Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
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
2.
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
3.
J Clin Invest ; 129(8): 3140-3152, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31162142

ABSTRACT

Elevated blood glucose (hyperglycemia) is a hallmark metabolic abnormality in diabetes. Hyperglycemia is associated with protein kinase A (PKA)-mediated stimulation of L-type Ca2+ channels in arterial myocytes resulting in increased vasoconstriction. However, the mechanisms by which glucose activates PKA remain unclear. Here, we showed that elevating extracellular glucose stimulates cAMP production in arterial myocytes, and that this was specifically dependent on adenylyl cyclase 5 (AC5) activity. Super-resolution imaging suggested nanometer proximity between subpopulations of AC5 and the L-type Ca2+ channel pore-forming subunit CaV1.2. In vitro, in silico, ex vivo and in vivo experiments revealed that this close association is critical for stimulation of L-type Ca2+ channels in arterial myocytes and increased myogenic tone upon acute hyperglycemia. This pathway supported the increase in L-type Ca2+ channel activity and myogenic tone in two animal models of diabetes. Our collective findings demonstrate a unique role for AC5 in PKA-dependent modulation of L-type Ca2+ channel activity and vascular reactivity during acute hyperglycemia and diabetes.


Subject(s)
Adenylyl Cyclases/metabolism , Cerebral Arteries/enzymology , Cyclic AMP/metabolism , Diabetes Mellitus, Experimental/enzymology , Hyperglycemia/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Adenylyl Cyclases/genetics , Animals , Calcium Channels, L-Type/biosynthesis , Calcium Channels, L-Type/genetics , Cerebral Arteries/pathology , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Hyperglycemia/genetics , Hyperglycemia/pathology , Mice , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology
4.
J Neuropathol Exp Neurol ; 78(2): 191-194, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30590671

ABSTRACT

Cerebral small vessel disease in deep penetrating arteries is a major cause of lacunar infarcts, white matter lesions and vascular cognitive impairment. Local cerebral blood flow in these small vessels is controlled by endothelial-derived nitric oxide, which exerts a primary vasodilator stimulus on vascular myocytes, via cytoplasmic cyclic GMP. Here, we investigated whether the cGMP-degrading enzyme phosphodiesterase-5 (PDE5) is present in small penetrating arteries in the deep subcortical white matter of older people. Frontal cortical tissue blocks were examined from donated brains of older people (n = 42, 24 male: 18 female, median age 81, range: 59-100 years). PDE5, detected by immunohistochemical labeling, was graded as absent, sparse, or abundant in vascular cells within small arteries in subcortical white matter (vessel outer diameter: 20-100 µm). PDE5 labeling within arterial myocytes was detected in all cases. Degree of PDE5 expression (absent, sparse, or abundant) was not associated with age or with neuropathological diagnosis of small vessel disease. In conclusion, PDE5 is present in vascular myocytes within small penetrating arteries in older people. This is a potential molecular target for pharmacological interventions.


Subject(s)
Brain/enzymology , Cerebral Arteries/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Muscle, Smooth, Vascular/enzymology , White Matter/enzymology , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Muscle Cells/enzymology
5.
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
6.
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
7.
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
8.
J Ethnopharmacol ; 191: 169-179, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27301615

ABSTRACT

ETHNOBOTANICAL RELEVANCE: Catalpol is the main active component of the radix from Rehmannia glutinosa Libosch, which has pleiotropic protective effects in neurodegenerative diseases, ischemic stroke, metabolic disorders and others AIM: Catalpol has been shown to have neuroprotective, neurorepair, and angiogenesis effects following ischemic brain injury. However, its molecular mechanisms are still poorly understood. In previous studies, the JAK2/STAT3 signaling pathway was found to play a role in neuroprotection and angiogenesis. This study investigated the role of catalpol in stimulating angiogenesis via the JAK2/STAT3 pathway after permanent focal cerebral ischemia (pMCAO). METHODS: Rats were subjected to right middle cerebral artery occlusion through electrocoagulation and were treated with catalpol (5mg/kg), AG490 was also used to inhibit STAT3 phosphorylation (pSTAT3). RESULTS: Following stroke, Catalpol improved the neuroethology deficit, increased the cerebral blood flow (CBF) of infarcted brain and upregulated EPO and EPOR. AG490 suppressed the phosphorylation of signal transducer and activator of transcription 3 (STAT3), ultimately inhibited VEGF mRNA expression, which reduced VEGF protein expression and inhibited stroke-induced angiogenesis. However, Catalpol enhanced stroke-induced STAT3 activation and subsequently restored STAT3 activity through the recovery of STAT3 binding to VEGF. Moreover, Catalpol reversed the effect of AG490 on STAT3 activation and nuclear translocation, restored the transcriptional activity of the VEGF promoter by recruiting STAT3 to the VEGF promoter, improved VEGF mRNA and protein expression, increased angiogenesis, reduced the difference in CBF between the infarcted and intact brain and ameliorated the neuroethology behaviors after stroke. CONCLUSION: Catalpol affects neuroprotection and angiogenesis via the JAK2/STAT3 signaling pathway, which is mediated by STAT3 activation and VEGF expression. Catalpol may be used as a potential therapeutic drug for stroke.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Brain/drug effects , Cerebral Arteries/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Iridoid Glucosides/pharmacology , Janus Kinase 2/metabolism , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/pharmacology , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Brain/enzymology , Brain/pathology , Brain/physiopathology , Cerebral Arteries/enzymology , Cerebral Arteries/pathology , Cerebral Arteries/physiopathology , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Erythropoietin/metabolism , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Phosphorylation , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Receptors, Erythropoietin/metabolism , Signal Transduction/drug effects , Time Factors , Transcriptional Activation , Up-Regulation , Vascular Endothelial Growth Factor A/genetics
9.
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
10.
PLoS One ; 10(12): e0145335, 2015.
Article in English | MEDLINE | ID: mdl-26683993

ABSTRACT

Cerebral arterial myogenic and autoregulatory responses are impaired in Fawn Hooded hypertensive (FHH) rats. Cerebral autoregulatory responses are restored in the congenic rat strain in which a segment of chromosome 1 from the Brown Norway (BN) rat was transferred into the FHH genetic background (FHH.1BN). The impact of this region on cerebral arterial dilator responses remains unknown. Aminopeptidase is a gene that was transferred into the FHH genetic background to generate the FHH.1BN rats and is responsible for degradation of the vasodilator bradykinin. Thus, we hypothesized that FHH rats will have increased aminopeptidase P levels with impaired cerebral arterial responses to bradykinin compared to BN and FHH.1BN rats. We demonstrated higher cerebral arterial expression of aminopeptidase P in FHH compared to BN rats. Accordingly, we demonstrated markedly impaired cerebral arterial dilation to bradykinin in FHH compared to BN rats. Interestingly, aminopeptidase P expression was lower in FHH.1BN compared to FHH rats. Decreased aminopeptidase P levels in FHH.1BN rats were associated with increased cerebral arterial bradykinin-induced dilator responses. Aminopeptidase P inhibition by apstatin improved cerebral arterial bradykinin dilator responses in FHH rats to a level similar to FHH.1BN rats. Unlike bradykinin, cerebral arterial responses to acetylcholine were similar between FHH and FHH.1BN groups. These findings indicate decreased bradykinin bioavailability contributes to impaired cerebral arterial dilation in FHH rats. Overall, these data indicate an important role of aminopeptidase P in the impaired cerebral arterial function in FHH rat.


Subject(s)
Aminopeptidases/metabolism , Bradykinin/pharmacology , Cerebral Arteries/physiopathology , Hypertension/enzymology , Vasodilator Agents/pharmacology , Acetylcholine/pharmacology , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/genetics , Animals , Antihypertensive Agents/pharmacology , Cerebral Arteries/drug effects , Cerebral Arteries/enzymology , Gene Expression , Hypertension/drug therapy , Male , Peptides/pharmacology , Rats, Inbred Strains , Vasodilation
11.
J Am Heart Assoc ; 4(7)2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26126547

ABSTRACT

BACKGROUND: P.R4810K of RNF213 (mysterin: rs112735431), which is an AAA(+) ATPase, is the susceptibility polymorphism for moyamoya disease (MMD) in East Asians. However, the role of RNF213 R4810K in the etiology of MMD is unknown. METHODS AND RESULTS: To clarify the role of RNF213 in known angiogenic pathways, RNF213 expression was analyzed in endothelial cells (ECs) treated with several angiogenic and antiangiogenic factors, including interferons (IFNs). RNF213 was upregulated by IFN-ß through signal transducer and activator of transcription x in the promoter and mediated antiangiogenic activity of IFN-ß. RNF213 wild-type (WT) overexpression could not lower angiogenesis without IFN-ß, but RNF213 R4810K overexpression could. To correlate biochemical function as ATPase and the role of RNF213 oligomer formation with antiangiogenic activity, we investigated the effects of mutations in the AAA(+) module. A mutation of the Walker B motif (WEQ), which stabilizes oligomerization, inhibited angiogenesis, but AAA(+) module deletion, which cannot initiate oligomerization, did not. Intriguingly, R4810K, similar to WEQ, decreased ATPase activity, suggesting its antiangiogenic activity through stabilizing oligomers. To confirm the antiangiogenic effect of RNF213 upregulation in vivo, vascular EC- or smooth muscle cell-specific Rnf213 R4757K (R4810K ortholog) or WT transgenic (Tg) mice were exposed to hypoxia. Cerebral angiogenesis by hypoxia was suppressed in EC-specific Rnf213 R4757K Tg mice, whereas it was not suppressed in other mice. CONCLUSIONS: This study suggests the importance of inflammatory signals as environmental factors and R4810K carriers for susceptibility to cerebral hypoxia. A specific inhibitor of ATP binding to the first AAA(+) could be a promising therapeutic candidate for MMD.


Subject(s)
Cerebral Arteries/enzymology , Endothelial Cells/enzymology , Moyamoya Disease/enzymology , Neovascularization, Physiologic , Ubiquitin-Protein Ligases/metabolism , Adenosine Triphosphatases , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Binding Sites , Binding, Competitive , Cell Movement , Cell Proliferation , Cerebral Arteries/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Genetic Predisposition to Disease , HEK293 Cells , HeLa Cells , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hydrolysis , Hypoxia/complications , Inflammation Mediators/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Moyamoya Disease/genetics , Moyamoya Disease/physiopathology , Mutation , Myocytes, Smooth Muscle/enzymology , Neovascularization, Physiologic/drug effects , Phenotype , Time Factors , Transfection , Ubiquitin-Protein Ligases/genetics
12.
Fiziol Zh (1994) ; 61(5): 11-20, 2015.
Article in English | MEDLINE | ID: mdl-26845839

ABSTRACT

Functional as well as structural reorganization of brain tissues takes place in the surrounding and remotes brain areas after focal ischemic lesions. In particular, reactive or regenerative processes have been described to occur in the infarction areas and the contralateral hemisphere. Experiments were performed on 63 rats, divided into 3 groups (each consisted of 21 animals): sham operated, short-term occlusion of the right middle cerebral artery (MCAO) group, and long-term MCAO group. We have studied changes in proteasome proteolysis during transient occlusion of the middle cerebral artery using method of Koizumi J., duration 2 and 60 min and made the comparison between changes in different types of proteasome activity and severity of ischemic injury and showed three types of decrease inproteolytic activity (trypsin-, chymotrypsin-like, peptidylglutamyl peptide-hydrolyzing) in the brain tissues. Chymotrypsin-like activity of ischemic areas of the brain for short-term MCAO decreased 4.1 times compared with controls (P > 0.05), for long-term MCAO decreased 5.8 times compared with controls (P < 0.05). Trypsin-like activity of ischemic areas of brain for short-term MCAO decreased 7.1 times compared with controls (P > 0.05), for long-term MCAO decreased 12.5 times compared with controls (P < 0.05). PGPH activity of ischemic areas for short-term MCAO decreased 8 times compared with controls (P > 0.05), for long-term MCAO decreased 2.8 times compared with controls (P < 0.05). The similar dynamics was observed also in the penumbra and the core zone of the brain at 6 h of reperfusion, in the long run there is no significant difference between the core and contralateral zones. Our results suggest that proteasome activity may play also a role in contralateral cortical plasticity occurring after focal cerebral ischemia.


Subject(s)
Brain Ischemia/enzymology , Chymotrypsin/metabolism , Endopeptidases/metabolism , Reperfusion Injury/enzymology , Stroke/enzymology , Trypsin/metabolism , Animals , Brain/blood supply , Brain/enzymology , Brain/pathology , Brain Chemistry , Brain Ischemia/pathology , Cerebral Arteries/enzymology , Male , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats , Rats, Wistar , Reperfusion Injury/pathology , Stroke/pathology
13.
Curr Vasc Pharmacol ; 13(2): 161-6, 2015.
Article in English | MEDLINE | ID: mdl-24188490

ABSTRACT

Matrix metalloproteinases (MMP) comprise a family of at least 25 zinc-dependent endopeptidases that play a pivotal role in the physiopathology of the mammalian central nervous system. In the first phases after stroke, the dysregulation of MMPs has been described to increase acute neurovascular disruption and cerebral injury. In particular, MMP-mediated alterations lead to blood-brain barrier (BBB) leakage, cerebral edema, hemorrhage, leukocyte infiltration and progressive inflammatory reactions underlying brain tissue loss. In addition, MMPs have been also shown to play critical activities during the repair phases of cerebral ischemia, particularly during angiogenesis and reestablishment of cerebral blood flow. The aim of this narrative review is to elucidate the mechanisms by which MMPs may provide detrimental and/or beneficial effects during the post-stroke injury and repair phases in animal models.


Subject(s)
Brain Ischemia/enzymology , Cerebral Arteries/enzymology , Matrix Metalloproteinases/metabolism , Stroke/enzymology , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/physiopathology , Brain Ischemia/physiopathology , Brain Ischemia/therapy , Capillary Permeability , Cerebral Arteries/physiopathology , Cerebrovascular Circulation , Disease Models, Animal , Humans , Signal Transduction , Stroke/physiopathology , Stroke/therapy , Vascular Remodeling
14.
J Am Heart Assoc ; 3(6): e001474, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25527626

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) has been reported to increase the concentration of nitric oxide (NO) in the brain and can lead to loss of cerebrovascular tone; however, the sources, amounts, and consequences of excess NO on the cerebral vasculature are unknown. Our objective was to elucidate the mechanism of decreased cerebral artery tone after TBI. METHODS AND RESULTS: Cerebral arteries were isolated from rats 24 hours after moderate fluid­percussion TBI. Pressure­induced increases in vasoconstriction (myogenic tone) and smooth muscle Ca2+ were severely blunted in cerebral arteries after TBI. However, myogenic tone and smooth muscle Ca2+ were restored by inhibition of NO synthesis or endothelium removal, suggesting that TBI increased endothelial NO levels. Live native cell NO, indexed by 4,5­diaminofluorescein (DAF­2 DA) fluorescence, was increased in endothelium and smooth muscle of cerebral arteries after TBI. Clamped concentrations of 20 to 30 nmol/L NO were required to simulate the loss of myogenic tone and increased (DAF­2T) fluorescence observed following TBI. In comparison, basal NO in control arteries was estimated as 0.4 nmol/L. Consistent with TBI causing enhanced NO­mediated vasodilation, inhibitors of guanylyl cyclase, protein kinase G, and large­conductance Ca2+­activated potassium (BK) channel restored function of arteries from animals with TBI. Expression of the inducible isoform of NO synthase was upregulated in cerebral arteries isolated from animals with TBI, and the inducible isoform of NO synthase inhibitor 1400W restored myogenic responses following TBI. CONCLUSIONS: The mechanism of profound cerebral artery vasodilation after TBI is a gain of function in vascular NO production by 60­fold over controls, resulting from upregulation of the inducible isoform of NO synthase in the endothelium.


Subject(s)
Brain Injuries/enzymology , Cerebral Arteries/enzymology , Endothelium, Vascular/enzymology , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , Vasodilation , Animals , Arterial Pressure , Brain Injuries/physiopathology , Calcium/metabolism , Calcium Signaling , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Mechanotransduction, Cellular , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Rats, Sprague-Dawley , Time Factors , Up-Regulation , Vasoconstriction , Vasodilation/drug effects
15.
J Chem Neuroanat ; 61-62: 161-8, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25462386

ABSTRACT

Extracellular acidification activates a family of proteins known as acid-sensing ion channels (ASICs). One ASIC subtype, ASIC type 1 (ASIC1), may play an important role in synaptic plasticity, memory, fear conditioning and ischemic brain injury. ASIC1 is found primarily in neurons, but one report showed its expression in isolated mouse cerebrovascular cells. In this study, we sought to determine if ASIC1 is present in intact rat and human major cerebral arteries. A potential physiological significance of such a finding is suggested by studies showing that nitric oxide (NO), which acts as a powerful vasodilator, may modulate proton-gated currents in cultured cells expressing ASIC1s. Because both constitutive NO synthesizing enzymes, neuronal nitric oxide synthase (nNOS) and endothelial NOS (eNOS), are expressed in cerebral arteries we also studied the anatomical relationship between ASIC1 and nNOS or eNOS in both rat and human cerebral arteries. Western blot analysis demonstrated ASIC1 in cerebral arteries from both species. Immunofluorescent histochemistry and confocal microscopy also showed that ASIC1-immunoreactivity (IR), colocalized with the smooth muscle marker alpha-smooth muscle actin (SMA), was present in the anterior cerebral artery (ACA), middle cerebral artery (MCA), posterior cerebral artery (PCA) and basilar artery (BA) of rat and human. Expression of ASIC1 in cerebral arteries is consistent with a role for ASIC1 in modulating cerebrovascular tone both in rat and human. Potential interactions between smooth muscle ASIC1 and nNOS or eNOS were supported by the presence of nNOS-IR in the neighboring adventitial layer and the presence of nNOS-IR and eNOS-IR in the adjacent endothelial layer of the cerebral arteries.


Subject(s)
Acid Sensing Ion Channels/biosynthesis , Cerebral Arteries/enzymology , Nitric Oxide Synthase/biosynthesis , Acid Sensing Ion Channels/analysis , Adolescent , Aged , Aged, 80 and over , Animals , Blotting, Western , Cadaver , Fluorescent Antibody Technique , Humans , Male , Microscopy, Confocal , Middle Aged , Muscle, Smooth, Vascular/metabolism , Nitric Oxide Synthase/analysis , Rats , Rats, Sprague-Dawley
16.
J Mol Med (Berl) ; 92(9): 983-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24858946

ABSTRACT

UNLABELLED: Arteriogenesis involves the rapid proliferation of preexisting arterioles to fully functional arteries as a compensatory mechanism to overcome circulatory deficits. Stimulation of arteriogenesis has therefore been considered a treatment concept in arterial occlusive disease. Here, we investigated the impact of inhibition of protein tyrosine phosphatases (PTPs) on cerebral arteriogenesis in rats. Arteriogenesis was induced by occlusion of one carotid and both vertebral arteries (three-vessel occlusion (3-VO)). Collateral growth and functional vessel perfusion was assessed 3-35 days following 3-VO. Furthermore, animals underwent 3-VO surgery and were treated with the pan-PTP inhibitor BMOV, the SHP-1 inhibitor sodium stibogluconate (SSG), or the PTP1B inhibitor AS279. Cerebral vessel diameters and cerebrovascular reserve capacity (CVRC) were determined, together with immunohistochemistry analyses and proximity ligation assays (PLA) for determination of tissue proliferation and phosphorylation patterns after 7 days. The most significant changes in vessel diameter increase were present in the ipsilateral posterior cerebral artery (PCA), with proliferative markers (PCNA) being time-dependently increased. The CVRC was lost in the early phase after 3-VO and partially recovered after 21 days. PTP inhibition resulted in a significant increase in the ipsilateral PCA diameter in BMOV-treated animals and rats subjected to PTP1B inhibition. Furthermore, CVRC was significantly elevated in AS279-treated rats compared to control animals, along with hyperphosphorylation of the platelet-derived growth factor-ß receptor in the vascular wall in vivo. In summary, our data indicate PTPs as hitherto unrecognized negative regulators in cerebral arteriogenesis. Further, PTP inhibition leading to enhanced collateral growth and blood perfusion suggests PTPs as novel targets in anti-ischemic treatment. KEY MESSAGES: PTPs exhibit negative regulatory function in cerebral collateral growth in rats. Inhibition of pan-PTP/PTP1B increases vessel PDGF-ß receptor phosphorylation. PTP1B inhibition enhances arteriogenesis and cerebrovascular reserve capacity.


Subject(s)
Brain/blood supply , Cerebral Arteries/growth & development , Enzyme Inhibitors/pharmacology , Neovascularization, Physiologic/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Brain/enzymology , Cerebral Arteries/drug effects , Cerebral Arteries/enzymology , Male , Phosphorylation , Protein Tyrosine Phosphatases/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/metabolism
17.
PLoS One ; 9(4): e95916, 2014.
Article in English | MEDLINE | ID: mdl-24759683

ABSTRACT

Exposure to microgravity results in post-flight cardiovascular deconditioning and orthostatic intolerance in astronauts. Vascular oxidative stress injury and mitochondrial dysfunction have been indicated in this process. To elucidate the mechanism for this condition, we investigated whether mitochondria regulated NADPH oxidase in hindlimb unweighting (HU) rat cerebral and mesenteric arteries. Four-week HU was used to simulate microgravity in rats. Vascular superoxide generation, protein and mRNA levels of Nox2/Nox4, and the activity of NADPH oxidase were examined in the present study. Compared with control rats, the levels of superoxide increased in cerebral (P<0.001) but not in mesenteric vascular smooth muscle cells. The protein and mRNA levels of Nox2 and Nox4 were upregulated significantly (P<0.001 and P<0.001 for Nox2, respectively; P<0.001 and P<0.001 for Nox4, respectively) in HU rat cerebral arteries but not in mesenteric arteries. NADPH oxidases were activated significantly by HU (P<0.001) in cerebral arteries but not in mesenteric arteries. Chronic treatment with mitochondria-targeted antioxidant mitoTEMPO attenuated superoxide levels (P<0.001), decreased the protein and mRNA expression levels of Nox2/Nox4 (P<0.01 and P<0.05 for Nox2, respectively; P<0.001 and P<0.001 for Nox4, respectively) and the activity of NADPH oxidase (P<0.001) in HU rat cerebral arteries, but exerted no effects on HU rat mesenteric arteries. Therefore, mitochondria regulated the expression and activity of NADPH oxidases during simulated microgravity. Both mitochondria and NADPH oxidase participated in vascular redox status regulation.


Subject(s)
Antioxidants/administration & dosage , Cerebral Arteries/enzymology , Hindlimb Suspension/methods , Mitochondria/enzymology , NADPH Oxidases/metabolism , Organophosphorus Compounds/administration & dosage , Oxidative Stress/drug effects , Piperidines/administration & dosage , Animals , Gene Expression Regulation , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesenteric Arteries/enzymology , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/genetics , Rats , Rats, Sprague-Dawley , Superoxides/metabolism , Weightlessness Simulation/methods
18.
Neurology ; 82(19): 1707-15, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24739782

ABSTRACT

OBJECTIVE: A significant cause of spontaneous hemorrhages in the elderly is cerebral amyloid angiopathy (CAA), which causes degeneration of cerebral vessels, but the mechanisms are unclear. METHODS: We isolated leptomeningeal vessels from rapidly autopsied brains (the average of postmortem intervals was 3.28 hours) from 9 patients with CAA and 10 age-matched controls, and used molecular, cell biology, and immunohistochemical approaches to examine ß-site APP-cleaving enzyme 1 (BACE1) protein expression and enzymatic activities as well as tight junction molecular components in small- and medium-sized arteries of the cerebral cortex and leptomeninges. RESULTS: We not only identified that the cerebral vessels, including leptomeningeal and cortical vessels, synthesize and express BACE1, but also found a significant elevation of both BACE1 protein levels and enzymatic activities in leptomeningeal vessels from patients with CAA. Moreover, overexpression of BACE1 in endothelial cells resulted in a significant reduction of occludin, a tight junction protein in blood vessels. CONCLUSION: These findings suggest that in addition to neurons, cerebral vascular cells express functional BACE1. Moreover, elevated vascular BACE1 may contribute to deficiency of occludin in cerebral vessels, which ultimately has a critical role in pathogenesis of CAA and its related hemorrhage.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Cerebral Amyloid Angiopathy/enzymology , Occludin/deficiency , Aged , Aged, 80 and over , Case-Control Studies , Cerebral Amyloid Angiopathy/metabolism , Cerebral Arteries/enzymology , Cerebral Arteries/metabolism , Cerebral Cortex/blood supply , Cerebral Cortex/enzymology , Female , Humans , Male , Meninges/blood supply , Meninges/enzymology
19.
Free Radic Biol Med ; 65: 1012-1022, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24017972

ABSTRACT

Disruption of the blood-brain barrier (BBB) and cerebral edema are the major pathogenic mechanisms leading to neurological dysfunction and death after ischemic stroke. The brain protects itself against infarction via activation of endogenous antioxidant defense mechanisms, and we here report the first evidence that sulforaphane-mediated preactivation of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target heme oxygenase-1 (HO-1) in the cerebral vasculature protects the brain against stroke. To induce ischemic stroke, Sprague-Dawley rats were subjected to 70 min middle cerebral artery occlusion (MCAo) followed by 4, 24, or 72 h reperfusion. Nrf2 and HO-1 protein expression was upregulated in cerebral microvessels of peri-infarct regions after 4-72 h, with HO-1 preferentially associated with perivascular astrocytes rather than the cerebrovascular endothelium. In naïve rats, treatment with sulforaphane increased Nrf2 expression in cerebral microvessels after 24h. Upregulation of Nrf2 by sulforaphane treatment prior to transient MCAo (1h) was associated with increased HO-1 expression in perivascular astrocytes in peri-infarct regions and cerebral endothelium in the infarct core. BBB disruption, lesion progression, as analyzed by MRI, and neurological deficits were reduced by sulforaphane pretreatment. As sulforaphane pretreatment led to a moderate increase in peroxynitrite generation, we suggest that hormetic preconditioning underlies sulforaphane-mediated protection against stroke. In conclusion, we propose that pharmacological or dietary interventions aimed to precondition the brain via activation of the Nrf2 defense pathway in the cerebral microvasculature provide a novel therapeutic approach for preventing BBB breakdown and neurological dysfunction in stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Heme Oxygenase-1/metabolism , Infarction, Middle Cerebral Artery/metabolism , Isothiocyanates/pharmacology , Membrane Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Psychomotor Disorders/prevention & control , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Cerebral Arteries/drug effects , Cerebral Arteries/enzymology , Gene Expression , Heme Oxygenase-1/genetics , Infarction, Middle Cerebral Artery/drug therapy , Isothiocyanates/therapeutic use , Male , Membrane Proteins/genetics , Microvessels/enzymology , NF-E2-Related Factor 2/genetics , Neuroprotective Agents/therapeutic use , Oxidative Stress , Psychomotor Disorders/metabolism , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Signal Transduction , Sulfoxides , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...