Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 37(6): 1959-1970, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27389178

ABSTRACT

Alterations of the renin-angiotensin system have been implicated in the pathogenesis of Alzheimer's disease. We tested the efficacy of losartan (10 mg/kg/day for three months), a selective angiotensin II type 1 receptor antagonist, in alleviating cerebrovascular and cognitive deficits in double-transgenic mice (six months at endpoint) that overexpress a mutated form of the human amyloid precursor protein (APPSwe,Ind) and a constitutively active form of the transforming growth factor-ß1, thereafter named A/T mice. Losartan rescued cerebrovascular reactivity, particularly the dilatory responses, but failed to attenuate astroglial activation and to normalize the neurovascular uncoupling response to sensory stimulation. The cognitive deficits of A/T mice were not restored by losartan nor were the increased brain levels of soluble and insoluble Aß1-40 and Aß1-42 peptides normalized. Our results are the first to demonstrate the capacity of losartan to improve cerebrovascular reactivity in an Alzheimer's disease mouse model of combined Aß-induced vascular oxidative stress and transforming growth factor-ß1-mediated vascular fibrosis. These data suggest that losartan may be promising for restoring cerebrovascular function in patients with vascular diseases at risk for vascular dementia or Alzheimer's disease. However, a combined therapy may be warranted for rescuing both vascular and cognitive deficits in a multifaceted pathology like Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/biosynthesis , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Cerebrovascular Circulation/drug effects , Losartan/therapeutic use , Transforming Growth Factor beta1/biosynthesis , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Blood Flow Velocity/drug effects , Blood Pressure/drug effects , Humans , Losartan/administration & dosage , Maze Learning/drug effects , Memory/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Oxidative Stress/drug effects , Renin-Angiotensin System/drug effects , Transforming Growth Factor beta1/genetics
2.
J Exp Med ; 212(10): 1529-49, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26347470

ABSTRACT

Inactivating mutations of the NF-κB essential modulator (NEMO), a key component of NF-κB signaling, cause the genetic disease incontinentia pigmenti (IP). This leads to severe neurological symptoms, but the mechanisms underlying brain involvement were unclear. Here, we show that selectively deleting Nemo or the upstream kinase Tak1 in brain endothelial cells resulted in death of endothelial cells, a rarefaction of brain microvessels, cerebral hypoperfusion, a disrupted blood-brain barrier (BBB), and epileptic seizures. TAK1 and NEMO protected the BBB by activating the transcription factor NF-κB and stabilizing the tight junction protein occludin. They also prevented brain endothelial cell death in a NF-κB-independent manner by reducing oxidative damage. Our data identify crucial functions of inflammatory TAK1-NEMO signaling in protecting the brain endothelium and maintaining normal brain function, thus explaining the neurological symptoms associated with IP.


Subject(s)
Brain/blood supply , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Cerebrovascular Circulation/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epilepsy/genetics , Female , I-kappa B Kinase/metabolism , Incontinentia Pigmenti/metabolism , Incontinentia Pigmenti/pathology , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinases/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Occludin/metabolism , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelA/metabolism
3.
Neurobiol Dis ; 68: 126-36, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24807206

ABSTRACT

Angiotensin II (AngII) receptor blockers that bind selectively AngII type 1 (AT1) receptors may protect from Alzheimer's disease (AD). We studied the ability of the AT1 receptor antagonist losartan to cure or prevent AD hallmarks in aged (~18months at endpoint, 3months treatment) or adult (~12months at endpoint, 10months treatment) human amyloid precursor protein (APP) transgenic mice. We tested learning and memory with the Morris water maze, and evaluated neurometabolic and neurovascular coupling using [(18)F]fluoro-2-deoxy-D-glucose-PET and laser Doppler flowmetry responses to whisker stimulation. Cerebrovascular reactivity was assessed with on-line videomicroscopy. We measured protein levels of oxidative stress enzymes (superoxide dismutases SOD1, SOD2 and NADPH oxidase subunit p67phox), and quantified soluble and deposited amyloid-ß (Aß) peptide, glial fibrillary acidic protein (GFAP), AngII receptors AT1 and AT2, angiotensin IV receptor AT4, and cortical cholinergic innervation. In aged APP mice, losartan did not improve learning but it consolidated memory acquisition and recall, and rescued neurovascular and neurometabolic coupling and cerebrovascular dilatory capacity. Losartan normalized cerebrovascular p67phox and SOD2 protein levels and up-regulated those of SOD1. Losartan attenuated astrogliosis, normalized AT1 and AT4 receptor levels, but failed to rescue the cholinergic deficit and the Aß pathology. Given preventively, losartan protected cognitive function, cerebrovascular reactivity, and AT4 receptor levels. Like in aged APP mice, these benefits occurred without a decrease in soluble Aß species or plaque load. We conclude that losartan exerts potent preventive and restorative effects on AD hallmarks, possibly by mitigating AT1-initiated oxidative stress and normalizing memory-related AT4 receptors.


Subject(s)
Alzheimer Disease/complications , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Cerebrovascular Circulation/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Losartan/therapeutic use , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Blood Pressure/drug effects , Blood Pressure/genetics , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition Disorders/pathology , Disease Models, Animal , Endothelin-1/pharmacology , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Losartan/pharmacology , Male , Mice , Mice, Transgenic , Mutation/genetics
4.
Neurobiol Aging ; 35(1): 203-12, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23954171

ABSTRACT

Cognitive and cerebrovascular deficits are 2 landmarks of Alzheimer's disease (AD) to target for effective therapy. Here, we evaluated the efficacy of simvastatin in bitransgenic A/T mice overexpressing a mutated form of the human amyloid precursor protein (APP(Swe,Ind)) and a constitutively active form of transforming growth factor-ß1. These mice feature the AD amyloid beta (Aß) and cerebrovascular pathology. Simvastatin significantly decreased insoluble Aß peptide levels and Aß plaque load despite no effect on ß-site amyloid precursor protein-cleaving enzyme and Aß-degrading enzyme neprilysin protein levels. However, simvastatin failed to improve spatial learning and memory deficits and the decreased baseline levels of the memory-related protein early growth response-1 (Egr-1) in the hippocampus CA1 area. The impaired hyperemic response to whisker stimulation in A/T mice was not improved with treatment, but simvastatin fully restored constitutive nitric oxide synthesis in vessel walls and exacerbated agonist-mediated dilatory deficits. These findings point to the efficacy of simvastatin on selective AD features in a complex model of the disease, likely reflecting the challenges faced by recent clinical trials in assessing statin efficacy.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Gene Expression/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mutation , Simvastatin/pharmacology , Simvastatin/therapeutic use , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Disease Models, Animal , Early Growth Response Protein 1/metabolism , Learning/drug effects , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Nitric Oxide/biosynthesis
5.
Br J Pharmacol ; 170(3): 661-70, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23889563

ABSTRACT

BACKGROUND AND PURPOSE: Transient receptor potential vanilloid type 4 (TRPV4) channels are expressed in brain endothelial cells, but their role in regulating cerebrovascular tone under physiological and pathological conditions is still largely unknown. EXPERIMENTAL APPROACH: Wild-type (WT) mice and mice that overexpress a mutated form of the human amyloid precursor protein (APP mice, model of increased amyloid ß), a constitutively active form of TGF-ß1 (TGF mice, model of cerebrovascular fibrosis) or both (APP/TGF mice) were used. Dilations to the selective TRPV4 channel opener GSK1016790A (GSK) or to ACh were measured in posterior cerebral artery segments. KEY RESULTS: Both GSK- and ACh-induced dilations virtually disappeared following endothelium denudation in WT mice. These responses were impaired in vessels from APP, TGF and APP/TGF mice compared with WT. Pre-incubation of WT vessels with the selective TRPV4 channel blocker HC-067047, or with small-conductance (SK channel, apamin) and/or intermediate-conductance (IK channel, charybdotoxin, ChTx) Ca(2+) -sensitive K(+) channel blocker abolished GSK-induced dilations and massively decreased those induced by ACh. These treatments had no or limited effects on ACh-induced dilation in vessels from APP, TGF or APP/TGF mice, and IK and SK channel function was preserved in transgenic mice. Antioxidant superoxide dismutase or catalase normalized GSK- and ACh-mediated dilations only in APP brain arteries. CONCLUSION AND IMPLICATIONS: We conclude that endothelial TRPV4 channels mediate ACh-induced dilation in cerebral arteries, that they are impaired in models of cerebrovascular pathology and that they are sensitive, albeit in the reversible manner, to amyloid ß-induced oxidative stress.


Subject(s)
Alzheimer Disease/metabolism , Cerebral Arteries/metabolism , Cerebrovascular Disorders/metabolism , Endothelium, Vascular/metabolism , TRPV Cation Channels/metabolism , Vasodilation , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Antioxidants/pharmacology , Cerebral Arteries/drug effects , Cerebral Arteries/physiopathology , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Oxidative Stress , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
6.
PLoS One ; 8(7): e68612, 2013.
Article in English | MEDLINE | ID: mdl-23874687

ABSTRACT

Animal models of Alzheimer's disease (AD) are invaluable in dissecting the pathogenic mechanisms and assessing the efficacy of potential new therapies. Here, we used the peroxisome proliferator-activated receptor gamma agonist pioglitazone in an attempt to rescue the pathogenic phenotype in adult (12 months) and aged (>18 months) bitransgenic A/T mice that overexpress a mutated human amyloid precursor protein (APPSwe,Ind) and a constitutively active form of transforming growth factor-ß1 (TGF-ß1). A/T mice recapitulate the AD-related cognitive deficits, amyloid beta (Aß) and cerebrovascular pathologies, as well as the altered metabolic and vascular coupling responses to increased neuronal activity. Pioglitazone normalized neurometabolic and neurovascular coupling responses to sensory stimulation, and reduced cortical astroglial and hippocampal microglial activation in both age groups. Spatial learning and memory deficits in the Morris water maze were not rescued by pioglitazone, but reversal learning was improved in the adult cohort notwithstanding a progressing Aß pathology. While pioglitazone preserved the constitutive nitric oxide synthesis in the vessel wall, it unexpectedly failed to restore cerebrovascular reactivity in A/T mice and even exacerbated the dilatory deficits. These data demonstrate pioglitazone's efficacy on selective AD hallmarks in a complex AD mouse model of comorbid amyloidosis and cerebrovascular pathology. They further suggest a potential benefit of pioglitazone in managing neuroinflammation, cerebral perfusion and glucose metabolism in AD patients devoid of cerebrovascular pathology.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Cerebrovascular Circulation/drug effects , Reversal Learning/drug effects , Thiazolidinediones/pharmacology , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Blotting, Western , Cerebrovascular Circulation/physiology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Laser-Doppler Flowmetry , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Transgenic , PPAR gamma/agonists , Pioglitazone , Reversal Learning/physiology , Thiazolidinediones/therapeutic use
7.
Am J Pathol ; 177(6): 3071-80, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21088218

ABSTRACT

High brain levels of amyloid-ß (Aß) and transforming growth factor-ß1 (TGF-ß1) have been implicated in the cognitive and cerebrovascular alterations of Alzheimer's disease (AD). We sought to investigate the impact of combined increases in Aß and TGF-ß1 on cerebrovascular, neuronal, and mnemonic function using transgenic mice overproducing these peptides (A/T mice). In particular, we measured cerebrovascular reactivity, evoked cerebral blood flow and glucose uptake during brain activation, cholinergic status, and spatial memory, along with cerebrovascular fibrosis, amyloidosis, and astrogliosis, and their evolution with age. An assessment of perfusion and metabolic responses was considered timely, given ongoing efforts for their validation as AD biomarkers. Relative to wild-type littermates, A/T mice displayed an early progressive decline in cerebrovascular dilatory ability, preserved contractility, and reduction in constitutive nitric oxide synthesis that establishes resting vessel tone. Altered levels of vasodilator-synthesizing enzymes and fibrotic proteins, resistance to antioxidant treatment, and unchanged levels of the antioxidant enzyme, superoxide dismutase-2, accompanied these impairments. A/T mice featured deficient neurovascular and neurometabolic coupling to whisker stimulation, cholinergic denervation, cerebral and cerebrovascular Aß deposition, astrocyte activation, and impaired Morris water maze performance, which gained severity with age. The combined Aß- and TGF-ß1-driven pathology recapitulates salient cerebrovascular, neuronal, and cognitive AD landmarks and yields a versatile model toward highly anticipated diagnostic and therapeutic tools for patients featuring Aß and TGF-ß1 increments.


Subject(s)
Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Cerebrovascular Circulation/physiology , Cognition/physiology , Transforming Growth Factor beta1/genetics , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Biomarkers/analysis , Cerebrovascular Circulation/genetics , Disease Models, Animal , Female , Humans , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Swimming , Transforming Growth Factor beta1/metabolism , Validation Studies as Topic
8.
Can J Physiol Pharmacol ; 88(6): 652-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20628431

ABSTRACT

Increased levels of transforming growth factor-beta1 (TGF-beta1) induce a vascular pathology that shares similarities with that seen in Alzheimer's disease, and which possibly contributes to the cognitive decline. In aged transgenic mice that overexpress TGF-beta1 (TGF mice), we previously found reduced dilatory function and selectively impaired endothelin-1 (ET-1)-induced contraction. Here we studied the effects of chronic treatments with selective ETA (ABT-627) or ETB (A-192621) receptor antagonist on cerebrovascular reactivity, cerebral perfusion, or memory performance. The dilatory deficit of TGF mice was not improved by either treatment, but both ET-1 contraction and basal nitric oxide (NO) production were distinctly altered. Although ABT-627 was devoid of any effect in TGF mice, it virtually abolished the ET-1-induced contraction and NO release in wild-type (WT) littermates. In contrast, A-192621 only acted upon TGF mice with full recovery of ET-1 contraction and baseline NO synthesis. TGF mice, treated or not, had no cognitive deficit in the Morris water maze, nor did ABT-627-treated WT controls despite severely impaired vasoreactivity. These findings confirm that ETA receptors primarily mediate the ET-1-induced contraction. Further, they suggest that ETB receptors play a detrimental role in conditions of increased TGF-beta1 and that vascular dysfunction does not inevitably lead to cognitive deficit.


Subject(s)
Alzheimer Disease/physiopathology , Blood Vessels/physiopathology , Brain/blood supply , Endothelin Receptor Antagonists , Transforming Growth Factor beta1/genetics , Aging/physiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Atrasentan , Blood Vessels/drug effects , Blood Vessels/metabolism , Brain/physiopathology , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Cerebral Arteries/physiopathology , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Endothelin A Receptor Antagonists , Endothelin B Receptor Antagonists , Endothelin-1/pharmacology , Female , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide/metabolism , Nitroarginine/pharmacology , Pyrrolidines/pharmacology , Transforming Growth Factor beta1/metabolism , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasodilation/drug effects , Vasodilation/physiology , Vasodilator Agents/pharmacology , Vibrissae/physiology
9.
Atherosclerosis ; 204(2): 395-404, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19111831

ABSTRACT

Urotensin II (UII) and its receptor UT are upregulated in the pathological setting of various cardiovascular diseases including atherosclerosis. However, their exact role in atherosclerosis remains to be determined. In the present study we used four strains of mice; wild-type (WT), UT(+) (a transgenic strain expressing human UT driven by the alpha-smooth muscle-specific, SM22, promoter), ApoE knockout (ko), and UT(+)/ApoE ko. All animals were fed high fat diet for 12 weeks. Western blot analysis revealed a significant increase in aortic UT expression in UT(+) relative to WT mice (P<0.05). Aortas of ApoE ko mice expressed comparable UT protein level to that of UT(+). Immunohistochemistry revealed the presence of strong expression of UT and UII proteins in the atheroma of UT(+), ApoE ko and UT(+)/ApoE ko mice, particularly in foam cells. Serum cholesterol and triglyceride levels were significantly increased in ApoE ko and in UT(+)/ApoE ko but not in UT(+) mice when compared to WT mice (P<0.0001). Analysis of aortas showed a significant increase in atherosclerotic lesion in the UT(+), ApoE ko and UT(+)/ApoE ko compared to WT mice (P<0.05). Oral administration of the UT receptor antagonist SB-657510A (30 microg/Kg/day gavage) for 10 weeks in a group of ApoE ko mice fed on high fat diet resulted in a significant reduction of lesion (P<0.001). SB-657510A also significantly reduced ACAT-1 protein expression in the atherosclerotic lesion of ApoE ko mice (P<0.05). The present findings demonstrate an important role for UT in the pathogenesis of atherosclerosis. The use of UT receptor antagonists may provide a beneficial tool in the management of this debilitating disease process.


Subject(s)
Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Cardiovascular Agents/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , Sulfonamides/pharmacology , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Aorta/drug effects , Aorta/metabolism , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blotting, Western , Cholesterol, Dietary/blood , Disease Models, Animal , Foam Cells/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Promoter Regions, Genetic , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Triglycerides/blood , Urotensins/metabolism
10.
Peptides ; 29(5): 764-9, 2008 May.
Article in English | MEDLINE | ID: mdl-17988761

ABSTRACT

Urotensin-II (U-II), a cyclic undecapeptide, and its receptor, UT, have been linked to vascular and cardiac remodeling. In patients with coronary artery disease (CAD), it has been shown that U-II plasma levels are significantly greater than in normal patients and the severity of the disease is increased proportionally to the U-II plasma levels. We showed that U-II protein and mRNA levels were significantly elevated in the arteries of patients with coronary atherosclerosis in comparison to healthy arteries. We observed U-II expression in endothelial cells, foam cells, and myointimal and medial vSMCs of atherosclerotic human coronary arteries. Recent studies have demonstrated that U-II acts in synergy with mildly oxidized LDL inducing vascular smooth muscle cell (vSMC) proliferation. Additionally, U-II has been shown to induce cardiac fibrosis and cardiomyocyte hypertrophy leading to cardiac remodeling. When using a selective U-II antagonist, SB-611812, we demonstrated a decrease in cardiac dysfunction including a reduction in cardiomyocyte hypertrophy and cardiac fibrosis. These findings suggest that U-II is undoubtedly a potential therapeutic target in treating cardiovascular remodeling.


Subject(s)
Cardiovascular System , Coronary Artery Disease/metabolism , Urotensins/metabolism , Ventricular Remodeling/physiology , Cardiovascular System/anatomy & histology , Cardiovascular System/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Humans , Sulfonamides/metabolism , Urotensins/antagonists & inhibitors , Urotensins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...