Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 123: 208-215, 2023 03.
Article in English | MEDLINE | ID: mdl-36586737

ABSTRACT

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and retinal vasculopathy with cerebral leukodystrophy and systemic manifestations (RVCL-S) are the most common forms of rare monogenic early-onset cerebral small vessel disease and share clinical, and, to different extents, neuroradiological and neuropathological features. However, whether CADASIL and RVCL-S overlapping phenotype may be explained by shared genetic risk or causative factors such as TREX1 coding variants remains poorly understood. To investigate this intriguing hypothesis, we used exome sequencing to screen TREX1 protein-coding variability in a large multi-ethnic cohort of 180 early-onset independent familial and apparently sporadic CADASIL-like Caucasian patients from the USA, Portugal, Finland, Serbia and Turkey. We report 2 very rare and likely pathogenic TREX1 mutations: a loss of function mutation (p.Ala129fs) clustering in the catalytic domain, in an apparently sporadic 46-year-old patient from the USA and a missense mutation (p.Tyr305Cys) in the well conserved C-terminal region, in a 57-year-old patient with positive family history from Serbia. In concert with recent findings, our study expands the clinical spectrum of diseases associated with TREX1 mutations.


Subject(s)
CADASIL , Cerebral Small Vessel Diseases , Leukoencephalopathies , Humans , CADASIL/genetics , Cerebral Infarction , Cerebral Small Vessel Diseases/complications , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/genetics , Mutation/genetics , Receptor, Notch3/genetics
2.
Stroke ; 53(5): 1735-1745, 2022 05.
Article in English | MEDLINE | ID: mdl-35105183

ABSTRACT

BACKGROUND: Connectome analysis of neuroimaging data is a rapidly expanding field that offers the potential to diagnose, characterize, and predict neurological disease. Animal models provide insight into biological mechanisms that underpin disease, but connectivity approaches are currently lagging in the rodent. METHODS: We present a pipeline adapted for structural and functional connectivity analysis of the mouse brain, and we tested it in a mouse model of vascular dementia. RESULTS: We observed lacunar infarctions, microbleeds, and progressive white matter change across 6 months. For the first time, we report that default mode network activity is disrupted in the mouse model. We also identified specific functional circuitry that was vulnerable to vascular stress, including perturbations in a sensorimotor, visual resting state network that were accompanied by deficits in visual and spatial memory tasks. CONCLUSIONS: These findings advance our understanding of the mouse connectome and provide insight into how it can be altered by vascular insufficiency.


Subject(s)
Connectome , Dementia, Vascular , Animals , Brain/diagnostic imaging , Connectome/methods , Dementia, Vascular/diagnostic imaging , Disease Models, Animal , Humans , Magnetic Resonance Imaging/methods , Mice , Nerve Net
3.
Neurobiol Aging ; 104: 122.e1-122.e17, 2021 08.
Article in English | MEDLINE | ID: mdl-33875290

ABSTRACT

Huntington's disease and subcortical vascular dementia display similar dementing features, shaped by different degrees of striatal atrophy, deep white matter degeneration and tau pathology. To investigate the hypothesis that Huntington's disease transcriptomic hallmarks may provide a window into potential protective genes upregulated during brain acute and subacute ischemia, we compared RNA sequencing signatures in the most affected brain areas of 2 widely used experimental mouse models: Huntington's disease, (R6/2, striatum and cortex and Q175, hippocampus) and brain ischemia-subcortical vascular dementia (BCCAS, striatum, cortex and hippocampus). We identified a cluster of 55 shared genes significantly differentially regulated in both models and we screened these in 2 different mouse models of Alzheimer's disease, and 96 early-onset familial and apparently sporadic small vessel ischemic disease patients. Our data support the prevalent role of transcriptional regulation upon genetic coding variability of known neuroprotective genes (Egr2, Fos, Ptgs2, Itga5, Cdkn1a, Gsn, Npas4, Btg2, Cebpb) and provide a list of potential additional ones likely implicated in different dementing disorders and worth further investigation.


Subject(s)
Brain Ischemia/genetics , Cyclooxygenase 2/genetics , Early Growth Response Protein 2/genetics , Huntington Disease/genetics , Proto-Oncogene Proteins c-fos/genetics , Transcriptome/genetics , Animals , Brain/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Dementia, Vascular/genetics , Dementia, Vascular/pathology , Disease Models, Animal , Huntington Disease/pathology , Integrins/genetics , Male , Mice, Inbred C57BL , Nerve Degeneration/genetics , Nerve Degeneration/pathology
4.
Sci Rep ; 11(1): 6072, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33727568

ABSTRACT

Recently, several genome-wide association studies identified PHACTR1 as key locus for five diverse vascular disorders: coronary artery disease, migraine, fibromuscular dysplasia, cervical artery dissection and hypertension. Although these represent significant risk factors or comorbidities for ischemic stroke, PHACTR1 role in brain small vessel ischemic disease and ischemic stroke most important survival mechanism, such as the recruitment of brain collateral arteries like posterior communicating arteries (PcomAs), remains unknown. Therefore, we applied exome and genome sequencing in a multi-ethnic cohort of 180 early-onset independent familial and apparently sporadic brain small vessel ischemic disease and CADASIL-like Caucasian patients from US, Portugal, Finland, Serbia and Turkey and in 2 C57BL/6J stroke mouse models (bilateral common carotid artery stenosis [BCCAS] and middle cerebral artery occlusion [MCAO]), characterized by different degrees of PcomAs patency. We report 3 very rare coding variants in the small vessel ischemic disease-CADASIL-like cohort (p.Glu198Gln, p.Arg204Gly, p.Val251Leu) and a stop-gain mutation (p.Gln273*) in one MCAO mouse. These coding variants do not cluster in PHACTR1 known pathogenic domains and are not likely to play a critical role in small vessel ischemic disease or brain collateral circulation. We also exclude the possibility that copy number variants (CNVs) or a variant enrichment in Phactr1 may be associated with PcomA recruitment in BCCAS mice or linked to diverse vascular traits (cerebral blood flow pre-surgery, PcomA size, leptomeningeal microcollateral length and junction density during brain hypoperfusion) in C57BL/6J mice, respectively. Genetic variability in PHACTR1 is not likely to be a common susceptibility factor influencing small vessel ischemic disease in patients and PcomA recruitment in C57BL/6J mice. Nonetheless, rare variants in PHACTR1 RPEL domains may influence the stroke outcome and are worth investigating in a larger cohort of small vessel ischemic disease patients, different ischemic stroke subtypes and with functional studies.


Subject(s)
Brain Ischemia , Microfilament Proteins , Mutation, Missense , Stroke , Aged , Amino Acid Substitution , Animals , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Middle Aged , Stroke/genetics , Stroke/metabolism , Stroke/pathology
5.
Sci Rep ; 11(1): 2126, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483548

ABSTRACT

Placental hypoperfusion and hypoxia are key drivers in complications during fetal development such as fetal growth restriction and preeclampsia. In order to study the mechanisms of disease in mouse models, the development of quantitative biomarkers of placental hypoxia is a prerequisite. The goal of this exploratory study was to establish a technique to noninvasively characterize placental partial pressure of oxygen (PO2) in vivo in the Lgals1 (lectin, galactoside-binding, soluble, 1) deficient mouse model of preeclampsia using fluorine magnetic resonance imaging. We hypothesized a decrease in placental oxygenation in knockout mice. Wildtype and knockout animals received fluorescently labeled perfluoro-5-crown-15-ether nanoemulsion i.v. on day E14-15 during pregnancy. Placental PO2 was assessed via calibrated 19F MRI saturation recovery T1 mapping. A gas challenge with varying levels of oxygen in breathing air (30%, 60% and 100% O2) was used to validate that changes in oxygenation can be detected in freely breathing, anesthetized animals. At the end of the experiment, fluorophore-coupled lectin was injected i.v. to label the vasculature for histology. Differences in PO2 between breathing conditions and genotype were statistically analyzed with linear mixed-effects modeling. As expected, a significant increase in PO2 with increasing oxygen in breathing air was found. PO2 in Lgals1 knockout animals was decreased but this effect was only present at 30% oxygen in breathing air, not at 60% and 100%. Histological examinations showed crossing of the perfluorocarbon nanoemulsion to the fetal blood pool but the dominating contribution of 19F MR signal is estimated at > 70% from maternal plasma based on volume fraction measurements of previous studies. These results show for the first time that 19F MRI can characterize oxygenation in mouse models of placental malfunction.


Subject(s)
Galectin 1/genetics , Magnetic Resonance Imaging/methods , Oxygen/metabolism , Placenta/metabolism , Algorithms , Animals , Crown Ethers/metabolism , Disease Models, Animal , Female , Fluorine/metabolism , Galectin 1/deficiency , Hypoxia , Mice, 129 Strain , Mice, Knockout , Partial Pressure , Phenotype , Pregnancy , Respiration
6.
Behav Brain Res ; 396: 112875, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32858115

ABSTRACT

Monoclonal anti-proprotein convertase subtilisin/kexin type 9 (PSCK9) neutralizing antibodies effectively lower plasma cholesterol levels and decrease cardiovascular events but also raised some concern that cognitive function could worsen as a side effect. Here, we performed experiments in mice to characterize the effect of anti-PCSK9 antibodies on behavior and cognitive function in detail. APOE*3Leiden.CETP mice and B6129SF1/J wildtype mice were fed a Western type diet and treated with the fully human anti-PCSK9 antibody CmAb1 (PL-45134; 10mg*kg-1 s.c.) or vehicle for 6 weeks. Locomotor activity, anxiety levels, recognition memory, and spatial learning were investigated using the open field, novel object recognition test, and Morris water maze, respectively. Serum cholesterol levels in APOE*3Leiden.CETP mice after treatment with anti-PCSK9 antibody were significantly lower compared to controls whereas cholesterol levels in B6129SF1/J wildtype mice remained unchanged at low levels. No apparent differences were found regarding locomotor activity, anxiety, recognition memory, and spatial learning between animals treated with anti-PCSK9 antibody or vehicle in APOE*3Leiden.CETP and B6129SF1/J wildtype mice. In this study, we found no evidence that treatment with anti-PCSK9 antibodies lead to differences in behavior or changes of cognition in mice.


Subject(s)
Behavior, Animal/drug effects , Locomotion/drug effects , PCSK9 Inhibitors , Protease Inhibitors/pharmacology , Recognition, Psychology/drug effects , Spatial Learning/drug effects , Animals , Antibodies , Mice , Proprotein Convertase 9/immunology
8.
Sci Rep ; 10(1): 7103, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32345996

ABSTRACT

Alzheimer's disease and small vessel ischemic disease frequently co-exist in the aging brain. However, pathogenic links between these 2 disorders are yet to be identified. Therefore we used Taqman genotyping, exome and RNA sequencing to investigate Alzheimer's disease known pathogenic variants and pathways: APOE ε4 allele, APP-Aß metabolism and late-onset Alzheimer's disease main genome-wide association loci (APOE, BIN1, CD33, MS4A6A, CD2AP, PICALM, CLU, CR1, EPHA1, ABCA7) in 96 early-onset small vessel ischemic disease Caucasian patients and 368 elderly neuropathologically proven controls (HEX database) and in a mouse model of cerebral hypoperfusion. Only a minority of patients (29%) carried APOE ε4 allele. We did not detect any pathogenic mutation in APP, PSEN1 and PSEN2 and report a burden of truncating mutations in APP-Aß degradation genes. The single-variant association test identified 3 common variants with a likely protective effect on small vessel ischemic disease (0.54>OR > 0.32, adj. p-value <0.05) (EPHA1 p.M900V and p.V160A and CD33 p.A14V). Moreover, 5/17 APP-Aß catabolism genes were significantly upregulated (LogFC > 1, adj. p-val<0.05) together with Apoe, Ms4a cluster and Cd33 during brain hypoperfusion and their overexpression correlated with the ischemic lesion size. Finally, the detection of Aß oligomers in the hypoperfused hippocampus supported the link between brain ischemia and Alzheimer's disease pathology.


Subject(s)
Alleles , Alzheimer Disease , Amyloid beta-Protein Precursor , Apolipoproteins E , Brain Ischemia , Brain , Genetic Loci , Aged , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Apolipoproteins E/genetics , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Ischemia/genetics , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Models, Animal , Female , Genome-Wide Association Study , Humans , Male , Mice , Mice, Transgenic , Middle Aged
9.
J Cereb Blood Flow Metab ; 40(2): 276-287, 2020 02.
Article in English | MEDLINE | ID: mdl-31549895

ABSTRACT

Brain collateral circulation is an essential compensatory mechanism in response to acute brain ischemia. To study the temporal evolution of brain macro and microcollateral recruitment and their reciprocal interactions in response to different ischemic conditions, we applied a combination of complementary techniques (T2-weighted magnetic resonance imaging [MRI], time of flight [TOF] angiography [MRA], cerebral blood flow [CBF] imaging and histology) in two different mouse models. Hypoperfusion was either induced by permanent bilateral common carotid artery stenosis (BCCAS) or 60-min transient unilateral middle cerebral artery occlusion (MCAO). In both models, collateralization is a very dynamic phenomenon with a global effect affecting both hemispheres. Patency of ipsilateral posterior communicating artery (PcomA) represents the main variable survival mechanism and the main determinant of stroke lesion volume and recovery in MCAO, whereas the promptness of external carotid artery retrograde flow recruitment together with PcomA patency, critically influence survival, brain ischemic lesion volume and retinopathy in BCCAS mice. Finally, different ischemic gradients shape microcollateral density and size.


Subject(s)
Brain Ischemia , Cerebral Arteries , Cerebrovascular Circulation , Magnetic Resonance Angiography , Stroke , Animals , Brain Ischemia/diagnostic imaging , Brain Ischemia/physiopathology , Cerebral Arteries/diagnostic imaging , Cerebral Arteries/physiopathology , Disease Models, Animal , Mice , Retinal Diseases/diagnostic imaging , Retinal Diseases/physiopathology , Stroke/diagnostic imaging , Stroke/physiopathology
10.
J Cereb Blood Flow Metab ; 39(2): 313-323, 2019 02.
Article in English | MEDLINE | ID: mdl-28829217

ABSTRACT

Lesion volume measurements with magnetic resonance imaging are widely used to assess outcome in rodent models of stroke. In this study, we improved a mathematical framework to correct lesion size for edema which is based on manual delineation of the lesion and hemispheres. Furthermore, a novel MATLAB toolbox to register mouse brain MR images to the Allen brain atlas is presented. Its capability to calculate edema-corrected lesion size was compared to the manual approach. Automated image registration performed equally well in in a mouse middle cerebral artery occlusion model (Pearson r = 0.976, p = 2.265e-11). Information encapsulated in the registration was used to generate maps of edema induced tissue volume changes. These showed discrepancies to simplified tissue models underlying the manual approach. The presented techniques provide biologically more meaningful, voxel-wise biomarkers of vasogenic edema after stroke.


Subject(s)
Blood-Brain Barrier/diagnostic imaging , Brain Edema , Magnetic Resonance Imaging , Stroke , Animals , Brain Edema/diagnostic imaging , Brain Edema/etiology , Disease Models, Animal , Male , Mice , Stroke/complications , Stroke/diagnostic imaging
11.
MAGMA ; 32(1): 105-114, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30421249

ABSTRACT

INTRODUCTION: We explored the use of a perfluoro-15-crown-5 ether nanoemulsion (PFC) for measuring tissue oxygenation using a mouse model of vascular cognitive impairment. METHODS: Seventeen C57BL/6 mice underwent stereotactic injection of PFC coupled to a fluorophore into the striatum and corpus callosum. Combined 1H/19F magnetic resonance imaging (MRI) to localize the PFC and R1 mapping to assess pO2 were performed. The effect of gas challenges on measured R1 was investigated. All mice then underwent bilateral implantation of microcoils around the common carotid arteries to induce global cerebral hypoperfusion. 19F-MRI and R1 mapping were performed 1 day, 1 week, and 4 weeks after microcoil implantation. In vivo R1 values were converted to pO2 through in vitro calibration. Tissue reaction to the PFC was assessed through ex vivo immunohistochemistry of microglial infiltration. RESULTS: R1 increased with increasing oxygen concentrations both in vitro and in vivo and the strength of the 19F signal remained largely stable over 4 weeks. In the two mice that received all four scans, tissue pO2 decreased after microcoil implantation and recovered 4 weeks later. We observed infiltration of the PFC deposits by microglia. DISCUSSION: Despite remaining technical challenges, intracerebrally injected PFC is suitable for monitoring brain oxygenation in vivo.


Subject(s)
Brain/metabolism , Cognition Disorders/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Fluorine-19 Magnetic Resonance Imaging/instrumentation , Fluorine/chemistry , Oxygen/metabolism , Animals , Calibration , Corpus Callosum/diagnostic imaging , Corpus Striatum/diagnostic imaging , Crown Ethers , Disease Models, Animal , Emulsions , Fluorine-19 Magnetic Resonance Imaging/methods , Fluorocarbons/chemistry , Image Processing, Computer-Assisted , Lung/chemistry , Male , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Radio Waves , Reproducibility of Results
13.
PLoS One ; 13(3): e0193961, 2018.
Article in English | MEDLINE | ID: mdl-29547662

ABSTRACT

Animal models of disease are an indispensable element in our quest to understand pathophysiology and develop novel therapies. Ex vivo studies have severe limitations, in particular their inability to study individual disease progression over time. In this respect, non-invasive in vivo technologies offer multiple advantages. We here used bilateral common carotid artery occlusion (BCCAO) in mice, an established model for ischemic retinopathy, and performed a multimodal in vivo and ex vivo follow-up. We used scanning laser ophthalmoscopy (SLO), ocular coherence tomography (OCT) and electroretinography (ERG) over 6 weeks followed by ex vivo analyses. BCCAO leads to vascular remodeling with thickening of veins starting at 4 weeks, loss of photoreceptor synapses with concomitant reduced b-waves in the ERG and thinning of the retina. Mononuclear phagocytes showed fluctuation of activity over time. There was large inter-individual variation in the severity of neuronal degeneration and cellular inflammatory responses. Ex vivo analysis confirmed these variable features of vascular remodeling, neurodegeneration and inflammation. In summary, we conclude that multimodal follow-up and subgroup analysis of retinal changes in BCCAO further calls into question the use of ex vivo studies with distinct single end-points. We propose that our approach can foster the understanding of retinal disease as well as the clinical translation of emerging therapeutic strategies.


Subject(s)
Arterial Occlusive Diseases/pathology , Carotid Artery Diseases/pathology , Retina/pathology , Retinal Vessels/pathology , Animals , Carotid Artery, Common/pathology , Disease Models, Animal , Disease Progression , Ischemia/pathology , Male , Mice , Mice, Inbred C57BL , Ophthalmoscopy/methods , Retinal Degeneration/pathology , Tomography, Optical Coherence/methods
14.
Behav Brain Res ; 321: 201-208, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28087280

ABSTRACT

Folates are B-vitamins that are vital for normal brain function. Deficiencies in folates either genetic (methylenetetrahydrofolate reductase, MTHFR) or dietary intake of folic acid result in elevated levels of homocysteine. Clinical studies have shown that elevated levels of homocysteine (Hcy) may be associated with the development of dementia, however this link remains unclear. The purpose of this study was to evaluate the impact of increased Hcy levels on a mouse model of vascular cognitive impairment (VCI) produced by chronic hypoperfusion. Male and female Mthfr+/+ and Mthfr+/- mice were placed on either control (CD) or folic acid deficient (FADD) diets after which all animals underwent microcoil implantation around each common carotid artery or a sham procedure. Post-operatively animals were tested on the Morris water maze (MWM), y-maze, and rotarod. Animals had no motor impairments on the rotarod, y-maze, and could learn the location of the platform on the MWM. However, on day 8 of testing of MWM testing during the probe trial, Mthfr+/- FADD microcoil mice spent significantly less time in the target quadrant when compared to Mthfr+/- CD sham mice, suggesting impaired reference memory. All FADD mice had elevated levels of plasma homocysteine. MRI analysis revealed arterial remodeling was present in Mthfr+/- microcoil mice not Mthfr+/+ mice. Acetylcholine and related metabolites were reduced in cortical tissue because of microcoil implantation and elevated levels of homocysteine. Deficiencies in folate metabolism resulting in increased Hcy levels yield a metabolic profile that increases susceptibility to neurodegeneration in a mouse model of VCI.


Subject(s)
Acetylcholine/metabolism , Cerebral Cortex/metabolism , Dementia, Vascular/metabolism , Homocysteine/metabolism , Memory/physiology , Animals , Cerebral Cortex/blood supply , Cerebral Cortex/diagnostic imaging , Dementia, Vascular/diagnostic imaging , Disease Models, Animal , Female , Folic Acid Deficiency/diagnostic imaging , Folic Acid Deficiency/metabolism , Folic Acid Deficiency/psychology , Male , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/psychology , Random Allocation
15.
Stroke ; 48(2): 468-475, 2017 02.
Article in English | MEDLINE | ID: mdl-28070001

ABSTRACT

BACKGROUND AND PURPOSE: Chronic hypoperfusion in the mouse brain has been suggested to mimic aspects of vascular cognitive impairment, such as white matter damage. Although this model has attracted attention, our group has struggled to generate a reliable cognitive and pathological phenotype. This study aimed to identify neuroimaging biomarkers of brain pathology in aged, more severely hypoperfused mice. METHODS: We used magnetic resonance imaging to characterize brain degeneration in mice hypoperfused by refining the surgical procedure to use the smallest reported diameter microcoils (160 µm). RESULTS: Acute cerebral blood flow decreases were observed in the hypoperfused group that recovered over 1 month and coincided with arterial remodeling. Increasing hypoperfusion resulted in a reduction in spatial learning abilities in the water maze that has not been previously reported. We were unable to observe severe white matter damage with histology, but a novel approach to analyze diffusion tensor imaging data, graph theory, revealed substantial reorganization of the hypoperfused brain network. A logistic regression model from the data revealed that 3 network parameters were particularly efficient at predicting group membership (global and local efficiency and degrees), and clustering coefficient was correlated with performance in the water maze. CONCLUSIONS: Overall, these findings suggest that, despite the autoregulatory abilities of the mouse brain to compensate for a sudden decrease in blood flow, there is evidence of change in the brain networks that can be used as neuroimaging biomarkers to predict outcome.


Subject(s)
Brain/diagnostic imaging , Cerebrovascular Circulation/physiology , Cognitive Dysfunction/diagnostic imaging , Disease Models, Animal , Neuroimaging , Animals , Brain/physiology , Cognitive Dysfunction/physiopathology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Neural Pathways/diagnostic imaging , Neural Pathways/physiology , Neuroimaging/methods , Predictive Value of Tests
16.
J Cereb Blood Flow Metab ; 37(5): 1687-1705, 2017 May.
Article in English | MEDLINE | ID: mdl-26994042

ABSTRACT

In rats, spreading depolarization induces vasodilation/hyperemia in naïve tissue but the inverse response when artificial cerebrospinal fluid is topically applied to the brain containing (a) a nitric oxide-lowering agent and (b) elevated K+. The inverse response is characterized by severe vasoconstriction/ischemia. The perfusion deficit runs together with the depolarization in the tissue (=spreading ischemia). Here, we found in male Wistar rats that pre-treatment with artificial cerebrospinal fluid containing elevated K+ in vivo led to a selective decline in α2/α3 Na+/K+-ATPase activity, determined spectrophotometrically ex vivo. Moreover, spreading ischemia, recorded with laser-Doppler flowmetry and electrocorticography, resulted from artificial cerebrospinal fluid containing a nitric oxide-lowering agent in combination with the Na+/K+-ATPase inhibitor ouabain at a concentration selectively inhibiting α2/α3 activity. Decline in α2/α3 activity results in increased Ca2+ uptake by internal stores of astrocytes, vascular myocytes, and pericytes since Ca2+ outflux via plasmalemmal Na+/Ca2+-exchanger declines. Augmented Ca2+ mobilization from internal stores during spreading depolarization might enhance vasoconstriction, thus, contributing to spreading ischemia. Accordingly, spreading ischemia was significantly shortened when intracellular Ca2+ stores were emptied by pre-treatment with thapsigargin, an inhibitor of the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA). These findings might have relevance for clinical conditions, in which spreading ischemia occurs such as delayed cerebral ischemia after subarachnoid hemorrhage.


Subject(s)
Brain Ischemia/physiopathology , Cerebrovascular Circulation/physiology , Cortical Spreading Depression/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Vasoconstriction/physiology , Animals , Brain Ischemia/metabolism , Calcium/metabolism , Cerebrospinal Fluid/chemistry , Cerebrovascular Circulation/drug effects , Electrocorticography , Laser-Doppler Flowmetry , Male , Potassium Chloride/pharmacology , Rats, Wistar , Spectrophotometry , Vasoconstriction/drug effects
17.
J Neurosci ; 36(31): 8132-48, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27488634

ABSTRACT

UNLABELLED: The aim of this study was to explore the signaling and neuroprotective effect of transactivator of transcription (TAT) protein transduction of the apoptosis repressor with CARD (ARC) in in vitro and in vivo models of cerebral ischemia in mice. In mice, transient focal cerebral ischemia reduced endogenous ARC protein in neurons in the ischemic striatum at early reperfusion time points, and in primary neuronal cultures, RNA interference resulted in greater neuronal susceptibility to oxygen glucose deprivation (OGD). TAT.ARC protein delivery led to a dose-dependent better survival after OGD. Infarct sizes 72 h after 60 min middle cerebral artery occlusion (MCAo) were on average 30 ± 8% (mean ± SD; p = 0.005; T2-weighted MRI) smaller in TAT.ARC-treated mice (1 µg intraventricularly during MCAo) compared with controls. TAT.ARC-treated mice showed better performance in the pole test compared with TAT.ß-Gal-treated controls. Importantly, post-stroke treatment (3 h after MCAo) was still effective in affording reduced lesion volume by 20 ± 7% (mean ± SD; p < 0.05) and better functional outcome compared with controls. Delayed treatment in mice subjected to 30 min MCAo led to sustained neuroprotection and functional behavior benefits for at least 28 d. Functionally, TAT.ARC treatment inhibited DAXX-ASK1-JNK signaling in the ischemic brain. ARC interacts with DAXX in a CARD-dependent manner to block DAXX trafficking and ASK1-JNK activation. Our work identifies for the first time ARC-DAXX binding to block ASK1-JNK activation as an ARC-specific endogenous mechanism that interferes with neuronal cell death and ischemic brain injury. Delayed delivery of TAT.ARC may present a promising target for stroke therapy. SIGNIFICANCE STATEMENT: Up to now, the only successful pharmacological target of human ischemic stroke is thrombolysis. Neuroprotective pharmacological strategies are needed to accompany therapies aiming to achieve reperfusion. We describe that apoptosis repressor with CARD (ARC) interacts and inhibits DAXX and proximal signals of cell death. In a murine stroke model mimicking human malignant infarction in the territory of the middle cerebral artery, TAT.ARC salvages brain tissue when given during occlusion or 3 h delayed with sustained functional benefits (28 d). This is a promising novel therapeutic approach because it appears to be effective in a model producing severe injury by interfering with an array of proximal signals and effectors of the ischemic cascade, upstream of JNK, caspases, and BIM and BAX activation.


Subject(s)
Apoptosis , Brain Ischemia/metabolism , Brain Ischemia/pathology , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Gene Products, tat/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Co-Repressor Proteins , Male , Mice , Mice, Inbred C57BL , Molecular Chaperones , Protein Binding , Protein Interaction Maps
18.
Behav Brain Res ; 283: 215-26, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25655513

ABSTRACT

Dietary deficiencies in folic acid result in elevated levels of plasma homocysteine, which has been associated with the development of dementia and other neurodegenerative disorders. Previously, we have shown that elevated levels of plasma homocysteine in mice deficient for a DNA repair enzyme, uracil-DNA glycosylase (UNG), result in neurodegeneration. The goal of this study was to evaluate how deficiencies in folic acid and UNG along with elevated levels of homocysteine affect vascular cognitive impairment, via chronic hypoperfursion in an animal model. Ung(+/+) and Ung(-/-) mice were placed on either control (CD) or folic acid deficient (FADD) diets. Six weeks later, the mice either underwent implantation of microcoils around both common carotid arteries. Post-operatively, behavioral tests began at 3-weeks, angiography was measured after 5-weeks using MRI to assess vasculature and at completion of study plasma and brain tissue was collected for analysis. Learning impairments in the Morris water maze (MWM) were observed only in hypoperfused Ung(-/-) FADD mice and these mice had significantly higher plasma homocysteine concentrations. Interestingly, Ung(+/+) FADD produced significant remodeling of the basilar artery and arterial vasculature. Increased expression of GFAP was observed in the dentate gyrus of Ung(-/-) hypoperfused and FADD sham mice. Chronic hypoperfusion resulted in increased cortical MMP-9 protein levels of FADD hypoperfused mice regardless of genotypes. These results suggest that elevated levels of homocysteine only, as a result of dietary folic acid deficiency, don't lead to memory impairments and neurobiochemical changes. Rather a combination of either chronic hypoperfusion or UNG deficiency is required.


Subject(s)
Cerebrovascular Disorders/physiopathology , Cognition Disorders/physiopathology , Folic Acid Deficiency/physiopathology , Homocysteine/blood , Learning Disabilities/physiopathology , Uracil-DNA Glycosidase/deficiency , Animals , Basilar Artery/pathology , Basilar Artery/physiopathology , Brain/blood supply , Brain/pathology , Brain/physiopathology , Carotid Artery Diseases , Cerebrovascular Disorders/pathology , Chronic Disease , Cognition Disorders/pathology , Diet , Disease Models, Animal , Female , Folic Acid Deficiency/pathology , Glial Fibrillary Acidic Protein , Gliosis/pathology , Gliosis/physiopathology , Learning Disabilities/pathology , Male , Matrix Metalloproteinase 9/metabolism , Maze Learning/physiology , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Random Allocation , Uracil-DNA Glycosidase/genetics
19.
J Cereb Blood Flow Metab ; 35(3): 476-84, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25492118

ABSTRACT

Our aims were to assess the spatiotemporal development of brain pathology in a mouse model of chronic hypoperfusion using magnetic resonance imaging (MRI), and to test whether the renin-angiotensin system (RAS) can offer therapeutic benefit. For the first time, different patterns of cerebral blood flow alterations were observed in hypoperfused mice that ranged from an immediate and dramatic to a delayed decrease in cerebral perfusion. Diffusion tensor imaging revealed increases in several quantitative parameters in different brain regions that are indicative of white-matter degeneration; this began around 3 weeks after induction of hypoperfusion. While this model may be more variable than previously reported, neuroimaging tools represent a promising way to identify surrogate markers of pathology. Vascular remodelling was observed in hypoperfused mice, particularly in the anterior part of the Circle of Willis. While the angiotensin II receptor type 2 agonist, Compound 21 (C21), did not influence this response, it did promote expansion of the basilar artery in microcoil animals. Furthermore, C21-treated animals exhibited increased brain lymphocyte infiltration, and importantly, C21 had opposing effects on spatial reference memory in hypoperfused and sham mice. These results suggest that the RAS may have a role in vascular cognitive impairment.


Subject(s)
Brain/pathology , Cerebrovascular Circulation/physiology , Dementia, Vascular/pathology , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Cerebrovascular Circulation/drug effects , Dementia, Vascular/metabolism , Disease Models, Animal , Flow Cytometry , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL
20.
Nat Commun ; 5: 5422, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25406732

ABSTRACT

Astrocytic network alterations have been reported in Alzheimer's disease (AD), but the underlying pathways have remained undefined. Here we measure astrocytic calcium, cerebral blood flow and amyloid-ß plaques in vivo in a mouse model of AD using multiphoton microscopy. We find that astrocytic hyperactivity, consisting of single-cell transients and calcium waves, is most pronounced in reactive astrogliosis around plaques and is sometimes associated with local blood flow changes. We show that astroglial hyperactivity is reduced after P2 purinoreceptor blockade or nucleotide release through connexin hemichannels, but is augmented by increasing cortical ADP concentration. P2X receptor blockade has no effect, but inhibition of P2Y1 receptors, which are strongly expressed by reactive astrocytes surrounding plaques, completely normalizes astrocytic hyperactivity. Our data suggest that astroglial network dysfunction is mediated by purinergic signalling in reactive astrocytes, and that intervention aimed at P2Y1 receptors or hemichannel-mediated nucleotide release may help ameliorate network dysfunction in AD.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , Plaque, Amyloid/metabolism , Receptors, Purinergic P2Y1/metabolism , Adenosine Triphosphate/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Calcium Signaling , Cerebrovascular Circulation , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...