Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Proc Natl Acad Sci U S A ; 118(45)2021 11 09.
Article in English | MEDLINE | ID: mdl-34728566

ABSTRACT

Drainage of interstitial fluid and solutes from the brainstem has not been well studied. To map one drainage pathway in the human brainstem, we took advantage of the focal blood-brain barrier disruption occurring in a multiple sclerosis brainstem lesion, coupled with intravenous injection of gadolinium, which simulates an intraparenchymal injection of gadolinium tracer within the restricted confines of this small brain region. Using high-resolution MRI, we show how it is possible for interstitial fluid to drain into the adjacent trigeminal and oculomotor nerves, in keeping with a pathway of communication between the extracellular spaces of the brainstem and cranial nerve parenchyma.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain Stem/physiology , Cranial Nerves/physiology , Extracellular Fluid/physiology , Multiple Sclerosis/physiopathology , Adult , Blood-Brain Barrier/diagnostic imaging , Cranial Nerves/diagnostic imaging , Gadolinium , Humans , Magnetic Resonance Imaging , Male
2.
Acta Neuropathol Commun ; 9(1): 171, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34674769

ABSTRACT

The extracellular matrix (ECM) of the cerebral vasculature provides a pathway for the flow of interstitial fluid (ISF) and solutes out of the brain by intramural periarterial drainage (IPAD). Failure of IPAD leads to protein elimination failure arteriopathies such as cerebral amyloid angiopathy (CAA). The ECM consists of a complex network of glycoproteins and proteoglycans that form distinct basement membranes (BM) around different vascular cell types. Astrocyte endfeet that are localised against the walls of blood vessels are tethered to these BMs by dystrophin associated protein complex (DPC). Alpha-dystrobrevin (α-DB) is a key dystrophin associated protein within perivascular astrocyte endfeet; its deficiency leads to a reduction in other dystrophin associated proteins, loss of AQP4 and altered ECM. In human dementia cohorts there is a positive correlation between dystrobrevin gene expression and CAA. In the present study, we test the hypotheses that (a) the positive correlation between dystrobrevin gene expression and CAA is associated with elevated expression of α-DB at glial-vascular endfeet and (b) a deficiency in α-DB results in changes to the ECM and failure of IPAD. We used human post-mortem brain tissue with different severities of CAA and transgenic α-DB deficient mice. In human post-mortem tissue we observed a significant increase in vascular α-DB with CAA (CAA vrs. Old p < 0.005, CAA vrs. Young p < 0.005). In the mouse model of α-DB deficiency, there was early modifications to vascular ECM (collagen IV and BM thickening) that translated into reduced IPAD efficiency. Our findings highlight the important role of α-DB in maintaining structure and function of ECM, particularly as a pathway for the flow of ISF and solutes out of the brain by IPAD.


Subject(s)
Cerebral Amyloid Angiopathy/metabolism , Cerebral Amyloid Angiopathy/pathology , Dystrophin-Associated Proteins/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Adult , Aged , Aged, 80 and over , Animals , Cerebrovascular Circulation/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
3.
Free Neuropathol ; 22021 Jan.
Article in English | MEDLINE | ID: mdl-37284630
4.
Pharmaceuticals (Basel) ; 13(9)2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32971843

ABSTRACT

Drainage of interstitial fluid from the brain occurs via the intramural periarterial drainage (IPAD) pathways along the basement membranes of cerebral capillaries and arteries against the direction of blood flow into the brain. The cerebrovascular smooth muscle cells (SMCs) provide the motive force for driving IPAD, and their decrease in function may explain the deposition of amyloid-beta as cerebral amyloid angiopathy (CAA), a key feature of Alzheimer's disease. The α-adrenoceptor subtype α1A is abundant in the brain, but its distribution in the cerebral vessels is unclear. We analysed cultured human cerebrovascular SMCs and young, old and CAA human brains for (a) the presence of α1A receptor and (b) the distribution of the α1A receptor within the cerebral vessels. The α1A receptor was present on the wall of cerebrovascular SMCs. No significant changes were observed in the vascular expression of the α1A-adrenergic receptor in young, old and CAA cases. The pattern of vascular staining appeared less punctate and more diffuse with ageing and CAA. Our results show that the α1A-adrenergic receptor is preserved in cerebral vessels with ageing and in CAA and is expressed on cerebrovascular smooth muscle cells, suggesting that vascular adrenergic receptors may hold potential for therapeutic targeting of IPAD.

5.
Alzheimers Dement (Amst) ; 12(1): e12070, 2020.
Article in English | MEDLINE | ID: mdl-32782922

ABSTRACT

INTRODUCTION: Accumulation of amyloid beta (Aß), α-synuclein (αSyn), and tau in dementias indicates their age-related failure of elimination from the brain. Aß is eliminated along basement membranes in walls of cerebral arterioles and leptomeningeal arteries (intramural peri-arterial drainage [IPAD]); IPAD is impaired with age. We test the hypothesis that αSyn and tau are also eliminated from the normal brain along IPAD pathways. METHODS: Soluble αSyn or tau was injected into mouse hippocampus. Animals were perfused 5 minutes to 7 days post-injection. Blood vessels were identified by ROX-SE for light-sheet and immunolabeling for confocal microscopy. IPAD was quantified by measuring the proportion of arterioles with αSyn/tau. RESULTS: αSyn and tau are eliminated from the brain by IPAD but with different dynamics. DISCUSSION: Age-related failure of IPAD may play a role in the pathogenesis of synucleinopathies and tauopathies. αSyn persists within IPAD at 24 hours, which may affect immunotherapy for αSyn.

6.
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.

7.
Acta Neuropathol Commun ; 8(1): 131, 2020 08 08.
Article in English | MEDLINE | ID: mdl-32771063

ABSTRACT

White matter hyperintensities (WMH) occur in association with dementia but the aetiology is unclear. Here we test the hypothesis that there is a combination of impaired elimination of interstitial fluid from the white matter together with a degree of hypoxia in WMH. One of the mechanisms for the elimination of amyloid-ß (Aß) from the brain is along the basement membranes in the walls of capillaries and arteries (Intramural Peri-Arterial Drainage - IPAD). We compared the dynamics of IPAD in the grey matter of the hippocampus and in the white matter of the corpus callosum in 10 week old C57/B16 mice by injecting soluble Aß as a tracer. The dynamics of IPAD in the white matter were significantly slower compared with the grey matter and this was associated with a lower density of capillaries in the white matter. Exposing cultures of smooth muscle cells to hypercapnia as a model of cerebral hypoperfusion resulted in a reduction in fibronectin and an increase in laminin in the extracellular matrix. Similar changes were detected in the white matter in human WMH suggesting that hypercapnia/hypoxia may play a role in WMH. Employing therapies to enhance both IPAD and blood flow in the white matter may reduce WMH in patients with dementia.


Subject(s)
Dementia/pathology , Extracellular Fluid/metabolism , Hypoxia, Brain/pathology , Muscle, Smooth, Vascular/metabolism , White Matter/pathology , Aged , Aged, 80 and over , Aging/pathology , Amyloid beta-Peptides/metabolism , Animals , Female , Fibronectins/metabolism , Glymphatic System/pathology , Humans , Laminin/metabolism , Male , Mice , Mice, Inbred C57BL
8.
Brain ; 143(7): 1975-1998, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32408345

ABSTRACT

Interest in neurofilaments has risen sharply in recent years with recognition of their potential as biomarkers of brain injury or neurodegeneration in CSF and blood. This is in the context of a growing appreciation for the complexity of the neurobiology of neurofilaments, new recognition of specialized roles for neurofilaments in synapses and a developing understanding of mechanisms responsible for their turnover. Here we will review the neurobiology of neurofilament proteins, describing current understanding of their structure and function, including recently discovered evidence for their roles in synapses. We will explore emerging understanding of the mechanisms of neurofilament degradation and clearance and review new methods for future elucidation of the kinetics of their turnover in humans. Primary roles of neurofilaments in the pathogenesis of human diseases will be described. With this background, we then will review critically evidence supporting use of neurofilament concentration measures as biomarkers of neuronal injury or degeneration. Finally, we will reflect on major challenges for studies of the neurobiology of intermediate filaments with specific attention to identifying what needs to be learned for more precise use and confident interpretation of neurofilament measures as biomarkers of neurodegeneration.


Subject(s)
Biomarkers , Intermediate Filaments , Nerve Degeneration , Synapses , Animals , Humans
9.
J Magn Reson Imaging ; 52(5): 1293-1305, 2020 11.
Article in English | MEDLINE | ID: mdl-32246802

ABSTRACT

The unexpected appearance of T1 hyperintensities, mostly in the dentate nucleus and the globus pallidus, during nonenhanced MRI was reported in 2014. This effect is associated with prior repeated administrations of gadolinium (Gd)-based contrast agents (GBCAs) in patients with a functional blood-brain barrier (BBB). It is widely assumed that GBCAs do not cross the intact BBB, but the observation of these hypersignals raises questions regarding this assumption. This review critically discusses the mechanisms of Gd accumulation in the brain with regard to access pathways, Gd species, tissue distribution, and subcellular location. We propose the hypothesis that there is early access of Gd species to cerebrospinal fluid, followed by passive diffusion into the brain parenchyma close to the cerebral ventricles. When accessing areas rich in endogenous metals or phosphorus, the less kinetically stable GBCAs would dissociate, and Gd would bind to endogenous macromolecules, and/or precipitate within the brain tissue. It is also proposed that Gd species enter the brain parenchyma along penetrating cortical arteries in periarterial pial-glial basement membranes and leave the brain along intramural peri-arterial drainage (IPAD) pathways. Lastly, Gd/GBCAs may access the brain parenchyma directly from the blood through the BBB in the walls of capillaries. It is crucial to distinguish between the physiological distribution and drainage pathways for GBCAs and the possible dissociation of less thermodynamically/kinetically stable GBCAs that lead to long-term Gd deposition in the brain. LEVEL OF EVIDENCE: 5. TECHNICAL EFFICACY STAGE: 3.


Subject(s)
Brain , Gadolinium , Brain/diagnostic imaging , Contrast Media , Gadolinium DTPA , Humans , Magnetic Resonance Imaging
10.
Int J Mol Sci ; 21(4)2020 Feb 12.
Article in English | MEDLINE | ID: mdl-32059400

ABSTRACT

In the absence of lymphatics, fluid and solutes such as amyloid-ß (Aß) are eliminated from the brain along basement membranes in the walls of cerebral capillaries and arteries-the Intramural Peri-Arterial Drainage (IPAD) pathway. IPAD fails with age and insoluble Aß is deposited as plaques in the brain and in IPAD pathways as cerebral amyloid angiopathy (CAA); fluid accumulates in the white matter as reflected by hyperintensities (WMH) on MRI. Within the brain, fluid uptake by astrocytes is regulated by aquaporin 4 (AQP4). We test the hypothesis that expression of astrocytic AQP4 increases in grey matter and decreases in white matter with onset of CAA. AQP4 expression was quantitated by immunocytochemistry and confocal microscopy in post-mortem occipital grey and white matter from young and old non-demented human brains, in CAA and in WMH. Results: AQP4 expression tended to increase with normal ageing but AQP4 expression in severe CAA was significantly reduced when compared to moderate CAA (p = 0.018). AQP4 expression tended to decline in the white matter with CAA and WMH, both of which are associated with impaired IPAD. Adjusting the level of AQP4 activity may be a valid therapeutic target for restoring homoeostasis in the brain as IPAD fails with age and CAA.


Subject(s)
Aging/metabolism , Aquaporin 4/metabolism , Brain/metabolism , Cerebral Amyloid Angiopathy/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Aquaporin 4/genetics , Astrocytes/metabolism , Brain/diagnostic imaging , Brain/pathology , Cerebral Amyloid Angiopathy/diagnostic imaging , Cerebral Amyloid Angiopathy/genetics , Cerebral Amyloid Angiopathy/pathology , Gray Matter/metabolism , Humans , Magnetic Resonance Imaging , Middle Aged , White Matter/metabolism
11.
Neuron ; 105(3): 400-401, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32027828

ABSTRACT

In this issue of Neuron, van Veluw et al. (2020) show that elimination of solutes from the brain along arterial walls is driven by low-frequency arteriolar oscillations and suggest that age-related reduction of this vasomotion may contribute to impaired clearance of Aß.


Subject(s)
Brain , Wakefulness , Animals , Arterioles , Drainage , Mice
12.
Acta Neuropathol ; 139(2): 383-401, 2020 02.
Article in English | MEDLINE | ID: mdl-31696318

ABSTRACT

The vertebrate CNS is surrounded by the meninges, a protective barrier comprised of the outer dura mater and the inner leptomeninges, which includes the arachnoid and pial layers. While the dura mater contains lymphatic vessels, no conventional lymphatics have been found within the brain or leptomeninges. However, non-lumenized cells called Brain/Mural Lymphatic Endothelial Cells or Fluorescent Granule Perithelial cells (muLECs/BLECs/FGPs) that share a developmental program and gene expression with peripheral lymphatic vessels have been described in the meninges of zebrafish. Here we identify a structurally and functionally similar cell type in the mammalian leptomeninges that we name Leptomeningeal Lymphatic Endothelial Cells (LLEC). As in zebrafish, LLECs express multiple lymphatic markers, containing very large, spherical inclusions, and develop independently from the meningeal macrophage lineage. Mouse LLECs also internalize macromolecules from the cerebrospinal fluid, including Amyloid-ß, the toxic driver of Alzheimer's disease progression. Finally, we identify morphologically similar cells co-expressing LLEC markers in human post-mortem leptomeninges. Given that LLECs share molecular, morphological, and functional characteristics with both lymphatics and macrophages, we propose they represent a novel, evolutionary conserved cell type with potential roles in homeostasis and immune organization of the meninges.


Subject(s)
Brain/pathology , Endothelial Cells/pathology , Endothelial Cells/physiology , Lymphatic System/pathology , Meninges/pathology , Adult , Aged , Aged, 80 and over , Amyloid beta-Peptides , Animals , Female , Humans , Male , Mice , Zebrafish
13.
Front Aging Neurosci ; 11: 1, 2019.
Article in English | MEDLINE | ID: mdl-30740048

ABSTRACT

The human brain is the organ with the highest metabolic activity but it lacks a traditional lymphatic system responsible for clearing waste products. We have demonstrated that the basement membranes of cerebral capillaries and arteries represent the lymphatic pathways of the brain along which intramural periarterial drainage (IPAD) of soluble metabolites occurs. Failure of IPAD could explain the vascular deposition of the amyloid-beta protein as cerebral amyloid angiopathy (CAA), which is a key pathological feature of Alzheimer's disease. The underlying mechanisms of IPAD, including its motive force, have not been clarified, delaying successful therapies for CAA. Although arterial pulsations from the heart were initially considered to be the motive force for IPAD, they are not strong enough for efficient IPAD. This study aims to unravel the driving force for IPAD, by shifting the perspective of a heart-driven clearance of soluble metabolites from the brain to an intrinsic mechanism of cerebral arteries (e.g., vasomotion-driven IPAD). We test the hypothesis that the cerebrovascular smooth muscle cells, whose cycles of contraction and relaxation generate vasomotion, are the drivers of IPAD. A novel multiscale model of arteries, in which we treat the basement membrane as a fluid-filled poroelastic medium deformed by the contractile cerebrovascular smooth muscle cells, is used to test the hypothesis. The vasomotion-induced intramural flow rates suggest that vasomotion-driven IPAD is the only mechanism postulated to date capable of explaining the available experimental observations. The cerebrovascular smooth muscle cells could represent valuable drug targets for prevention and early interventions in CAA.

14.
PLoS One ; 13(10): e0205276, 2018.
Article in English | MEDLINE | ID: mdl-30286191

ABSTRACT

The mechanisms behind the clearance of soluble waste from deep within the parenchyma of the brain remain unclear. Experimental evidence reveals that one pathway for clearance of waste, termed intra-mural peri-arterial drainage (IPAD), is the rapid drainage of interstitial fluid along basement membranes (BM) of the smooth muscle cells of cerebral arteries; failure of IPAD is closely associated with the pathology of Alzheimer's disease (AD), but its driving mechanism remains unclear. We have previously shown that arterial pulsations generated by the heart beat are not strong enough to drive IPAD. Here we present computational evidence for a mechanism for clearance of waste from the brain that is driven by functional hyperaemia, that is, the dilatation of cerebral arterioles as a consequence of increased nutrient demand from neurons. This mechanism is based on our model for the flow of fluid through the vascular BM. It accounts for clearance rates observed in mouse experiments, and aligns with pathological observations and recommendations to lower the individual risk of AD, such as mental and physical activity. Thus, our neurovascular hypothesis should act as the new working hypothesis for the driving force behind IPAD.


Subject(s)
Alzheimer Disease/therapy , Brain/pathology , Cerebral Amyloid Angiopathy , Drainage/methods , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Capillaries/metabolism , Capillaries/pathology , Cerebral Arteries/metabolism , Cerebral Arteries/pathology , Extracellular Fluid/metabolism , Humans , Mice , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neurons
15.
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
16.
Acta Neuropathol ; 135(3): 363-385, 2018 03.
Article in English | MEDLINE | ID: mdl-29368214

ABSTRACT

Meninges that surround the CNS consist of an outer fibrous sheet of dura mater (pachymeninx) that is also the inner periosteum of the skull. Underlying the dura are the arachnoid and pia mater (leptomeninges) that form the boundaries of the subarachnoid space. In this review we (1) examine the development of leptomeninges and their role as barriers and facilitators in the foetal CNS. There are two separate CSF systems during early foetal life, inner CSF in the ventricles and outer CSF in the subarachnoid space. As the foramina of Magendi and Luschka develop, one continuous CSF system evolves. Due to the lack of arachnoid granulations during foetal life, it is most likely that CSF is eliminated by lymphatic drainage pathways passing through the cribriform plate and nasal submucosa. (2) We then review the fine structure of the adult human and rodent leptomeninges to establish their roles as barriers and facilitators for the movement of fluid, cells and pathogens. Leptomeningeal cells line CSF spaces, including arachnoid granulations and lymphatic drainage pathways, and separate elements of extracellular matrix from the CSF. The leptomeningeal lining facilitates the traffic of inflammatory cells within CSF but also allows attachment of bacteria such as Neisseria meningitidis and of tumour cells as CSF metastases. Single layers of leptomeningeal cells extend into the brain closely associated with the walls of arteries so that there are no perivascular spaces around arteries in the cerebral cortex. Perivascular spaces surrounding arteries in the white matter and basal ganglia relate to their two encompassing layers of leptomeninges. (3) Finally we examine the roles of ligands expressed by leptomeningeal cells for the attachment of inflammatory cells, bacteria and tumour cells as understanding these roles may aid the design of therapeutic strategies to manage developmental, autoimmune, infectious and neoplastic diseases relating to the CSF, the leptomeninges and the associated CNS.


Subject(s)
Meninges/cytology , Meninges/metabolism , Animals , Humans , Meninges/blood supply , Meninges/microbiology , Rodentia
17.
Clin Sci (Lond) ; 131(22): 2745-2752, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29021222

ABSTRACT

Although there are no conventional lymphatic vessels in the brain, fluid and solutes drain along basement membranes (BMs) of cerebral capillaries and arteries towards the subarachnoid space and cervical lymph nodes. Convective influx/glymphatic entry of the cerebrospinal fluid (CSF) into the brain parenchyma occurs along the pial-glial BMs of arteries. This project tested the hypotheses that pial-glial BM of arteries are thicker in the midbrain, allowing more glymphatic entry of CSF. The in vivo MRI and PET images were obtained from a 4.2-year-old dog, whereas the post-mortem electron microscopy was performed in a 12-year-old dog. We demonstrated a significant increase in the thickness of the pial-glial BM in the midbrain compared with the same BM in different regions of the brain and an increase in the convective influx of fluid from the subarachnoid space. These results are highly significant for the intrathecal drug delivery into the brain, indicating that the midbrain is better equipped for convective influx/glymphatic entry of the CSF.


Subject(s)
Cerebrospinal Fluid/metabolism , Mesencephalon/blood supply , Animals , Arteries/ultrastructure , Basement Membrane/ultrastructure , Dogs , Endothelium/ultrastructure , Magnetic Resonance Imaging , Mesencephalon/ultrastructure , Muscle, Smooth/ultrastructure , Neuroglia/ultrastructure , Pia Mater/ultrastructure , Positron-Emission Tomography , Time Factors
18.
Clin Sci (Lond) ; 131(22): 2737-2744, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28982724

ABSTRACT

Dilatation of periarteriolar spaces in MRI of the ageing human brains occurs in white matter (WM), basal ganglia and midbrain but not in cerebral cortex. Perivenous collagenous occurs in periventricular but not in subcortical WM.Here we test the hypotheses that (a) the capacity for dilatation of periarteriolar spaces correlates with the anatomical distribution of leptomeningeal cells coating intracerebral arteries and (b) the regional development of perivenous collagenous in the WM correlates with the population of intramural cells in the walls of veins.The anatomical distribution of leptomeningeal and intramural cells related to cerebral blood vessels is best documented by electron microscopy, requiring perfusion-fixed tissue not available in human material. We therefore analysed perfusion-fixed brain from a 12-year-old Beagle dog as the canine brain represents the anatomical arrangement in the human brain. Results showed regional variation in the arrangement of leptomeningeal cells around blood vessels. Arterioles are enveloped by one complete layer of leptomeninges often with a second incomplete layer in the WM. Venules showed incomplete layers of leptomeningeal cells. Intramural cell expression was higher in the post-capillary venules of the subcortical WM when compared with periventricular WM, suggesting that periventricular collagenosis around venules may be due to a lower resistance in the venular walls. It appears that the regional variation in the capacity for dilatation of arteriolar perivascular spaces in the white WM may be related to the number of perivascular leptomeningeal cells surrounding vessels in different areas of the brain.


Subject(s)
Aging/physiology , Brain/anatomy & histology , Brain/blood supply , Animals , Arterioles/cytology , Arterioles/ultrastructure , Brain/cytology , Dogs , White Matter/anatomy & histology , White Matter/blood supply
19.
Nat Immunol ; 18(2): 123-131, 2017 02.
Article in English | MEDLINE | ID: mdl-28092374

ABSTRACT

Discoveries leading to an improved understanding of immune surveillance of the central nervous system (CNS) have repeatedly provoked dismissal of the existence of immune privilege of the CNS. Recent rediscoveries of lymphatic vessels within the dura mater surrounding the brain, made possible by modern live-cell imaging technologies, have revived this discussion. This review emphasizes the fact that understanding immune privilege of the CNS requires intimate knowledge of its unique anatomy. Endothelial, epithelial and glial brain barriers establish compartments in the CNS that differ strikingly with regard to their accessibility to immune-cell subsets. There is a unique system of lymphatic drainage from the CNS to the peripheral lymph nodes. We summarize current knowledge on the cellular and molecular mechanisms involved in immune-cell trafficking and lymphatic drainage from the CNS, and we take into account differences in rodent and human CNS anatomy.


Subject(s)
Central Nervous System/immunology , Dura Mater/immunology , Immune Tolerance , Microglia/immunology , Physiology, Comparative , Animals , Autoimmunity , Cell Movement/immunology , Central Nervous System/anatomy & histology , Humans , Immunologic Surveillance , Mice , Rats
20.
Methods Mol Biol ; 1559: 343-365, 2017.
Article in English | MEDLINE | ID: mdl-28063056

ABSTRACT

In this chapter we describe in detail the surgical and imaging techniques employed for the study of the anatomical routes of drainage of cerebrospinal fluid (CSF) and interstitial fluid (ISF) from the brain. The types of tracers, sites of injection, and volumes injected are crucial. For example, when testing the drainage of ISF from the parenchyma, volumes larger than 0.5 µL result in spillage of ISF into the ventricular CSF.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Extracellular Fluid/metabolism , Immunohistochemistry/methods , Lymphatic Vessels/ultrastructure , Microscopy, Fluorescence/methods , Stereotaxic Techniques , Alzheimer Disease/pathology , Animals , Cisterna Magna , Extracellular Fluid/chemistry , Fixatives/chemistry , Fluorescent Dyes/chemistry , Formaldehyde/chemistry , Hippocampus , Humans , Injections, Intraventricular , Lymphatic Vessels/metabolism , Mice , Parenchymal Tissue , Perfusion/methods , Polymers/chemistry , Tissue Embedding/methods , Xanthenes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...