Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cereb Blood Flow Metab ; 42(4): 686-693, 2022 04.
Article in English | MEDLINE | ID: mdl-34822744

ABSTRACT

There is growing evidence for the vascular contribution to cognitive impairment and dementia in Alzheimer's disease (AD) and other neurodegenerative diseases. While perfusion deficits have been observed in patients with Alzheimer's disease and tauopaties, little is known about the role of tau in vascular dysfunction. In the present study, regional cerebral blood (rCBF) was characterized in P301L mice with arterial spin labeling. No differences in rCBF in P301L mice compared to their age-matched non-transgenic littermates at mid (10-12 months of age) and advanced (19-21 months of age) disease stages. This was concomitant with preservation of cortical brain structure as assessed with structural T2-weighted magnetic resonance imaging. These results show that hypoperfusion and neurodegeneration are not a phenotype of P301L mice. More studies are thus needed to understand the relationship of tau, neurodegeneration and vascular dysfunction and its modulators in AD and primary tauopathies.


Subject(s)
Alzheimer Disease , Tauopathies , Alzheimer Disease/metabolism , Animals , Cerebrovascular Circulation , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Spin Labels , Tauopathies/diagnostic imaging , Tauopathies/metabolism , tau Proteins/genetics , tau Proteins/metabolism
2.
Angiogenesis ; 24(4): 823-842, 2021 11.
Article in English | MEDLINE | ID: mdl-34046769

ABSTRACT

Pericytes play essential roles in blood-brain barrier integrity and their dysfunction is implicated in neurological disorders such as stroke although the underlying mechanisms remain unknown. Hypoxia-inducible factor-1 (HIF-1), a master regulator of injury responses, has divergent roles in different cells especially during stress scenarios. On one hand HIF-1 is neuroprotective but on the other it induces vascular permeability. Since pericytes are critical for barrier stability, we asked if pericyte HIF-1 signaling impacts barrier integrity and injury severity in a mouse model of ischemic stroke. We show that pericyte HIF-1 loss of function (LoF) diminishes ischemic damage and barrier permeability at 3 days reperfusion. HIF-1 deficiency preserved barrier integrity by reducing pericyte death thereby maintaining vessel coverage and junctional protein organization, and suppressing vascular remodeling. Importantly, considerable improvements in sensorimotor function were observed in HIF-1 LoF mice indicating that better vascular functionality post stroke improves outcome. Thus, boosting vascular integrity by inhibiting pericytic HIF-1 activation and/or increasing pericyte survival may be a lucrative option to accelerate recovery after severe brain injury.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Animals , Blood-Brain Barrier , Hypoxia , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Pericytes
3.
Front Aging Neurosci ; 11: 27, 2019.
Article in English | MEDLINE | ID: mdl-30890928

ABSTRACT

Functional magnetic resonance imaging (fMRI) techniques can be used to assess cerebrovascular dysfunction in Alzheimer's disease, an important and early contributor to pathology. We hypothesized that bradykinin receptor inhibition alleviates the vascular dysfunction in a transgenic arcAß mouse model of cerebral amyloidosis and that fMRI techniques can be used to monitor the treatment response. Transgenic arcAß mice, and non-transgenic littermates of 14 months-of-age were either treated with the bradykinin receptors 1 and 2 blocker noscapine or received normal drinking water as control over 3 months (n = 8-11/group) and all mice were assessed using fMRI at the end of the treatment period. Perfusion MRI using an arterial spin labeling technique showed regional hypoperfusion in arcAß compared to non-transgenic controls, which was alleviated by noscapine treatment. Similarly, measuring cerebral blood volume changes upon pharmacological stimulation using vessel dilator acetazolamide revealed recovery of regional impairment of cerebral vascular reactivity in arcAß mice upon noscapine treatment. In addition, we assessed with immunohistochemistry beta-amyloid (Aß) and inflammation levels in brain sections. Immunohistological stainings for Aß deposition (6E10) and related microgliosis (Iba1) in the cortex and hippocampus were found comparable between noscapine-treated and untreated arcAß mice. In addition, levels of soluble and insoluble Aß38, Aß40, Aß42 were found to be similar in brain tissue homogenates of noscapine-treated and untreated arcAß mice using electro-chemiluminescent based immunoassay. In summary, bradykinin receptors blockade recovered cerebral vascular dysfunction in a mouse model of cerebral amyloidosis. fMRI methods revealed the functional deficit in disease condition and were useful tools to monitor the treatment response.

4.
Brain ; 142(4): 885-902, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30805583

ABSTRACT

Brain calcifications are commonly detected in aged individuals and accompany numerous brain diseases, but their functional importance is not understood. In cases of primary familial brain calcification, an autosomally inherited neuropsychiatric disorder, the presence of bilateral brain calcifications in the absence of secondary causes of brain calcification is a diagnostic criterion. To date, mutations in five genes including solute carrier 20 member 2 (SLC20A2), xenotropic and polytropic retrovirus receptor 1 (XPR1), myogenesis regulating glycosidase (MYORG), platelet-derived growth factor B (PDGFB) and platelet-derived growth factor receptor ß (PDGFRB), are considered causal. Previously, we have reported that mutations in PDGFB in humans are associated with primary familial brain calcification, and mice hypomorphic for PDGFB (Pdgfbret/ret) present with brain vessel calcifications in the deep regions of the brain that increase with age, mimicking the pathology observed in human mutation carriers. In this study, we characterize the cellular environment surrounding calcifications in Pdgfbret/ret animals and show that cells around vessel-associated calcifications express markers for osteoblasts, osteoclasts and osteocytes, and that bone matrix proteins are present in vessel-associated calcifications. Additionally, we also demonstrate the osteogenic environment around brain calcifications in genetically confirmed primary familial brain calcification cases. We show that calcifications cause oxidative stress in astrocytes and evoke expression of neurotoxic astrocyte markers. Similar to previously reported human primary familial brain calcification cases, we describe high interindividual variation in calcification load in Pdgfbret/ret animals, as assessed by ex vivo and in vivo quantification of calcifications. We also report that serum of Pdgfbret/ret animals does not differ in calcification propensity from control animals and that vessel calcification occurs only in the brains of Pdgfbret/ret animals. Notably, ossification of vessels and astrocytic neurotoxic response is associated with specific behavioural and cognitive alterations, some of which are associated with primary familial brain calcification in a subset of patients.


Subject(s)
Astrocytes/metabolism , Ossification, Heterotopic/pathology , Proto-Oncogene Proteins c-sis/metabolism , Aged , Animals , Brain/pathology , Brain Diseases/genetics , Calcinosis/pathology , Female , Humans , Male , Mice , Mutation , Osteogenesis/physiology , Oxidative Stress , Pedigree , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics , Sodium-Phosphate Cotransporter Proteins, Type III/genetics , Xenotropic and Polytropic Retrovirus Receptor
5.
Arch Toxicol ; 90(12): 3017-3027, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26838043

ABSTRACT

Microsomal epoxide hydrolase (mEH) is a detoxifying enzyme for xenobiotic compounds. Enzymatic activity of mEH can be greatly increased by a point mutation, leading to an E404D amino acid exchange in its catalytic triad. Surprisingly, this variant is not found in any vertebrate species, despite the obvious advantage of accelerated detoxification. We hypothesized that this evolutionary avoidance is due to the fact that the mEH plays a dualistic role in detoxification and control of endogenous vascular signaling molecules. To test this, we generated mEH E404D mice and assessed them for detoxification capacity and vascular dynamics. In liver microsomes from these mice, turnover of the xenobiotic compound phenanthrene-9,10-oxide was four times faster compared to WT liver microsomes, confirming accelerated detoxification. mEH E404D animals also showed faster metabolization of a specific class of endogenous eicosanoids, arachidonic acid-derived epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids (DHETs). Significantly higher DHETs/EETs ratios were found in mEH E404D liver, urine, plasma, brain and cerebral endothelial cells compared to WT controls, suggesting a broad impact of the mEH mutant on endogenous EETs metabolism. Because EETs are strong vasodilators in cerebral vasculature, hemodynamics were assessed in mEH E404D and WT cerebral cortex and hippocampus using cerebral blood volume (CBV)-based functional magnetic resonance imaging (fMRI). Basal CBV0 levels were similar between mEH E404D and control mice in both brain areas. But vascular reactivity and vasodilation in response to the vasodilatory drug acetazolamide were reduced in mEH E404D forebrain compared to WT controls by factor 3 and 2.6, respectively. These results demonstrate a critical role for mEH E404D in vasodynamics and suggest that deregulation of endogenous signaling pathways is the undesirable gain of function associated with the E404D variant.


Subject(s)
Cerebrovascular Circulation , Cerebrovascular Disorders/metabolism , Epoxide Hydrolases/metabolism , Microsomes, Liver/enzymology , Point Mutation , Xenobiotics/pharmacokinetics , Amino Acid Substitution , Animals , Catalytic Domain , Cerebral Cortex/blood supply , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cerebrovascular Circulation/drug effects , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/physiopathology , Eicosanoids/blood , Eicosanoids/metabolism , Eicosanoids/urine , Epoxide Hydrolases/chemistry , Epoxide Hydrolases/genetics , Hippocampus/blood supply , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Inactivation, Metabolic , Mice , Mice, Mutant Strains , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Phenanthrenes/metabolism , Vascular Resistance/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Xenobiotics/metabolism
6.
J Cereb Blood Flow Metab ; 36(9): 1614-24, 2016 09.
Article in English | MEDLINE | ID: mdl-26661253

ABSTRACT

Magnetic resonance imaging employing administration of iron oxide-based contrast agents is widely used to visualize cellular and molecular processes in vivo. In this study, we investigated the ability of [Formula: see text] and quantitative susceptibility mapping to quantitatively assess the accumulation of ultrasmall superparamagnetic iron oxide (USPIO) particles in the arcAß mouse model of cerebral amyloidosis. Gradient-echo data of mouse brains were acquired at 9.4 T after injection of USPIO. Focal areas with increased magnetic susceptibility and [Formula: see text] values were discernible across several brain regions in 12-month-old arcAß compared to 6-month-old arcAß mice and to non-transgenic littermates, indicating accumulation of particles after USPIO injection. This was concomitant with higher [Formula: see text] and increased magnetic susceptibility differences relative to cerebrospinal fluid measured in USPIO-injected compared to non-USPIO-injected 12-month-old arcAß mice. No differences in [Formula: see text] and magnetic susceptibility were detected in USPIO-injected compared to non-injected 12-month-old non-transgenic littermates. Histological analysis confirmed focal uptake of USPIO particles in perivascular macrophages adjacent to small caliber cerebral vessels with radii of 2-8 µm that showed no cerebral amyloid angiopathy. USPIO-enhanced [Formula: see text] and quantitative susceptibility mapping constitute quantitative tools to monitor such functional microvasculopathies.


Subject(s)
Cerebral Amyloid Angiopathy/diagnostic imaging , Ferric Compounds/pharmacokinetics , Magnetic Resonance Imaging/methods , Microvessels/pathology , Animals , Cerebral Amyloid Angiopathy/metabolism , Cerebrovascular Circulation , Contrast Media , Macrophages/metabolism , Mice
7.
Ann Neurol ; 58(5): 706-19, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16173073

ABSTRACT

Spinal cord trauma leads to loss of motor, sensory and autonomic functions below the lesion. Recovery is very restricted, due in part to neurite growth inhibitory myelin proteins, in particular Nogo-A. Two neutralizing antibodies against Nogo-A were used to study recovery and axonal regeneration after spinal cord lesions. Three months old Lewis rats were tested in sensory-motor tasks (open field locomotion, crossing of ladder rungs and narrow beams, the CatWalk(R) runway, reactions to heat and von Frey hairs). A T-shaped lesion was made at T8, and an intrathecal catheter delivered highly purified anti-Nogo-A monoclonal IgGs or unspecific IgGs for 2 weeks. A better outcome in motor behavior was obtained as early as two weeks after lesion in the animals receiving the Nogo-A antibodies. Withdrawal responses to heat and mechanical stimuli were not different between the groups. Histology showed enhanced regeneration of corticospinal axons in the anti-Nogo-A antibody groups. fMRI revealed significant cortical responses to stimulation of the hindpaw exclusively in anti-Nogo-A animals. These results demonstrate that neutralization of the neurite growth inhibitor Nogo-A by intrathecal antibodies leads to enhanced regeneration and reorganization of the injured CNS, resulting in improved recovery of compromised functions in the absence of dysfunctions.


Subject(s)
Immunoglobulin G/therapeutic use , Locomotion/drug effects , Myelin Proteins/immunology , Nerve Regeneration/drug effects , Spinal Cord Injuries/drug therapy , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Female , Functional Laterality , Image Processing, Computer-Assisted/methods , Immunoglobulin G/cerebrospinal fluid , Locomotion/physiology , Magnetic Resonance Imaging/methods , Nerve Regeneration/physiology , Nogo Proteins , Psychomotor Performance/drug effects , Rats , Rats, Inbred Lew , Recovery of Function/drug effects , Reflex/drug effects , Spinal Cord Injuries/cerebrospinal fluid , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Swimming , Time Factors , Urinary Bladder/drug effects , Urinary Bladder/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...