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










Database
Language
Publication year range
1.
JACC Basic Transl Sci ; 4(8): 940-958, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31909302

ABSTRACT

Heart failure (HF) and subarachnoid hemorrhage (SAH) chronically reduce cerebral perfusion, which negatively affects clinical outcome. This work demonstrates a strong relationship between cerebral artery cystic fibrosis transmembrane conductance regulator (CFTR) expression and altered cerebrovascular reactivity in HF and SAH. In HF and SAH, CFTR corrector compounds (C18 or lumacaftor) normalize pathological alterations in cerebral artery CFTR expression, vascular reactivity, and cerebral perfusion, without affecting systemic hemodynamic parameters. This normalization correlates with reduced neuronal injury. Therefore, CFTR therapeutics have emerged as valuable clinical tools to manage cerebrovascular dysfunction, impaired cerebral perfusion, and neuronal injury.

2.
Stroke ; 46(8): 2260-70, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26138121

ABSTRACT

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is a complex stroke subtype characterized by an initial brain injury, followed by delayed cerebrovascular constriction and ischemia. Current therapeutic strategies nonselectively curtail exacerbated cerebrovascular constriction, which necessarily disrupts the essential and protective process of cerebral blood flow autoregulation. This study identifies a smooth muscle cell autocrine/paracrine signaling network that augments myogenic tone in a murine model of experimental SAH: it links tumor necrosis factor-α (TNFα), the cystic fibrosis transmembrane conductance regulator, and sphingosine-1-phosphate signaling. METHODS: Mouse olfactory cerebral resistance arteries were isolated, cannulated, and pressurized for in vitro vascular reactivity assessments. Cerebral blood flow was measured by speckle flowmetry and magnetic resonance imaging. Standard Western blot, immunohistochemical techniques, and neurobehavioral assessments were also used. RESULTS: We demonstrate that targeting TNFα and sphingosine-1-phosphate signaling in vivo has potential therapeutic application in SAH. Both interventions (1) eliminate the SAH-induced myogenic tone enhancement, but otherwise leave vascular reactivity intact; (2) ameliorate SAH-induced neuronal degeneration and apoptosis; and (3) improve neurobehavioral performance in mice with SAH. Furthermore, TNFα sequestration with etanercept normalizes cerebral perfusion in SAH. CONCLUSIONS: Vascular smooth muscle cell TNFα and sphingosine-1-phosphate signaling significantly enhance cerebral artery tone in SAH; anti-TNFα and anti-sphingosine-1-phosphate treatment may significantly improve clinical outcome.


Subject(s)
Lysophospholipids/biosynthesis , Sphingosine/analogs & derivatives , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/physiopathology , Tumor Necrosis Factor-alpha/biosynthesis , Vasoconstriction/physiology , Animals , Cerebral Arteries/drug effects , Cerebral Arteries/physiology , Gene Targeting/methods , Lysophospholipids/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Organ Culture Techniques , Phenylephrine/administration & dosage , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/biosynthesis , Sphingosine/deficiency , Subarachnoid Hemorrhage/therapy , Tumor Necrosis Factor-alpha/deficiency , Vasoconstriction/drug effects , Vasomotor System/drug effects , Vasomotor System/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...