Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 15573, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666540

ABSTRACT

Cigarette smoking is a risk factor for stroke and is linked to stroke severity. Previous studies have shown that cigarette smoke extract (CSE) triggers endothelial dysfunction in vitro by initiating oxidative stress and/or an inflammatory response. In addition, cerebral endothelial dysfunction (particularly at the level of the blood-brain barrier [BBB]) contributes to stroke pathogenesis. Therefore, we hypothesized that cigarette smoking may influence stroke, at least in part, by exacerbating ischaemia-induced BBB disruption. To test this, we examined the effect of CSE on the permeability of cerebral endothelial cells exposed to oxygen glucose deprivation and reoxygenation (OGD + RO). We found that the loss of BBB integrity following ischaemic/reperfusion-like conditions was significantly worsened by CSE. Despite this being associated with increased mRNA expression of Nox catalytic subunits, reactive oxygen species (ROS) levels were however markedly lower. Furthermore, this occurred in association with elevated expression of antioxidant enzymes (SOD1, SOD2, and Gpx-1), suggesting an antioxidant defence response. Lastly, we found that CSE significantly upregulated mRNA expression of cytokines (IL-6 and TGF-ß). Collectively, these results show that acute exposure to CSE worsens BBB disruption caused by OGD + RO, however, this is not linked to elevated ROS levels but may involve inflammatory mechanisms.


Subject(s)
Brain/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glucose/deficiency , Oxygen/metabolism , Smoke/adverse effects , Tobacco Products/adverse effects , Animals , Cell Line , Cell Survival/drug effects , Cytokines/genetics , Endothelial Cells/cytology , Gene Expression Regulation/drug effects , Hydrogen Peroxide/metabolism , Mice , Permeability/drug effects , RNA, Messenger/genetics , Superoxides/metabolism
2.
Sci Rep ; 9(1): 3622, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842652

ABSTRACT

Stroke is a major cause of death worldwide and ischemic stroke is the most common subtype accounting for approximately 80% of all cases. Pulmonary complications occur in the first few days to weeks following ischemic stroke and are a major contributor to morbidity and mortality. Acute lung injury (ALI) occurs in up to 30% of patients with subarachnoid haemorrhage but the incidence of ALI after ischemic stroke is unclear. As ischemic stroke is the most common subtype of stroke, it is important to understand the development of ALI following the initial ischemic injury to the brain. Therefore, this study investigated whether focal ischemic stroke causes lung inflammation and ALI in mice. Ischemic stroke caused a significant increase in bronchoalveolar lavage fluid (BALF) macrophages and neutrophils and whole lung tissue proinflammatory IL-1ß mRNA expression but this did not translate into histologically evident ALI. Thus, it appears that lung inflammation, but not ALI, occurs after experimental ischemic stroke in mice. This has significant implications for organ donors as the lungs from patient's dying of ischemic stroke are not severely damaged and could thus be used for transplantation in people awaiting this life-saving therapy.


Subject(s)
Brain Ischemia/complications , Pneumonia/pathology , Stroke/complications , Acute Lung Injury , Animals , Disease Models, Animal , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/pathology , Pneumonia/etiology
3.
Endocrinology ; 158(10): 3162-3173, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28938442

ABSTRACT

The metabolic syndrome is associated with an increase in the activation of the renin angiotensin system, whose inhibition reduces the incidence of new-onset diabetes. Importantly, angiotensin II (AngII), independently of its vasoconstrictor action, causes ß-cell inflammation and dysfunction, which may be an early step in the development of type 2 diabetes. The aim of this study was to determine how AngII causes ß-cell dysfunction. Islets of Langerhans were isolated from C57BL/6J mice that had been infused with AngII in the presence or absence of taurine-conjugated ursodeoxycholic acid (TUDCA) and effects on endoplasmic reticulum (ER) stress, inflammation, and ß-cell function determined. The mechanism of action of AngII was further investigated using isolated murine islets and clonal ß cells. We show that AngII triggers ER stress, an increase in the messenger RNA expression of proinflammatory cytokines, and promotes ß-cell dysfunction in murine islets of Langerhans both in vivo and ex vivo. These effects were significantly attenuated by TUDCA, an inhibitor of ER stress. We also show that AngII-induced ER stress is required for the increased expression of proinflammatory cytokines and is caused by reactive oxygen species and IP3 receptor activation. These data reveal that the induction of ER stress is critical for AngII-induced ß-cell dysfunction and indicates how therapies that promote ER homeostasis may be beneficial in the prevention of type 2 diabetes.


Subject(s)
Angiotensin II/pharmacology , Endoplasmic Reticulum Stress/physiology , Inflammation/physiopathology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Animals , Cell Line, Tumor , Cytokines/genetics , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/prevention & control , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/antagonists & inhibitors , Endoribonucleases/genetics , Endoribonucleases/physiology , Gene Expression/drug effects , Gene Knockdown Techniques , Glucose/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/physiology , Insulinoma , Islets of Langerhans/drug effects , Islets of Langerhans/physiopathology , Male , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/physiology , Taurine/pharmacology , Ursodeoxycholic Acid/pharmacology , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/physiology
4.
Diab Vasc Dis Res ; 14(3): 246-253, 2017 05.
Article in English | MEDLINE | ID: mdl-28467198

ABSTRACT

Hydrogen sulphide (H2S) is endogenously produced in vascular tissue and has anti-oxidant and vasoprotective properties. This study investigates whether chronic treatment using the fast H2S donor NaHS could elicit a vasoprotective effect in diabetes. Diabetes was induced in male C57BL6/J mice with streptozotocin (60 mg/kg daily, ip for 2 weeks) and confirmed by elevated blood glucose and glycated haemoglobin levels. Diabetic mice were then treated with NaHS (100 µmol/kg/day) for 4 weeks, and aortae collected for functional and biochemical analyses. In the diabetic group, both endothelium-dependent vasorelaxation and basal nitric oxide (NO•) bioactivity were significantly reduced ( p < 0.05), and maximal vasorelaxation to the NO• donor sodium nitroprusside was impaired ( p < 0.05) in aorta compared to control mice. Vascular superoxide generation via nicotine adenine dinucleotide phosphate (NADPH) oxidase ( p < 0.05) was elevated in aorta from diabetic mice which was associated with increased expression of NOX2 ( p < 0.05). NaHS treatment of diabetic mice restored endothelial function and exogenous NO• efficacy back to control levels. NaHS treatment also reduced the diabetes-induced increase in NADPH oxidase activity, but did not affect NOX2 protein expression. These data show that chronic NaHS treatment reverses diabetes-induced vascular dysfunction by restoring NO• efficacy and reducing superoxide production in the mouse aorta.


Subject(s)
Antioxidants/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Diabetic Angiopathies/prevention & control , Endothelium, Vascular/drug effects , Oxidative Stress/drug effects , Sulfides/administration & dosage , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/complications , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/physiopathology , Dose-Response Relationship, Drug , Drug Administration Schedule , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Glycated Hemoglobin/metabolism , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , NADPH Oxidase 2/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase Type III/metabolism , Superoxides/metabolism , Time Factors , Vasodilation/drug effects , Vasodilator Agents/pharmacology
5.
Clin Sci (Lond) ; 130(17): 1545-58, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27303049

ABSTRACT

The major ghrelin forms, acylated ghrelin and des-acylated ghrelin, are novel gastrointestinal hormones. Moreover, emerging evidence indicates that these peptides may have other functions including neuro- and vaso-protection. Here, we investigated whether post-stroke treatment with acylated ghrelin or des-acylated ghrelin could improve functional and histological endpoints of stroke outcome in mice after transient middle cerebral artery occlusion (tMCAo). We found that des-acylated ghrelin (1 mg/kg) improved neurological and functional performance, reduced infarct and swelling, and decreased apoptosis. In addition, it reduced blood-brain barrier (BBB) disruption in vivo and attenuated the hyper-permeability of mouse cerebral microvascular endothelial cells after oxygen glucose deprivation and reoxygenation (OGD + RO). By contrast, acylated ghrelin (1 mg/kg or 5 mg/kg) had no significant effect on these endpoints of stroke outcome. Next we found that des-acylated ghrelin's vasoprotective actions were associated with increased expression of tight junction proteins (occludin and claudin-5), and decreased cell death. Moreover, it attenuated superoxide production, Nox activity and expression of 3-nitrotyrosine. Collectively, these results demonstrate that post-stroke treatment with des-acylated ghrelin, but not acylated ghrelin, protects against ischaemia/reperfusion-induced brain injury and swelling, and BBB disruption, by reducing oxidative and/or nitrosative damage.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Ghrelin/metabolism , Protective Agents/metabolism , Stroke/complications , Acylation , Animals , Brain Injuries/drug therapy , Brain Injuries/etiology , Endothelial Cells/metabolism , Ghrelin/administration & dosage , Humans , Male , Mice , Mice, Inbred C57BL , Oxygen/metabolism , Protective Agents/administration & dosage
6.
Clin Exp Pharmacol Physiol ; 43(8): 777-86, 2016 08.
Article in English | MEDLINE | ID: mdl-27128638

ABSTRACT

Studies have utilised immortalised mouse cerebral endothelial cells (bEnd.3) exposed to oxygen glucose deprivation (OGD) to study blood-brain barrier (BBB) disruption after ischaemia. However, there is a paucity of literature describing the duration of OGD (and reoxygenation [RO]) required to best simulate BBB disruption in vivo. In this study we assessed BBB disruption in bEnd.3 cells after exposure to a range of OGD periods, and also after OGD + RO. Exposure of bEnd.3 monolayers to 4, 6, 16, or 24 hours of OGD resulted in a significant increase in permeability. The hyperpermeability after 16 or 24 hours was associated with decreased expression of tight junction proteins (occludin and claudin-5). Furthermore, there was a decrease in cell viability and increased expression of the pro-apoptotic protein, cleaved caspase-3. Exposure of bEnd.3 monolayers to 1 hour OGD+ 23 hours RO exacerbated hyperpermeability relative to 1 hour OGD, which was associated with decreased expression levels of occludin and ZO-1, but no change in cell viability or caspase-3. 4 hours OGD + 23 hours RO exacerbated hyperpermeability, decreased expression levels of tight junction proteins, decreased cell viability, and increased caspase-3 expression. Thus, bEnd.3 cells exhibit hyperpermeability, a loss of tight junction proteins, and undergo cell death, after exposure to prolonged periods of OGD. Moreover, they exhibit exacerbated hyperpermeability, a loss of tight junction proteins, and increased expression of caspase-3 after OGD + RO. These findings will facilitate the use of this cell line in studies of BBB disruption and for the testing of therapeutics.


Subject(s)
Capillary Permeability/physiology , Cerebrovascular Circulation/physiology , Endothelial Cells/metabolism , Glucose/deficiency , Microvessels/metabolism , Oxygen/metabolism , Animals , Blood-Brain Barrier/metabolism , Cell Hypoxia/physiology , Cell Line, Transformed , Cell Survival/physiology , Mice
7.
Clin Exp Pharmacol Physiol ; 43(4): 468-75, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26800483

ABSTRACT

The ghrelin gene is expressed in the stomach where it ultimately encodes up to three peptides, namely, acylated ghrelin, des-acylated ghrelin and obestatin, which all have neuroendocrine roles. Recently, the authors' reported that these peptides have important physiological roles in positively regulating vasodilator nitric oxide (NO) production in the cerebral circulation, and may normally suppress superoxide production by the pro-oxidant enzyme, Nox2-NADPH oxidase. To date, the majority of studies using exogenous peptides infer that they may have similar roles in the systemic circulation. Therefore, this study examined whether exogenous and endogenous ghrelin-related peptides modulate NO production and superoxide levels in mouse mesenteric arteries and/or thoracic aorta. Using wire myography, it was found that application of exogenous acylated ghrelin, des-acylated ghrelin or obestatin to mouse thoracic aorta or mesenteric arteries failed to elicit a vasorelaxation response, whereas all three peptides elicited vasorelaxation responses of rat thoracic aorta. Also, none of the peptides modulated mouse aortic superoxide levels as measured by L-012-enhanced chemiluminescence. Next, it was found that NO bioactivity and superoxide levels were unaffected in the thoracic aorta from ghrelin-deficient mice when compared with wild-type mice. Lastly, using novel GHSR-eGFP reporter mice in combination with double-labelled immunofluorescence, no evidence was found for the growth hormone secretagogue receptor (GHSR1a) in the throracic aorta, which is the only functional ghrelin receptor identified to date. Collectively these findings demonstrate that, in contrast to systemic vessels of other species (e.g. rat and human) and mouse cerebral vessels, ghrelin-related peptides do not modulate vasodilator NO production or superoxide levels in mouse systemic arteries.


Subject(s)
Aorta, Thoracic/drug effects , Ghrelin/pharmacology , Mesenteric Arteries/drug effects , Nitric Oxide/biosynthesis , Superoxides/metabolism , Vasodilation/drug effects , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiology , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiology , Mice , Nitric Oxide/metabolism , Rats , Receptors, Ghrelin/metabolism
8.
Endocrinology ; 156(1): 280-90, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25322462

ABSTRACT

The ghrelin-related peptides, acylated ghrelin, des-acylated ghrelin, and obestatin, are novel gastrointestinal hormones. We firstly investigated whether the ghrelin gene, ghrelin O-acyltransferase, and the ghrelin receptor (GH secretagogue receptor 1a [GHSR1a]) are expressed in mouse cerebral arteries. Secondly, we assessed the cerebrovascular actions of ghrelin-related peptides by examining their effects on vasodilator nitric oxide (NO) and superoxide production. Using RT-PCR, we found the ghrelin gene and ghrelin O-acyltransferase to be expressed at negligible levels in cerebral arteries from male wild-type mice. mRNA expression of GHSR1a was also found to be low in cerebral arteries, and GHSR protein was undetectable in GHSR-enhanced green fluorescent protein mice. We next found that exogenous acylated ghrelin had no effect on the tone of perfused cerebral arteries or superoxide production. By contrast, exogenous des-acylated ghrelin or obestatin elicited powerful vasodilator responses (EC50 < 10 pmol/L) that were abolished by the NO synthase inhibitor N(ω)-nitro-L-arginine methyl ester. Furthermore, exogenous des-acylated ghrelin suppressed superoxide production in cerebral arteries. Consistent with our GHSR expression data, vasodilator effects of des-acylated ghrelin or obestatin were sustained in the presence of YIL-781 (GHSR1a antagonist) and in arteries from Ghsr-deficient mice. Using ghrelin-deficient (Ghrl(-/-)) mice, we also found that endogenous production of ghrelin-related peptides regulates NO bioactivity and superoxide levels in the cerebral circulation. Specifically, we show that NO bioactivity was markedly reduced in Ghrl(-/-) vs wild-type mice, and superoxide levels were elevated. These findings reveal protective actions of exogenous and endogenous ghrelin-related peptides in the cerebral circulation and show the existence of a novel ghrelin receptor(s) in the cerebral endothelium.


Subject(s)
Cerebral Arteries/drug effects , Cerebrum/blood supply , Ghrelin/analogs & derivatives , Ghrelin/pharmacology , Receptors, Ghrelin/metabolism , Animals , Gene Expression Regulation/physiology , Male , Mice , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Piperidines/pharmacology , Quinazolinones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/genetics , Superoxides
9.
Free Radic Biol Med ; 60: 264-71, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23459072

ABSTRACT

Nox2 oxidase activity underlies the oxidative stress and vascular dysfunction associated with several vascular-related diseases. We have reported that nitric oxide (NO) decreases reactive oxygen species production by endothelial Nox2. This study tested the hypothesis that nitroxyl (HNO), the redox sibling of NO, also suppresses vascular Nox2 oxidase activity. Specifically, we examined the influence of two well-characterized HNO donors, Angeli's salt and isopropylamine NONOate (IPA/NO), on Nox2-dependent responses to angiotensin II (reactive oxygen species production and vasoconstriction) in mouse cerebral arteries. Angiotensin II (0.1µmol/L)-stimulated superoxide (measured by lucigenin-enhanced chemiluminescence) and hydrogen peroxide (Amplex red fluorescence) levels in cerebral arteries (pooled basilar and middle cerebral (MCA)) from wild-type (WT) mice were ~60% lower (P<0.05) in the presence of either Angeli's salt (1µmol/L) or IPA/NO (1µmol/L). Similarly, phorbyl 12,13-dibutyrate (10µmol/L; Nox2 activator)-stimulated hydrogen peroxide levels were ~40% lower in the presence of IPA/NO (1µmol/L; P<0.05). The ability of IPA/NO to decrease superoxide levels was reversible and abolished by the HNO scavenger l-cysteine (3mmol/L; P<0.05), but was unaffected by hydroxocobalamin (100µmol/L; NO scavenger), ODQ (10µmol/L; soluble guanylyl cyclase (sGC) inhibitor), or Rp-8-pCPT-cGMPS (10µmol/L; cyclic guanosine monophosphate (cGMP)-dependent protein kinase inhibitor). Angiotensin II-stimulated superoxide was substantially less in arteries from Nox2-deficient (Nox2(-/y)) versus WT mice (P<0.05). In contrast to WT, IPA/NO (1µmol/L) had no effect on superoxide levels in arteries from Nox2(-/y) mice. Finally, angiotensin II (1-1000µmol/L)-induced constriction of WT MCA was virtually abolished by IPA/NO (1µmol/L), whereas constrictor responses to either the thromboxane A2 mimetic U46619 (1-100 nmol/L) or high potassium (122.7mmol/L) were unaffected. In conclusion, HNO suppresses vascular Nox2 oxidase activity via a sGC-cGMP-independent pathway. Thus, HNO donors might be useful therapeutic agents to limit and/or prevent Nox2-dependent vascular dysfunction.


Subject(s)
Hydrazines/administration & dosage , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Nitrogen Oxides/metabolism , Vascular Diseases/enzymology , Angiotensin II/metabolism , Animals , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Humans , Hydrazines/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Mice , Molecular Targeted Therapy , NADPH Oxidase 2 , NADPH Oxidases/antagonists & inhibitors , Nitric Oxide/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Vascular Diseases/drug therapy , Vascular Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...