1
|
Pham C, Komaki Y, Deàs-Just A, Le Gac B, Mouffle C, Franco C, Chaperon A, Vialou V, Tsurugizawa T, Cauli B, Li D. Astrocyte aquaporin mediates a tonic water efflux maintaining brain homeostasis. eLife 2024; 13:RP95873. [PMID: 39508543 DOI: 10.7554/elife.95873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Brain water homeostasis not only provides a physical protection, but also determines the diffusion of chemical molecules key for information processing and metabolic stability. As a major type of glia in brain parenchyma, astrocytes are the dominant cell type expressing aquaporin water channel. How astrocyte aquaporin contributes to brain water homeostasis in basal physiology remains to be understood. We report that astrocyte aquaporin 4 (AQP4) mediates a tonic water efflux in basal conditions. Acute inhibition of astrocyte AQP4 leads to intracellular water accumulation as optically resolved by fluorescence-translated imaging in acute brain slices, and in vivo by fiber photometry in mobile mice. We then show that aquaporin-mediated constant water efflux maintains astrocyte volume and osmotic equilibrium, astrocyte and neuron Ca2+ signaling, and extracellular space remodeling during optogenetically induced cortical spreading depression. Using diffusion-weighted magnetic resonance imaging (DW-MRI), we observed that in vivo inhibition of AQP4 water efflux heterogeneously disturbs brain water homeostasis in a region-dependent manner. Our data suggest that astrocyte aquaporin, though bidirectional in nature, mediates a tonic water outflow to sustain cellular and environmental equilibrium in brain parenchyma.
Collapse
Affiliation(s)
- Cuong Pham
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Yuji Komaki
- Central Institute for Experimental Medicine and Life Science, Kawasaki, Japan
| | - Anna Deàs-Just
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Benjamin Le Gac
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Christine Mouffle
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Clara Franco
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Agnès Chaperon
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Vincent Vialou
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Tomokazu Tsurugizawa
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
- Faculty of Engineering, University of Tsukuba, Tsukuba, Japan
| | - Bruno Cauli
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| | - Dongdong Li
- Sorbonne Université - CNRS - INSERM, Institut de Biologie Paris Seine, Neuroscience Paris Seine, Paris, France
| |
Collapse
|
2
|
Hladky SB, Barrand MA. Alterations in brain fluid physiology during the early stages of development of ischaemic oedema. Fluids Barriers CNS 2024; 21:51. [PMID: 38858667 PMCID: PMC11163777 DOI: 10.1186/s12987-024-00534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/22/2024] [Indexed: 06/12/2024] Open
Abstract
Oedema occurs when higher than normal amounts of solutes and water accumulate in tissues. In brain parenchymal tissue, vasogenic oedema arises from changes in blood-brain barrier permeability, e.g. in peritumoral oedema. Cytotoxic oedema arises from excess accumulation of solutes within cells, e.g. ischaemic oedema following stroke. This type of oedema is initiated when blood flow in the affected core region falls sufficiently to deprive brain cells of the ATP needed to maintain ion gradients. As a consequence, there is: depolarization of neurons; neural uptake of Na+ and Cl- and loss of K+; neuronal swelling; astrocytic uptake of Na+, K+ and anions; swelling of astrocytes; and reduction in ISF volume by fluid uptake into neurons and astrocytes. There is increased parenchymal solute content due to metabolic osmolyte production and solute influx from CSF and blood. The greatly increased [K+]isf triggers spreading depolarizations into the surrounding penumbra increasing metabolic load leading to increased size of the ischaemic core. Water enters the parenchyma primarily from blood, some passing into astrocyte endfeet via AQP4. In the medium term, e.g. after three hours, NaCl permeability and swelling rate increase with partial opening of tight junctions between blood-brain barrier endothelial cells and opening of SUR1-TPRM4 channels. Swelling is then driven by a Donnan-like effect. Longer term, there is gross failure of the blood-brain barrier. Oedema resolution is slower than its formation. Fluids without colloid, e.g. infused mock CSF, can be reabsorbed across the blood-brain barrier by a Starling-like mechanism whereas infused serum with its colloids must be removed by even slower extravascular means. Large scale oedema can increase intracranial pressure (ICP) sufficiently to cause fatal brain herniation. The potentially lethal increase in ICP can be avoided by craniectomy or by aspiration of the osmotically active infarcted region. However, the only satisfactory treatment resulting in retention of function is restoration of blood flow, providing this can be achieved relatively quickly. One important objective of current research is to find treatments that increase the time during which reperfusion is successful. Questions still to be resolved are discussed.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, Tennis Court Rd., Cambridge, CB2 1PD, UK.
| | - Margery A Barrand
- Department of Pharmacology, Tennis Court Rd., Cambridge, CB2 1PD, UK
| |
Collapse
|
3
|
Li J, Jia S, Song Y, Xu W, Lin J. Ginkgolide B can alleviate spinal cord glymphatic system dysfunction and provide neuroprotection in painful diabetic neuropathy rats by inhibiting matrix metalloproteinase-9. Neuropharmacology 2024; 250:109907. [PMID: 38492884 DOI: 10.1016/j.neuropharm.2024.109907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
The glymphatic system plays a crucial role in maintaining optimal central nervous system (CNS) function by facilitating the removal of metabolic wastes. Aquaporin-4 (AQP4) protein, predominantly located on astrocyte end-feet, is a key pathway for metabolic waste excretion. β-Dystroglycan (β-DG) can anchor AQP4 protein to the end-feet membrane of astrocytes and can be cleaved by matrix metalloproteinase (MMP)-9 protein. Studies have demonstrated that hyperglycemia upregulates MMP-9 expression in the nervous system, leading to neuropathic pain. Ginkgolide B (GB) exerts an inhibitory effect on the MMP-9 protein. In this study, we investigated whether inhibition of MMP-9-mediated β-DG cleavage by GB is involved in the regulation of AQP4 polarity within the glymphatic system in painful diabetic neuropathy (PDN) and exerts neuroprotective effects. The PDN model was established by injecting streptozotocin (STZ). Functional changes in the glymphatic system were observed using magnetic resonance imaging (MRI). The paw withdrawal threshold (PWT) was measured to assess mechanical allodynia. The protein expressions of MMP-9, β-DG, and AQP4 were detected by Western blotting and immunofluorescence. Our findings revealed significant decreases in the efficiency of contrast agent clearance within the spinal glymphatic system of the rats, accompanied by decreased PWT, increased MMP-9 protein expression, decreased β-DG protein expression, and loss of AQP4 polarity. Notably, GB treatment demonstrated the capacity to ameliorate spinal cord glymphatic function by modulating AQP4 polarity through MMP-9 inhibition, offering a promising therapeutic avenue for PDN.
Collapse
Affiliation(s)
- Jiang Li
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | - Shuaiying Jia
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | | | - Wenmei Xu
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| | - Jingyan Lin
- Department of Anesthesiology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
4
|
Tong XJ, Akdemir G, Wadhwa M, Verkman AS, Smith AJ. Large molecules from the cerebrospinal fluid enter the optic nerve but not the retina of mice. Fluids Barriers CNS 2024; 21:1. [PMID: 38178155 PMCID: PMC10768282 DOI: 10.1186/s12987-023-00506-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
It has been proposed that cerebrospinal fluid (CSF) can enter and leave the retina and optic nerve along perivascular spaces surrounding the central retinal vessels as part of an aquaporin-4 (AQP4) dependent ocular 'glymphatic' system. Here, we injected fluorescent dextrans and antibodies into the CSF of mice at the cisterna magna and measured their distribution in the optic nerve and retina. We found that uptake of dextrans in the perivascular spaces and parenchyma of the optic nerve is highly sensitive to the cisternal injection rate, where high injection rates, in which dextran disperses fully in the sub-arachnoid space, led to uptake along the full length of the optic nerve. Accumulation of dextrans in the optic nerve did not differ significantly in wild-type and AQP4 knockout mice. Dextrans did not enter the retina, even when intracranial pressure was greatly increased over intraocular pressure. However, elevation of intraocular pressure reduced accumulation of fluorescent dextrans in the optic nerve head, and intravitreally injected dextrans left the retina via perivascular spaces surrounding the central retinal vessels. Human IgG distributed throughout the perivascular and parenchymal areas of the optic nerve to a similar extent as dextran following cisternal injection. However, uptake of a cisternally injected AQP4-IgG antibody, derived from a seropositive neuromyelitis optica spectrum disorder subject, was limited by AQP4 binding. We conclude that large molecules injected in the CSF can accumulate along the length of the optic nerve if they are fully dispersed in the optic nerve sub-arachnoid space but that they do not enter the retina.
Collapse
Affiliation(s)
- Xiao J Tong
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, 94131, USA
| | - Gokhan Akdemir
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA, 94131, USA
| | - Meetu Wadhwa
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA, 94131, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA, 94131, USA
| | - Alex J Smith
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, 94131, USA.
| |
Collapse
|
5
|
Vinje V, Zapf B, Ringstad G, Eide PK, Rognes ME, Mardal KA. Human brain solute transport quantified by glymphatic MRI-informed biophysics during sleep and sleep deprivation. Fluids Barriers CNS 2023; 20:62. [PMID: 37596635 PMCID: PMC10439559 DOI: 10.1186/s12987-023-00459-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/21/2023] [Indexed: 08/20/2023] Open
Abstract
Whether you are reading, running or sleeping, your brain and its fluid environment continuously interacts to distribute nutrients and clear metabolic waste. Yet, the precise mechanisms for solute transport within the human brain have remained hard to quantify using imaging techniques alone. From multi-modal human brain MRI data sets in sleeping and sleep-deprived subjects, we identify and quantify CSF tracer transport parameters using forward and inverse subject-specific computational modelling. Our findings support the notion that extracellular diffusion alone is not sufficient as a brain-wide tracer transport mechanism. Instead, we show that human MRI observations align well with transport by either by an effective diffusion coefficent 3.5[Formula: see text] that of extracellular diffusion in combination with local clearance rates corresponding to a tracer half-life of up to 5 h, or by extracellular diffusion augmented by advection with brain-wide average flow speeds on the order of 1-9 [Formula: see text]m/min. Reduced advection fully explains reduced tracer clearance after sleep-deprivation, supporting the role of sleep and sleep deprivation on human brain clearance.
Collapse
Affiliation(s)
- Vegard Vinje
- Simula Research Laboratory, Kristian Augusts gate 23, 0164, Oslo, Norway
- Expert Analytics AS, Møllergata 8, 0179, Oslo, Norway
| | - Bastian Zapf
- Department of Mathematics, University of Oslo, Oslo, Norway
| | - Geir Ringstad
- Department of Radiology, Oslo University Hospital - Rikshospitalet, Oslo, Norway
- Department of Geriatrics and Internal Medicine, Sørlandet Hospital, Arendal, Norway
| | - Per Kristian Eide
- Department of Neurosurgery, Oslo University Hospital - Rikshospitalet, Oslo, Norway
| | - Marie E Rognes
- Simula Research Laboratory, Kristian Augusts gate 23, 0164, Oslo, Norway
| | - Kent-Andre Mardal
- Simula Research Laboratory, Kristian Augusts gate 23, 0164, Oslo, Norway.
- Department of Mathematics, University of Oslo, Oslo, Norway.
| |
Collapse
|
6
|
Poulain A, Riseth J, Vinje V. Multi-compartmental model of glymphatic clearance of solutes in brain tissue. PLoS One 2023; 18:e0280501. [PMID: 36881576 PMCID: PMC9990927 DOI: 10.1371/journal.pone.0280501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/02/2023] [Indexed: 03/08/2023] Open
Abstract
The glymphatic system is the subject of numerous pieces of research in biology. Mathematical modelling plays a considerable role in this field since it can indicate the possible physical effects of this system and validate the biologists' hypotheses. The available mathematical models that describe the system at the scale of the brain (i.e. the macroscopic scale) are often solely based on the diffusion equation and do not consider the fine structures formed by the perivascular spaces. We therefore propose a mathematical model representing the time and space evolution of a mixture flowing through multiple compartments of the brain. We adopt a macroscopic point of view in which the compartments are all present at any point in space. The equations system is composed of two coupled equations for each compartment: One equation for the pressure of a fluid and one for the mass concentration of a solute. The fluid and solute can move from one compartment to another according to certain membrane conditions modelled by transfer functions. We propose to apply this new modelling framework to the clearance of 14C-inulin from the rat brain.
Collapse
Affiliation(s)
- Alexandre Poulain
- Laboratoire Paul Painlevé, UMR 8524 CNRS, Université de Lille, Lille, France
- Department for Numerical Analysis and Scientific Computing, Simula Research Laboratory, Oslo, Norway
| | - Jørgen Riseth
- Department of Mathematics, University of Oslo, Oslo, Norway
- Department for Numerical Analysis and Scientific Computing, Simula Research Laboratory, Oslo, Norway
| | - Vegard Vinje
- Department for Numerical Analysis and Scientific Computing, Simula Research Laboratory, Oslo, Norway
| |
Collapse
|
7
|
Han W, Song Y, Rocha M, Shi Y. Ischemic brain edema: Emerging cellular mechanisms and therapeutic approaches. Neurobiol Dis 2023; 178:106029. [PMID: 36736599 DOI: 10.1016/j.nbd.2023.106029] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/14/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Brain edema is one of the most devastating consequences of ischemic stroke. Malignant cerebral edema is the main reason accounting for the high mortality rate of large hemispheric strokes. Despite decades of tremendous efforts to elucidate mechanisms underlying the formation of ischemic brain edema and search for therapeutic targets, current treatments for ischemic brain edema remain largely symptom-relieving rather than aiming to stop the formation and progression of edema. Recent preclinical research reveals novel cellular mechanisms underlying edema formation after brain ischemia and reperfusion. Advancement in neuroimaging techniques also offers opportunities for early diagnosis and prediction of malignant brain edema in stroke patients to rapidly adopt life-saving surgical interventions. As reperfusion therapies become increasingly used in clinical practice, understanding how therapeutic reperfusion influences the formation of cerebral edema after ischemic stroke is critical for decision-making and post-reperfusion management. In this review, we summarize these research advances in the past decade on the cellular mechanisms, and evaluation, prediction, and intervention of ischemic brain edema in clinical settings, aiming to provide insight into future preclinical and clinical research on the diagnosis and treatment of brain edema after stroke.
Collapse
Affiliation(s)
- Wenxuan Han
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Yang Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Marcelo Rocha
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Yejie Shi
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States of America.
| |
Collapse
|
8
|
Semyachkina-Glushkovskaya OV, Postnov DE, Khorovodov AP, Navolokin NA, Kurthz JHG. Lymphatic Drainage System of the Brain: a New Player in Neuroscience. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s0022093023010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
|
9
|
Investigating molecular transport in the human brain from MRI with physics-informed neural networks. Sci Rep 2022; 12:15475. [PMID: 36104360 PMCID: PMC9474534 DOI: 10.1038/s41598-022-19157-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
In recent years, a plethora of methods combining neural networks and partial differential equations have been developed. A widely known example are physics-informed neural networks, which solve problems involving partial differential equations by training a neural network. We apply physics-informed neural networks and the finite element method to estimate the diffusion coefficient governing the long term spread of molecules in the human brain from magnetic resonance images. Synthetic testcases are created to demonstrate that the standard formulation of the physics-informed neural network faces challenges with noisy measurements in our application. Our numerical results demonstrate that the residual of the partial differential equation after training needs to be small for accurate parameter recovery. To achieve this, we tune the weights and the norms used in the loss function and use residual based adaptive refinement of training points. We find that the diffusion coefficient estimated from magnetic resonance images with physics-informed neural networks becomes consistent with results from a finite element based approach when the residuum after training becomes small. The observations presented here are an important first step towards solving inverse problems on cohorts of patients in a semi-automated fashion with physics-informed neural networks.
Collapse
|
10
|
Kimball EC, Quillen S, Pease ME, Keuthan C, Nagalingam A, Zack DJ, Johnson TV, Quigley HA. Aquaporin 4 is not present in normal porcine and human lamina cribrosa. PLoS One 2022; 17:e0268541. [PMID: 35709078 PMCID: PMC9202842 DOI: 10.1371/journal.pone.0268541] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/03/2022] [Indexed: 02/02/2023] Open
Abstract
Aquaporin 4 is absent from astrocytes in the rodent optic nerve head, despite high expression in the retina and myelinated optic nerve. The purpose of this study was to quantify regional aquaporin channel expression in astrocytes of the porcine and human mouse optic nerve (ON). Ocular tissue sections were immunolabeled for aquaporins 1(AQP1), 4(AQP4), and 9(AQP9), myelin basic protein (MBP), glial fibrillary acidic protein (GFAP) and alpha-dystroglycan (αDG) for their presence in retina, lamina, myelin transition zone (MTZ, region just posterior to lamina) and myelinated ON (MON). Semi- quantification of AQP4 labeling & real-time quantitative PCR (qPCR) data were analyzed in retina and ON tissue. Porcine and control human eyes had abundant AQP4 in Müller cells, retinal astrocytes, and myelinated ON (MON), but minimal expression in the lamina cribrosa. AQP1 and AQP9 were present in retina, but not in the lamina. Immunolabeling of GFAP and αDG was similar in lamina, myelin transition zone (MTZ) and MON regions. Semi-quantitative AQP4 labeling was at background level in lamina, increasing in the MTZ, and highest in the MON (lamina vs MTZ, MON; p≤0.05, p≤0.01, respectively). Expression of AQP4 mRNA was minimal in lamina and substantial in MTZ and MON, while GFAP mRNA expression was uniform among the lamina, MTZ, and MON regions. Western blot assay showed AQP4 protein expression in the MON samples, but none was detected in the lamina tissue. The minimal presence of AQP4 in the lamina is a specific regional phenotype of astrocytes in the mammalian optic nerve head.
Collapse
Affiliation(s)
- Elizabeth C. Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sarah Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mary E. Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Casey Keuthan
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Aru Nagalingam
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Donald J. Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Thomas V. Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Harry A. Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
11
|
Kedarasetti RT, Drew PJ, Costanzo F. Arterial vasodilation drives convective fluid flow in the brain: a poroelastic model. Fluids Barriers CNS 2022; 19:34. [PMID: 35570287 PMCID: PMC9107702 DOI: 10.1186/s12987-022-00326-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/29/2022] [Indexed: 01/26/2023] Open
Abstract
The movement of fluid into, through, and out of the brain plays an important role in clearing metabolic waste. However, there is controversy regarding the mechanisms driving fluid movement in the fluid-filled paravascular spaces (PVS), and whether the movement of metabolic waste in the brain extracellular space (ECS) is primarily driven by diffusion or convection. The dilation of penetrating arterioles in the brain in response to increases in neural activity (neurovascular coupling) is an attractive candidate for driving fluid circulation, as it drives deformation of the brain tissue and of the PVS around arteries, resulting in fluid movement. We simulated the effects of vasodilation on fluid movement into and out of the brain ECS using a novel poroelastic model of brain tissue. We found that arteriolar dilations could drive convective flow through the ECS radially outward from the arteriole, and that this flow is sensitive to the dynamics of the dilation. Simulations of sleep-like conditions, with larger vasodilations and increased extracellular volume in the brain showed enhanced movement of fluid from the PVS into the ECS. Our simulations suggest that both sensory-evoked and sleep-related arteriolar dilations can drive convective flow of cerebrospinal fluid not just in the PVS, but also into the ECS through the PVS around arterioles.
Collapse
Affiliation(s)
- Ravi Teja Kedarasetti
- grid.29857.310000 0001 2097 4281Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| | - Patrick J. Drew
- grid.29857.310000 0001 2097 4281Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Center for Neural Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Neurosurgery, Pennsylvania State University, University Park, PA USA
| | - Francesco Costanzo
- grid.29857.310000 0001 2097 4281Department of Engineering Science and Mechanics, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Center for Neural Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA ,grid.29857.310000 0001 2097 4281Department of Mathematics, Pennsylvania State University, University Park, PA USA
| |
Collapse
|
12
|
Salman MM, Kitchen P, Halsey A, Wang MX, Törnroth-Horsefield S, Conner AC, Badaut J, Iliff JJ, Bill RM. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis. Brain 2022; 145:64-75. [PMID: 34499128 PMCID: PMC9088512 DOI: 10.1093/brain/awab311] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 06/28/2021] [Accepted: 07/31/2021] [Indexed: 11/25/2022] Open
Abstract
Aquaporin channels facilitate bidirectional water flow in all cells and tissues. AQP4 is highly expressed in astrocytes. In the CNS, it is enriched in astrocyte endfeet, at synapses, and at the glia limitans, where it mediates water exchange across the blood-spinal cord and blood-brain barriers (BSCB/BBB), and controls cell volume, extracellular space volume, and astrocyte migration. Perivascular enrichment of AQP4 at the BSCB/BBB suggests a role in glymphatic function. Recently, we have demonstrated that AQP4 localization is also dynamically regulated at the subcellular level, affecting membrane water permeability. Ageing, cerebrovascular disease, traumatic CNS injury, and sleep disruption are established and emerging risk factors in developing neurodegeneration, and in animal models of each, impairment of glymphatic function is associated with changes in perivascular AQP4 localization. CNS oedema is caused by passive water influx through AQP4 in response to osmotic imbalances. We have demonstrated that reducing dynamic relocalization of AQP4 to the BSCB/BBB reduces CNS oedema and accelerates functional recovery in rodent models. Given the difficulties in developing pore-blocking AQP4 inhibitors, targeting AQP4 subcellular localization opens up new treatment avenues for CNS oedema, neurovascular and neurodegenerative diseases, and provides a framework to address fundamental questions about water homeostasis in health and disease.
Collapse
Affiliation(s)
- Mootaz M Salman
- Department of Physiology, Anatomy and Genetics,
University of Oxford, Oxford OX1 3PT, UK
| | - Philip Kitchen
- School of Biosciences, College of Health and Life
Sciences, Aston University, Aston Triangle,
Birmingham B4 7ET, UK
| | - Andrea Halsey
- Institute of Clinical Sciences, College of Medical
and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham B15 2TT, UK
| | - Marie Xun Wang
- Department of Psychiatry and Behavioral Sciences,
University of Washington School of Medicine, Seattle, WA, USA
| | | | - Alex C Conner
- Institute of Clinical Sciences, College of Medical
and Dental Sciences, University of Birmingham,
Edgbaston, Birmingham B15 2TT, UK
| | - Jerome Badaut
- CNRS-UMR 5536-Centre de Résonance
Magnétique des systèmes Biologiques, Université de
Bordeaux, 33076 Bordeaux, France
| | - Jeffrey J Iliff
- Department of Psychiatry and Behavioral Sciences,
University of Washington School of Medicine, Seattle, WA, USA
- Department of Neurology, University of Washington
School of Medicine, Seattle, WA, USA
- VISN 20 Mental Illness Research, Education and
Clinical Center, VA Puget Sound Health Care System, Seattle, WA,
USA
| | - Roslyn M Bill
- School of Biosciences, College of Health and Life
Sciences, Aston University, Aston Triangle,
Birmingham B4 7ET, UK
| |
Collapse
|
13
|
Hladky SB, Barrand MA. The glymphatic hypothesis: the theory and the evidence. Fluids Barriers CNS 2022; 19:9. [PMID: 35115036 PMCID: PMC8815211 DOI: 10.1186/s12987-021-00282-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022] Open
Abstract
The glymphatic hypothesis proposes a mechanism for extravascular transport into and out of the brain of hydrophilic solutes unable to cross the blood-brain barrier. It suggests that there is a circulation of fluid carrying solutes inwards via periarterial routes, through the interstitium and outwards via perivenous routes. This review critically analyses the evidence surrounding the mechanisms involved in each of these stages. There is good evidence that both influx and efflux of solutes occur along periarterial routes but no evidence that the principal route of outflow is perivenous. Furthermore, periarterial inflow of fluid is unlikely to be adequate to provide the outflow that would be needed to account for solute efflux. A tenet of the hypothesis is that flow sweeps solutes through the parenchyma. However, the velocity of any possible circulatory flow within the interstitium is too small compared to diffusion to provide effective solute movement. By comparison the earlier classical hypothesis describing extravascular transport proposed fluid entry into the parenchyma across the blood-brain barrier, solute movements within the parenchyma by diffusion, and solute efflux partly by diffusion near brain surfaces and partly carried by flow along "preferred routes" including perivascular spaces, white matter tracts and subependymal spaces. It did not suggest fluid entry via periarterial routes. Evidence is still incomplete concerning the routes and fate of solutes leaving the brain. A large proportion of the solutes eliminated from the parenchyma go to lymph nodes before reaching blood but the proportions delivered directly to lymph or indirectly via CSF which then enters lymph are as yet unclear. In addition, still not understood is why and how the absence of AQP4 which is normally highly expressed on glial endfeet lining periarterial and perivenous routes reduces rates of solute elimination from the parenchyma and of solute delivery to it from remote sites of injection. Neither the glymphatic hypothesis nor the earlier classical hypothesis adequately explain how solutes and fluid move into, through and out of the brain parenchyma. Features of a more complete description are discussed. All aspects of extravascular transport require further study.
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
|
14
|
Abstract
The accumulation of neurotoxic amyloid-beta (Aβ) in the brain is one of the characteristic hallmarks of Alzheimer's disease (AD). Aβ-peptide brain homeostasis is governed by its production and various clearance mechanisms. The blood-brain barrier provides a large surface area for influx and efflux mechanisms into and out of the brain. Different transporters and receptors have been implicated to play crucial roles in Aβ clearance from brain. Besides Aβ transport, the blood-brain barrier tightly regulates the brain's microenvironment; however, vascular alterations have been shown in patients with AD. Here, we summarize how the blood-brain barrier changes during aging and in disease and focus on recent findings of how the ABC transporter P-glycoprotein (ABCB1/P-gp) and the receptor low-density lipoprotein receptor-related protein 1 (LRP1) play a role in Aβ clearance from brain.
Collapse
|
15
|
Omileke D, Bothwell SW, Pepperall D, Beard DJ, Coupland K, Patabendige A, Spratt NJ. Decreased Intracranial Pressure Elevation and Cerebrospinal Fluid Outflow Resistance: A Potential Mechanism of Hypothermia Cerebroprotection Following Experimental Stroke. Brain Sci 2021; 11:brainsci11121589. [PMID: 34942890 PMCID: PMC8699790 DOI: 10.3390/brainsci11121589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Elevated intracranial pressure (ICP) occurs 18–24 h after ischaemic stroke and is implicated as a potential cause of early neurological deterioration. Increased resistance to cerebrospinal fluid (CSF) outflow after ischaemic stroke is a proposed mechanism for ICP elevation. Ultra-short duration hypothermia prevents ICP elevation 24 h post-stroke in rats. We aimed to determine whether hypothermia would reduce CSF outflow resistance post-stroke. Methods: Transient middle cerebral artery occlusion was performed, followed by gradual cooling to 33 °C. At 18 h post-stroke, CSF outflow resistance was measured using a steady-state infusion method. Results: Hypothermia to 33 °C prevented ICP elevation 18 h post-stroke (hypothermia ∆ICP = 0.8 ± 3.6 mmHg vs. normothermia ∆ICP = 4.4 ± 2.0 mmHg, p = 0.04) and reduced infarct volume 24 h post-stroke (hypothermia = 78.6 ± 21.3 mm3 vs. normothermia = 108.1 ± 17.8 mm3; p = 0.01). Hypothermia to 33 °C did not result in a significant reduction in CSF outflow resistance compared with normothermia controls (0.32 ± 0.36 mmHg/µL/min vs. 1.07 ± 0.99 mmHg/µL/min, p = 0.06). Conclusions: Hypothermia treatment was protective in terms of ICP rise prevention, infarct volume reduction, and may be implicated in CSF outflow resistance post-stroke. Further investigations are warranted to elucidate the mechanisms of ICP elevation and hypothermia treatment.
Collapse
Affiliation(s)
- Daniel Omileke
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (D.O.); (S.W.B.); (D.P.); (D.J.B.); (K.C.)
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Steven W. Bothwell
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (D.O.); (S.W.B.); (D.P.); (D.J.B.); (K.C.)
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Debbie Pepperall
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (D.O.); (S.W.B.); (D.P.); (D.J.B.); (K.C.)
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Daniel J. Beard
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (D.O.); (S.W.B.); (D.P.); (D.J.B.); (K.C.)
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Kirsten Coupland
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (D.O.); (S.W.B.); (D.P.); (D.J.B.); (K.C.)
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Adjanie Patabendige
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (D.O.); (S.W.B.); (D.P.); (D.J.B.); (K.C.)
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Wirral CH64 7TE, UK
- Department of Biology, Edge Hill University, Ormskirk L39 4QP, UK
- Correspondence: (A.P.); (N.J.S.)
| | - Neil J. Spratt
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia; (D.O.); (S.W.B.); (D.P.); (D.J.B.); (K.C.)
- Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
- Hunter New England Local Health District, New Lambton Heights, Newcastle, NSW 2305, Australia
- Correspondence: (A.P.); (N.J.S.)
| |
Collapse
|
16
|
Wafford KA. Aberrant waste disposal in neurodegeneration: why improved sleep could be the solution. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100025. [PMID: 36324713 PMCID: PMC9616228 DOI: 10.1016/j.cccb.2021.100025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 06/16/2023]
Abstract
Sleep takes up a large percentage of our lives and the full functions of this state are still not understood. However, over the last 10 years a new and important function has emerged as a mediator of brain clearance. Removal of toxic metabolites and proteins from the brain parenchyma generated during waking activity and high levels of synaptic processing is critical to normal brain function and only enabled during deep sleep. Understanding of this process is revealing how impaired sleep contributes an important and likely causative role in the accumulation and aggregation of aberrant proteins such as β-amyloid and phosphorylated tau, as well as inflammation and neuronal damage. We are also beginning to understand how brain slow-wave activity interacts with vascular function allowing the flow of CSF and interstitial fluid to drain into the body's lymphatic system. New methodology is enabling visualization of this process in both animals and humans and is revealing how these processes break down during ageing and disease. With this understanding we can begin to envisage novel therapeutic approaches to the treatment of neurodegeneration, and how reversing sleep impairment in the correct manner may provide a way to slow these processes and improve brain function.
Collapse
Key Words
- AQP4, aquaporin-4
- Alzheimer's disease
- Amyloid
- Aquaporin-4
- Astrocyte
- Aβ, beta amyloid
- BOLD, blood-oxygen level dependent imaging
- CAA, cerebral amyloid angiopathy
- CSF, Cerebrospinal fluid
- Clearance
- EEG, electroencephalography
- EMG, electromyography
- Glymphatic
- ISF, interstitial fluid
- MCI, mild cognitive impairment
- MRI, magnetic resonance imaging
- NOS, nitric oxide synthase
- NREM, non-rapid eye movement
- OSA, obstructive sleep apnea
- PET, positron emission tomography
- REM, rapid-eye movement
- SWA, slow wave activity
- SWS, slow-wave sleep
- Slow-wave sleep
- iNPH, idiopathic normal pressure hydrocephalus
Collapse
|
17
|
Smith AJ, Akdemir G, Wadhwa M, Song D, Verkman AS. Application of fluorescent dextrans to the brain surface under constant pressure reveals AQP4-independent solute uptake. J Gen Physiol 2021; 153:212385. [PMID: 34128962 PMCID: PMC8210582 DOI: 10.1085/jgp.202112898] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/11/2021] [Indexed: 12/26/2022] Open
Abstract
Extracellular solutes in the central nervous system are exchanged between the interstitial fluid, the perivascular compartment, and the cerebrospinal fluid (CSF). The "glymphatic" mechanism proposes that the astrocyte water channel aquaporin-4 (AQP4) is a major determinant of solute transport between the CSF and the interstitial space; however, this is controversial in part because of wide variance in experimental data on interstitial uptake of cisternally injected solutes. Here, we investigated the determinants of solute uptake in brain parenchyma following cisternal injection and reexamined the role of AQP4 using a novel constant-pressure method. In mice, increased cisternal injection rate, which modestly increased intracranial pressure, remarkably increased solute dispersion in the subarachnoid space and uptake in the cortical perivascular compartment. To investigate the role of AQP4 in the absence of confounding variations in pressure and CSF solute concentration over time and space, solutes were applied directly onto the brain surface after durotomy under constant external pressure. Pressure elevation increased solute penetration into the perivascular compartment but had little effect on parenchymal solute uptake. Solute penetration and uptake did not differ significantly between wild-type and AQP4 knockout mice. Our results offer an explanation for the variability in cisternal injection studies and indicate AQP4-independent solute transfer from the CSF to the interstitial space in mouse brain.
Collapse
Affiliation(s)
- Alex J Smith
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA.,Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA
| | - Gokhan Akdemir
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA
| | - Meetu Wadhwa
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA
| | - Dan Song
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA
| |
Collapse
|
18
|
Lymphatics in Eye Fluid Homeostasis: Minor Contributors or Significant Actors? BIOLOGY 2021; 10:biology10070582. [PMID: 34201989 PMCID: PMC8301034 DOI: 10.3390/biology10070582] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023]
Abstract
Lymphatic vessels exert major effects on the maintenance of interstitial fluid homeostasis, immune cell trafficking, lipid absorption, tumor progression and metastasis. Recently, novel functional roles for the lymphatic vasculature have emerged, which can be associated with pathological situations. Among them, lymphatics have been proposed to participate in eye aqueous humor drainage, with potential consequences on intraocular pressure, a main risk factor for progression of glaucoma disease. In this review, after the description of eye fluid dynamics, we provide an update on the data concerning the distribution of ocular lymphatics. Particular attention is given to the results of investigations allowing the three dimensional visualization of the ocular surface vasculature, and to the molecular mechanisms that have been characterized to regulate ocular lymphatic vessel development. The studies concerning the potential role of lymphatics in aqueous humor outflow are reported and discussed. We also considered the novel studies mentioning the existence of an ocular glymphatic system which may have, in connection with lymphatics, important repercussions in retinal clearance and in diseases affecting the eye posterior segment. Some remaining unsolved questions and new directions to explore are proposed to improve the knowledge about both lymphatic and glymphatic system interactions with eye fluid homeostasis.
Collapse
|
19
|
Ries M, Watts H, Mota BC, Lopez MY, Donat CK, Baxan N, Pickering JA, Chau TW, Semmler A, Gurung B, Aleksynas R, Abelleira-Hervas L, Iqbal SJ, Romero-Molina C, Hernandez-Mir G, d’Amati A, Reutelingsperger C, Goldfinger MH, Gentleman SM, Van Leuven F, Solito E, Sastre M. Annexin A1 restores cerebrovascular integrity concomitant with reduced amyloid-β and tau pathology. Brain 2021; 144:1526-1541. [PMID: 34148071 PMCID: PMC8262982 DOI: 10.1093/brain/awab050] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/27/2020] [Accepted: 12/09/2020] [Indexed: 12/05/2022] Open
Abstract
Alzheimer's disease, characterized by brain deposits of amyloid-β plaques and neurofibrillary tangles, is also linked to neurovascular dysfunction and blood-brain barrier breakdown, affecting the passage of substances into and out of the brain. We hypothesized that treatment of neurovascular alterations could be beneficial in Alzheimer's disease. Annexin A1 (ANXA1) is a mediator of glucocorticoid anti-inflammatory action that can suppress microglial activation and reduce blood-brain barrier leakage. We have reported recently that treatment with recombinant human ANXA1 (hrANXA1) reduced amyloid-β levels by increased degradation in neuroblastoma cells and phagocytosis by microglia. Here, we show the beneficial effects of hrANXA1 in vivo by restoring efficient blood-brain barrier function and decreasing amyloid-β and tau pathology in 5xFAD mice and Tau-P301L mice. We demonstrate that young 5xFAD mice already suffer cerebrovascular damage, while acute pre-administration of hrANXA1 rescued the vascular defects. Interestingly, the ameliorated blood-brain barrier permeability in young 5xFAD mice by hrANXA1 correlated with reduced brain amyloid-β load, due to increased clearance and degradation of amyloid-β by insulin degrading enzyme (IDE). The systemic anti-inflammatory properties of hrANXA1 were also observed in 5xFAD mice, increasing IL-10 and reducing TNF-α expression. Additionally, the prolonged treatment with hrANXA1 reduced the memory deficits and increased synaptic density in young 5xFAD mice. Similarly, in Tau-P301L mice, acute hrANXA1 administration restored vascular architecture integrity, affecting the distribution of tight junctions, and reduced tau phosphorylation. The combined data support the hypothesis that blood-brain barrier breakdown early in Alzheimer's disease can be restored by hrANXA1 as a potential therapeutic approach.
Collapse
Affiliation(s)
- Miriam Ries
- Department of Brain Sciences, Imperial College London, London, UK
| | - Helena Watts
- Department of Brain Sciences, Imperial College London, London, UK
| | - Bibiana C Mota
- Department of Brain Sciences, Imperial College London, London, UK
| | | | | | - Nicoleta Baxan
- Biological Imaging Centre, Imperial College London, London, UK
| | | | - Tsz Wing Chau
- Department of Brain Sciences, Imperial College London, London, UK
| | - Annika Semmler
- Department of Brain Sciences, Imperial College London, London, UK
| | - Brinda Gurung
- Department of Brain Sciences, Imperial College London, London, UK
| | | | | | | | | | | | - Antonio d’Amati
- William Harvey Research Institute, Queen Mary University London SMD, London, UK
| | - Chris Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | | | | | - Fred Van Leuven
- Experimental Genetics Group-LEGTEGG, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Egle Solito
- William Harvey Research Institute, Queen Mary University London SMD, London, UK
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Universitá degli Studi di Napoli “Federico II”, Naples, Italy
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, London, UK
| |
Collapse
|
20
|
Alshuhri MS, Gallagher L, Work LM, Holmes WM. Direct imaging of glymphatic transport using H217O MRI. JCI Insight 2021; 6:141159. [PMID: 33857020 PMCID: PMC8262348 DOI: 10.1172/jci.insight.141159] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
The recently proposed glymphatic pathway for solute transport and waste clearance from the brain has been the focus of intense debate. By exploiting an isotopically enriched MRI tracer, H217O, we directly imaged glymphatic water transport in the rat brain in vivo. Our results reveal glymphatic transport that is dramatically faster and more extensive than previously thought and unlikely to be explained by diffusion alone. Moreover, we confirm the critical role of aquaporin-4 channels in glymphatic transport.
Collapse
Affiliation(s)
- Mohammed S Alshuhri
- Institute of Neuroscience and Psychology, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow, United Kingdom.,Radiology and Medical Imaging Department, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
| | - Lindsay Gallagher
- Institute of Neuroscience and Psychology, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow, United Kingdom
| | - Lorraine M Work
- Institute of Cardiovascular and Medical Sciences, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow, United Kingdom
| | - William M Holmes
- Institute of Neuroscience and Psychology, College of Medicine, Veterinary and Life Science, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
21
|
Verheggen ICM, Freeze WM, de Jong JJA, Jansen JFA, Postma AA, van Boxtel MPJ, Verhey FRJ, Backes WH. Application of contrast-enhanced magnetic resonance imaging in the assessment of blood-cerebrospinal fluid barrier integrity. Neurosci Biobehav Rev 2021; 127:171-183. [PMID: 33930471 DOI: 10.1016/j.neubiorev.2021.04.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/15/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
VERHEGGEN, I.C.M., W. Freeze, J. de Jong, J. Jansen, A. Postma, M. van Boxtel, F. Verhey and W. Backes. The application of contrast-enhanced MRI in the assessment of blood-cerebrospinal fluid barrier integrity. Choroid plexus epithelial cells form a barrier that enables active, bidirectional exchange between the blood plasma and cerebrospinal fluid (CSF), known as the blood-CSF barrier (BCSFB). Through its involvement in CSF composition, the BCSFB maintains homeostasis in the central nervous system. While the relation between blood-brain barrier disruption, aging and neurodegeneration is extensively studied using contrast-enhanced MRI, applying this technique to investigate BCSFB disruption in age-related neurodegeneration has received little attention. This review provides an overview of the current status of contrast-enhanced MRI to assess BCSFB permeability. Post-contrast ventricular gadolinium enhancement has been used to indicate BCSFB permeability. Moreover, new techniques highly sensitive to low gadolinium concentrations in the CSF, for instance heavily T2-weighted imaging with cerebrospinal fluid suppression, seem promising. Also, attempts are made at using other contrast agents, such as manganese ions or very small superparamagnetic iron oxide particles, that seem to be cleared from the brain at the choroid plexus. Advancing and applying new developments such as these could progress the assessment of BCSFB integrity.
Collapse
Affiliation(s)
- Inge C M Verheggen
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands.
| | - Whitney M Freeze
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Department of Radiology, Leiden University Medical Center, Leiden, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Joost J A de Jong
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Jacobus F A Jansen
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Alida A Postma
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands
| | - Martin P J van Boxtel
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Frans R J Verhey
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Walter H Backes
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| |
Collapse
|
22
|
Steward CE, Venkatraman VK, Lui E, Malpas CB, Ellis KA, Cyarto EV, Vivash L, O'Brien TJ, Velakoulis D, Ames D, Masters CL, Lautenschlager NT, Bammer R, Desmond PM. Assessment of the DTI-ALPS Parameter Along the Perivascular Space in Older Adults at Risk of Dementia. J Neuroimaging 2021; 31:569-578. [PMID: 33556226 DOI: 10.1111/jon.12837] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Recently, there has been growing interest in the glymphatic system (the functional waste clearance pathway for the central nervous system and its role in flushing solutes (such as amyloid ß and tau), metabolic, and other cellular waste products in the brain. Herein, we investigate a recent potential biomarker for glymphatic activity (the diffusion tensor imaging along the perivascular space [DTI-ALPS] parameter) using diffusion MRI imaging in an elderly cohort comprising 10 cognitively normal, 10 mild cognitive impairment (MCI), and 16 Alzheimer's disease (AD). METHODS All 36 participants imaged on a Siemens 3.0T Tim Trio. Single-SE diffusion weighted Echo-planar imaging scans were acquired as well as T1 magnetization prepared rapid gradient echo, T2 axial, and susceptibility weighted imaging. Three millimeter regions of interest were drawn in the projection and association fibers adjacent to the medullary veins at the level of the lateral ventricle. The DTI-ALPS parameter was calculated in these regions and correlated with cognitive status, Mini-Mental State Examination (MMSE), and ADASCog11 measures. RESULTS Significant correlations were found between DTI-ALPS and MMSE and ADASCog11 in the right hemisphere adjusting for age, sex, and APoE ε4 status. Significant differences were also found in the right DTI-ALPS indices between cognitively normal and AD groups (P < .026) and MCI groups (P < .025) in a univariate general linear model corrected for age, sex, and APoE ε4. Significant differences in apparent diffusion coefficient between cognitively normal and AD groups were found in the right projection fibers (P = .028). CONCLUSION Further work is needed to determine the utility of DTI-ALPS index in larger elderly cohorts and whether it measures glymphatic activity.
Collapse
Affiliation(s)
- Christopher E Steward
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Vijay K Venkatraman
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Elaine Lui
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Charles B Malpas
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Kathryn A Ellis
- Academic Unit for Psychiatry of Old Age, Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia.,Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Lucy Vivash
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurology, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Neurosciences, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurosciences, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Dennis Velakoulis
- Neuropsychiatry Unit, Royal Melbourne Hospital & Melbourne Neuropsychiatry Centre, University of Melbourne, Melbourne, Victoria, Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia.,National Ageing and Research Institute, Melbourne, Victoria, Australia.,St George's Hospital, Kew, Victoria, Australia
| | - Colin L Masters
- Florey Institute for Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Nicola T Lautenschlager
- Academic Unit for Psychiatry of Old Age, Department of Psychiatry, University of Melbourne, Melbourne, Victoria, Australia.,NorthWestern Mental Health, Melbourne Health, Melbourne, Victoria, Australia
| | - Roland Bammer
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Patricia M Desmond
- Department of Medicine and Radiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Kimball E, Schaub J, Quillen S, Keuthan C, Pease ME, Korneva A, Quigley H. The role of aquaporin-4 in optic nerve head astrocytes in experimental glaucoma. PLoS One 2021; 16:e0244123. [PMID: 33529207 PMCID: PMC7853498 DOI: 10.1371/journal.pone.0244123] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/11/2021] [Indexed: 02/04/2023] Open
Abstract
PURPOSE To study aquaporin channel expression in astrocytes of the mouse optic nerve (ON) and the response to IOP elevation in mice lacking aquaporin 4 (AQP4 null). METHODS C57BL/6 (B6) and AQP4 null mice were exposed to bead-induced IOP elevation for 3 days (3D-IOP), 1 and 6 weeks. Mouse ocular tissue sections were immunolabeled against aquaporins 1(AQP1), 4(AQP4), and 9(AQP9). Ocular tissue was imaged to identify normal AQP distribution, ON changes, and axon loss after IOP elevation. Ultrastructure examination, cell proliferation, gene expression, and transport block were also analyzed. RESULTS B6 mice had abundant AQP4 expression in Müller cells, astrocytes of retina and myelinated ON (MON), but minimal AQP4in prelaminar and unmyelinated ON (UON). MON of AQP4 nulls had smaller ON area, smaller axon diameter, higher axon density, and larger proportionate axon area than B6 (all p≤0.05). Bead-injection led to comparable 3D-IOP elevation (p = 0.42) and axonal transport blockade in both strains. In B6, AQP4 distribution was unchanged after 3D-IOP. At baseline, AQP1 and AQP9 were present in retina, but not in UON and this was unaffected after IOP elevation in both strains. In 3D-IOP mice, ON astrocytes and microglia proliferated, more in B6 than AQP4 null. After 6 week IOP elevation, axon loss occurred equally in the two mouse types (24.6%, AQP4 null vs. 23.3%, B6). CONCLUSION Lack of AQP4 was neither protective nor detrimental to the effects of IOP elevation. The minimal presence of AQP4 in UON may be a vital aspect of the regionally specific phenotype of astrocytes in the mouse optic nerve head.
Collapse
Affiliation(s)
- Elizabeth Kimball
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Julie Schaub
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sarah Quillen
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Casey Keuthan
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mary Ellen Pease
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Arina Korneva
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Harry Quigley
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
24
|
Bourasset F, Auvity S, Thorne RG, Scherrmann JM. Brain Distribution of Drugs: Brain Morphology, Delivery Routes, and Species Differences. Handb Exp Pharmacol 2021; 273:97-120. [PMID: 33474672 DOI: 10.1007/164_2020_402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Neuropharmacokinetics considers cerebral drug distribution as a critical process for central nervous system drug action as well as for drug penetration through the CNS barriers. Brain distribution of small molecules obeys classical rules of drug partition, permeability, binding to fluid proteins or tissue components, and tissue perfusion. The biodistribution of all drugs, including both small molecules and biologics, may also be influenced by specific brain properties related to brain anatomy and physiological barriers, fluid dynamics, and cellular and biochemical composition, each of which can exhibit significant interspecies differences. All of these properties contribute to select optimal dosing paradigms and routes of drug delivery to reach brain targets for classical small molecule drugs as well as for biologics. The importance of these properties for brain delivery and exposure also highlights the need for efficient new analytical technologies to more comprehensively investigate drug distribution in the CNS, a complex multi-compartmentalized organ system.
Collapse
Affiliation(s)
- Fanchon Bourasset
- Faculty of Pharmacy, University of Paris, Paris, France.,INSERM UMR-S1144, Paris, France
| | - Sylvain Auvity
- Faculty of Pharmacy, University of Paris, Paris, France.,INSERM UMR-S1144, Paris, France
| | - Robert G Thorne
- Denali Therapeutics, Inc., South San Francisco, CA, USA. .,Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA.
| | - Jean-Michel Scherrmann
- Faculty of Pharmacy, University of Paris, Paris, France. .,INSERM UMR-S1144, Paris, France.
| |
Collapse
|
25
|
Daversin-Catty C, Vinje V, Mardal KA, Rognes ME. The mechanisms behind perivascular fluid flow. PLoS One 2020; 15:e0244442. [PMID: 33373419 PMCID: PMC7771676 DOI: 10.1371/journal.pone.0244442] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/09/2020] [Indexed: 11/26/2022] Open
Abstract
Flow of cerebrospinal fluid (CSF) in perivascular spaces (PVS) is one of the key concepts involved in theories concerning clearance from the brain. Experimental studies have demonstrated both net and oscillatory movement of microspheres in PVS (Mestre et al. (2018), Bedussi et al. (2018)). The oscillatory particle movement has a clear cardiac component, while the mechanisms involved in net movement remain disputed. Using computational fluid dynamics, we computed the CSF velocity and pressure in a PVS surrounding a cerebral artery subject to different forces, representing arterial wall expansion, systemic CSF pressure changes and rigid motions of the artery. The arterial wall expansion generated velocity amplitudes of 60-260 μm/s, which is in the upper range of previously observed values. In the absence of a static pressure gradient, predicted net flow velocities were small (<0.5 μm/s), though reaching up to 7 μm/s for non-physiological PVS lengths. In realistic geometries, a static systemic pressure increase of physiologically plausible magnitude was sufficient to induce net flow velocities of 20-30 μm/s. Moreover, rigid motions of the artery added to the complexity of flow patterns in the PVS. Our study demonstrates that the combination of arterial wall expansion, rigid motions and a static CSF pressure gradient generates net and oscillatory PVS flow, quantitatively comparable with experimental findings. The static CSF pressure gradient required for net flow is small, suggesting that its origin is yet to be determined.
Collapse
Affiliation(s)
- Cécile Daversin-Catty
- Simula Research Laboratory, Department of Numerical Analysis and Scientific Computing, Lysaker, Norway
| | - Vegard Vinje
- Simula Research Laboratory, Department of Numerical Analysis and Scientific Computing, Lysaker, Norway
| | - Kent-André Mardal
- Simula Research Laboratory, Department of Numerical Analysis and Scientific Computing, Lysaker, Norway
- Department of Mathematics, University of Oslo, Oslo, Norway
| | - Marie E. Rognes
- Simula Research Laboratory, Department of Numerical Analysis and Scientific Computing, Lysaker, Norway
| |
Collapse
|
26
|
Brain Glymphatic/Lymphatic Imaging by MRI and PET. Nucl Med Mol Imaging 2020; 54:207-223. [PMID: 33088350 DOI: 10.1007/s13139-020-00665-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/09/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023] Open
Abstract
Since glymphatic was proposed and meningeal lymphatic was discovered, MRI and even PET were introduced to investigate brain parenchymal interstitial fluid (ISF), cerebrospinal fluid (CSF), and lymphatic outflow in rodents and humans. Previous findings by ex vivo fluorescent microscopic, and in vivo two-photon imaging in rodents were reproduced using intrathecal contrast (gadobutrol and the similar)-enhanced MRI in rodents and further in humans. On dynamic MRI of meningeal lymphatics, in contrast to rodents, humans use mainly dorsal meningeal lymphatic pathways of ISF-CSF-lymphatic efflux. In mice, ISF-CSF exchange was examined thoroughly using an intra-cistern injection of fluorescent tracers during sleep, aging, and neurodegeneration yielding many details. CSF to lymphatic efflux is across arachnoid barrier cells over the dorsal dura in rodents and in humans. Meningeal lymphatic efflux to cervical lymph nodes and systemic circulation is also well-delineated especially in humans onintrathecal contrast MRI. Sleep- or anesthesia-related changes of glymphatic-lymphatic flow and the coupling of ISF-CSF-lymphatic drainage are major confounders ininterpreting brain glymphatic/lymphatic outflow in rodents. PET imaging in humans should be interpreted based on human anatomy and physiology, different in some aspects, using MRI recently. Based on the summary in this review, we propose non-invasive and longer-term intrathecal SPECT/PET or MRI studies to unravel the roles of brain glymphatic/lymphatic in diseases.
Collapse
|
27
|
Kedarasetti RT, Turner KL, Echagarruga C, Gluckman BJ, Drew PJ, Costanzo F. Functional hyperemia drives fluid exchange in the paravascular space. Fluids Barriers CNS 2020; 17:52. [PMID: 32819402 PMCID: PMC7441569 DOI: 10.1186/s12987-020-00214-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/09/2020] [Indexed: 12/20/2022] Open
Abstract
The brain lacks a conventional lymphatic system to remove metabolic waste. It has been proposed that directional fluid movement through the arteriolar paravascular space (PVS) promotes metabolite clearance. We performed simulations to examine if arteriolar pulsations and dilations can drive directional CSF flow in the PVS and found that arteriolar wall movements do not drive directional CSF flow. We propose an alternative method of metabolite clearance from the PVS, namely fluid exchange between the PVS and the subarachnoid space (SAS). In simulations with compliant brain tissue, arteriolar pulsations did not drive appreciable fluid exchange between the PVS and the SAS. However, when the arteriole dilated, as seen during functional hyperemia, there was a marked exchange of fluid. Simulations suggest that functional hyperemia may serve to increase metabolite clearance from the PVS. We measured blood vessels and brain tissue displacement simultaneously in awake, head-fixed mice using two-photon microscopy. These measurements showed that brain deforms in response to pressure changes in PVS, consistent with our simulations. Our results show that the deformability of the brain tissue needs to be accounted for when studying fluid flow and metabolite transport.
Collapse
Affiliation(s)
- Ravi Teja Kedarasetti
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, USA
| | - Kevin L Turner
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Christina Echagarruga
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Bruce J Gluckman
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, USA
- Department of Neurosurgery, The Pennsylvania State University, University Park, PA, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA
| | - Patrick J Drew
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA.
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, USA.
- Department of Neurosurgery, The Pennsylvania State University, University Park, PA, USA.
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA.
| | - Francesco Costanzo
- Center for Neural Engineering, The Pennsylvania State University, University Park, PA, USA.
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, USA.
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, USA.
- Department of Mathematics, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
28
|
Berggreen E, Wiig H, Virtej A. Fluid transport from the dental pulp revisited. Eur J Oral Sci 2020; 128:365-368. [PMID: 32794278 DOI: 10.1111/eos.12733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2020] [Indexed: 11/30/2022]
Abstract
In the dental pulp surrounded by rigid dentinal walls, an increase in fluid volume will be followed by a rapid increase in interstitial fluid pressure. To maintain pressure homeostasis, a fluid drainage system is required. The dental pulp and apical periodontal ligament lack lymphatic vessels, and the questions are how the transport can take place inside the pulp and where the lymphatic vessels draining fluid from the apical periodontal ligament are located. The drainage of fluid within the pulp must be governed by a tissue pressure gradient (driving pressure) and the fluid is likely transported in loose connective tissue (gaps) surrounding vessels and nerve fibers. We suggest that aging of the pulp tissue characterized by fibrosis will reduce the draining capacity and make it more vulnerable to circulatory failure. When the fluid leaves the pulp, it will follow the nerve bundles and vessels through the periapical ligament into bone channels, where lymphatic vessels are found. In the mandibular canal, lymphatic vessels are localized and the fluid washout rate from the canal is slow, but chewing may speed it up by increasing the fluid pressure. In acute apical periodontitis, inflammatory mediators and bacterial components can be spread to regional lymph nodes via lymphatic vessels inside the jaw bone.
Collapse
Affiliation(s)
- Ellen Berggreen
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anca Virtej
- Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Oral and Maxillofacial Surgery, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
29
|
Nasution RA, Islam AA, Hatta M, Prihantono, Warsinggih, Ludong DH, Ismail, Wangi H, Massi MN, Nasution KI. Effects of caffeic acid phenethyl ester in reducing cerebral edema in rat subjects experiencing brain injury: An in vivo study. Ann Med Surg (Lond) 2020; 57:328-333. [PMID: 32874565 PMCID: PMC7452142 DOI: 10.1016/j.amsu.2020.08.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 02/08/2023] Open
Abstract
Background A head injury is a very dangerous condition that threatens human life. This study examines the use of caffeic acid phenethyl ester (CAPE) in reducing cerebral edema in cases of head injury. The purpose of this study is to demonstrate whether CAPE can improve various parameters related to the expression of Aquaporin-4 (AQP4) mRNA and the serum AQP4 levels in rat subjects. Methods This is a randomized controlled study using a posttest-only control group design that uses experimental animals-specifically, male Rattus norvegicus (Sprague Dawley strain) rats aged 10-12 weeks and weighing 200-300 g. This study used a head injury model according to Marmarou (1994) with minor modifications to the animal model fixation tool. The parameters of the AQP4 mRNA were examined with real-time PCR, while serum AQP4 levels were examined with sandwich ELISA. Results The AQP4 mRNA expression in rats that were given CAPE was lower than those not given CAPE, both on the fourth and seventh days; serum AQP4 levels in rats that were given CAPE were also lower than those not given CAPE, both on the fourth and seventh days. Conclusion Administration of CAPE in a rat model with head injury can reduce cerebral edema, mediated by AQP4.
Collapse
Affiliation(s)
| | - Andi Asadul Islam
- Department of Neurosurgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Mochammad Hatta
- Clinical Microbiologist Program, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Prihantono
- Department of Surgery Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Warsinggih
- Department of Surgery Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Dany H Ludong
- Doctoral Program of Medical Sciences, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Ismail
- Doctoral Program of Medical Sciences, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Harakati Wangi
- Department of Interna Medicine, Pelamonia Hospital, Makassar, Indonesia
| | - Muh Nassrum Massi
- Departement of Microbiology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | | |
Collapse
|
30
|
Keep RF, Jones HC, Drewes LR. This was the year that was: brain barriers and brain fluid research in 2019. Fluids Barriers CNS 2020; 17:20. [PMID: 32138786 PMCID: PMC7059280 DOI: 10.1186/s12987-020-00181-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This editorial highlights advances in brain barrier and brain fluid research published in 2019, as well as addressing current controversies and pressing needs. Topics include recent advances related to: the cerebral endothelium and the neurovascular unit; the choroid plexus, arachnoid membrane; cerebrospinal fluid and the glymphatic hypothesis; the impact of disease states on brain barriers and brain fluids; drug delivery to the brain; and translation of preclinical data to the clinic. This editorial also mourns the loss of two important figures in the field, Malcolm B. Segal and Edward G. Stopa.
Collapse
Affiliation(s)
- Richard F. Keep
- Department of Neurosurgery, University of Michigan, R5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200 USA
| | | | - Lester R. Drewes
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, Duluth, MN 55812 USA
| |
Collapse
|
31
|
Koundal S, Elkin R, Nadeem S, Xue Y, Constantinou S, Sanggaard S, Liu X, Monte B, Xu F, Van Nostrand W, Nedergaard M, Lee H, Wardlaw J, Benveniste H, Tannenbaum A. Optimal Mass Transport with Lagrangian Workflow Reveals Advective and Diffusion Driven Solute Transport in the Glymphatic System. Sci Rep 2020; 10:1990. [PMID: 32029859 PMCID: PMC7004986 DOI: 10.1038/s41598-020-59045-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/22/2020] [Indexed: 02/05/2023] Open
Abstract
The glymphatic system (GS) hypothesis states that advective driven cerebrospinal fluid (CSF) influx from the perivascular spaces into the interstitial fluid space rapidly transport solutes and clear waste from brain. However, the presence of advection in neuropil is contested and solutes are claimed to be transported by diffusion only. To address this controversy, we implemented a regularized version of the optimal mass transport (rOMT) problem, wherein the advection/diffusion equation is the only a priori assumption required. rOMT analysis with a Lagrangian perspective of GS transport revealed that solute speed was faster in CSF compared to grey and white matter. Further, rOMT analysis also demonstrated 2-fold differences in regional solute speed within the brain. Collectively, these results imply that advective transport dominates in CSF while diffusion and advection both contribute to GS transport in parenchyma. In a rat model of cerebral small vessel disease (cSVD), solute transport in the perivascular spaces (PVS) and PVS-to-tissue transfer was slower compared to normal rats. Thus, the analytical framework of rOMT provides novel insights in the local dynamics of GS transport that may have implications for neurodegenerative diseases. Future studies should apply the rOMT analysis approach to confirm GS transport reductions in humans with cSVD.
Collapse
Affiliation(s)
- Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Rena Elkin
- Departments of Computer Science/Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA
| | - Saad Nadeem
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, NY, NY, USA
| | - Yuechuan Xue
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Simon Sanggaard
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Xiaodan Liu
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Brittany Monte
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Feng Xu
- George & Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Rhode Island, RI, USA
| | - William Van Nostrand
- George & Anne Ryan Institute for Neuroscience, Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Rhode Island, RI, USA
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Joanna Wardlaw
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Allen Tannenbaum
- Departments of Computer Science/Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
32
|
Valenza M, Facchinetti R, Steardo L, Scuderi C. Altered Waste Disposal System in Aging and Alzheimer's Disease: Focus on Astrocytic Aquaporin-4. Front Pharmacol 2020; 10:1656. [PMID: 32063858 PMCID: PMC7000422 DOI: 10.3389/fphar.2019.01656] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/17/2019] [Indexed: 12/15/2022] Open
Abstract
Among the diverse cell types included in the general population named glia, astrocytes emerge as being the focus of a growing body of research aimed at characterizing their heterogeneous and complex functions. Alterations of both their morphology and activities have been linked to a variety of neurological diseases. One crucial physiological need satisfied by astrocytes is the cleansing of the cerebral tissue from waste molecules. Several data demonstrate that aquaporin-4 (AQP-4), a protein expressed by astrocytes, is crucially important for facilitating the removal of waste products from the brain. Aquaporins are water channels found in all district of the human organism and the most abundant isoform in the brain is AQP-4. This protein is involved in a myriad of astrocytic activities, including calcium signal transduction, potassium buffering, synaptic plasticity, astrocyte migration, glial scar formation and neuroinflammation. The highest density of AQP-4 is found at the astrocytic domains closest to blood vessels, the endfeet that envelop brain vessels, with low to zero expression in other astrocytic membrane regions. Increased AQP-4 expression and loss of polarization have recently been documented in altered physiological conditions. Here we review the latest findings related to aging and Alzheimer’s disease (AD) on this topic, as well as the available knowledge on pharmacological tools to target AQP-4.
Collapse
Affiliation(s)
- Marta Valenza
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy.,Epitech Group SpA, Saccolongo, Italy
| | - Roberta Facchinetti
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Luca Steardo
- Università Telematica Giustino Fortunato, Benevento, Italy
| | - Caterina Scuderi
- Department Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Affiliation(s)
- Matthew Simon
- Oregon Health & Science University, Portland, OR, USA
| | | |
Collapse
|
34
|
Affiliation(s)
- Alex J Smith
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Alan S Verkman
- Departments of Medicine and Physiology, University of California San Francisco, San Francisco, CA, 94143, USA
| |
Collapse
|