by dilation), both of which were conducted to the remote site (distance 1142+/-68 microm). Endothelial impairment inhibited conducted but not local biphasic responses. Extraluminal ouabain attenuated local and conducted secondary dilation but not initial constriction. The local biphasic response was unaffected by extraluminal or intraluminal BaCl2. Extraluminal but not intraluminal BaCl2 impaired both conducted constriction and dilation. CONCLUSIONS: In rat penetrating arteriole, (1) [K+]o (3 to 5 mmol/L) strongly regulates arteriolar tone and causes conducted vasomotor responses; (2) local responses to elevated [K+]o are endothelium independent but conducted responses are dependent on an intact endothelium; (3) smooth muscle Na+-K+-ATPase activation is the generator of conducted dilation; and (4) smooth muscle inward rectifier potassium channels sustain conduction. Our findings suggest that potassium-induced conducted vasomotor responses may link local neuronal activity to microvascular regulation, which may be attenuated in pathological conditions.


Subject(s)
Arterioles/physiology , Brain/blood supply , Extracellular Space/metabolism , Potassium/metabolism , Potassium/pharmacology , Animals , Arterioles/drug effects , Arterioles/physiopathology , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Embolism, Air/physiopathology , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Male , Microcirculation/drug effects , Microcirculation/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Ouabain/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Vascular Patency/drug effects , Vasomotor System/drug effects , Vasomotor System/physiology
18.
Stroke ; 33(3): 844-9, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11872913

ABSTRACT

BACKGROUND AND PURPOSE: Potassium channels or nitric oxide or both are major mediators of acidosis-induced dilation in the cerebral circulation. However, these contributions depend on a variety of factors such as species and vessel location. The present study was designed to clarify whether potassium channels and endothelial nitric oxide are involved in acidosis-induced dilation of isolated rat cerebral arterioles. METHODS: Cerebral arterioles were cannulated and monitored with an inverted microscope. Acidosis (pH 6.8 to 7.4) produced by adding hydrogen ions mediated dilation of the cerebral arterioles in a concentration-dependent manner. The role of nitric oxide and potassium channels in response to acidosis was examined with several specific inhibitors and endothelial damage. RESULTS: The dilation was significantly inhibited by potassium chloride (30 mmol/L) and glibenclamide (3 micromol/L; ATP-sensitive potassium channel inhibitor). We found that 30 micromol/L BaCl2 (concentration-dependent potassium channel inhibitor) also affected the dilation; however, an additional treatment of 3 micromol/L glibenclamide did not produce further inhibition. Tetraethylammonium ion (1 mmol/L; calcium-activated potassium channel inhibitor) and 4-aminopyridine (100 micromol/L; voltage-dependent potassium channel inhibitor) as well as ouabain (10 micromol/L; Na-K ATPase inhibitor) and N-methylsulphonyl-6-(2-proparglyloxyphenyl) hexanamide (1 micromol/L; cytochrome P450 epoxygenase inhibitor) did not alter acidotic dilation. N(omega)-Monomethyl-L-arginine (10 micromol/L) and N(omega)-nitro-L-arginine (10 micromol/L) as nitric oxide synthase inhibitor blunted the dilation. Furthermore, the dilation was significantly attenuated after the endothelial impairment. Additional treatment with glibenclamide (3 micromol/L) further reduced the dilation in response to acidosis. CONCLUSIONS: Endothelial nitric oxide and smooth muscle ATP-sensitive potassium channels contribute to acidosis-induced dilation of rat cerebral arterioles. Endothelial damage caused by pathological conditions such as subarachnoid hemorrhage or traumatic brain injury may contribute to reduced blood flow despite injury-induced cerebral acidosis.


Subject(s)
Arterioles/metabolism , Cerebrovascular Circulation/physiology , Muscle, Smooth, Vascular/metabolism , Nitric Oxide/metabolism , Potassium Channels/metabolism , Acidosis/chemically induced , Acidosis/metabolism , Amides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arterioles/drug effects , Cerebrovascular Circulation/drug effects , Embolism, Air/metabolism , Enzyme Inhibitors/pharmacology , Glyburide/pharmacology , In Vitro Techniques , Muscle, Smooth, Vascular/drug effects , Potassium/metabolism , Potassium/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...