201
|
Abstract
Neuroimmunologists seek to understand the interactions between the central nervous system (CNS) and the immune system, both under homeostatic conditions and in diseases. Unanswered questions include those relating to the diversity and specificity of the meningeal T cell repertoire; the routes taken by immune cells that patrol the meninges under healthy conditions and invade the parenchyma during pathology; the opposing effects (beneficial or detrimental) of these cells on CNS function; the role of immune cells after CNS injury; and the evolutionary link between the two systems, resulting in their tight interaction and interdependence. This Review summarizes the current standing of and challenging questions related to interactions between adaptive immunity and the CNS and considers the possible directions in which these aspects of neuroimmunology will be heading over the next decade.
Collapse
Affiliation(s)
- Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
202
|
Bhat NR. Vasculoprotection as a Convergent, Multi-Targeted Mechanism of Anti-AD Therapeutics and Interventions. J Alzheimers Dis 2016; 46:581-91. [PMID: 26402511 DOI: 10.3233/jad-150098] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Using a variety of animal models of Alzheimer's disease (AD), there have been a number of recent studies reporting varying degrees of success with anti-AD therapeutics. The efficacies are often discussed in terms of the modulatory effects of the compounds tested on identified or assumed targets among the known (or proposed) pathogenic and neuroprotective mechanisms, largely within the context of the dominant amyloid cascade hypothesis. However, it is clear that several of the relatively more efficacious treatments tend to be multifunctional and target multiple pathological processes associated with AD including most commonly, oxidative and metabolic stress and neuroinflammation. Increasing evidence suggests that vascular and neurodegenerative pathologies often co-exist and that neurovascular dysfunction plays a critical role in the development or progression of AD. In this review, we will discuss the significance of vasculoprotection or neurovascular unit integrity as a common, multi-targeted mechanism underlying the reported efficacy of a majority of anti-AD therapeutics--amyloid-targeted or otherwise--while providing a strong support for future neurovascular-based treatment strategies and interventions.
Collapse
|
203
|
Roth C, Stitz H, Roth C, Ferbert A, Deinsberger W, Pahl R, Engel H, Kleffmann J. Craniocervical manual lymphatic drainage and its impact on intracranial pressure - a pilot study. Eur J Neurol 2016; 23:1441-6. [PMID: 27238738 DOI: 10.1111/ene.13055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 04/21/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND PURPOSE Theoretical considerations and the results of animal studies indicate that manual lymphatic drainage (MLD) might have an impact on intracranial pressure (ICP). There is a lack of clinically qualitative investigations on patients with severe cerebral diseases. METHODS Between April 2013 and January 2015 a prospective observational study was performed on patients who were undergoing intracranial pressure measurement and treatment with MLD. ICP, cerebral perfusion pressure, mean arterial pressure (MAP), heart rate and oxygen saturation were recorded continuously 15 min before the procedure, during MLD (22 min) and for 15 min after the procedure. For analysis the data treatment units were divided into two groups: patients with a mean baseline ICP <15 mmHg (group 1) and patients with a mean ICP ≥15 mmHg before MLD (group 2). RESULTS A total of 133 treatment units (61 patients) were analysed (group 1 n = 99; group 2 n = 34). The mean baseline ICP was 10.4 mmHg overall, and 8.3 mmHg and 18.6 mmHg respectively in group 1 and group 2; ICP significantly decreased during therapy with MLD and this persisted during the follow-up period in group 2. MAP did not show any significant differences between the different periods. CONCLUSIONS Our data showed a significant reduction of ICP during therapy with craniocervical MLD in patients with severe cerebral diseases.
Collapse
Affiliation(s)
- C Roth
- Department of Neurology, Klinikum Kassel, Kassel, Germany
| | - H Stitz
- Department of Physiotherapy, Klinikum Kassel, Kassel, Germany
| | - C Roth
- Department of Physiotherapy, Klinikum Kassel, Kassel, Germany
| | - A Ferbert
- Department of Neurology, Klinikum Kassel, Kassel, Germany
| | - W Deinsberger
- Department of Neurosurgery, Klinikum Kassel, Kassel, Germany
| | - R Pahl
- Institute of Medical Biometry and Epidemiology (IMBE), Philipps University Marburg, Marburg, Germany
| | - H Engel
- Department of Hand, Plastic and Reconstructive Surgery, Klinikum Kassel, Kassel, Germany
| | - J Kleffmann
- Department of Neurosurgery, Klinikum Kassel, Kassel, Germany
| |
Collapse
|
204
|
Buss L, Fisher E, Hardy J, Nizetic D, Groet J, Pulford L, Strydom A. Intracerebral haemorrhage in Down syndrome: protected or predisposed? F1000Res 2016; 5. [PMID: 27239286 PMCID: PMC4870990 DOI: 10.12688/f1000research.7819.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2016] [Indexed: 01/08/2023] Open
Abstract
Down syndrome (DS), which arises from trisomy of chromosome 21, is associated with deposition of large amounts of amyloid within the central nervous system. Amyloid accumulates in two compartments: as plaques within the brain parenchyma and in vessel walls of the cerebral microvasculature. The parenchymal plaque amyloid is thought to result in an early onset Alzheimer’s disease (AD) dementia, a phenomenon so common amongst people with DS that it could be considered a defining feature of the condition. The amyloid precursor protein (
APP) gene lies on chromosome 21 and its presence in three copies in DS is thought to largely drive the early onset AD. In contrast, intracerebral haemorrhage (ICH), the main clinical consequence of vascular amyloidosis, is a more poorly defined feature of DS. We review recent epidemiological data on stroke (including haemorrhagic stroke) in order to make comparisons with a rare form of familial AD due to duplication (i.e. having three copies) of the
APP region on chromosome 21, here called ‘dup-APP’, which is associated with more frequent and severe ICH. We conclude that although people with DS are at increased risk of ICH, this is less common than in dup-APP, suggesting the presence of mechanisms that act protectively. We review these mechanisms and consider comparative research into DS and dup-APP that may yield further pathophysiological insight.
Collapse
Affiliation(s)
- Lewis Buss
- Division of Psychiatry, University College London, London, UK; London Down Syndrome (LonDownS) Consortium, University College London, London, UK
| | - Elizabeth Fisher
- Institute of Neurology, University College London, London, UK; London Down Syndrome (LonDownS) Consortium, University College London, London, UK
| | - John Hardy
- Institute of Neurology, University College London, London, UK; London Down Syndrome (LonDownS) Consortium, University College London, London, UK
| | - Dean Nizetic
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Blizard Institute, Barts and the London School of Medicine, Queen Mary, University of London, London, UK; London Down Syndrome (LonDownS) Consortium, University College London, London, UK
| | - Jurgen Groet
- Blizard Institute, Barts and the London School of Medicine, Queen Mary, University of London, London, UK; London Down Syndrome (LonDownS) Consortium, University College London, London, UK
| | - Laura Pulford
- Institute of Neurology, University College London, London, UK; London Down Syndrome (LonDownS) Consortium, University College London, London, UK
| | - André Strydom
- Division of Psychiatry, University College London, London, UK; London Down Syndrome (LonDownS) Consortium, University College London, London, UK
| |
Collapse
|
205
|
Manukhina EB, Downey HF, Shi X, Mallet RT. Intermittent hypoxia training protects cerebrovascular function in Alzheimer's disease. Exp Biol Med (Maywood) 2016; 241:1351-63. [PMID: 27190276 DOI: 10.1177/1535370216649060] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a leading cause of death and disability among older adults. Modifiable vascular risk factors for AD (VRF) include obesity, hypertension, type 2 diabetes mellitus, sleep apnea, and metabolic syndrome. Here, interactions between cerebrovascular function and development of AD are reviewed, as are interventions to improve cerebral blood flow and reduce VRF. Atherosclerosis and small vessel cerebral disease impair metabolic regulation of cerebral blood flow and, along with microvascular rarefaction and altered trans-capillary exchange, create conditions favoring AD development. Although currently there are no definitive therapies for treatment or prevention of AD, reduction of VRFs lowers the risk for cognitive decline. There is increasing evidence that brief repeated exposures to moderate hypoxia, i.e. intermittent hypoxic training (IHT), improve cerebral vascular function and reduce VRFs including systemic hypertension, cardiac arrhythmias, and mental stress. In experimental AD, IHT nearly prevented endothelial dysfunction of both cerebral and extra-cerebral blood vessels, rarefaction of the brain vascular network, and the loss of neurons in the brain cortex. Associated with these vasoprotective effects, IHT improved memory and lessened AD pathology. IHT increases endothelial production of nitric oxide (NO), thereby increasing regional cerebral blood flow and augmenting the vaso- and neuroprotective effects of endothelial NO. On the other hand, in AD excessive production of NO in microglia, astrocytes, and cortical neurons generates neurotoxic peroxynitrite. IHT enhances storage of excessive NO in the form of S-nitrosothiols and dinitrosyl iron complexes. Oxidative stress plays a pivotal role in the pathogenesis of AD, and IHT reduces oxidative stress in a number of experimental pathologies. Beneficial effects of IHT in experimental neuropathologies other than AD, including dyscirculatory encephalopathy, ischemic stroke injury, audiogenic epilepsy, spinal cord injury, and alcohol withdrawal stress have also been reported. Further research on the potential benefits of IHT in AD and other brain pathologies is warranted.
Collapse
Affiliation(s)
- Eugenia B Manukhina
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA Institute of General Pathology and Pathophysiology, Moscow 125315, Russian Federation
| | - H Fred Downey
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Xiangrong Shi
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| | - Robert T Mallet
- University of North Texas Health Science Center, Fort Worth, TX 76107-2699, USA
| |
Collapse
|
206
|
Deckert M, Brunn A, Montesinos-Rongen M, Siebert R. Absence of Lymphatic Vessels in PCNSL May Contribute to Confinement of Tumor Cells to the Central Nervous System. J Neuropathol Exp Neurol 2016; 75:499-502. [PMID: 27142645 DOI: 10.1093/jnen/nlw027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/01/2016] [Indexed: 12/25/2022] Open
Abstract
Primary central nervous system (CNS) lymphoma (PCNSL) is a mature lymphoma of the diffuse large B-cell lymphoma (DLBCL) type confined to the CNS. Despite cytomorphological similarities between PCNSL and systemic DLBCL, molecular differences between both entities have been identified. The exclusively topographical restriction of PCNSL to the CNS is an unexplained mystery. To address the question of whether the unique lymphatic drainage system of the CNS, which differs from that of other organs, may play a role for this peculiar behavior, we investigated a series of 20 PCNSLs for the presence of lymphatic vessels by immunohistochemistry for Lyve-1, podoplanin, and Prox-1 expression. All PCNSLs lacked lymphatic vessels and, in this regard, were similar to 20 glioblastoma multiforme samples. In contrast to these tumors, all of which were located in the deep brain parenchyma, dural and meningeal DLBCL harbored lymphatic vessels that expressed Lyve-1 (3/8 tumors), podoplanin (5/8 tumors), and Prox-1 (5/8 tumors) in areas where the tumors had invaded the fibrous tissue of the dura. These data indicate that local topographical characteristics of the specific lymphatic drainage system may contribute to confinement of the tumor cells in PCNSL and malignant gliomas.
Collapse
Affiliation(s)
- Martina Deckert
- From the Department of Neuropathology, University Hospital of Cologne, Cologne, Germany (MD, AB, MMR); and Institute of Human Genetics, Christian-Albrechts-University Kiel, Kiel, Germany (RS)
| | - Anna Brunn
- From the Department of Neuropathology, University Hospital of Cologne, Cologne, Germany (MD, AB, MMR); and Institute of Human Genetics, Christian-Albrechts-University Kiel, Kiel, Germany (RS)
| | - Manuel Montesinos-Rongen
- From the Department of Neuropathology, University Hospital of Cologne, Cologne, Germany (MD, AB, MMR); and Institute of Human Genetics, Christian-Albrechts-University Kiel, Kiel, Germany (RS)
| | - Reiner Siebert
- From the Department of Neuropathology, University Hospital of Cologne, Cologne, Germany (MD, AB, MMR); and Institute of Human Genetics, Christian-Albrechts-University Kiel, Kiel, Germany (RS)
| |
Collapse
|
207
|
Tau pathology-dependent remodelling of cerebral arteries precedes Alzheimer's disease-related microvascular cerebral amyloid angiopathy. Acta Neuropathol 2016; 131:737-52. [PMID: 26988843 PMCID: PMC4835519 DOI: 10.1007/s00401-016-1560-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 03/05/2016] [Accepted: 03/06/2016] [Indexed: 01/18/2023]
Abstract
Alzheimer’s disease (AD) is characterised by pathologic cerebrovascular remodelling. Whether this occurs already before disease onset, as may be indicated by early Braak tau-related cerebral hypoperfusion and blood–brain barrier (BBB) impairment found in previous studies, remains unknown. Therefore, we systematically quantified Braak tau stage- and cerebral amyloid angiopathy (CAA)-dependent alterations in the alpha-smooth muscle actin (α-SMA), collagen, and elastin content of leptomeningeal arterioles, small arteries, and medium-sized arteries surrounding the gyrus frontalis medialis (GFM) and hippocampus (HIPP), including the sulci, of 17 clinically and pathologically diagnosed AD subjects (Braak stage IV–VI) and 28 non-demented control subjects (Braak stage I–IV). GFM and HIPP paraffin sections were stained for general collagen and elastin with the Verhoeff–van Gieson stain; α-SMA and CAA/amyloid β (Aβ) were detected using immunohistochemistry. Significant arterial elastin degradation was observed from Braak stage III onward and correlated with Braak tau pathology (ρ = 0.909, 95 % CI 0.370 to 0.990, p < 0.05). This was accompanied by an increase in neutrophil elastase expression by α-SMA-positive cells in the vessel wall. Small and medium-sized arteries exhibited significant CAA-independent α-SMA loss starting between Braak stage I and II–III, along with accumulation of phosphorylated paired helical filament (PHF) tau in the perivascular space of intraparenchymal vessels. α-SMA remained at the decreased level throughout the later Braak stages. In contrast, arterioles exhibited significant α-SMA loss only at Braak stage V and VI/in AD subjects, which was CAA-dependent/correlated with CAA burden (ρ = −0.422, 95 % CI −0.557 to −0.265, p < 0.0001). Collagen content was only significantly changed in small arteries. Our data indicate that vessel wall remodelling of leptomeningeal arteries is an early-onset, Braak tau pathology-dependent process unrelated to CAA and AD, which potentially may contribute to downstream CAA-dependent microvascular pathology in AD.
Collapse
|
208
|
Nelson AR, Sweeney MD, Sagare AP, Zlokovic BV. Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:887-900. [PMID: 26705676 PMCID: PMC4821735 DOI: 10.1016/j.bbadis.2015.12.016] [Citation(s) in RCA: 404] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/10/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023]
Abstract
Vascular insults can initiate a cascade of molecular events leading to neurodegeneration, cognitive impairment, and dementia. Here, we review the cellular and molecular mechanisms in cerebral blood vessels and the pathophysiological events leading to cerebral blood flow dysregulation and disruption of the neurovascular unit and the blood-brain barrier, which all may contribute to the onset and progression of dementia and Alzheimer's disease (AD). Particularly, we examine the link between neurovascular dysfunction and neurodegeneration including the effects of AD genetic risk factors on cerebrovascular functions and clearance of Alzheimer's amyloid-β peptide toxin, and the impact of vascular risk factors, environment, and lifestyle on cerebral blood vessels, which in turn may affect synaptic, neuronal, and cognitive functions. Finally, we examine potential experimental treatments for dementia and AD based on the neurovascular model, and discuss some critical questions to be addressed by future studies. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Amy R Nelson
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Melanie D Sweeney
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Abhay P Sagare
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics and the Zilkha Neurogenetic Institute, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
209
|
Lowenstein PR, Castro MG. The Long and Winding Road: From the High-Affinity Choline Uptake Site to Clinical Trials for Malignant Brain Tumors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 76:147-73. [PMID: 27288077 DOI: 10.1016/bs.apha.2016.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malignant brain tumors are one of the most lethal cancers. They originate from glial cells which infiltrate throughout the brain. Current standard of care involves surgical resection, radiotherapy, and chemotherapy; median survival is currently ~14-20 months postdiagnosis. Given that the brain immune system is deficient in priming systemic immune responses to glioma antigens, we proposed to reconstitute the brain immune system to achieve immunological priming from within the brain. Two adenoviral vectors are injected into the resection cavity or remaining tumor. One adenoviral vector expresses the HSV-1-derived thymidine kinase which converts ganciclovir into a compound only cytotoxic to dividing glioma cells. The second adenovirus expresses the cytokine fms-like tyrosine kinase 3 ligand (Flt3L). Flt3L differentiates precursors into dendritic cells and acts as a chemokine that attracts dendritic cells to the brain. HSV-1/ganciclovir killing of tumor cells releases tumor antigens that are taken up by dendritic cells within the brain tumor microenvironment. Tumor killing also releases HMGB1, an endogenous TLR2 agonist that activates dendritic cells. HMGB1-activated dendritic cells, loaded with glioma antigens, migrate to cervical lymph nodes to stimulate a systemic CD8+ T cells cytotoxic immune response against glioma. This immune response is specific to glioma tumors, induces immunological memory, and does neither cause brain toxicity nor autoimmune responses. An IND was granted by the FDA on 4/7/2011. A Phase I, first in person trial, to test whether reengineering the brain immune system is potentially therapeutic is ongoing.
Collapse
Affiliation(s)
- P R Lowenstein
- The Medical School, The University of Michigan, Ann Arbor, MI, United States.
| | - M G Castro
- The Medical School, The University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
210
|
Solomos AC, Rall GF. Get It through Your Thick Head: Emerging Principles in Neuroimmunology and Neurovirology Redefine Central Nervous System "Immune Privilege". ACS Chem Neurosci 2016; 7:435-41. [PMID: 26854733 DOI: 10.1021/acschemneuro.5b00336] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The central nervous system (CNS) coordinates all aspects of life, autonomic and sentient, though how it has evolved to contend with pathogenic infections remains, to a great degree, a mystery. The skull and cerebrospinal fluid (CSF) provide protection from blunt force contacts, and it was once thought that the blood-brain barrier (BBB) was a fortress that restricted pathogen entry and limited inflammation. Recent studies, however, have caused a revision of this viewpoint: the CNS is monitored by blood-borne lymphocytes, but can use alternative strategies to prevent or resolve many pathogenic challenges. In this Review, we discuss emerging principles that indicate how the CNS is immunologically unique from peripheral tissues. We focus on developments that include glymphatics, recently characterized brain lymphatic vessels, distinctions in innate and adaptive immune strategies, novel points of entry for neurotropic viruses, and, finally, how the periphery can influence CNS homeostasis and immune responses within the brain. Collectively, these attributes demand a re-evaluation of immunity in the brain: not privileged, but distinct.
Collapse
Affiliation(s)
- Andreas C. Solomos
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, Pennsylvania 19111, United States
- Department
of Microbiology and Immunology, Drexel University College of Medicine, 2900 W Queen Ln, Philadelphia, Pennsylvania 19129, United States
| | - Glenn F. Rall
- Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, Pennsylvania 19111, United States
| |
Collapse
|
211
|
Ramirez J, McNeely AA, Berezuk C, Gao F, Black SE. Dynamic Progression of White Matter Hyperintensities in Alzheimer's Disease and Normal Aging: Results from the Sunnybrook Dementia Study. Front Aging Neurosci 2016; 8:62. [PMID: 27047377 PMCID: PMC4805606 DOI: 10.3389/fnagi.2016.00062] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/10/2016] [Indexed: 12/03/2022] Open
Abstract
Although white matter hyperintensities (WMH), markers of cerebral small vessel disease (SVD), are believed to generally increase over time, some studies have shown sharp decreases after therapeutic intervention, suggesting that WMH progression may be more dynamic than previously thought. Our primary goal was to examine dynamic progression of WMH in a real-world sample of Alzheimer’s disease (AD) patients and normal elderly (NC), with varying degrees of SVD. WMH volumes from serial magnetic resonance imaging (MRI; mean = 1.8 years) were measured from NC (n = 44) and AD patients (n = 113) with high and low SVD burden. Dynamic progression for each individual was measured using spatial overlap images to assess shrinkage, growth, and stable WMH volumes. Significant group differences were found for shrinkage (p < 0.001), growth (p < 0.001) and stable (p < 0.001) WMH, where the AD high SVD group showed the largest changes relative to low SVD and NC. Our results suggest spatial progression measured at the individual patient level may be more sensitive to the dynamic nature of WMH.
Collapse
Affiliation(s)
- Joel Ramirez
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research InstituteToronto, ON, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences CentreToronto, ON, Canada
| | - Alicia A McNeely
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research InstituteToronto, ON, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences CentreToronto, ON, Canada
| | - Courtney Berezuk
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research InstituteToronto, ON, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences CentreToronto, ON, Canada; Graduate Department of Psychological Clinical Science, University of Toronto ScarboroughToronto, ON, Canada
| | - Fuqiang Gao
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research InstituteToronto, ON, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences CentreToronto, ON, Canada
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research InstituteToronto, ON, Canada; Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences CentreToronto, ON, Canada; Graduate Department of Psychological Clinical Science, University of Toronto ScarboroughToronto, ON, Canada; Faculty of Medicine, School of Graduate Studies, University of TorontoToronto, ON, Canada; Department of Medicine, Neurology, University of Toronto and Sunnybrook Health Sciences CentreToronto, ON, Canada
| |
Collapse
|
212
|
Lymphatic Clearance of the Brain: Perivascular, Paravascular and Significance for Neurodegenerative Diseases. Cell Mol Neurobiol 2016; 36:181-94. [PMID: 26993512 PMCID: PMC4844641 DOI: 10.1007/s10571-015-0273-8] [Citation(s) in RCA: 274] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/18/2015] [Indexed: 01/04/2023]
Abstract
The lymphatic clearance pathways of the brain are different compared to the other organs of the body and have been the subject of heated debates. Drainage of brain extracellular fluids, particularly interstitial fluid (ISF) and cerebrospinal fluid (CSF), is not only important for volume regulation, but also for removal of waste products such as amyloid beta (Aβ). CSF plays a special role in clinical medicine, as it is available for analysis of biomarkers for Alzheimer's disease. Despite the lack of a complete anatomical and physiological picture of the communications between the subarachnoid space (SAS) and the brain parenchyma, it is often assumed that Aβ is cleared from the cerebral ISF into the CSF. Recent work suggests that clearance of the brain mainly occurs during sleep, with a specific role for peri- and para-vascular spaces as drainage pathways from the brain parenchyma. However, the direction of flow, the anatomical structures involved and the driving forces remain elusive, with partially conflicting data in literature. The presence of Aβ in the glia limitans in Alzheimer's disease suggests a direct communication of ISF with CSF. Nonetheless, there is also the well-described pathology of cerebral amyloid angiopathy associated with the failure of perivascular drainage of Aβ. Herein, we review the role of the vasculature and the impact of vascular pathology on the peri- and para-vascular clearance pathways of the brain. The different views on the possible routes for ISF drainage of the brain are discussed in the context of pathological significance.
Collapse
|
213
|
Experimental Cerebral Malaria Spreads along the Rostral Migratory Stream. PLoS Pathog 2016; 12:e1005470. [PMID: 26964100 PMCID: PMC4786214 DOI: 10.1371/journal.ppat.1005470] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 02/03/2016] [Indexed: 12/25/2022] Open
Abstract
It is poorly understood how progressive brain swelling in experimental cerebral malaria (ECM) evolves in space and over time, and whether mechanisms of inflammation or microvascular sequestration/obstruction dominate the underlying pathophysiology. We therefore monitored in the Plasmodium berghei ANKA-C57BL/6 murine ECM model, disease manifestation and progression clinically, assessed by the Rapid-Murine-Coma-and-Behavioral-Scale (RMCBS), and by high-resolution in vivo MRI, including sensitive assessment of early blood-brain-barrier-disruption (BBBD), brain edema and microvascular pathology. For histological correlation HE and immunohistochemical staining for microglia and neuroblasts were obtained. Our results demonstrate that BBBD and edema initiated in the olfactory bulb (OB) and spread along the rostral-migratory-stream (RMS) to the subventricular zone of the lateral ventricles, the dorsal-migratory-stream (DMS), and finally to the external capsule (EC) and brainstem (BS). Before clinical symptoms (mean RMCBS = 18.5±1) became evident, a slight, non-significant increase of quantitative T2 and ADC values was observed in OB+RMS. With clinical manifestation (mean RMCBS = 14.2±0.4), T2 and ADC values significantly increased along the OB+RMS (p = 0.049/p = 0.01). Severe ECM (mean RMCBS = 5±2.9) was defined by further spread into more posterior and deeper brain structures until reaching the BS (significant T2 elevation in DMS+EC+BS (p = 0.034)). Quantitative automated histological analyses confirmed microglial activation in areas of BBBD and edema. Activated microglia were closely associated with the RMS and neuroblasts within the RMS were severely misaligned with respect to their physiological linear migration pattern. Microvascular pathology and ischemic brain injury occurred only secondarily, after vasogenic edema formation and were both associated less with clinical severity and the temporal course of ECM. Altogether, we identified a distinct spatiotemporal pattern of microglial activation in ECM involving primarily the OB+RMS axis, a distinct pathway utilized by neuroblasts and immune cells. Our data suggest significant crosstalk between these two cell populations to be operative in deeper brain infiltration and further imply that the manifestation and progression of cerebral malaria may depend on brain areas otherwise serving neurogenesis. Brain swelling is difficult to detect ex vivo and has recently been identified as a strong predictor of death not only in experimental cerebral malaria (ECM), but also in human cerebral malaria. As whole-brain in-vivo imaging methods have been widely underutilized in this disease model, little is known about the spatiotemporal evolution of brain swelling. To unravel this question, we monitored the evolution of ECM in vivo using high-field magnetic resonance imaging (MRI) with whole-brain coverage and have identified a distinct pattern of cerebral disease spread. Inflammatory disruption of the blood-brain-barrier and consecutive brain swelling initiates in the olfactory bulb and spreads from there along the rostral migratory stream—a neurogenic niche—deeper into the brain. When the brainstem is eventually reached, mice start to fall into a comatose state. Those findings correlate with previously published human MRI findings, which also show brain swelling of the brainstem in comatose children with cerebral malaria as well as early involvement of the striatum—recently recognized to serve neurogenesis in humans. Our study provides a novel link between neurogenic areas specifically permitting the spatiotemporal expansion of activated microglia, blood-brain-barrier disruption and consequent brain edema. Finally, the dominant role of the neurogenic axis in the transmission of inflammation may provide an explanation why children are more vulnerable to cerebral malaria.
Collapse
|
214
|
Ramirez J, Berezuk C, McNeely AA, Gao F, McLaurin J, Black SE. Imaging the Perivascular Space as a Potential Biomarker of Neurovascular and Neurodegenerative Diseases. Cell Mol Neurobiol 2016; 36:289-99. [PMID: 26993511 PMCID: PMC11482437 DOI: 10.1007/s10571-016-0343-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/03/2016] [Indexed: 12/11/2022]
Abstract
Although the brain lacks conventional lymphatic vessels found in peripheral tissue, evidence suggests that the space surrounding the vasculature serves a similar role in the clearance of fluid and metabolic waste from the brain. With aging, neurodegeneration, and cerebrovascular disease, these microscopic perivascular spaces can become enlarged, allowing for visualization and quantification on structural MRI. The purpose of this review is to: (i) describe some of the recent pre-clinical findings from basic science that shed light on the potential neurophysiological mechanisms driving glymphatic and perivascular waste clearance, (ii) review some of the pathobiological etiologies that may lead to MRI-visible enlarged perivascular spaces (ePVS), (iii) describe the possible clinical implications of ePVS, (iv) evaluate existing qualitative and quantitative techniques used for measuring ePVS burden, and (v) propose future avenues of research that may improve our understanding of this potential clinical neuroimaging biomarker for fluid and metabolic waste clearance dysfunction in neurodegenerative and neurovascular diseases.
Collapse
Affiliation(s)
- Joel Ramirez
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada, M4N 3M5.
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre (SHSC), Toronto, ON, Canada.
| | - Courtney Berezuk
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada, M4N 3M5
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre (SHSC), Toronto, ON, Canada
| | - Alicia A McNeely
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada, M4N 3M5
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre (SHSC), Toronto, ON, Canada
| | - Fuqiang Gao
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada, M4N 3M5
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre (SHSC), Toronto, ON, Canada
| | - JoAnne McLaurin
- Department of Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada, M4N 3M5
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre (SHSC), Toronto, ON, Canada
- Department of Medicine, Neurology (SHSC), Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
215
|
Ueno M, Chiba Y, Murakami R, Matsumoto K, Kawauchi M, Fujihara R. Blood-brain barrier and blood-cerebrospinal fluid barrier in normal and pathological conditions. Brain Tumor Pathol 2016; 33:89-96. [PMID: 26920424 DOI: 10.1007/s10014-016-0255-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/16/2016] [Indexed: 01/13/2023]
Abstract
Blood-borne substances can invade into the extracellular spaces of the brain via endothelial cells in sites without the blood-brain barrier (BBB), and can travel through the interstitial fluid (ISF) of the brain parenchyma adjacent to non-BBB sites. It has been shown that cerebrospinal fluid (CSF) drains directly into the blood via the arachnoid villi and also into lymph nodes via the subarachnoid spaces of the brain, while ISF drains into the cervical lymph nodes through perivascular drainage pathways. In addition, the glymphatic pathway of fluids, characterized by para-arterial pathways, aquaporin4-dependent passage through astroglial cytoplasm, interstitial spaces, and paravenous routes, has been established. Meningeal lymphatic vessels along the superior sagittal sinus were very recently discovered. It is known that, in mice, blood-borne substances can be transferred to areas with intact BBB function, such as the medial regions of the hippocampus, presumably through leaky vessels in non-BBB sites. In the present paper, we review the clearance mechanisms of interstitial substances, such as amyloid-β peptides, as well as summarize models of BBB deterioration in response to different types of insults, including acute ischemia followed by reperfusion, hypertension, and chronic hypoperfusion. Lastly, we discuss the relationship between perivascular clearance and brain disorders.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan.
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Machi Kawauchi
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Ryuji Fujihara
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| |
Collapse
|
216
|
Lawley JS, Levine BD, Williams MA, Malm J, Eklund A, Polaner DM, Subudhi AW, Hackett PH, Roach RC. Cerebral spinal fluid dynamics: effect of hypoxia and implications for high-altitude illness. J Appl Physiol (1985) 2016; 120:251-62. [DOI: 10.1152/japplphysiol.00370.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/17/2015] [Indexed: 12/24/2022] Open
Abstract
The pathophysiology of acute mountain sickness and high-altitude cerebral edema, the cerebral forms of high-altitude illness, remain uncertain and controversial. Persistently elevated or pathological fluctuations in intracranial pressure are thought to cause symptoms similar to those reported by individuals suffering cerebral forms of high-altitude illness. This review first focuses on the basic physiology of the craniospinal system, including a detailed discussion of the long-term and dynamic regulation of intracranial pressure. Thereafter, we critically examine the available literature, based primarily on invasive pressure monitoring, that suggests intracranial pressure is acutely elevated at altitude due to brain swelling and/or elevated sagittal sinus pressure, but normalizes over time. We hypothesize that fluctuations in intracranial pressure occur around a slightly elevated or normal mean intracranial pressure, in conjunction with oscillations in arterial Po2 and arterial blood pressure. Then these modest fluctuations in intracranial pressure, in concert with direct vascular stretch due to dilatation and/or increased blood pressure transmission, activate the trigeminal vascular system and cause symptoms of acute mountain sickness. Elevated brain water (vasogenic edema) may be due to breakdown of the blood-brain barrier. However, new information suggests cerebral spinal fluid flux into the brain may be an important factor. Regardless of the source (or mechanisms responsible) for the excess brain water, brain swelling occurs, and a “tight fit” brain would be a major risk factor to produce symptoms; activities that produce large changes in brain volume and cause fluctuations in blood pressure are likely contributing factors.
Collapse
Affiliation(s)
- Justin S. Lawley
- Institute for Exercise and Environmental Medicine, Presbyterian Hospital of Dallas, Dallas, Texas
- UT Southwestern Medical Center, Dallas, Texas
| | - Benjamin D. Levine
- Institute for Exercise and Environmental Medicine, Presbyterian Hospital of Dallas, Dallas, Texas
- UT Southwestern Medical Center, Dallas, Texas
| | - Michael A. Williams
- Sandra and Malcolm Berman Brain & Spine Institute, Dept. of Neurology, Sinai Hospital, Baltimore, Maryland
| | - Jon Malm
- Department of Clinical Neuroscience, Umeå University, Umeå, Sweden
| | - Anders Eklund
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - David M. Polaner
- Departments of Anesthesiology and Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, Colorado
| | - Andrew W. Subudhi
- Department of Biology, University of Colorado, Colorado Springs, Colorado
- Altitude Research Center, Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; and
| | | | - Robert C. Roach
- Altitude Research Center, Department of Emergency Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; and
| |
Collapse
|
217
|
Hughes TM, Craft S, Lopez OL. Review of 'the potential role of arterial stiffness in the pathogenesis of Alzheimer's disease'. Neurodegener Dis Manag 2016; 5:121-35. [PMID: 25894876 DOI: 10.2217/nmt.14.53] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Arterial stiffness is emerging as an important risk marker for poor brain aging and dementia through its associations with cerebral small vessel disease, stroke, β-amyloid deposition, brain atrophy and cognitive impairment. Arterial stiffness directly relates the detrimental effects of hypertension on peripheral organs with dire consequences for the extensive microvasculature structure of the kidneys and brain. In this review, we discuss the evidence linking arterial stiffness, hypertension and brain structural abnormalities in older adults. In particular, we discuss the potential mechanisms linking arterial stiffness to brain β-amyloid deposition and dementia and potential therapeutic strategies to prevent hypertension's adverse effects on the brain.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Division of Gerontology & Geriatric Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157-1207, USA
| | | | | |
Collapse
|
218
|
Abstract
In autoimmune neurologic disorders, the blood-brain barrier (BBB) plays a central role in immunopathogenesis, since this vascular interface is an entry path for cells and effector molecules of the peripheral immune system to reach the target organ, the central nervous system (CNS). The BBB's unique anatomic structure and the tightly regulated interplay of its cellular and acellular components allow for maintenance of brain homeostasis, regulation of influx and efflux, and protection from harm; these ensure an optimal environment for the neuronal network to function properly. In both health and disease, the BBB acts as mediator between the periphery and the CNS. For example, immune cell trafficking through the cerebral vasculature is essential to clear microbes or cell debris from neural tissues, while poorly regulated cellular transmigration can underlie or worsen CNS pathology. In this chapter, we focus on the specialized multicellular structure and function of the BBB/neurovascular unit and discuss how BBB breakdown can precede or be a consequence of neuroinflammation. We introduce the blood-cerebrospinal fluid barrier and include a brief aside about evolutionary aspects of barrier formation and refinements. Lastly, since restoration of barrier function is considered key to ameliorate neurologic disease, we speculate about new therapeutic avenues to repair a damaged BBB.
Collapse
Affiliation(s)
| | - Ajay Verma
- Biomarkers and Experimental Medicine, Biogen, Cambridge, MA, USA
| | | |
Collapse
|
219
|
Hughes TM, Craft S. The role of insulin in the vascular contributions to age-related dementia. Biochim Biophys Acta Mol Basis Dis 2015; 1862:983-91. [PMID: 26657615 DOI: 10.1016/j.bbadis.2015.11.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 11/29/2015] [Indexed: 01/12/2023]
Abstract
In addition to its well-known role in energy metabolism in the body, insulin is a vasoactive hormone that regulates peripheral and cerebral blood flow and neuronal function. Vascular and metabolic dysfunctions are emerging risk factors for Alzheimer's disease (AD) and age-related dementias, and recent evidence suggests that the two pathways are constitutive and interrelated. As a result, an emphasis on correcting metabolic disorders is emerging as an important strategy in the treatment and prevention of age-related cognitive impairment and AD. We review the evidence regarding the unique and interactive effects of vascular and metabolic disorders in pathological brain aging, with special consideration of the role of insulin dysregulation in promoting AD pathologic processes and vascular brain injury. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA.
| |
Collapse
|
220
|
The Role of the Craniocervical Junction in Craniospinal Hydrodynamics and Neurodegenerative Conditions. Neurol Res Int 2015; 2015:794829. [PMID: 26770824 PMCID: PMC4681798 DOI: 10.1155/2015/794829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/07/2015] [Accepted: 09/17/2015] [Indexed: 02/07/2023] Open
Abstract
The craniocervical junction (CCJ) is a potential choke point for craniospinal hydrodynamics and may play a causative or contributory role in the pathogenesis and progression of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, MS, and ALS, as well as many other neurological conditions including hydrocephalus, idiopathic intracranial hypertension, migraines, seizures, silent-strokes, affective disorders, schizophrenia, and psychosis. The purpose of this paper is to provide an overview of the critical role of the CCJ in craniospinal hydrodynamics and to stimulate further research that may lead to new approaches for the prevention and treatment of the above neurodegenerative and neurological conditions.
Collapse
|
221
|
Ueno M, Chiba Y, Matsumoto K, Murakami R, Fujihara R, Kawauchi M, Miyanaka H, Nakagawa T. Blood-brain barrier damage in vascular dementia. Neuropathology 2015; 36:115-24. [DOI: 10.1111/neup.12262] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/12/2015] [Accepted: 09/13/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine; Kagawa University; Kagawa Japan
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine; Kagawa University; Kagawa Japan
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine; Kagawa University; Kagawa Japan
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine; Kagawa University; Kagawa Japan
| | - Ryuji Fujihara
- Department of Pathology and Host Defense, Faculty of Medicine; Kagawa University; Kagawa Japan
| | - Machi Kawauchi
- Department of Pathology and Host Defense, Faculty of Medicine; Kagawa University; Kagawa Japan
| | | | | |
Collapse
|
222
|
Adalbert R, Coleman MP. Review: Axon pathology in age-related neurodegenerative disorders. Neuropathol Appl Neurobiol 2015; 39:90-108. [PMID: 23046254 DOI: 10.1111/j.1365-2990.2012.01308.x] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 09/24/2012] [Indexed: 12/12/2022]
Abstract
'Dying back' axon degeneration is a prominent feature of many age-related neurodegenerative disorders and is widespread in normal ageing. Although the mechanisms of disease- and age-related losses may differ, both contribute to symptoms. Here, we review recent advances in understanding axon pathology in age-related neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and glaucoma. In particular, we highlight the importance of axonal transport, autophagy, traumatic brain injury and mitochondrial quality control. We then place these disease mechanisms in the context of changes to axons and dendrites that occur during normal ageing. We discuss what makes ageing such an important risk factor for many neurodegenerative disorders and conclude that the processes of normal ageing and disease combine at the molecular, cellular or systems levels in a range of disorders to produce symptoms. Pathology identical to disease also occurs at the cellular level in most elderly individuals. Thus, normal ageing and age-related disease are inextricably linked and the term 'healthy ageing' downplays the important contributions of cellular pathology. For a full understanding of normal ageing or age-related disease we must study both processes.
Collapse
Affiliation(s)
- R Adalbert
- Signalling Programme, The Babraham Institute, Babraham, Cambridge, UK
| | - M P Coleman
- Signalling Programme, The Babraham Institute, Babraham, Cambridge, UK
| |
Collapse
|
223
|
Nakada T. The Molecular Mechanisms of Neural Flow Coupling: A New Concept. J Neuroimaging 2015; 25:861-5. [PMID: 25704766 PMCID: PMC5023998 DOI: 10.1111/jon.12219] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 12/10/2014] [Indexed: 11/29/2022] Open
Abstract
The phenomenon known as neural flow coupling (NFC) occurs at the capillary level where there are no known pressure controlling structures. Recent developments in advanced magnetic resonance imaging technologies have made possible in vivo direct investigations of water physiology that have shed new insight on the water dynamics of the cortical pericapillary space and their complex functionality in relation to NFC. Neural activities initiate a chain of events that ultimately affect NFC. First, neural activities generate extracellular acidification. Extracellular acidosis in turn produces inhibition of aquaporin-4 (AQP-4) located at the end feet of pericapillary astrocytes, the water channel which regulates water influx into the pericapillary space and, hence, interstitial flow. Reduction of pericapillary water pressure results in a negative balance between pericapillary and intraluminal capillary pressure, allowing for capillary caliber expansion. Proton permeability through the tight junctions of the blood brain barrier is significantly high owing to the Grotthuss proton "tunneling" mechanism and, therefore, carbonic anhydrase (CA) type IV (CA-IV) anchored to the luminal surface of brain capillaries functions as scavenger of extracellular protons. CA-IV inhibition by acetazolamide or carbon dioxide results in the accumulation of extracellular protons, causing AQP-4 inhibition and a secondary increase in rCBF.
Collapse
Affiliation(s)
- Tsutomu Nakada
- Center for Integrated Human Brain Science, Brain Research InstituteUniversity of NiigataNiigataJapan
| |
Collapse
|
224
|
Simon MJ, Iliff JJ. Regulation of cerebrospinal fluid (CSF) flow in neurodegenerative, neurovascular and neuroinflammatory disease. Biochim Biophys Acta Mol Basis Dis 2015; 1862:442-51. [PMID: 26499397 DOI: 10.1016/j.bbadis.2015.10.014] [Citation(s) in RCA: 221] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/23/2015] [Accepted: 10/19/2015] [Indexed: 12/20/2022]
Abstract
Cerebrospinal fluid (CSF) circulation and turnover provides a sink for the elimination of solutes from the brain interstitium, serving an important homeostatic role for the function of the central nervous system. Disruption of normal CSF circulation and turnover is believed to contribute to the development of many diseases, including neurodegenerative conditions such as Alzheimer's disease, ischemic and traumatic brain injury, and neuroinflammatory conditions such as multiple sclerosis. Recent insights into CSF biology suggesting that CSF and interstitial fluid exchange along a brain-wide network of perivascular spaces termed the 'glymphatic' system suggest that CSF circulation may interact intimately with glial and vascular function to regulate basic aspects of brain function. Dysfunction within this glial vascular network, which is a feature of the aging and injured brain, is a potentially critical link between brain injury, neuroinflammation and the development of chronic neurodegeneration. Ongoing research within this field may provide a powerful new framework for understanding the common links between neurodegenerative, neurovascular and neuroinflammatory disease, in addition to providing potentially novel therapeutic targets for these conditions. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
Affiliation(s)
- Matthew J Simon
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA; Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey J Iliff
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA; Neuroscience Graduate Program, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
225
|
Kida S. [Progress in diagnosis of and therapy for idiopathic normal-pressure hydrocephalus--Lymphatic drainage of CSF and ISF from the brain: recent concept and hypothesis]. Rinsho Shinkeigaku 2015; 54:1187-9. [PMID: 25672741 DOI: 10.5692/clinicalneurol.54.1187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cerebrospinal fluid (CSF) drains via the cribriform plate and nasal mucosa to cervical lymph nodes. There are no conventional lymphatics in the brain but physiological studies have revealed a substantial and immunologically significant lymphatic drainage from brain to cervical lymph nodes. Interstitial fluid (ISF) and solutes from brain parenchyma drain along capillary and perivascular space of artery, and path through the skull base, then reach to the cervical lymph nodes. CSF and ISF appear to drain by separate routes from the brain, especially in humans. However, there are interrelationships between the two fluid compartments that become more significant when drainage of CSF or ISF is impaired by disease processes. Vessel pulsations appear to be the driving force for the perivascular lymphatic drainage along artery walls, and as vessels stiffen with age, amyloid peptides (Aβ) deposit in the drainage pathways as cerebral amyloid angiopathy (CAA). Blockage of lymphatic drainage of ISF and solutes from the brain by CAA may result in loss of homeostasis of the neuronal environment that may contribute to neuronal malfunction and dementia. Such failure of perivascular drainage may associated with the pathoetiology of Alzheimer's disease, cerebral small artery disease and idiopathic normal pressure hydrocephalus (iNPH).
Collapse
Affiliation(s)
- Shinya Kida
- Department of Neurosurgery, Fukui Prefectural Hospital
| |
Collapse
|
226
|
Immunohistochemical analysis of transporters related to clearance of amyloid-β peptides through blood–cerebrospinal fluid barrier in human brain. Histochem Cell Biol 2015; 144:597-611. [DOI: 10.1007/s00418-015-1366-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2015] [Indexed: 01/25/2023]
|
227
|
Louveau A, Harris TH, Kipnis J. Revisiting the Mechanisms of CNS Immune Privilege. Trends Immunol 2015; 36:569-577. [PMID: 26431936 PMCID: PMC4593064 DOI: 10.1016/j.it.2015.08.006] [Citation(s) in RCA: 483] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 08/13/2015] [Accepted: 08/13/2015] [Indexed: 12/25/2022]
Abstract
Whereas the study of the interactions between the immune system and the central nervous system (CNS) has often focused on pathological conditions, the importance of neuroimmune communication in CNS homeostasis and function has become clear over that last two decades. Here we discuss the progression of our understanding of the interaction between the peripheral immune system and the CNS. We examine the notion of immune privilege of the CNS in light of both earlier findings and recent studies revealing a functional meningeal lymphatic system that drains cerebrospinal fluid (CSF) to the deep cervical lymph nodes, and consider the implications of a revised perspective on the immune privilege of the CNS on the etiology and pathology of different neurological disorders.
Collapse
Affiliation(s)
- Antoine Louveau
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Tajie H Harris
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia, Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
228
|
Peristalsis with Oscillating Flow Resistance: A Mechanism for Periarterial Clearance of Amyloid Beta from the Brain. Ann Biomed Eng 2015; 44:1553-65. [PMID: 26399987 DOI: 10.1007/s10439-015-1457-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022]
Abstract
Alzheimer's disease is characterized by accumulation of amyloid-β (Aβ) in the brain and in the walls of cerebral arteries. The focus of this work is on clearance of Aβ along artery walls, the failure of which may explain the accumulation of Aβ in Alzheimer's disease. Periarterial basement membranes form continuous channels from cerebral capillaries to major arteries on the surface of the brain. Arterial pressure pulses drive peristaltic flow in the basement membranes in the same direction as blood flow. Here we forward the hypothesis that flexible structures within the basement membrane, if oriented such they present greater resistance to forward than retrograde flow, may cause net reverse flow, advecting Aβ along with it. A solution was obtained for peristaltic flow with low Reynolds number, long wavelength compared to channel height and small channel height compared to vessel radius in a Darcy-Brinkman medium representing a square array of cylinders. Results show that retrograde flow is promoted by high cylinder volume fraction and low peristaltic amplitude. A decrease in cylinder concentration and/or an increase in amplitude, both of which may occur during ageing, can reduce retrograde flow or even cause a transition from retrograde to forward flow. Such changes may explain the accumulation of Aβ in the brain and in artery walls in Alzheimer's disease.
Collapse
|
229
|
Weller RO, Hawkes CA, Kalaria RN, Werring DJ, Carare RO. White matter changes in dementia: role of impaired drainage of interstitial fluid. Brain Pathol 2015; 25:63-78. [PMID: 25521178 DOI: 10.1111/bpa.12218] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/08/2014] [Indexed: 12/22/2022] Open
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.
Collapse
Affiliation(s)
- Roy O Weller
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | | | | | | |
Collapse
|
230
|
Hawkes CA, Jayakody N, Johnston DA, Bechmann I, Carare RO. Failure of perivascular drainage of β-amyloid in cerebral amyloid angiopathy. Brain Pathol 2015; 24:396-403. [PMID: 24946077 DOI: 10.1111/bpa.12159] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 01/18/2023] Open
Abstract
In Alzheimer's disease, amyloid-β (Aβ) accumulates as insoluble plaques in the brain and deposits in blood vessel walls as cerebral amyloid angiopathy (CAA). The severity of CAA correlates with the degree of cognitive decline in dementia. The distribution of Aβ in the walls of capillaries and arteries in CAA suggests that Aβ is deposited in the perivascular pathways by which interstitial fluid drains from the brain. Soluble Aβ from the extracellular spaces of gray matter enters the basement membranes of capillaries and drains along the arterial basement membranes that surround smooth muscle cells toward the leptomeningeal arteries. The motive force for perivascular drainage is derived from arterial pulsations combined with the valve effect of proteins present in the arterial basement membranes. Physical and biochemical changes associated with arteriosclerosis, aging and possession of apolipoprotein E4 genotype lead to a failure of perivascular drainage of soluble proteins, including Aβ. Perivascular cells associated with arteries and the lymphocytes recruited in the perivenous spaces contribute to the clearance of Aβ. The failure of perivascular clearance of Aβ may be a major factor in the accumulation of Aβ in CAA and may have significant implications for the design of therapeutics for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Cheryl A Hawkes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | | | | | | |
Collapse
|
231
|
Abstract
The central nervous system (CNS) possesses powerful local and global immunosuppressive capabilities that modulate unwanted inflammatory reactions in nervous tissue. These same immune-modulatory mechanisms are also co-opted by malignant brain tumors and pose a formidable challenge to brain tumor immunotherapy. Routes by which malignant gliomas coordinate immunosuppression include the mechanical and functional barriers of the CNS; immunosuppressive cytokines and catabolites; immune checkpoint molecules; tumor-infiltrating immune cells; and suppressor immune cells. The challenges to overcoming tumor-induced immunosuppression, however, are not unique to the brain, and several analogous immunosuppressive mechanisms also exist for primary tumors outside of the CNS. Ultimately, the immune responses in the CNS are linked and complementary to immune processes in the periphery, and advances in tumor immunotherapy in peripheral sites may therefore illuminate novel approaches to brain tumor immunotherapy, and vice versa.
Collapse
Affiliation(s)
- Powell Perng
- Department of Neurosurgery, School of Medicine, Johns Hopkins University , Baltimore, MD , USA
| | - Michael Lim
- Department of Neurosurgery, School of Medicine, Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|
232
|
Vandal M, Bourassa P, Calon F. Can insulin signaling pathways be targeted to transport Aβ out of the brain? Front Aging Neurosci 2015; 7:114. [PMID: 26136681 PMCID: PMC4468380 DOI: 10.3389/fnagi.2015.00114] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 05/29/2015] [Indexed: 12/11/2022] Open
Abstract
Although the causal role of Amyloid-β (Aβ) in Alzheimer’s disease (AD) is unclear, it is still reasonable to expect that lowering concentrations of Aβ in the brain may decrease the risk of developing the neurocognitive symptoms of the disease. Brain capillary endothelial cells forming the blood-brain barrier (BBB) express transporters regulating the efflux of Aβ out of the cerebral tissue. Age-related BBB dysfunctions, that have been identified in AD patients, might impair Aβ clearance from the brain. Thus, targeting BBB outward transport systems has been suggested as a way to stimulate the clearance of Aβ from the brain. Recent data indicate that the increase in soluble brain Aβ and behavioral impairments in 3×Tg-AD mice generated by months of intake of a high-fat diet can be acutely reversed by the administration of a single dose of insulin. A concomitant increase in plasma Aβ suggests that clearance from the brain through the BBB is a likely mechanism for this rapid effect of insulin. Here, we review how BBB insulin response pathways could be stimulated to decrease brain Aβ concentrations and improve cognitive performance, at least on the short term.
Collapse
Affiliation(s)
- Milene Vandal
- Faculté de Pharmacie, Université Laval Quebec, QC, Canada ; Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL) Québec, QC, Canada ; Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval Québec, QC, Canada
| | - Philippe Bourassa
- Faculté de Pharmacie, Université Laval Quebec, QC, Canada ; Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL) Québec, QC, Canada ; Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval Québec, QC, Canada
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval Quebec, QC, Canada ; Axe Neurosciences, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUL) Québec, QC, Canada ; Institut des Nutraceutiques et des Aliments Fonctionnels, Université Laval Québec, QC, Canada
| |
Collapse
|
233
|
Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, Wiig H, Alitalo K. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. ACTA ACUST UNITED AC 2015; 212:991-9. [PMID: 26077718 PMCID: PMC4493418 DOI: 10.1084/jem.20142290] [Citation(s) in RCA: 1480] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 06/04/2015] [Indexed: 12/16/2022]
Abstract
Aspelund et al. discover the presence of a lymphatic vessel network in the dura mater of the mouse brain and show that these dural lymphatic vessels are important for the clearance of macromolecules from the brain. The central nervous system (CNS) is considered an organ devoid of lymphatic vasculature. Yet, part of the cerebrospinal fluid (CSF) drains into the cervical lymph nodes (LNs). The mechanism of CSF entry into the LNs has been unclear. Here we report the surprising finding of a lymphatic vessel network in the dura mater of the mouse brain. We show that dural lymphatic vessels absorb CSF from the adjacent subarachnoid space and brain interstitial fluid (ISF) via the glymphatic system. Dural lymphatic vessels transport fluid into deep cervical LNs (dcLNs) via foramina at the base of the skull. In a transgenic mouse model expressing a VEGF-C/D trap and displaying complete aplasia of the dural lymphatic vessels, macromolecule clearance from the brain was attenuated and transport from the subarachnoid space into dcLNs was abrogated. Surprisingly, brain ISF pressure and water content were unaffected. Overall, these findings indicate that the mechanism of CSF flow into the dcLNs is directly via an adjacent dural lymphatic network, which may be important for the clearance of macromolecules from the brain. Importantly, these results call for a reexamination of the role of the lymphatic system in CNS physiology and disease.
Collapse
Affiliation(s)
- Aleksanteri Aspelund
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland
| | - Salli Antila
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland
| | - Steven T Proulx
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH Zurich), CH-8093 Zurich, Switzerland
| | | | - Sinem Karaman
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH Zurich), CH-8093 Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH Zurich), CH-8093 Zurich, Switzerland
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, 5009 Bergen, Norway
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
234
|
Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, Harris TH, Kipnis J. Structural and functional features of central nervous system lymphatic vessels. Nature 2015; 523:337-41. [PMID: 26030524 PMCID: PMC4506234 DOI: 10.1038/nature14432] [Citation(s) in RCA: 2994] [Impact Index Per Article: 299.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/20/2015] [Indexed: 12/15/2022]
Abstract
One of the characteristics of the central nervous system is the lack of a classical lymphatic drainage system. Although it is now accepted that the central nervous system undergoes constant immune surveillance that takes place within the meningeal compartment, the mechanisms governing the entrance and exit of immune cells from the central nervous system remain poorly understood. In searching for T-cell gateways into and out of the meninges, we discovered functional lymphatic vessels lining the dural sinuses. These structures express all of the molecular hallmarks of lymphatic endothelial cells, are able to carry both fluid and immune cells from the cerebrospinal fluid, and are connected to the deep cervical lymph nodes. The unique location of these vessels may have impeded their discovery to date, thereby contributing to the long-held concept of the absence of lymphatic vasculature in the central nervous system. The discovery of the central nervous system lymphatic system may call for a reassessment of basic assumptions in neuroimmunology and sheds new light on the aetiology of neuroinflammatory and neurodegenerative diseases associated with immune system dysfunction.
Collapse
Affiliation(s)
- Antoine Louveau
- 1] Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Igor Smirnov
- 1] Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Timothy J Keyes
- 1] Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Jacob D Eccles
- 1] Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Beirne B. Carter Center for Immunology Research, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [3] Department of Medicine (Division of Allergy), School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Sherin J Rouhani
- 1] Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Beirne B. Carter Center for Immunology Research, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [3] Department of Microbiology, Immunology, and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - J David Peske
- 1] Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Beirne B. Carter Center for Immunology Research, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [3] Department of Microbiology, Immunology, and Cancer Biology, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Noel C Derecki
- 1] Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - David Castle
- Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - James W Mandell
- Department of Pathology (Neuropathology), School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Kevin S Lee
- 1] Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [3] Department of Neurosurgery, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Tajie H Harris
- 1] Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Jonathan Kipnis
- 1] Center for Brain Immunology and Glia, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [2] Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA [3] Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA
| |
Collapse
|
235
|
Berezuk C, Ramirez J, Gao F, Scott CJM, Huroy M, Swartz RH, Murray BJ, Black SE, Boulos MI. Virchow-Robin Spaces: Correlations with Polysomnography-Derived Sleep Parameters. Sleep 2015; 38:853-8. [PMID: 26163465 DOI: 10.5665/sleep.4726] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/24/2014] [Indexed: 12/14/2022] Open
Abstract
STUDY OBJECTIVES To test the hypothesis that enlarged Virchow-Robin space volumes (VRS) are associated with objective measures of poor quality sleep. DESIGN Retrospective cross-sectional study. SETTING Sunnybrook Health Sciences Centre. PATIENTS Twenty-six patients being evaluated for cerebrovascular disease were assessed using polysomnography and high-resolution structural magnetic resonance imaging. MEASUREMENTS AND RESULTS Regionalized VRS were quantified from three-dimensional high-resolution magnetic resonance imaging and correlated with measures of polysomnography-derived sleep parameters while controlling for age, stroke volume, body mass index, systolic blood pressure, and ventricular cerebrospinal fluid volume. Sleep efficiency was negatively correlated with total VRS (rho = -0.47, P = 0.03) and basal ganglia VRS (rho = -0.54, P = 0.01), whereas wake after sleep onset was positively correlated with basal ganglia VRS (rho = 0.52, P = 0.02). Furthermore, VRS in the basal ganglia were negatively correlated with duration of N3 (rho = -0.53, P = 0.01). CONCLUSIONS These preliminary results suggest that sleep may play a role in perivascular clearance in ischemic brain disease, and invite future research into the potential relevance of Virchow-Robin spaces as an imaging biomarker for nocturnal metabolite clearance.
Collapse
Affiliation(s)
- Courtney Berezuk
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada
| | - Joel Ramirez
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada
| | - Fuqiang Gao
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada
| | - Christopher J M Scott
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada
| | - Menal Huroy
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada
| | - Richard H Swartz
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada.,Institute of Medical Science, Faculty of Medicine, School of Graduate Studies University of Toronto, Toronto, Canada.,Department of Medicine (Neurology), University of Toronto and Sunnybrook HSC, Toronto, Canada
| | - Brian J Murray
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada.,Department of Medicine (Neurology), University of Toronto and Sunnybrook HSC, Toronto, Canada
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada.,Institute of Medical Science, Faculty of Medicine, School of Graduate Studies University of Toronto, Toronto, Canada.,Department of Medicine (Neurology), University of Toronto and Sunnybrook HSC, Toronto, Canada
| | - Mark I Boulos
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute (SRI), University of Toronto, Canada.,Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada.,Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre (HSC), Toronto, Canada.,Department of Medicine (Neurology), University of Toronto and Sunnybrook HSC, Toronto, Canada
| |
Collapse
|
236
|
Suzuki Y, Nakamura Y, Yamada K, Igarashi H, Kasuga K, Yokoyama Y, Ikeuchi T, Nishizawa M, Kwee IL, Nakada T. Reduced CSF Water Influx in Alzheimer's Disease Supporting the β-Amyloid Clearance Hypothesis. PLoS One 2015; 10:e0123708. [PMID: 25946191 PMCID: PMC4422624 DOI: 10.1371/journal.pone.0123708] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 03/05/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To investigate whether water influx into cerebrospinal fluid (CSF) space is reduced in Alzheimer's patients as previously shown in the transgenic mouse model for Alzheimer's disease. METHODS Ten normal young volunteers (young control, 21-30 years old), ten normal senior volunteers (senior control, 60-78 years old, MMSE ≥ 29), and ten Alzheimer's disease (AD) patients (study group, 59-84 years old, MMSE: 13-19) participated in this study. All AD patients were diagnosed by neurologists specializing in dementia based on DSM-IV criteria. CSF dynamics were analyzed using positron emission tomography (PET) following an intravenous injection of 1,000 MBq [15O]H2O synthesized on-line. RESULTS Water influx into CSF space in AD patients, expressed as influx ratio, (0.755 ± 0.089) was significantly reduced compared to young controls (1.357 ± 0.185; p < 0.001) and also compared to normal senior controls (0.981 ± 0.253, p < 0.05). Influx ratio in normal senior controls was significantly reduced compared to young controls (p < 0.01). CONCLUSION Water influx into the CSF is significantly reduced in AD patients. β-amyloid clearance has been shown to be dependent on interstitial flow and CSF production. The current study indicates that reduction in water influx into the CSF may disturb the clearance rate of β-amyloid, and therefore be linked to the pathogenesis of AD. TRIAL REGISTRATION UMIN Clinical Trials Registry UMIN000011939.
Collapse
Affiliation(s)
- Yuji Suzuki
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Yukihiro Nakamura
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Kenichi Yamada
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Hironaka Igarashi
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Yuichi Yokoyama
- Department of Psychiatry, Faculty of Medicine, University of Niigata, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Masatoyo Nishizawa
- Department of Neurology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Ingrid L. Kwee
- Department of Neurology, University of California Davis, Davis, California, United States of America
| | - Tsutomu Nakada
- Center for Integrated Human Brain Science, Brain Research Institute, University of Niigata, Niigata, Japan
- Department of Neurology, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
237
|
van Keulen LJM, Langeveld JPM, Dolstra CH, Jacobs J, Bossers A, van Zijderveld FG. TSE strain differentiation in mice by immunohistochemical PrP(Sc) profiles and triplex Western blot. Neuropathol Appl Neurobiol 2015; 41:756-79. [PMID: 25201447 DOI: 10.1111/nan.12181] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 08/25/2014] [Indexed: 11/27/2022]
Abstract
UNLABELLED TSE strains are routinely identified by their incubation period and vacuolation profile in the brain after intracerebral inoculation and serial passaging in inbred mouse lines. There are some major drawbacks to this method that are related to the variation in vacuolation that exists in the brains of mice infected with the same TSE strain and to variation between observers and laboratories in scoring vacuolation and determining the final incubation period. AIM We investigated the potential of PrP(Sc) immunohistochemistry and triplex Western blotting as possible alternative methods to differentiate between TSE strains. METHODS TSE reference strains ME7, 87A/87V, 22A/22C, 79A/79V and 301C/301V were intracerebrally inoculated in RIII or VM inbred mice that differ in their PrP genotype. Immunohistochemical PrP(Sc) profiles were drawn up by scanning light microscopy both on coronal and sagittal sections. RESULTS On the basis of the localization of PrP(Sc) in the cerebral cortex, hippocampus, and cerebellar cortex and the overall type of PrP(Sc) staining, all TSE strains could be well differentiated from each other through their typical strain dependent characteristics. In addition, Western blot showed that the combination of glycosylation profile and 12B2 epitope content of PrP(Sc) allowed to distinguish between all reference strains except for ME7 and 22A in VM mice. CONCLUSION TSE strains in mice can be identified on the basis of their PrP(Sc) profile alone. The potential to identify TSE strains in ruminants with these PrP(Sc) profiles after a single primary passage in mice will be the topic of future studies.
Collapse
Affiliation(s)
- Lucien J M van Keulen
- Department of Infection Biology, Central Veterinary Institute of Wageningen UR, Lelystad, The Netherlands
| | - Jan P M Langeveld
- Department of Infection Biology, Central Veterinary Institute of Wageningen UR, Lelystad, The Netherlands
| | - Corry H Dolstra
- Department of Infection Biology, Central Veterinary Institute of Wageningen UR, Lelystad, The Netherlands
| | - Jorg Jacobs
- Department of Infection Biology, Central Veterinary Institute of Wageningen UR, Lelystad, The Netherlands
| | - Alex Bossers
- Department of Infection Biology, Central Veterinary Institute of Wageningen UR, Lelystad, The Netherlands
| | - Fred G van Zijderveld
- Department of Bacteriology and TSEs, Central Veterinary Institute of Wageningen UR, Lelystad, The Netherlands
| |
Collapse
|
238
|
Charidimou A, Hong YT, Jäger HR, Fox Z, Aigbirhio FI, Fryer TD, Menon DK, Warburton EA, Werring DJ, Baron JC. White matter perivascular spaces on magnetic resonance imaging: marker of cerebrovascular amyloid burden? Stroke 2015; 46:1707-9. [PMID: 25908461 DOI: 10.1161/strokeaha.115.009090] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/23/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE We investigated the relationship between magnetic resonance imaging-visible centrum semiovale perivascular spaces (CSO-PVS), a biomarker of impaired interstitial fluid drainage, and positron emission tomography-based amyloid-β burden across a wide range of cerebrovascular amyloid deposition. METHODS Thirty-one nondemented subjects (11 probable cerebral amyloid angiopathy patients and 10 healthy subjects≥60 years; 10 older individuals, <60 years) had brain magnetic resonance imaging and Pittsburgh compound B-positron emission tomography. CSO-PVS was evaluated on T2-magnetic resonance imaging using a 4-point scale. The association between Pittsburgh compound B and CSO-PVS was assessed in linear regression. RESULTS In multivariable analyses adjusted for age, microbleeds and white matter hyperintensities, whole cortex Pittsburgh compound B binding was associated with CSO-PVS degree both as continuous (coefficient, 0.11; 95% confidence interval, 0.01-0.22; P=0.040) and as dichotomous variable (coefficient, 0.27; 95% confidence interval, 0.11-0.44; P=0.002). The median Pittsburgh compound B retention was higher in high versus low CSO-PVS degree (P=0.0007). CONCLUSIONS This pilot study suggests a possible association between cerebrovascular amyloid deposition and CSO-PVS, with potential pathophysiological implications.
Collapse
Affiliation(s)
- Andreas Charidimou
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.).
| | - Young T Hong
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - Hans R Jäger
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - Zoe Fox
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - Franklin I Aigbirhio
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - Tim D Fryer
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - David K Menon
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - Elizabeth A Warburton
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - David J Werring
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| | - Jean-Claude Baron
- From the UCL Institute of Neurology, Queen Square, London, United Kingdom (A.C., H.R.J., Z.F., D.J.W.); Wolfson Brain Imaging Centre (Y.T.H., F.I.A., T.D.F.), Division of Anaesthesia (D.K.M.), and Stroke Research Group, Department of Clinical Neurosciences (E.A.W., J.-C.B.), University of Cambridge, Cambridge, United Kingdom; and INSERM U894, Centre Hospitalier Sainte-Anne, Sorbonne Paris Cité, Paris, France (J.-C.B.)
| |
Collapse
|
239
|
Awan M, Liu S, Sahgal A, Das S, Chao ST, Chang EL, Knisely JPS, Redmond K, Sohn JW, Machtay M, Sloan AE, Mansur DB, Rogers LR, Lo SS. Extra-CNS metastasis from glioblastoma: a rare clinical entity. Expert Rev Anticancer Ther 2015; 15:545-52. [DOI: 10.1586/14737140.2015.1028374] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
240
|
Moriya M, Miyajima M, Nakajima M, Ogino I, Arai H. Impact of cerebrospinal fluid shunting for idiopathic normal pressure hydrocephalus on the amyloid cascade. PLoS One 2015; 10:e0119973. [PMID: 25821958 PMCID: PMC4379026 DOI: 10.1371/journal.pone.0119973] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 02/03/2015] [Indexed: 11/30/2022] Open
Abstract
The aim of this study was to determine whether the improvement of cerebrospinal fluid (CSF) flow dynamics by CSF shunting, can suppress the oligomerization of amyloid β-peptide (Aβ), by measuring the levels of Alzheimer’s disease (AD)-related proteins in the CSF before and after lumboperitoneal shunting. Lumbar CSF from 32 patients with idiopathic normal pressure hydrocephalus (iNPH) (samples were obtained before and 1 year after shunting), 15 patients with AD, and 12 normal controls was analyzed for AD-related proteins and APLP1-derived Aβ-like peptides (APL1β) (a surrogate marker for Aβ). We found that before shunting, individuals with iNPH had significantly lower levels of soluble amyloid precursor proteins (sAPP) and Aβ38 compared to patients with AD and normal controls. We divided the patients with iNPH into patients with favorable (improvement ≥ 1 on the modified Rankin Scale) and unfavorable (no improvement on the modified Rankin Scale) outcomes. Compared to the unfavorable outcome group, the favorable outcome group showed significant increases in Aβ38, 40, 42, and phosphorylated-tau levels after shunting. In contrast, there were no significant changes in the levels of APL1β25, 27, and 28 after shunting. After shunting, we observed positive correlations between sAPPα and sAPPβ, Aβ38 and 42, and APL1β25 and 28, with shifts from sAPPβ to sAPPα, from APL1β28 to 25, and from Aβ42 to 38 in all patients with iNPH. Our results suggest that Aβ production remained unchanged by the shunt procedure because the levels of sAPP and APL1β were unchanged. Moreover, the shift of Aβ from oligomer to monomer due to the shift of Aβ42 (easy to aggregate) to Aβ38 (difficult to aggregate), and the improvement of interstitial-fluid flow, could lead to increased Aβ levels in the CSF. Our findings suggest that the shunting procedure can delay intracerebral deposition of Aβ in patients with iNPH.
Collapse
Affiliation(s)
- Masao Moriya
- Department of Neurosurgery, Juntendo University Graduate School of medicine, Tokyo, Japan
- * E-mail:
| | - Masakazu Miyajima
- Department of Neurosurgery, Juntendo University Graduate School of medicine, Tokyo, Japan
| | - Madoka Nakajima
- Department of Neurosurgery, Juntendo University Graduate School of medicine, Tokyo, Japan
| | - Ikuko Ogino
- Department of Neurosurgery, Juntendo University Graduate School of medicine, Tokyo, Japan
| | - Hajime Arai
- Department of Neurosurgery, Juntendo University Graduate School of medicine, Tokyo, Japan
| |
Collapse
|
241
|
Liu H, Ni Z, Chen Y, Wang D, Qi Y, Zhang Q, Wang S. Olfactory route for cerebrospinal fluid drainage into the cervical lymphatic system in a rabbit experimental model. Neural Regen Res 2015; 7:766-71. [PMID: 25737700 PMCID: PMC4345659 DOI: 10.3969/j.issn.1673-5374.2012.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 11/22/2011] [Indexed: 12/24/2022] Open
Abstract
The present study analyzed the anatomical association between intracranial subarachnoid space and the cervical lymphatic system. X-ray contrast medium and Microfil(®) (Microfil compounds fill and opacify microvascular and other spaces of non-surviving animals and post-mortem tissue under physiological injection pressure) were injected into the cisterna magna of the rabbit, and perineural routes of cerebrospinal fluid outflow into the lymphatic system were visualized. Under a surgical operating microscope, Microfil was found within the subarachnoid space and along the olfactory nerves. At the nasal mucosa, a lymphatic network was identified near the olfactory nerves, which crossed the nasopharyngeal region and finally emptied into the superficial and deep cervical lymph nodes. Under a light microscope, Microfil was visible around the olfactory nerves and within lymphatic vessels. These results suggested that cerebrospinal fluid drained from the subarachnoid space along the olfactory nerves to nasal lymphatic vessels, which in turn, emptied into the cervical lymph nodes. This anatomical route, therefore, allowed connection between the central nervous system and the lymphatic system.
Collapse
Affiliation(s)
- Haisheng Liu
- Department of Neurosurgery, Yuquan Hospital, Tsinghua University, Beijing 100049, China
| | - Zhili Ni
- Department of Otorhinolaryngology and Head & Neck Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yetao Chen
- Department of Neurosurgery, Yuquan Hospital, Tsinghua University, Beijing 100049, China
| | - Dong Wang
- Department of Neurosurgery, Yuquan Hospital, Tsinghua University, Beijing 100049, China
| | - Yan Qi
- Department of Otorhinolaryngology and Head & Neck Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Qiuhang Zhang
- Department of Otorhinolaryngology and Head & Neck Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shijie Wang
- Department of Otorhinolaryngology and Head & Neck Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
242
|
Chui HC, Ramirez-Gomez L. Clinical and imaging features of mixed Alzheimer and vascular pathologies. ALZHEIMERS RESEARCH & THERAPY 2015; 7:21. [PMID: 25722748 PMCID: PMC4342006 DOI: 10.1186/s13195-015-0104-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The co-occurrence of both Alzheimer disease (AD) pathology and vascular brain injury (VBI) is very common, especially amongst the oldest of old. In neuropathologic studies, the prevalence of AD, VBI, and mixed AD/VBI lesions ranks ahead of Lewy bodies and hippocampal sclerosis. In the modern era of structural magnetic resonance imaging (MRI) and amyloid positron emission tomography (PET) imaging, this review examines 1) the prevalence of mixed AD and VBI pathology, 2) the significance of these pathologies for cognitive impairment (AD and vascular cognitive impairment (VCI)), and 3) the diagnosis and treatment of mixed AD/VCI. Although epidemiologic studies report that vascular risk factors for arteriosclerosis increase the risk of incident AD, both autopsy and amyloid PET studies indicate that AD and VBI contribute additively, but independently, to the risk of dementia. The literature confirms the malignancy of AD and highlights the adverse effects of microinfarcts on cognitive function. For the clinical diagnosis of mixed AD/VCI, the presence of AD can be recognized by neuropsychological profile, structural imaging, cerebrospinal fluid biomarkers, and glucose PET and amyloid PET imaging. The diagnosis of VBI, however, still hinges predominantly on the structural MRI findings. Severe amnesia and atrophy of the hippocampus are characteristic of early AD, whereas the cognitive profile for VCI is highly variable and dependent on size and location of VBI. The cognitive profile of mixed AD/VBI is dominated by AD. With the notable exception of microinfarcts (which elude in vivo detection), infarcts, hemorrhages, and white matter hyperintensities on structural MRI currently represent the best markers for the presence VBI. Better markers that reflect the health and reactivity of intracerebral blood vessels are needed. For prevention and treatment, the type of underlying cerebrovascular disease (for example, arteriosclerosis or cerebral amyloid angiopathy) should be considered. It is likely that reduction of vascular risk factors for arteriosclerosis can significantly reduce vascular contributions to mixed dementia.
Collapse
Affiliation(s)
- Helena C Chui
- Department of Neurology, University of Southern California, 1570 Alcazar Street, Suite 215, Los Angeles, CA 90033 USA
| | - Liliana Ramirez-Gomez
- Department of Neurology, University of Southern California, 1570 Alcazar Street, Suite 215, Los Angeles, CA 90033 USA
| |
Collapse
|
243
|
Lai AY, Dorr A, Thomason LAM, Koletar MM, Sled JG, Stefanovic B, McLaurin J. Venular degeneration leads to vascular dysfunction in a transgenic model of Alzheimer's disease. ACTA ACUST UNITED AC 2015; 138:1046-58. [PMID: 25688079 DOI: 10.1093/brain/awv023] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Most patients with Alzheimer's disease exhibit accumulation of amyloid-β peptide on leptomeningeal and cortical arterioles, or cerebral amyloid angiopathy, which is associated with impaired vascular reactivity and accelerated cognitive decline. Despite widespread recognition of the significance of vascular dysfunction in Alzheimer's disease aetiology and progression, much uncertainty still surrounds the mechanism underlying Alzheimer's disease vascular injury. Studies to date have focused on amyloid-β-induced damage to capillaries and plaque-associated arterioles, without examining effects across the entire vascular bed. In the present study, we investigated the structural and functional impairment of the feeding arteriolar versus draining venular vessels in a transgenic murine Alzheimer's disease model, with a particular focus on the mural cell populations that dictate these vessels' contractility. Although amyloid-β deposition was restricted to arterioles, we found that vascular impairment extended to the venules, which showed significant depletion of their mural cell coverage by the mid-stage of Alzheimer's disease pathophysiology. These structural abnormalities were accompanied by an abolishment of the normal vascular network flow response to hypercapnia: this functional impairment was so severe as to result in hypercapnia-induced flow decreases in the arterioles. Further pharmacological depletion of mural cells using SU6668, a platelet-derived growth factor receptor-β antagonist, resulted in profound structural abnormalities of the cortical microvasculature, including vessel coiling and short-range looping, increased tortuosity of the venules but not of the arterioles, increased amyloid-β deposition on the arterioles, and further alterations of the microvascular network cerebral blood flow response to hypercapnia. Together, this work shows hitherto unrecognized structural alterations in penetrating venules, demonstrates their functional significance and sheds light on the complexity of the relationship between vascular network structure and function in Alzheimer's disease.
Collapse
Affiliation(s)
- Aaron Y Lai
- 1 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada 2 Sunnybrook Research Institute, Toronto, ON, Canada
| | - Adrienne Dorr
- 2 Sunnybrook Research Institute, Toronto, ON, Canada
| | | | | | - John G Sled
- 3 Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada 4 Hospital for Sick Children, Toronto, ON, Canada
| | - Bojana Stefanovic
- 2 Sunnybrook Research Institute, Toronto, ON, Canada 3 Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - JoAnne McLaurin
- 1 Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada 2 Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
244
|
Insights into cognitive aging and Alzheimer’s disease using amyloid PET and structural MRI scans. Clin Transl Imaging 2015. [DOI: 10.1007/s40336-015-0110-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
245
|
Size-dependent long-term tissue response to biostable nanowires in the brain. Biomaterials 2014; 42:172-83. [PMID: 25542805 DOI: 10.1016/j.biomaterials.2014.11.051] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 11/13/2014] [Accepted: 11/25/2014] [Indexed: 01/03/2023]
Abstract
Nanostructured neural interfaces, comprising nanotubes or nanowires, have the potential to overcome the present hurdles of achieving stable communication with neuronal networks for long periods of time. This would have a strong impact on brain research. However, little information is available on the brain response to implanted high-aspect-ratio nanoparticles, which share morphological similarities with asbestos fibres. Here, we investigated the glial response and neuronal loss in the rat brain after implantation of biostable and structurally controlled nanowires of different lengths for a period up to one year post-surgery. Our results show that, as for lung and abdominal tissue, the brain is subject to a sustained, local inflammation when biostable and high-aspect-ratio nanoparticles of 5 μm or longer are present in the brain tissue. In addition, a significant loss of neurons was observed adjacent to the 10 μm nanowires after one year. Notably, the inflammatory response was restricted to a narrow zone around the nanowires and did not escalate between 12 weeks and one year. Furthermore, 2 μm nanowires did not cause significant inflammatory response nor significant loss of neurons nearby. The present results provide key information for the design of future neural implants based on nanomaterials.
Collapse
|
246
|
Hladky SB, Barrand MA. Mechanisms of fluid movement into, through and out of the brain: evaluation of the evidence. Fluids Barriers CNS 2014; 11:26. [PMID: 25678956 PMCID: PMC4326185 DOI: 10.1186/2045-8118-11-26] [Citation(s) in RCA: 423] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/21/2014] [Indexed: 01/08/2023] Open
Abstract
Interstitial fluid (ISF) surrounds the parenchymal cells of the brain and spinal cord while cerebrospinal fluid (CSF) fills the larger spaces within and around the CNS. Regulation of the composition and volume of these fluids is important for effective functioning of brain cells and is achieved by barriers that prevent free exchange between CNS and blood and by mechanisms that secrete fluid of controlled composition into the brain and distribute and reabsorb it. Structures associated with this regular fluid turnover include the choroid plexuses, brain capillaries comprising the blood-brain barrier, arachnoid villi and perineural spaces penetrating the cribriform plate. ISF flow, estimated from rates of removal of markers from the brain, has been thought to reflect rates of fluid secretion across the blood-brain barrier, although this has been questioned because measurements were made under barbiturate anaesthesia possibly affecting secretion and flow and because CSF influx to the parenchyma via perivascular routes may deliver fluid independently of blood-brain barrier secretion. Fluid secretion at the blood-brain barrier is provided by specific transporters that generate solute fluxes so creating osmotic gradients that force water to follow. Any flow due to hydrostatic pressures driving water across the barrier soon ceases unless accompanied by solute transport because water movements modify solute concentrations. CSF is thought to be derived primarily from secretion by the choroid plexuses. Flow rates measured using phase contrast magnetic resonance imaging reveal CSF movements to be more rapid and variable than previously supposed, even implying that under some circumstances net flow through the cerebral aqueduct may be reversed with net flow into the third and lateral ventricles. Such reversed flow requires there to be alternative sites for both generation and removal of CSF. Fluorescent tracer analysis has shown that fluid flow can occur from CSF into parenchyma along periarterial spaces. Whether this represents net fluid flow and whether there is subsequent flow through the interstitium and net flow out of the cortex via perivenous routes, described as glymphatic circulation, remains to be established. Modern techniques have revealed complex fluid movements within the brain. This review provides a critical evaluation of the data.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Margery A Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| |
Collapse
|
247
|
Aβ immunotherapy for Alzheimer's disease: effects on apoE and cerebral vasculopathy. Acta Neuropathol 2014; 128:777-89. [PMID: 25195061 DOI: 10.1007/s00401-014-1340-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/30/2022]
Abstract
Aβ immunotherapy for Alzheimer's disease (AD) results in the removal of Aβ plaques and increased cerebral amyloid angiopathy (CAA). In current clinical trials, amyloid-related imaging abnormalities (ARIAs), putatively due to exacerbation of CAA, are concerning side effects. We aimed to assess the role of the Aβ transporter apolipoprotein E (apoE) in the exacerbation of CAA and development of CAA-associated vasculopathy after Aβ immunotherapy. 12 Aβ42-immunized AD (iAD; AN1792, Elan Pharmaceuticals) cases were compared with 28 unimmunized AD (cAD) cases. Immunohistochemistry was quantified for Aβ42, apoE, apoE E4 and smooth muscle actin, and CAA-associated vasculopathy was analyzed. Aβ immunotherapy was associated with redistribution of apoE from cortical plaques to cerebral vessel walls, mirroring the altered distribution of Aβ42. Concentric vessel wall splitting was increased threefold in leptomeningeal vessels after immunotherapy (cAD 6.3 vs iAD 20.6 %, P < 0.001), but smooth muscle cell abnormalities did not differ. The findings suggest that apoE is involved in the removal of plaques and transport of Aβ to the cerebral vasculature induced by Aβ immunotherapy. Immunotherapy was not associated with CAA-related vascular smooth muscle damage, but was accompanied by increased splitting of the vessel wall, perhaps reflecting enhanced deposition and subsequent removal of Aβ. ARIA occurring in some current trials of Aβ immunotherapy may reflect an extreme form of these vascular changes.
Collapse
|
248
|
Capel C, Makki M, Gondry-Jouet C, Bouzerar R, Courtois V, Krejpowicz B, Balédent O. Insights into cerebrospinal fluid and cerebral blood flows in infants and young children. J Child Neurol 2014; 29:1608-15. [PMID: 24346313 DOI: 10.1177/0883073813511854] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
This study investigates the craniospinal flows of blood and cerebrospinal fluid using phase-contrast magnetic resonance imaging (MRI) on 23 control neonates and infants (5 d-68 mo old). Mean arterial cerebral blood flow increased with age of infant from 180 mL/min after birth to 1330 mL/min around 6 years of age. This corresponds to 51 mL/min/100 g and 95 mL/min/100 g, respectively. Cervical cerebrospinal fluid stroke volume increased from 38 × 10(-3) mL to 752 × 10(-3) mL per cardiac cycle. After arterial systolic blood inflow, we observed a delay of the venous outflow that was always preceded by cerebrospinal fluid flushing out through the spinal canal. These results highlighted the importance of compliance of the spinal compartment and the interaction of blood and cerebrospinal fluid dynamics. The capacity of the spinal compartment to receive intracranial cerebrospinal fluid in presence of fontanels was demonstrated. We provide reference values to understand the physiology of cerebrospinal fluid and cerebral blood.
Collapse
Affiliation(s)
- Cyrille Capel
- Image Processing Unit, University Hospital, Amiens, France Bio Flow Image, Research group of Picardie Jules Verne, France Neurosurgery Unit, University Hospital, Amiens, France
| | - Malek Makki
- MRI Research, University Children Hospital, Zurich, Switzerland
| | - Catherine Gondry-Jouet
- Bio Flow Image, Research group of Picardie Jules Verne, France Radiology Unit, University Hospital, Amiens, France
| | - Roger Bouzerar
- Image Processing Unit, University Hospital, Amiens, France Bio Flow Image, Research group of Picardie Jules Verne, France
| | - Véronique Courtois
- Ostéobio, Ecole supérieure d'ostéopathie et de biomécanique, Paris, France
| | | | - Olivier Balédent
- Image Processing Unit, University Hospital, Amiens, France Bio Flow Image, Research group of Picardie Jules Verne, France
| |
Collapse
|
249
|
Recent advances in the role of toll-like receptors and TLR agonists in immunotherapy for human glioma. Protein Cell 2014; 5:899-911. [PMID: 25411122 PMCID: PMC4259890 DOI: 10.1007/s13238-014-0112-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 09/30/2014] [Indexed: 02/06/2023] Open
Abstract
Gliomas are extremely aggressive brain tumors with a very poor prognosis. One of the more promising strategies for the treatment of human gliomas is targeted immunotherapy where antigens that are unique to the tumors are exploited to generate vaccines. The approach, however, is complicated by the fact that human gliomas escape immune surveillance by creating an immune suppressed microenvironment. In order to oppose the glioma imposed immune suppression, molecules and pathways involved in immune cell maturation, expansion, and migration are under intensive clinical investigation as adjuvant therapy. Toll-like receptors (TLRs) mediate many of these functions in immune cell types, and TLR agonists, thus, are currently primary candidate molecules to be used as important adjuvants in a variety of cancers. In animal models for glioma, TLR agonists have exhibited antitumor properties by facilitating antigen presentation and stimulating innate and adaptive immunity. In clinical trials, several TLR agonists have achieved survival benefit, and many more trials are recruiting or ongoing. However, a second complicating factor is that TLRs are also expressed on cancer cells where they can participate instead in a variety of tumor promoting activities including cell growth, proliferation, invasion, migration, and even stem cell maintenance. TLR agonists can, therefore, possibly play dual roles in tumor biology. Here, how TLRs and TLR agonists function in glioma biology and in anti-glioma therapies is summarized in an effort to provide a current picture of the sophisticated relationship of glioma with the immune system and the implications for immunotherapy.
Collapse
|
250
|
Nelson ES, Mulugeta L, Myers JG. Microgravity-induced fluid shift and ophthalmic changes. Life (Basel) 2014; 4:621-65. [PMID: 25387162 PMCID: PMC4284461 DOI: 10.3390/life4040621] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 09/17/2014] [Accepted: 10/17/2014] [Indexed: 11/16/2022] Open
Abstract
Although changes to visual acuity in spaceflight have been observed in some astronauts since the early days of the space program, the impact to the crew was considered minor. Since that time, missions to the International Space Station have extended the typical duration of time spent in microgravity from a few days or weeks to many months. This has been accompanied by the emergence of a variety of ophthalmic pathologies in a significant proportion of long-duration crewmembers, including globe flattening, choroidal folding, optic disc edema, and optic nerve kinking, among others. The clinical findings of affected astronauts are reminiscent of terrestrial pathologies such as idiopathic intracranial hypertension that are characterized by high intracranial pressure. As a result, NASA has placed an emphasis on determining the relevant factors and their interactions that are responsible for detrimental ophthalmic response to space. This article will describe the Visual Impairment and Intracranial Pressure syndrome, link it to key factors in physiological adaptation to the microgravity environment, particularly a cephalad shifting of bodily fluids, and discuss the implications for ocular biomechanics and physiological function in long-duration spaceflight.
Collapse
Affiliation(s)
- Emily S Nelson
- NASA Glenn Research Center, 21000 Brookpark Rd., Cleveland, OH 44135, USA.
| | - Lealem Mulugeta
- Universities Space Research Association, Division of Space Life Sciences, 3600 Bay Area Boulevard, Houston, TX 77058, USA.
| | - Jerry G Myers
- NASA Glenn Research Center, 21000 Brookpark Rd., Cleveland, OH 44135, USA.
| |
Collapse
|