Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters

Database
Language
Journal subject
Publication year range
1.
Am J Physiol Cell Physiol ; 313(2): C207-C218, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28566491

ABSTRACT

In utero hypoxia influences the structure and function of most fetal arteries, including those of the developing cerebral circulation. Whereas the signals that initiate this hypoxic remodeling remain uncertain, these appear to be distinct from the mechanisms that maintain the remodeled vascular state. The present study explores the hypothesis that chronic hypoxia elicits sustained changes in fetal cerebrovascular reactivity to endothelin-1 (ET-1), a potent vascular contractant and mitogen. In fetal lambs, chronic hypoxia (3,820-m altitude for the last 110 days of gestation) had no significant effect on plasma ET-1 levels or ETA receptor density in cerebral arteries but enhanced contractile responses to ET-1 in an ETA-dependent manner. In organ culture (24 h), 10 nM ET-1 increased medial thicknesses less in hypoxic than in normoxic arteries, and these increases were ablated by inhibition of PKC (chelerythrine) in both normoxic and hypoxic arteries but were attenuated by inhibition of CaMKII (KN93) and p38 (SB203580) in normoxic but not hypoxic arteries. As indicated by Ki-67 immunostaining, ET-1 increased medial thicknesses via hypertrophy. Measurements of colocalization between MLCK and SMαA revealed that organ culture with ET-1 also promoted contractile dedifferentiation in normoxic, but not hypoxic, arteries through mechanisms attenuated by inhibitors of PKC, CaMKII, and p38. These results support the hypothesis that chronic hypoxia elicits sustained changes in fetal cerebrovascular reactivity to ET-1 through pathways dependent upon PKC, CaMKII, and p38 that cause increased ET-1-mediated contractility, decreased ET-1-mediated smooth muscle hypertrophy, and a depressed ability of ET-1 to promote contractile dedifferentiation.


Subject(s)
Cell Differentiation/genetics , Cerebral Arteries/metabolism , Endothelin-1/genetics , Hypoxia/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cerebral Arteries/embryology , Endothelin-1/administration & dosage , Endothelin-1/blood , Female , Fetus/blood supply , Fetus/metabolism , Hypoxia/blood , Hypoxia/physiopathology , Muscle Contraction/drug effects , Muscle Contraction/genetics , Organ Culture Techniques , Pregnancy , Protein Kinase C/genetics , Sheep , Vascular Remodeling/drug effects , Vascular Remodeling/genetics , Vasoconstriction/drug effects , Vasoconstriction/genetics , p38 Mitogen-Activated Protein Kinases/genetics
2.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1093-R1104, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27707720

ABSTRACT

This study explored the hypothesis that intracerebral hemorrhage (ICH) promotes release of diffusible factors that can significantly influence the structure and function of cerebral arteries remote from the site of injury, through action on platelet-derived growth factor (PDGF) receptors. Four groups of adult male Sprague-Dawley rats were studied (n = 8 each): 1) sham; 2) sham + 60 mg/kg ip imatinib; 3) ICH (collagenase method); and 4) ICH + 60 mg/kg ip imatinib given 60 min after injury. At 24 h after injury, sham artery passive diameters (+3 mM EGTA) averaged 244 ± 7 µm (at 60 mmHg). ICH significantly increased passive diameters up to 6.4% and decreased compliance up to 42.5%. For both pressure- and potassium-induced contractions, ICH decreased calcium mobilization up to 26.2% and increased myofilament calcium sensitivity up to 48.4%. ICH reduced confocal colocalization of smooth muscle α-actin (αActin) with nonmuscle myosin heavy chain (MHC) and increased its colocalization with smooth muscle MHC, suggesting that ICH promoted contractile differentiation. ICH also enhanced colocalization of myosin light chain kinase (MLCK) with both αActin and regulatory 20-kDa myosin light chain. All effects of ICH on passive diameter, compliance, contractility, and contractile protein colocalization were significantly reduced or absent in arteries from animals treated with imatinib. These findings support the hypothesis that ICH promotes release into the cerebrospinal fluid of vasoactive factors that can diffuse to and promote activation of cerebrovascular PDGF receptors, thereby altering the structure, contractile protein organization, contractility, and smooth muscle phenotype of cerebral arteries remote from the site of hemorrhage.


Subject(s)
Cerebral Arteries/physiopathology , Cerebral Hemorrhage/physiopathology , Cerebrovascular Disorders/prevention & control , Cerebrovascular Disorders/physiopathology , Imatinib Mesylate/administration & dosage , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Animals , Cerebral Arteries/drug effects , Cerebral Hemorrhage/drug therapy , Cerebrovascular Circulation/drug effects , Dose-Response Relationship, Drug , Male , Phenotype , Rats , Rats, Sprague-Dawley , Receptors, Platelet-Derived Growth Factor/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL