Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 298
1.
Neuromolecular Med ; 21(4): 325-343, 2019 12.
Article En | MEDLINE | ID: mdl-31055715

Intracranial aneurysms (IA) are local dilatations in cerebral arteries that predominantly affect the circle of Willis. Occurring in approximately 2-5% of adults, these weakened areas are susceptible to rupture, leading to subarachnoid hemorrhage (SAH), a type of hemorrhagic stroke. Due to its early age of onset and poor prognosis, SAH accounts for > 25% of years lost for all stroke victims under the age of 65. In this review, we describe the cerebrovascular pathology associated with intracranial aneurysms. To understand IA genetics, we summarize syndromes with elevated incidence, genome-wide association studies (GWAS), whole exome studies on IA-affected families, and recent research that established definitive roles for Thsd1 (Thrombospondin Type 1 Domain Containing Protein 1) and Sox17 (SRY-box 17) in IA using genetically engineered mouse models. Lastly, we discuss the underlying molecular mechanisms of IA, including defects in vascular endothelial and smooth muscle cells caused by dysfunction in mechanotransduction, Thsd1/FAK (Focal Adhesion Kinase) signaling, and the Transforming Growth Factor ß (TGF-ß) pathway. As illustrated by THSD1 research, cell adhesion may play a significant role in IA.


Intracranial Aneurysm , Aneurysm, Ruptured/complications , Animals , Arteritis/complications , Arteritis/pathology , Case-Control Studies , Cerebral Arteries/ultrastructure , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Focal Adhesions , Genetic Predisposition to Disease , Genome-Wide Association Study , Hemorheology , Humans , Incidence , Intracranial Aneurysm/complications , Intracranial Aneurysm/epidemiology , Intracranial Aneurysm/genetics , Intracranial Aneurysm/pathology , Mammals , Mechanotransduction, Cellular , Mice , Myocytes, Smooth Muscle/pathology , SOXF Transcription Factors/physiology , Subarachnoid Hemorrhage/etiology , Syndrome , Thrombospondins/physiology , Transforming Growth Factor beta/physiology , Exome Sequencing , Zebrafish
3.
J Vasc Res ; 54(2): 59-67, 2017.
Article En | MEDLINE | ID: mdl-28365703

BACKGROUND/AIMS: Endothelial glycocalyx refers to the proteoglycan or glycoprotein layer of vessel walls and has critical physiological functions. Cerebral glycocalyx may have additional functions considering the blood-brain barrier and other features. However, the assessment of it has only been performed ex vivo, which includes processes presumably damaging the glycocalyx layer. Here we visualize and characterize the cerebral endothelial glycocalyx in vivo. METHODS: We visualized and quantified the cerebral endothelial glycocalyx in vivo under a 2-photon microscope by tagging glycocalyx and vessel lumen with wheat germ agglutinin lectin and dextran, respectively. The radial intensity was analyzed to measure the thickness of the cerebral endothelial glycocalyx in each vessel type. RESULTS: Cerebral arteries and capillaries have an intact endothelial glycocalyx, but veins and venules do not. The thickness of the glycocalyx layer in pial arteries, penetrating arteries, and capillaries was different; however, it was not correlated with the vessel diameter within each vessel type. CONCLUSION: We characterized the distribution of the cerebral endothelial glycocalyx in vivo. Compared to the results from ex vivo studies, the layer is thicker, indicating that the layer may be damaged in ex vivo systems. We also observed an inhomogeneous cerebral endothelial glycocalyx distribution that might reflect the functional heterogeneity of the vessel type.


Brain/blood supply , Capillaries/chemistry , Cerebral Arteries/chemistry , Cerebral Veins/chemistry , Endothelial Cells/chemistry , Glycocalyx/chemistry , Microscopy, Fluorescence, Multiphoton/methods , Venules/chemistry , Animals , Capillaries/ultrastructure , Cerebral Arteries/ultrastructure , Cerebral Veins/ultrastructure , Endothelial Cells/ultrastructure , Fluorescein-5-isothiocyanate/analogs & derivatives , Glycocalyx/ultrastructure , Male , Mice, Inbred C57BL , Venules/ultrastructure , Wheat Germ Agglutinins , Xanthenes
4.
J Cereb Blood Flow Metab ; 37(7): 2539-2554, 2017 Jul.
Article En | MEDLINE | ID: mdl-27683449

Stroke-induced blood-brain barrier breakdown promotes complications like cerebral edema and hemorrhagic transformation, especially in association with therapeutical recanalization of occluded vessels. As arteries, capillaries and veins display distinct functional and morphological characteristics, we here investigated patterns of blood-brain barrier breakdown for each segment of the vascular tree in rodent models of embolic, permanent, and transient middle cerebral artery occlusion, added by analyses of human stroke tissue. Twenty-four hours after ischemia induction, loss of blood-brain barrier function towards FITC-albumin was equally observed for arteries, capillaries, and veins in rodent brains. Noteworthy, veins showed highest ratios of leaky vessels, whereas capillaries exhibited the most and arteries the least widespread perivascular tracer extravasation. In contrast, human autoptic stroke tissue exhibited pronounced extravasations of albumin around arteries and veins, while the pericapillary immunoreactivity appeared only faint. Although electron microscopy revealed comparable alterations of the arterial and capillary endothelium throughout the applied animal models, structural loss of arterial smooth muscle cells was only observed in the translationally relevant model of embolic middle cerebral artery occlusion. In light of the so far available concepts of stroke treatment, the consideration of a differential vascular pathophysiology along the cerebral vasculature is likely to allow development of novel effective treatment strategies.


Blood-Brain Barrier/ultrastructure , Capillaries/ultrastructure , Cerebral Arteries/ultrastructure , Stroke/pathology , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/physiopathology , Capillaries/diagnostic imaging , Capillaries/physiopathology , Cerebral Arteries/diagnostic imaging , Cerebral Arteries/physiopathology , Disease Models, Animal , Female , Humans , Male , Mice , Microscopy, Electron , Microscopy, Fluorescence , Rats, Wistar , Species Specificity , Stroke/diagnostic imaging , Stroke/physiopathology
5.
Childs Nerv Syst ; 30(5): 815-24, 2014 May.
Article En | MEDLINE | ID: mdl-24643709

The cerebral vasculature and the choroid plexus are innervated by peripheral nerves. The anatomy of the vascular supply to the brain and its related perivascular nerves is reviewed. Intracerebral and intraventricular schwannomas most likely come from neoplastic transformation of Schwann cells investing the perivascular nerves and nerves within the choroid plexus.


Brain Neoplasms/pathology , Brain/pathology , Neurilemmoma/pathology , Schwann Cells/pathology , Cerebral Arteries/pathology , Cerebral Arteries/ultrastructure , Choroid Plexus/pathology , Humans
6.
FASEB J ; 28(6): 2715-24, 2014 Jun.
Article En | MEDLINE | ID: mdl-24604081

Exposure to microgravity results in cardiovascular deconditioning, and cerebrovascular oxidative stress injury has been suggested to occur. To elucidate the mechanism for this condition, we investigated whether simulated microgravity induces mitochondrial dysfunction in rat arteries. Four-week hindlimb unweighting (HU) was used to simulate microgravity in rats. Mitochondrial reactive oxygen species (ROS), mitochondrial membrane potential (Δψm), mitochondrial permeability transition pore (mPTP) opening, mitochondrial respiratory control ratio (RCR), MnSOD/GPx activity and expression, and mitochondrial malondialdehyde (MDA) were examined in rat cerebral and mesenteric VSMCs. Compared with the control rats, mitochondrial ROS levels, mPTP opening, and MDA content increased significantly (P<0.001, P<0.01, and P<0.01, respectively), Δψm, RCR, MnSOD/GPx activity (P<0.001 for Δψm and RCR; P<0.05 for MnSOD; and P<0.001 for GPx activity) and protein abundance of mitochondrial MnSOD/GPx-1 decreased (P<0.001 for MnSOD and GPx-1) in HU rat cerebral but not mesenteric arteries. Chronic treatment with NADPH oxidase inhibitor apocynin and mitochondria-targeted antioxidant mitoTempol promoted recovery of mitochondrial function in HU rat cerebral arteries, but exerted no effects on HU rat mesenteric arteries. Therefore, simulated microgravity resulted in cerebrovascular mitochondrial dysfunction, and crosstalk between NADPH oxidase and mitochondria participated in the process.


Cerebral Arteries/physiopathology , Mitochondria/metabolism , Weightlessness Simulation/adverse effects , Acetophenones/pharmacology , Animals , Cerebral Arteries/ultrastructure , Glutathione Peroxidase/metabolism , Hindlimb Suspension , Male , Membrane Potential, Mitochondrial/physiology , Mesenteric Arteries/ultrastructure , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , NADPH Oxidases/metabolism , Organophosphorus Compounds/pharmacology , Piperidines/pharmacology , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
7.
Ultrastruct Pathol ; 37(6): 425-32, 2013 Dec.
Article En | MEDLINE | ID: mdl-24205927

This study is based on data analysis by light and transmission electron microscopy of the surgical cases in cerebral tumors, cerebrovascular malformations, thromboses in the carotid system, and other injuries such as perivascular hemorrhage. We examined cortical arteries and veins, perivascular areas with old hematic masses, vasculogenic foci, and broken large vessels. We identified, characterized, and compared both undifferentiated cells and well-differentiated cordocytes within periadventitial areas where these cells cooperate very well with precursor/stem cells to perform vital functions for cerebral vasculature with immediate effect on brain parenchyma. This useful cellular cooperation was observed by serial sections pointing out the main role of cordocytes during the entire process of collateral vessel formation after thrombosis and, respectively, in vascular wall repair after ruptures. This is the first cytohistopathological study which illustrates and explains some facets of cordocytes-stem cells cooperation around the vessels of human brain with emphasis on the fundamental role of cordocytes in response to vascular injuries. Our pioneering study will be completed for both basic science and modern medical care by further studies.


Brain Neoplasms/ultrastructure , Cell Communication , Cerebral Arteries/ultrastructure , Cerebral Veins/ultrastructure , Intracranial Arteriovenous Malformations/pathology , Intracranial Thrombosis/pathology , Stem Cells/ultrastructure , Cell Differentiation , Cerebral Arteries/abnormalities , Cerebral Veins/abnormalities , Humans , Microscopy, Electron, Transmission , Neovascularization, Physiologic , Regeneration
10.
Arkh Patol ; 74(2): 51-6, 2012.
Article Ru | MEDLINE | ID: mdl-22880419

Skin and muscles biopsy specimens of a patient harboring A3243G mutation in mitochondrial DNA, with dissection of internal carotid and vertebral arteries, associated with MELAS were studied using histochemical and electron-microscopy techniques. Ragged red fibers, regional variability of SDH histochemical reaction, two types of morphologically atypical mitochondria and their aggregation were found in muscle. There was correlation between SDH histochemical staining and number of mitochondria revealed by electron microscopy in muscle tissue. Similar mitochondrial abnormality, their distribution and cell lesions followed by extra-cellular matrix mineralization were found in the blood vessel walls. In line with generalization of cytopathy process caused by gene mutation it can be supposed that changes found in skin and muscle microvessels also exist in large cerebral vessels causing the vessel wall "weakness", predisposing them to dissection.


Carotid Artery Diseases/pathology , Cerebral Arteries/ultrastructure , MELAS Syndrome/pathology , Microvessels/ultrastructure , Mitochondria, Muscle/ultrastructure , Muscle, Skeletal/ultrastructure , Carotid Artery Diseases/genetics , Humans , MELAS Syndrome/genetics , Middle Aged , Mitochondria, Muscle/genetics , Rupture, Spontaneous , Skin/pathology
11.
Ultrastruct Pathol ; 36(3): 166-70, 2012 May.
Article En | MEDLINE | ID: mdl-22559043

The authors analyzed by transmission electron microscopy the modifications of plasmalemma and nuclear envelope in the cerebral arterial wall in humans. Their ultrastructural observations are performed on the tunica media and endothelium. During autoschizis, some smooth muscle cells showed deep invaginations of the nuclear envelope with multiple craters that disintegrate the nucleus, whereas in the endothelium repetitive invaginations of plasmalemma lead to cell demise by cytoplasmic self-excisions. During survival mechanism, a perinuclear constriction of plasmalemma occurs, which conserves nucleus and cytoskeleton, and only a segregated cytoplasmic area, without organelles, is removed in lumen.


Cerebral Arteries/ultrastructure , Intracellular Membranes/ultrastructure , Moyamoya Disease/pathology , Nuclear Envelope/ultrastructure , Tunica Media/ultrastructure , Carotid Artery Thrombosis/pathology , Cavernous Sinus Thrombosis/pathology , Cell Death , Cytoplasm/ultrastructure , Cytoskeleton/ultrastructure , Endothelium, Vascular/ultrastructure , Humans , Microscopy, Electron, Transmission , Muscle, Smooth, Vascular/ultrastructure
12.
Acta Neuropathol ; 122(3): 293-311, 2011 Sep.
Article En | MEDLINE | ID: mdl-21688176

Cerebrovascular lesions related to congophilic amyloid angiopathy (CAA) often accompany deposition of ß-amyloid (Aß) in Alzheimer's disease (AD), leading to disturbed cerebral blood flow and cognitive dysfunction, posing the question how cerebrovascular pathology contributes to the pathology of AD. To address this question, we characterised the morphology, biochemistry and functionality of brain blood vessels in transgenic arctic ß-amyloid (arcAß) mice expressing human amyloid precursor protein (APP) with both the familial AD-causing Swedish and Arctic mutations; these mice are characterised by strong CAA pathology. Mice were analysed at early, mid and late-stage pathology. Expression of the glucose transporter GLUT1 at the blood-brain barrier (BBB) was significantly decreased and paralleled by impaired in vivo blood-to-brain glucose transport and reduced cerebral lactate release during neuronal activation from mid-stage pathology onwards. Reductions in astrocytic GLUT1 and lactate transporters, as well as retraction of astrocyte endfeet and swelling consistent with neurovascular uncoupling, preceded wide-spread ß-amyloid plaque pathology. We show that CAA at later disease stages is accompanied by severe morphological alterations of brain blood vessels including stenoses, BBB leakages and the loss of vascular smooth muscle cells (SMCs). Together, our data establish that cerebrovascular and astrocytic pathology are paralleled by impaired cerebral metabolism in arcAß mice, and that astrocyte alterations occur already at premature stages of pathology, suggesting that astrocyte dysfunction can contribute to early behavioural and cognitive impairments seen in these mice.


Astrocytes/pathology , Cerebral Amyloid Angiopathy/complications , Cerebrovascular Disorders/etiology , Cerebrovascular Disorders/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/ultrastructure , Basement Membrane/metabolism , Basement Membrane/pathology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Brain/pathology , Cell Culture Techniques , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/pathology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Cerebral Arteries/ultrastructure , Disease Models, Animal , Disease Progression , Dystroglycans/metabolism , Endothelium/metabolism , Endothelium/pathology , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Hemorrhage/etiology , Hemorrhage/metabolism , Hemorrhage/pathology , Humans , Lactase/metabolism , Laminin/metabolism , Mice , Mice, Transgenic , Microdialysis/methods , Microscopy, Electron, Scanning/methods , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Muscle, Smooth/ultrastructure , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Symporters/genetics , Symporters/metabolism
13.
Acta Neurochir Suppl ; 109: 163-7, 2011.
Article En | MEDLINE | ID: mdl-20960337

Intraoperative computed tomography (iCT) has gained increasing impact among modern neurosurgical techniques. Multislice CT with a sliding gantry in the OR provides excellent diagnostic image quality in the visualization of vascular lesions as well as bony structures including skull base and spine. Due to short acquisition times and a high spatial and temporal resolution, various modalities such as iCT-angiography, iCT-cerebral perfusion and the integration of intraoperative navigation with automatic re-registration after scanning can be performed. This allows a variety of applications, e.g. intraoperative angiography, intraoperative cerebral perfusion studies, update of cerebral and spinal navigation, stereotactic procedures as well as resection control in tumour surgery. Its versatility promotes its use in a multidisciplinary setting. Radiation exposure is comparable to standard CT systems outside the OR. For neurosurgical purposes, however, new hardware components (e.g. a radiolucent headholder system) had to be developed. Having a different range of applications compared to intraoperative MRI, it is an attractive modality for intraoperative imaging being comparatively easy to install and cost efficient.


Neuronavigation/methods , Neurosurgical Procedures/methods , Spine/surgery , Tomography, X-Ray Computed/methods , Vascular Diseases/surgery , Cerebral Arteries/pathology , Cerebral Arteries/ultrastructure , Humans , Spine/diagnostic imaging , Vascular Diseases/diagnostic imaging
14.
J Cereb Blood Flow Metab ; 30(6): 1226-39, 2010 Jun.
Article En | MEDLINE | ID: mdl-20125181

Although dihydropyridines are widely used for the treatment of vasospasm, their effectiveness is questionable, suggesting that other voltage-dependent calcium channels (VDCCs) contribute to control of cerebrovascular tone. This study therefore investigated the role of dihydropyridine-insensitive VDCCs in cerebrovascular function. Using quantitative PCR and immunohistochemistry, we found mRNA and protein for L-type (Ca(V)1.2) and T-type (Ca(V)3.1 and Ca(V)3.2) channels in adult rat basilar and middle cerebral arteries and their branches. Immunoelectron microscopy revealed both L- and T-type channels in smooth muscle cell (SMC) membranes. Using patch clamp electrophysiology, we found that a high-voltage-activated calcium current, showing T-type channel kinetics and insensitivity to nifedipine and nimodipine, comprised approximately 20% of current in SMCs of the main arteries and approximately 45% of current in SMCs from branches. Both components were abolished by the T-type antagonists mibefradil, NNC 55-0396, and efonidipine. Although nifedipine completely blocked vasoconstriction in pressurized basilar arteries, a nifedipine-insensitive constriction was found in branches and this increased in magnitude as vessel size decreased. We conclude that a heterogeneous population of VDCCs contributes to cerebrovascular function, with dihydropyridine-insensitive channels having a larger role in smaller vessels. Sensitivity of these currents to nonselective T-type channel antagonists suggests that these drugs may provide a more effective treatment for therapy-refractory cerebrovascular constriction.


Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Calcium/metabolism , Cerebral Arteries/metabolism , Cerebrovascular Circulation/drug effects , Dihydropyridines/pharmacology , Animals , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cerebral Arteries/ultrastructure , Cerebrovascular Circulation/physiology , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Rats , Rats, Wistar , Vasoconstriction/drug effects , Vasoconstriction/physiology
15.
Neurosci Lett ; 470(1): 76-80, 2010 Feb 05.
Article En | MEDLINE | ID: mdl-20043978

Fasudil (a Rho-kinase inhibitor) has been shown to attenuate abdominal aortic aneurysm development, but any preventive effect against development of cerebral aneurysms is unclear. The effect of fasudil on the development of cerebral aneurysms was investigated in 55 female Sprague-Dawley rats divided into 4 groups: Group 1 (n=10) was the control group without treatment. Groups 2-4 (n=15 each) were subjected to cerebral aneurysm induction procedures plus 1% NaCl in the drinking water. Groups 3 and 4 were also treated with 0.5 or 1.0mg/mL of fasudil in the drinking water, respectively. Vascular corrosion casts of the cerebral arteries were prepared and examined using a scanning electron microscope after 2 months. No significant differences were observed in the degree of induced hypertension between Groups 2, 3 and 4. No aneurysms were found in Group 1. Examination of the left anterior cerebral-olfactory artery junction, which is the most susceptible site for aneurysm development, found significantly fewer aneurysmal lesions in Groups 3 (60%) and 4 (53%) compared to Group 2 (100%) (P<0.02). This study suggests that fasudil attenuated induction of cerebral aneurysms in the rat model.


1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Cerebral Arteries/pathology , Intracranial Aneurysm/drug therapy , Intracranial Aneurysm/pathology , Neuroprotective Agents/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Brain/blood supply , Brain/drug effects , Brain/pathology , Cerebral Arteries/drug effects , Cerebral Arteries/ultrastructure , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Female , Microscopy, Electron, Scanning , Neuroprotective Agents/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , rho-Associated Kinases/antagonists & inhibitors
16.
J Neurosci Res ; 88(7): 1457-74, 2010 May 15.
Article En | MEDLINE | ID: mdl-20025060

The blood-brain barrier (BBB) refers to the network of microvessels that selectively restricts the passage of substances between the circulation and the central nervous system (CNS). This microvascular network is comprised of arterioles, capillaries and venules, yet the respective contribution of each of these to the BBB awaits clarification. In this regard, it has been postulated that brain microvascular endothelial cells (BMEC) from these different tributaries might exhibit considerable heterogeneity in form and function, with such diversity underlying unique roles in physiological and pathophysiological processes. Means to begin exploring such endothelial differences in situ, free from caveats associated with cell isolation and culturing procedures, are crucial to comprehending the nature and treatment of CNS diseases with vascular involvement. Here, the recently validated approach of immuno-laser capture microdissection (immuno-LCM) coupled to quantitative real-time PCR (qRT-PCR) was used to analyze gene expression patterns of BMEC retrieved in situ from either capillaries or venules. From profiling 87 genes known to play a role in BBB function and/or be enriched in isolated brain microvessels, results imply that most BBB properties reside in both segments, but that capillaries preferentially express some genes related to solute transport, while venules tend toward higher expression of an assortment of genes involved in inflammatory-related tasks. Fuller appreciation of such heterogeneity will be critical for efficient therapeutic targeting of the endothelium and the management of CNS disease.


Blood-Brain Barrier/metabolism , Brain/blood supply , Cerebral Arteries/physiology , Endothelial Cells/metabolism , Gene Expression Regulation/physiology , Microcirculation/physiology , Animals , Blood-Brain Barrier/ultrastructure , Capillaries/metabolism , Capillaries/ultrastructure , Cerebral Arteries/ultrastructure , Cerebrovascular Circulation/genetics , Endothelial Cells/ultrastructure , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Microdissection , Proteins/genetics , Proteins/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Venules/metabolism , Venules/ultrastructure
17.
Ultrastruct Pathol ; 33(3): 102-11, 2009.
Article En | MEDLINE | ID: mdl-19479650

In a vascular anomaly showing moderate edema, the extracellular space appeared apparently normal, exhibiting a membrane to membrane space of about 20 nm in width. In congenital hydrocephalus, this space appeared notably enlarged and occupied by an electron transparent, nonproteinaceous interstitial edema fluid, due to abnormal accumulation of cerebrospinal fluid. In brain trauma, the distended extracellular space contained either electron-lucid nonproteinaceous or electron-dense proteinaceous edema fluid. Hemorrhagic foci, fibrinoid material, and non-nervous invading cells, such as macrophages and monocytes, were also found. In brain tumors, the widened extracellular space showed electron-dense proteinaceous edema fluid and bundles of fibrinoid material. The enlarged extracellular space found in congenital hydrocephalus, vascular anomalies, brain trauma, and tumors is closely related to the clinical symptoms exhibited by the patients under study.


Brain Edema/pathology , Cerebral Cortex/ultrastructure , Extracellular Space , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Brain/blood supply , Brain/ultrastructure , Brain Injuries/pathology , Brain Neoplasms/ultrastructure , Cell Membrane/ultrastructure , Cerebral Arteries/abnormalities , Cerebral Arteries/ultrastructure , Child , Child, Preschool , Female , Humans , Hydrocephalus/complications , Hydrocephalus/ultrastructure , Infant , Infant, Newborn , Male , Middle Aged , Young Adult
18.
J Neurochem ; 110(1): 58-71, 2009 Jul.
Article En | MEDLINE | ID: mdl-19457074

Hypoxic (low oxygen) and reperfusion (post-hypoxic reoxygenation) phases of stroke promote an increase in microvascular permeability at tight junctions (TJs) of the blood-brain barrier (BBB) that may lead to cerebral edema. To investigate the effect of hypoxia (Hx) and reoxygenation on oligomeric assemblies of the transmembrane TJ protein occludin, rats were subjected to either normoxia (Nx, 21% O(2), 60 min), Hx (6% O(2), 60 min), or hypoxia/reoxygenation (H/R, 6% O(2), 60 min followed by 21% O(2), 10 min). After treatment, cerebral microvessels were isolated, fractionated by detergent-free density gradient centrifugation, and occludin oligomeric assemblies associated with plasma membrane lipid rafts were solubilized by perfluoro-octanoic acid (PFO) exclusively as high molecular weight protein complexes. Analysis by non-reducing and reducing sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis/western blot of PFO-solubilized occludin revealed that occludin oligomeric assemblies co-localizing with 'TJ-associated' raft domains contained a high molecular weight 'structural core' that was resistant to disassembly by either SDS or a hydrophilic reducing agent ex vivo, and by Hx and H/R conditions in vivo. However, exposure of PFO-solubilized occludin oligomeric assemblies to SDS ex vivo revealed the non-covalent association of a significant amount of dimeric and monomeric occludin isoforms to the disulfide-bonded inner core, and dispersal of these non-covalently attached occludin subunits to lipid rafts of higher density in vivo was differentially promoted by Hx and H/R. Our data suggest a model of isoform interaction within occludin oligomeric assemblies at the BBB that enables occludin to simultaneously perform a structural role in inhibiting paracellular diffusion, and a signaling role involving interactions of dimeric and monomeric occludin isoforms with a variety of regulatory molecules within different plasma membrane lipid raft domains.


Blood-Brain Barrier/metabolism , Brain Edema/metabolism , Hypoxia, Brain/metabolism , Membrane Proteins/metabolism , Reperfusion Injury/metabolism , Tight Junctions/metabolism , Animals , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Blotting, Western , Brain Edema/pathology , Brain Edema/physiopathology , Cerebral Arteries/chemistry , Cerebral Arteries/metabolism , Cerebral Arteries/ultrastructure , Diffusion , Electrophoresis, Polyacrylamide Gel , Female , Hypoxia, Brain/pathology , Hypoxia, Brain/physiopathology , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Membrane Microdomains/ultrastructure , Membrane Proteins/analysis , Membrane Proteins/chemistry , Models, Molecular , Occludin , Protein Multimerization/physiology , Protein Subunits/chemistry , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Stress, Physiological/physiology , Subcellular Fractions/metabolism , Tight Junctions/chemistry , Tight Junctions/pathology
19.
Brain ; 132(Pt 6): 1601-12, 2009 Jun.
Article En | MEDLINE | ID: mdl-19293235

Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an autosomal dominant small-vessel disease of the brain caused by mutations in the NOTCH3 receptor. The highly stereotyped nature of the mutations, which alter the number of cysteine residues within the epidermal growth factor-like repeats (EGFR), predicts that all mutations share common mechanisms. Prior in vitro assays and genetic studies in the mouse support the hypothesis that common mutations do not compromise canonical Notch3 function but instead convey a non-physiological and deleterious activity to the receptor through the unpaired cysteine residue. Intriguingly, in vitro studies predict that mutations located in the Delta/Serrate/LAG-2 ligand binding domain-(EGFR10-11) may result in a loss of Notch3 receptor function. However, the in vivo relevance and functional significance of this with respect to the pathogenic mechanisms and clinical expression of the disease remain largely unexplored. To ascertain, in vivo, the functional significance of EGFR10-11 mutations, we generated transgenic mice with one representative mutation (C428S) in EGFR10 of Notch3. These mice, like those with a common R90C mutation, developed characteristic arterial accumulation of Notch3 protein and granular osmiophilic material upon aging. By introducing the mutant C428S transgene into a Notch3 null background, we found that, unlike the R90C mutant protein, the C428S mutant protein has lost wild-type Notch3 activity and exhibited mild dominant-negative activity in three different biological settings. From a large prospectively recruited cohort of 176 CADASIL patients, we identified 10 patients, from five distinct pedigrees carrying a mutation in EGFR10 or 11. These mutations were associated with significantly higher Mini-Mental State Examination and Mattis Dementia Rating Scale scores (P < 0.05), when compared with common mutations. Additionally, we found a strong effect of this genotype on the burden of white matter hyperintensities (P < 0.01). Collectively, these results highlight distinctive functional and phenotypic features of EGFR10-11 mutations relative to the common CADASIL mutations. Our findings are compatible with the hypothesis that EGFR10-11 mutations cause the disease through the same gain of novel function as the common mutations, and lead us to propose that reduced Notch3 signalling acts as a modifier of the CADASIL phenotype.


CADASIL/genetics , Mutation , Receptors, Notch/genetics , Adult , Aged , Animals , Brain/pathology , CADASIL/metabolism , CADASIL/pathology , Cerebral Arteries/metabolism , Cerebral Arteries/ultrastructure , Disease Models, Animal , Genotype , Humans , Ligands , Magnetic Resonance Imaging/methods , Mice , Mice, Transgenic , Microscopy, Electron , Middle Aged , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/ultrastructure , Phenotype , Prospective Studies , Receptor, Notch3 , Receptors, Notch/metabolism , Receptors, Notch/physiology , Reverse Transcriptase Polymerase Chain Reaction/methods
20.
Neurochem Int ; 54(3-4): 253-63, 2009.
Article En | MEDLINE | ID: mdl-19111869

Blood-brain barrier (BBB) characteristics are induced and maintained by cross-talk between brain microvessel endothelial cells and neighbouring elements of the neurovascular unit. While pericytes are the cells situated closest to brain endothelial cells morphologically and share a common basement membrane, they have not been used in co-culture BBB models for testing drug permeability. We have developed and characterized a new syngeneic BBB model using primary cultures of the three main cell types of cerebral microvessels. The co-culture of endothelial cells, pericytes and astrocytes mimick the anatomical situation in vivo. In the presence of both pericytes and astrocytes rat brain endothelial cells expressed enhanced levels of tight junction (TJ) proteins occludin, claudin-5 and ZO-1 with a typical localization at the cell borders. Further morphological evidence of the presence of interendothelial TJs was provided by electron microscopy. The transendothelial electrical resistance (TEER) of brain endothelial monolayers in triple co-culture, indicating the tightness of TJs reached 400Omegacm(2) on average, while the endothelial permeability coefficients (P(e)) for fluorescein was in the range of 3x10(-6)cm/s. Brain endothelial cells in the new model expressed glucose transporter-1, efflux transporters P-glycoprotein and multidrug resistance protein-1, and showed a polarized transport of rhodamine 123, a ligand for P-glycoprotein. To further characterize the model, drug permeability assays were performed using a set of 19 compounds with known in vivo BBB permeability. Good correlation (R(2)=0.89) was found between in vitroP(e) values obtained from measurements on the BBB model and in vivo BBB permeability data. The new BBB model, which is the first model to incorporate pericytes in a triple co-culture setting, can be a useful tool for research on BBB physiology and pathology and to test candidate compounds for centrally acting drugs.


Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Pericytes/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Astrocytes/ultrastructure , Blood-Brain Barrier/ultrastructure , Capillaries/metabolism , Capillaries/ultrastructure , Carrier Proteins/metabolism , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Cells, Cultured , Cerebral Arteries/metabolism , Cerebral Arteries/ultrastructure , Claudin-5 , Endothelial Cells/ultrastructure , Membrane Potentials/physiology , Membrane Proteins/metabolism , Microcirculation/physiology , Microscopy, Electron, Transmission , Models, Biological , Occludin , Pericytes/ultrastructure , Phosphoproteins/metabolism , Rats , Rhodamine 123/pharmacokinetics , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Zonula Occludens-1 Protein
...