Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Alzheimers Dement ; 20(2): 1421-1435, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37897797

ABSTRACT

This editorial summarizes advances from the Clearance of Interstitial Fluid and Cerebrospinal Fluid (CLIC) group, within the Vascular Professional Interest Area (PIA) of the Alzheimer's Association International Society to Advance Alzheimer's Research and Treatment (ISTAART). The overarching objectives of the CLIC group are to: (1) understand the age-related physiology changes that underlie impaired clearance of interstitial fluid (ISF) and cerebrospinal fluid (CSF) (CLIC); (2) understand the cellular and molecular mechanisms underlying intramural periarterial drainage (IPAD) in the brain; (3) establish novel diagnostic tests for Alzheimer's disease (AD), cerebral amyloid angiopathy (CAA), retinal amyloid vasculopathy, amyloid-related imaging abnormalities (ARIA) of spontaneous and iatrogenic CAA-related inflammation (CAA-ri), and vasomotion; and (4) establish novel therapies that facilitate IPAD to eliminate amyloid ß (Aß) from the aging brain and retina, to prevent or reduce AD and CAA pathology and ARIA side events associated with AD immunotherapy.


Subject(s)
Alzheimer Disease , Cerebral Amyloid Angiopathy , Cerebrovascular Disorders , Humans , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Extracellular Fluid , Cerebral Amyloid Angiopathy/therapy , Cerebral Amyloid Angiopathy/pathology , Brain/metabolism , Cerebrovascular Disorders/complications
2.
Nutrients ; 14(23)2022 Dec 04.
Article in English | MEDLINE | ID: mdl-36501191

ABSTRACT

Alterations in neural pathways that regulate appetitive motivation may contribute to increased obesity risk in offspring born to mothers fed a high fat (HF) diet. However, current findings on the impact of maternal obesity on motivation in offspring are inconclusive, and there is no information about the long-lasting effects in aged animals. This study examined the longitudinal effect of perinatal and chronic postnatal HF intake on appetitive motivation in young and aged offspring. Female C57Bl/6 were fed either a control (C) or HF diet before mating through to lactation. At weaning, offspring were maintained on the C or HF diet, generating the following four diet groups: C/C, C/HF, HF/C, and HF/HF based on the pre/post weaning diet. At 6 months, motivation was higher in HF/C females, but lower in male and female C/HF and HF/HF mice. By 12 months, this difference was lost, as C-fed animals became less motivated, while motivation increased in HF-fed mice. The mRNA levels of dopamine receptor 1 and 2 increased with age, while cannabinoid receptor 1 and µ-opioid receptor expression remained stable or decreased in mesolimbic and mesocortical dopaminergic pathways. Results from this study suggest that perinatal and chronic postnatal HF feeding produced opposite effects on appetitive motivation in young adult offspring mice, which was also reflected in the shift in motivation over time. These results have significant implications for patterns of hedonic eating across the life course and the relative risk of obesity at different time points.


Subject(s)
Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects , Animals , Mice , Female , Pregnancy , Male , Humans , Life Change Events , Diet, High-Fat/adverse effects , Obesity/metabolism , Lactation
3.
Acta Neuropathol Commun ; 10(1): 127, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36045406

ABSTRACT

α-Dystrobrevin (α-DB) is a major component of the dystrophin-associated protein complex (DAPC). Knockout (KO) of α-DB in the brain is associated with astrocytic abnormalities and loss of neuronal GABA receptor clustering. Mutations in DAPC proteins are associated with altered dopamine signaling and cognitive and psychiatric disorders, including schizophrenia. This study tested the hypothesis that motivation and associated underlying biological pathways are altered in the absence of α-DB expression. Male wildtype and α-DB KO mice were tested for measures of motivation, executive function and extinction in the rodent touchscreen apparatus. Subsequently, brain tissues were evaluated for mRNA and/or protein levels of dysbindin-1, dopamine transporter and receptor 1 and 2, mu opioid receptor 1 (mOR1) and cannabinoid receptor 1 (CB1). α-DB KO mice had significantly increased motivation for the appetitive reward, while measures of executive function and extinction were unaffected. No differences were observed between wildtype and KO animals on mRNA levels of dysbindin-1 or any of the dopamine markers. mRNA levels of mOR1were significantly decreased in the caudate-putamen and nucleus accumbens of α-DB KO compared to WT animals, but protein levels were unaltered. However, CB1 protein levels were significantly increased in the prefrontal cortex and decreased in the nucleus accumbens of α-DB KO mice. Triple-labelling immunohistochemistry confirmed that changes in CB1 were not specific to astrocytes. These results highlight a novel role for α-DB in the regulation of appetitive motivation that may have implications for other behaviours that involve the dopaminergic and endocannabinoid systems.


Subject(s)
Dopamine , Dystrophin-Associated Proteins , Motivation , Receptors, Cannabinoid , Animals , Brain/metabolism , Dopamine/metabolism , Dysbindin/metabolism , Dystrophin-Associated Proteins/genetics , Male , Mice , Mice, Knockout , RNA, Messenger/metabolism , Receptors, Cannabinoid/genetics , Receptors, Cannabinoid/metabolism , Reward
5.
Pharmaceutics ; 13(9)2021 Sep 21.
Article in English | MEDLINE | ID: mdl-34575601

ABSTRACT

Blood-brain barrier (BBB) dysfunction is a key hallmark in the pathology of many neuroinflammatory disorders. Extracellular vesicles (EVs) are lipid membrane-enclosed carriers of molecular cargo that are involved in cell-to-cell communication. Circulating endothelial EVs are increased in the plasma of patients with neurological disorders, and immune cell-derived EVs are known to modulate cerebrovascular functions. However, little is known about whether brain endothelial cell (BEC)-derived EVs themselves contribute to BBB dysfunction. Human cerebral microvascular cells (hCMEC/D3) were treated with TNFα and IFNy, and the EVs were isolated and characterised. The effect of EVs on BBB transendothelial resistance (TEER) and leukocyte adhesion in hCMEC/D3 cells was measured by electric substrate cell-substrate impedance sensing and the flow-based T-cell adhesion assay. EV-induced molecular changes in recipient hCMEC/D3 cells were analysed by RT-qPCR and Western blotting. A stimulation of naïve hCMEC/D3 cells with small EVs (sEVs) reduced the TEER and increased the shear-resistant T-cell adhesion. The levels of microRNA-155, VCAM1 and ICAM1 were increased in sEV-treated hCMEC/D3 cells. Blocking the expression of VCAM1, but not of ICAM1, prevented sEV-mediated T-cell adhesion to brain endothelia. These results suggest that sEVs derived from inflamed BECs promote cerebrovascular dysfunction. These findings may provide new insights into the mechanisms involving neuroinflammatory disorders.

6.
Neurobiol Aging ; 101: 273-284, 2021 05.
Article in English | MEDLINE | ID: mdl-33579556

ABSTRACT

Blood-brain barrier (BBB) breakdown occurs in aging and neurodegenerative diseases. Although age-associated alterations have previously been described, most studies focused in male brains; hence, little is known about BBB breakdown in females. This study measured ultrastructural features in the aging female BBB using transmission electron microscopy and 3-dimensional reconstruction of cortical and hippocampal capillaries from 6- and 24-month-old female C57BL/6J mice. Aged cortical capillaries showed more changes than hippocampal capillaries. Specifically, the aged cortex showed thicker basement membrane, higher number and volume of endothelial pseudopods, decreased endothelial mitochondrial number, larger pericyte mitochondria, higher pericyte-endothelial cell contact, and increased tight junction tortuosity compared with young animals. Only increased basement membrane thickness and pericyte mitochondrial volume were observed in the aged hippocampus. Regional comparison revealed significant differences in endothelial pseudopods and tight junctions between the cortex and hippocampus of 24-month-old mice. Therefore, the aging female BBB shows region-specific ultrastructural alterations that may lead to oxidative stress and abnormal capillary blood flow and barrier stability, potentially contributing to cerebrovascular diseases, particularly in postmenopausal women.


Subject(s)
Aging/pathology , Blood-Brain Barrier/ultrastructure , Capillaries/ultrastructure , Cerebral Cortex/blood supply , Cerebral Cortex/ultrastructure , Hippocampus/blood supply , Hippocampus/ultrastructure , Animals , Basement Membrane/pathology , Basement Membrane/ultrastructure , Blood-Brain Barrier/pathology , Capillaries/pathology , Cerebral Cortex/pathology , Female , Hippocampus/pathology , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Mitochondrial Size , Oxidative Stress , Pericytes/pathology , Pericytes/ultrastructure , Postmenopause
7.
Acta Neuropathol Commun ; 9(1): 12, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33413694

ABSTRACT

Vascular dysregulation and cholinergic basal forebrain degeneration are both early pathological events in the development of Alzheimer's disease (AD). Acetylcholine contributes to localised arterial dilatation and increased cerebral blood flow (CBF) during neurovascular coupling via activation of endothelial nitric oxide synthase (eNOS). Decreased vascular reactivity is suggested to contribute to impaired clearance of ß-amyloid (Aß) along intramural periarterial drainage (IPAD) pathways of the brain, leading to the development of cerebral amyloid angiopathy (CAA). However, the possible relationship between loss of cholinergic innervation, impaired vasoreactivity and reduced clearance of Aß from the brain has not been previously investigated. In the present study, intracerebroventricular administration of mu-saporin resulted in significant death of cholinergic neurons and fibres in the medial septum, cortex and hippocampus of C57BL/6 mice. Arterial spin labelling MRI revealed a loss of CBF response to stimulation of eNOS by the Rho-kinase inhibitor fasudil hydrochloride in the cortex of denervated mice. By contrast, the hippocampus remained responsive to drug treatment, in association with altered eNOS expression. Fasudil hydrochloride significantly increased IPAD in the hippocampus of both control and saporin-treated mice, while increased clearance from the cortex was only observed in control animals. Administration of mu-saporin in the TetOAPPSweInd mouse model of AD was associated with a significant and selective increase in Aß40-positive CAA. These findings support the importance of the interrelationship between cholinergic innervation and vascular function in the aetiology and/or progression of CAA and suggest that combined eNOS/cholinergic therapies may improve the efficiency of Aß removal from the brain and reduce its deposition as CAA.


Subject(s)
Acetylcholine/metabolism , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/physiopathology , Cerebral Cortex/blood supply , Cerebrovascular Circulation/physiology , Cholinergic Fibers/physiology , Cholinergic Neurons/physiology , Hippocampus/blood supply , Nitric Oxide Synthase Type III/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Cerebral Amyloid Angiopathy/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebrovascular Circulation/drug effects , Cholinergic Fibers/drug effects , Cholinergic Fibers/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Neurovascular Coupling/drug effects , Neurovascular Coupling/physiology , Saporins/toxicity , Septal Nuclei , Vasodilator Agents/pharmacology
8.
Alzheimers Dement (Amst) ; 12(1): e12053, 2020.
Article in English | MEDLINE | ID: mdl-32775596

ABSTRACT

Two of the key functions of arteries in the brain are (1) the well-recognized supply of blood via the vascular lumen and (2) the emerging role for the arterial walls as routes for the elimination of interstitial fluid (ISF) and soluble metabolites, such as amyloid beta (Aß), from the brain and retina. As the brain and retina possess no conventional lymphatic vessels, fluid drainage toward peripheral lymph nodes is mediated via transport along basement membranes in the walls of capillaries and arteries that form the intramural peri-arterial drainage (IPAD) system. IPAD tends to fail as arteries age but the mechanisms underlying the failure are unclear. In some people this is reflected in the accumulation of Aß plaques in the brain in Alzheimer's disease (AD) and deposition of Aß within artery walls as cerebral amyloid angiopathy (CAA). Knowledge of the dynamics of IPAD and why it fails with age is essential for establishing diagnostic tests for the early stages of the disease and for devising therapies that promote the clearance of Aß in the prevention and treatment of AD and CAA. This editorial is intended to introduce the rationale that has led to the establishment of the Clearance of Interstitial Fluid (ISF) and CSF (CLIC) group, within the Vascular Professional Interest Area of the Alzheimer's Association International Society to Advance Alzheimer's Research and Treatment.

9.
Fluids Barriers CNS ; 17(1): 10, 2020 Feb 10.
Article in English | MEDLINE | ID: mdl-32036786

ABSTRACT

BACKGROUND: Idiopathic intracranial hypertension (IIH) is a neurological disorder characterised by raised cerebrospinal fluid (CSF) pressure in the absence of any intracranial pathology. IIH mainly affects women with obesity between the ages of 15 and 45. Two possible mechanisms that could explain the increased CSF pressure in IIH are excessive CSF production by the choroid plexus (CP) epithelium or impaired CSF drainage from the brain. However, the molecular mechanisms controlling these mechanisms in IIH remain to be determined. METHODS: In vivo ventriculo-cisternal perfusion (VCP) and variable rate infusion (VRI) techniques were used to assess changes in rates of CSF secretion and resistance to CSF drainage in female and male Wistar rats fed either a control (C) or high-fat (HF) diet (under anaesthesia with 20 µl/100 g medetomidine, 50 µl/100 g ketamine i.p). In addition, CSF secretion and drainage were assessed in female rats following treatment with inflammatory mediators known to be elevated in the CSF of IIH patients: C-C motif chemokine ligand 2 (CCL2), interleukin (IL)-17 (IL-17), IL-6, IL-1ß, tumour necrosis factor-α (TNF-α), as well as glucocorticoid hydrocortisone (HC). RESULTS: Female rats fed the HF diet had greater CSF secretion compared to those on control diet (3.18 ± 0.12 µl/min HF, 1.49 ± 0.15 µl/min control). Increased CSF secretion was seen in both groups following HC treatment (by 132% in controls and 114% in HF) but only in control rats following TNF-α treatment (137% increase). The resistance to CSF drainage was not different between control and HF fed female rats (6.13 ± 0.44 mmH2O min/µl controls, and 7.09 ± 0.26 mmH2O min/µl HF). and when treated with CCL2, both groups displayed an increase in resistance to CSF drainage of 141% (controls) and 139% (HF) indicating lower levels of CSF drainage. CONCLUSIONS: Weight loss and therapies targeting HC, TNF-α and CCL2, whether separately or in combination, may be beneficial to modulate rates of CSF secretion and/or resistance to CSF drainage pathways, both factors likely contributing to the raised intracranial pressure (ICP) observed in female IIH patients with obesity.


Subject(s)
Cerebrospinal Fluid Leak/drug therapy , Cerebrospinal Fluid/drug effects , Cytokines/pharmacology , Diet , Animals , Brain/drug effects , Brain/physiopathology , Cytokines/metabolism , Female , Hydrodynamics , Intracranial Hypertension/drug therapy , Intracranial Pressure/drug effects , Male , Obesity/complications , Rats, Wistar
10.
Front Neurosci ; 13: 1045, 2019.
Article in English | MEDLINE | ID: mdl-31632236

ABSTRACT

Compelling experimental and clinical evidence supports a role for maternal obesity in offspring health. Adult children of obese mothers are at greater risk of obesity, diabetes, coronary heart disease and stroke. These offspring may also be at greater risk of age-related neurodegenerative diseases for which mid-life obesity is a risk factor. Rodent diet-induced obesity models have shown that high fat (HF) diet consumption damages the integrity of the blood-brain barrier (BBB) in the adult brain. However, there is currently little information about the effect of chronic HF feeding on the BBB of aged animals. Moreover, the long-term consequences of maternal obesity on the cerebrovasculature of aged offspring are not known. This study determined the impact of pre- and post-natal HF diet on the structure and integrity of cerebral blood vessels in aged male and female mice. Female C57Bl/6 mice were fed either a 10% fat control (C) or 45% HF diet before mating and during gestation and lactation. At weaning, male and female offspring were fed the C or HF diet until sacrifice at 16-months of age. Both dams and offspring fed the HF diet weighed significantly more than mice fed the C diet. Post-natal HF diet exposure increased hippocampal BBB leakiness in female offspring, in association with loss of astrocyte endfoot coverage of arteries. Markers of tight junctions, pericytes or smooth muscle cells were not altered by pre- or post-natal HF diet. Male offspring born to HF-fed mothers showed decreased parenchymal GFAP expression compared to offspring of mothers fed C diet, while microglial and macrophage markers were higher in the same female diet group. In addition, female offspring exposed to the HF diet for their entire lifespan showed more significant changes in vessel structure, BBB permeability and inflammation compared to male animals. These results suggest that the long-term impact of prenatal HF diet on the integrity of cerebral blood vessels differs between male and female offspring depending on the post-natal diet. This may have implications for the prevention and management of age- and obesity-related cerebrovascular diseases that differentially affect men and women.

11.
Front Aging Neurosci ; 11: 172, 2019.
Article in English | MEDLINE | ID: mdl-31333445

ABSTRACT

Increasing evidence supports a role for cerebrovasculature dysfunction in the etiology of Alzheimer's disease (AD). Blood vessels in the brain are composed of a collection of cells and acellular material that comprise the neurovascular unit (NVU). The NVU in the hippocampus and cortex receives innervation from cholinergic neurons that originate in the basal forebrain. Death of these neurons and their nerve fibers is an early feature of AD. However, the effect of the loss of cholinergic innervation on the NVU is not well characterized. The purpose of this study was to evaluate the effect of the loss of cholinergic innervation of components of the NVU at capillaries, arteries and veins in the hippocampus and cortex. Adult male C57BL/6 mice received an intracerebroventricular injection of the immunotoxin p75NTR mu-saporin to induce the loss of cholinergic neurons. Quadruple labeling immunohistochemistry and 3D reconstruction were carried out to characterize specific points of contact between cholinergic fibers and collagen IV, smooth muscle cells and astrocyte endfeet. Innate differences were observed between vessels of the hippocampus and cortex of control mice, including a greater amount of cholinergic contact with perivascular astrocytes in hippocampal capillaries and a thicker basement membrane in hippocampal veins. Saporin treatment induced a loss of cholinergic innervation at the arterial basement membrane and smooth muscle cells of both the hippocampus and the cortex. In the cortex, there was an additional loss of innervation at the astrocytic endfeet. The current results suggest that cortical arteries are more strongly affected by cholinergic denervation than arteries in the hippocampus. This regional variation may have implications for the etiology of the vascular pathology that develops in AD.

12.
Acta Neuropathol ; 136(1): 139-152, 2018 07.
Article in English | MEDLINE | ID: mdl-29754206

ABSTRACT

Tracers injected into CSF pass into the brain alongside arteries and out again. This has been recently termed the "glymphatic system" that proposes tracers enter the brain along periarterial "spaces" and leave the brain along the walls of veins. The object of the present study is to test the hypothesis that: (1) tracers from the CSF enter the cerebral cortex along pial-glial basement membranes as there are no perivascular "spaces" around cortical arteries, (2) tracers leave the brain along smooth muscle cell basement membranes that form the Intramural Peri-Arterial Drainage (IPAD) pathways for the elimination of interstitial fluid and solutes from the brain. 2 µL of 100 µM soluble, fluorescent fixable amyloid ß (Aß) were injected into the CSF of the cisterna magna of 6-10 and 24-30 month-old male mice and their brains were examined 5 and 30 min later. At 5 min, immunocytochemistry and confocal microscopy revealed Aß on the outer aspects of cortical arteries colocalized with α-2 laminin in the pial-glial basement membranes. At 30 min, Aß was colocalised with collagen IV in smooth muscle cell basement membranes in the walls of cortical arteries corresponding to the IPAD pathways. No evidence for drainage along the walls of veins was found. Measurements of the depth of penetration of tracer were taken from 11 regions of the brain. Maximum depths of penetration of tracer into the brain were achieved in the pons and caudoputamen. Conclusions drawn from the present study are that tracers injected into the CSF enter and leave the brain along separate periarterial basement membrane pathways. The exit route is along IPAD pathways in which Aß accumulates in cerebral amyloid angiopathy (CAA) in Alzheimer's disease. Results from this study suggest that CSF may be a suitable route for delivery of therapies for neurological diseases, including CAA.


Subject(s)
Amyloid beta-Peptides/metabolism , Basement Membrane/metabolism , Brain/metabolism , Cerebrospinal Fluid/metabolism , Extracellular Fluid/metabolism , Glymphatic System/metabolism , Actins/metabolism , Age Factors , Amyloid beta-Peptides/cerebrospinal fluid , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Blood Vessels/cytology , Blood Vessels/metabolism , Brain/cytology , Collagen Type IV/metabolism , Fluorescein-5-isothiocyanate/metabolism , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Parenchymal Tissue/metabolism , Receptors, Cell Surface/metabolism , Time Factors
13.
Clin Sci (Lond) ; 132(8): 851-868, 2018 04 30.
Article in English | MEDLINE | ID: mdl-29712883

ABSTRACT

Cerebral small vessel disease (SVD) is a major contributor to stroke, cognitive impairment and dementia with limited therapeutic interventions. There is a critical need to provide mechanistic insight and improve translation between pre-clinical research and the clinic. A 2-day workshop was held which brought together experts from several disciplines in cerebrovascular disease, dementia and cardiovascular biology, to highlight current advances in these fields, explore synergies and scope for development. These proceedings provide a summary of key talks at the workshop with a particular focus on animal models of cerebral vascular disease and dementia, mechanisms and approaches to improve translation. The outcomes of discussion groups on related themes to identify the gaps in knowledge and requirements to advance knowledge are summarized.


Subject(s)
Cerebral Small Vessel Diseases/etiology , Translational Research, Biomedical , Animals , Humans
14.
Clin Sci (Lond) ; 131(12): 1207-1214, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28566449

ABSTRACT

Neuronal death is a hallmark of Alzheimer's disease (AD) and considerable work has been done to understand how the loss of interconnectivity between neurons contributes to the associated dementia. Often overlooked however, is how the loss of neuronal innervation of blood vessels, termed perivascular innervation, may also contribute to the pathogenesis of AD. There is now considerable evidence supporting a crucial role for the neurovascular unit (NVU) in mediating the clearance of the ß-amyloid (Aß) peptide, one of the main pathological constituents of AD, from the brain. Moreover, efficient removal appears to be dependent on the communication of cells within the NVU to maintain adequate vascular tone and pulsatility. This review summarizes the composition of the NVU, including the sources of perivascular innervation and how the NVU mediates Aß clearance from the brain. It also explores evidence supporting the hypothesis that loss of neurally mediated vasoreactivity contributes to Aß pathology in the AD brain.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Blood Vessels/innervation , Brain/blood supply , Hemodynamics , Nerve Degeneration , Neurovascular Coupling , Plaque, Amyloid , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Brain/physiopathology , Cerebrovascular Circulation , Humans , Neurons/pathology
15.
Int J Mol Sci ; 18(5)2017 May 19.
Article in English | MEDLINE | ID: mdl-28534818

ABSTRACT

Globally, more than 20% of women of reproductive age are currently estimated to be obese. Children born to obese mothers are at higher risk of developing obesity, coronary heart disease, diabetes, stroke, and asthma in adulthood. Increasing clinical and experimental evidence suggests that maternal obesity also affects the health and function of the offspring brain across the lifespan. This review summarizes the current findings from human and animal studies that detail the impact of maternal obesity on aspects of learning, memory, motivation, affective disorders, attention-deficit hyperactivity disorder, autism spectrum disorders, and neurodegeneration in the offspring. Epigenetic mechanisms that may contribute to this mother-child interaction are also discussed.


Subject(s)
Mood Disorders/etiology , Neurodegenerative Diseases/etiology , Neurodevelopmental Disorders/etiology , Obesity/complications , Prenatal Exposure Delayed Effects/etiology , Animals , Attention Deficit Disorder with Hyperactivity/etiology , Autism Spectrum Disorder/etiology , Cognition , Diet, High-Fat/adverse effects , Female , Humans , Mental Health , Obesity/etiology , Pregnancy , Pregnancy Complications/etiology
16.
Acta Neuropathol ; 131(5): 725-36, 2016 May.
Article in English | MEDLINE | ID: mdl-26975356

ABSTRACT

In the absence of conventional lymphatics, drainage of interstitial fluid and solutes from the brain parenchyma to cervical lymph nodes is along basement membranes in the walls of cerebral capillaries and tunica media of arteries. Perivascular pathways are also involved in the entry of CSF into the brain by the convective influx/glymphatic system. The objective of this study is to differentiate the cerebral vascular basement membrane pathways by which fluid passes out of the brain from the pathway by which CSF enters the brain. Experiment 1: 0.5 µl of soluble biotinylated or fluorescent Aß, or 1 µl 15 nm gold nanoparticles was injected into the mouse hippocampus and their distributions determined at 5 min by transmission electron microscopy. Aß was distributed within the extracellular spaces of the hippocampus and within basement membranes of capillaries and tunica media of arteries. Nanoparticles did not enter capillary basement membranes from the extracellular spaces. Experiment 2: 2 µl of 15 nm nanoparticles were injected into mouse CSF. Within 5 min, groups of nanoparticles were present in the pial-glial basement membrane on the outer aspect of cortical arteries between the investing layer of pia mater and the glia limitans. The results of this study and previous research suggest that cerebral vascular basement membranes form the pathways by which fluid passes into and out of the brain but that different basement membrane layers are involved. The significance of these findings for neuroimmunology, Alzheimer's disease, drug delivery to the brain and the concept of the Virchow-Robin space are discussed.


Subject(s)
Basement Membrane/metabolism , Blood Vessels/cytology , Hippocampus/metabolism , Actins/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacokinetics , Animals , Basement Membrane/drug effects , Basement Membrane/ultrastructure , Biotinylation , Cerebrospinal Fluid/drug effects , Cerebrospinal Fluid/metabolism , Cisterna Magna/drug effects , Cisterna Magna/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Fluorescent Dyes/pharmacokinetics , Hippocampus/drug effects , Hippocampus/ultrastructure , Laminin/metabolism , Male , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Peptide Fragments/metabolism , Peptide Fragments/pharmacokinetics
17.
Sci Rep ; 6: 21981, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26911528

ABSTRACT

Maternal obesity is associated with increased risk of developing diabetes, obesity and premature death in adult offspring. Mid-life diabetes, hypertension and hypercholesterolaemia are risk factors for the development of sporadic Alzheimer's disease (AD). A key pathogenic feature of AD is the accumulation of ß-amyloid (Aß) in the brain. The purpose of this study was to investigate the effect of high fat diet feeding during early life on Aß pathology in the Tg2576 mouse model of AD. Female mice were fed a standard (C) or high fat (HF) diet before mating and during gestation and lactation. At weaning, male offspring were fed a C diet. Significantly higher levels of guanidine-soluble Aß and plaque loads were observed in the hippocampi of 11-month old Tg2576 mice born to mothers fed a HF diet. Changes in the extracellular matrix led to increased retention of Aß within the parenchyma. These data support a role for maternal and gestational health on the health of the aged brain and pathologies associated with AD and may provide a novel target for both the prevention and treatment of AD.


Subject(s)
Aging , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Diet, High-Fat/adverse effects , Maternal Exposure/adverse effects , Prenatal Exposure Delayed Effects , Alzheimer Disease/pathology , Animals , Biomarkers , Disease Models, Animal , Extracellular Matrix/metabolism , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Plaque, Amyloid , Pregnancy
19.
J Pathol ; 235(4): 619-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25345857

ABSTRACT

Alzheimer's disease (AD) is characterized by the accumulation of ß-amyloid (Aß) peptides in the extracellular spaces of the brain as plaques and in the walls of blood vessels as cerebral amyloid angiopathy (CAA). Failure of perivascular drainage of Aß along cerebrovascular basement membranes contributes to the development of CAA. Mid-life hypercholesterolaemia is a risk factor for the development of AD. Maternal obesity is associated with the development of obesity, hypertension and hypercholesterolaemia in adulthood, suggesting that the risk for AD and CAA may also be influenced by the early-life environment. In the present study, we tested the hypothesis that early-life exposure to a high-fat diet results in changes to the cerebrovasculature and failure of Aß clearance from the brain. We also assessed whether vascular Aß deposition is greater in the brains of aged humans with a history of hyperlipidaemia, compared to age-matched controls with normal lipidaemia. Using a mouse model of maternal obesity, we found that exposure to a high-fat diet during gestation and lactation induced changes in multiple components of the neurovascular unit, including a down-regulation in collagen IV, fibronectin and apolipoprotein E, an up-regulation in markers of astrocytes and perivascular macrophages and altered blood vessel morphology in the brains of adult mice. Sustained high-fat diet over the entire lifespan resulted in additional decreases in levels of pericytes and impaired perivascular clearance of Aß from the brain. In humans, vascular Aß load was significantly increased in the brains of aged individuals with a history of hypercholesterolaemia. These results support a critical role for early dietary influence on the brain vasculature across the lifespan, with consequences for the development of age-related cerebrovascular and neurodegenerative diseases.


Subject(s)
Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Animal Nutritional Physiological Phenomena , Brain/metabolism , Cerebral Amyloid Angiopathy/etiology , Cerebral Arteries/metabolism , Diet, High-Fat/adverse effects , Maternal Nutritional Physiological Phenomena , Obesity/complications , Prenatal Exposure Delayed Effects , Age Factors , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Basement Membrane/metabolism , Brain/pathology , Case-Control Studies , Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Cerebral Arteries/pathology , Cholesterol/metabolism , Extracellular Matrix Proteins/metabolism , Female , Gestational Age , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Mice, Inbred C57BL , Nutritional Status , Obesity/metabolism , Obesity/physiopathology , Plaque, Amyloid , Pregnancy , Risk Factors , Time Factors
20.
Brain Pathol ; 25(1): 63-78, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25521178

ABSTRACT

White matter abnormalities on magnetic resonance imaging (MRI) are associated with dementia and include white matter hyperintensities (WMH; also termed leukoaraiosis) and visible perivascular spaces (PVS). We review the potential role of impaired drainage of interstitial fluid in the pathogenesis of WMH and PVS. Whereas the volume of extracellular space in the grey matter is tightly controlled, fluid accumulates and expands the extracellular spaces of the white matter in acute hydrocephalus, vasogenic edema and WMH. Although there are no conventional lymphatic vessels in the brain, there is very effective lymphatic drainage for fluid and solutes along restricted pathways in the basement membranes of cerebral capillaries and arteries in young individuals. Lymphatic drainage of the brain is impaired with age and in association with apolipoprotein E ε4, risk factors for Alzheimer's disease and cerebral amyloid angiopathy (CAA). Deposition of proteins in the lymphatic drainage pathways in the walls of cerebral arteries with age is recognized as protein elimination failure angiopathy (PEFA), as in CAA and cerebral autosomal dominant arteriopathy and leukoencephalopathy (CADASIL). Facilitating perivascular lymphatic drainage from the aging brain may play a significant role in the prevention of CAA, WMH and Alzheimer's disease and may enhance the efficacy of immunotherapy for Alzheimer's disease.


Subject(s)
Brain/physiopathology , Dementia/physiopathology , Extracellular Fluid/metabolism , Leukoaraiosis/physiopathology , White Matter/physiopathology , Aging/pathology , Aging/physiology , Animals , Brain/pathology , Dementia/pathology , Dementia/therapy , Humans , Leukoaraiosis/pathology , Leukoaraiosis/therapy , White Matter/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...