1
|
Hochstetler A, Hehnly C, Dawes W, Harris D, Sadegh C, Mangano FT, Lanjewar SN, Chau MJ. Research priorities for non-invasive therapies to improve hydrocephalus outcomes. Fluids Barriers CNS 2025; 22:24. [PMID: 40033423 DOI: 10.1186/s12987-025-00632-1] [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: 12/21/2024] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
The Hydrocephalus Association organized two workshops with the support of the Rudi Schulte Research Institute and Cincinnati Children's Hospital Medical Center entitled "Developing Non-Invasive Hydrocephalus Therapies: Molecular and Cellular Targets", held September 27-29, 2023, in Dallas, TX, and "Developing Non-Invasive Hydrocephalus Therapies: Advancing Towards Clinical Trials", held April 12-13, 2024, in Cincinnati, OH. The goal of these workshops was to explore the frontiers of ongoing research for non-invasive therapies for the treatment of hydrocephalus across all etiologies and to improve patient outcomes at all stages of diagnosis and management. During the consensus-building discussions throughout the research workshops, basic, translational, and clinical scientists aimed to identify the next steps to develop novel treatments for hydrocephalus. This detailed report discusses the research priorities that emerged from these workshops to inspire researchers and guide studies towards better treatment for people living with hydrocephalus.
Collapse
Affiliation(s)
- Alexandra Hochstetler
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Christine Hehnly
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - William Dawes
- Department of Paediatric Neurosurgery, Oxford University Hospital, Oxford, UK
| | | | - Cameron Sadegh
- Department of Neurosurgery, University of California-Davis, Sacramento, CA, 95817, USA
| | - Francesco T Mangano
- Division of Neurosurgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | | | - Monica J Chau
- Research Department, Hydrocephalus Association, Bethesda, MD, 20814, USA.
| |
Collapse
|
2
|
Yoshida T, Yang ZH, Ashida S, Yu ZX, Shrivastav S, Vamsi Rojulpote K, Bahar P, Nguyen D, Springer DA, Munasinghe J, Starost MF, Hoffmann VJ, Rosenberg AZ, Bielekova B, Wen H, Remaley AT, Kopp JB. Apolipoprotein-L1 G1 variant contributes to hydrocephalus but not to atherosclerosis in apolipoprotein-E knock-out mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.28.630625. [PMID: 39803526 PMCID: PMC11722280 DOI: 10.1101/2024.12.28.630625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Introduction In USA, six million individuals with Sub-Saharan ancestry carry two APOL1 high-risk variants, which increase the risk for kidney diseases. Whether APOL1 high-risk variants are independent risk factors for cardiovascular diseases is unclear and requires further investigation. Methods We characterized a mouse model to investigate the role of APOL1 in dyslipidemia and cardiovascular diseases. Transgenic mice carrying APOL1 (G0 and G1 variants) on bacterial artificial chromosomes (BAC/APOL1 mice) were crossed with the ApoE knock-out (ApoE-KO) atherosclerosis mouse model. The compound transgenic mice were evaluated for the impact of APOL1 on systemic phenotypes. Results ApoE-KO mice carrying APOL1-G0 and APOL1-G1 did not show differences in the extent of atherosclerotic lesions or aortic calcification, as evaluated by Sudan IV staining and radiographic examination, respectively. However, ~20% of ApoE-KO; BAC/APOL1-G1 mice developed hydrocephalus and required euthanasia. The hydrocephalus was communicating and likely was due to excess cerebrospinal fluid produced by the choroid plexus, where epithelial cells expressed APOL1. Single-nuclear RNA-seq of choroid plexus identified solute transporter upregulation and mTORC2 pathway activation in APOL1-G1-expressing epithelial cells. Further, in the All of Us cohort, we found higher hydrocephalus prevalence among individuals with the APOL1-G1 variant in both recessive and dominant models, supporting the mouse findings. Conclusion While APOL1-G1 expression in ApoE-KO mice did not worsen cardiovascular disease phenotypes, we uncovered hydrocephalus as a novel APOL1 risk allele-mediated phenotype. These findings extend the spectrum of APOL1-associated pathologies.
Collapse
Affiliation(s)
- Teruhiko Yoshida
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD
- Graduate School of Medicine, The University of Tokyo, Tokyo, JAPAN
| | - Zhi-Hong Yang
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Shinji Ashida
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD
| | - Zu Xi Yu
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Shashi Shrivastav
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD
| | | | - Piroz Bahar
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - David Nguyen
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | | | - Jeeva Munasinghe
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD
| | | | | | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Bibi Bielekova
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD
| | - Han Wen
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Alan T. Remaley
- National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Jeffrey B. Kopp
- National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD
| |
Collapse
|
3
|
Rappe A, Vihinen HA, Suomi F, Hassinen AJ, Ehsan H, Jokitalo ES, McWilliams TG. Longitudinal autophagy profiling of the mammalian brain reveals sustained mitophagy throughout healthy aging. EMBO J 2024; 43:6199-6231. [PMID: 39367235 PMCID: PMC11612485 DOI: 10.1038/s44318-024-00241-y] [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: 11/17/2023] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 10/06/2024] Open
Abstract
Mitophagy neutralizes mitochondrial damage, thereby preventing cellular dysfunction and apoptosis. Defects in mitophagy have been strongly implicated in age-related neurodegenerative disorders such as Parkinson's and Alzheimer's disease. While mitophagy decreases throughout the lifespan of short-lived model organisms, it remains unknown whether such a decline occurs in the aging mammalian brain-a question of fundamental importance for understanding cell type- and region-specific susceptibility to neurodegeneration. Here, we define the longitudinal dynamics of basal mitophagy and macroautophagy across neuronal and non-neuronal cell types within the intact aging mouse brain in vivo. Quantitative profiling of reporter mouse cohorts from young to geriatric ages reveals cell- and tissue-specific alterations in mitophagy and macroautophagy between distinct subregions and cell populations, including dopaminergic neurons, cerebellar Purkinje cells, astrocytes, microglia and interneurons. We also find that healthy aging is hallmarked by the dynamic accumulation of differentially acidified lysosomes in several neural cell subsets. Our findings argue against any widespread age-related decline in mitophagic activity, instead demonstrating dynamic fluctuations in mitophagy across the aging trajectory, with strong implications for ongoing theragnostic development.
Collapse
Affiliation(s)
- Anna Rappe
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Helena A Vihinen
- Electron Microscopy Unit (EMBI), Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Fumi Suomi
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Antti J Hassinen
- High Content Imaging and Analysis Unit (FIMM-HCA), Institute for Molecular Medicine, Helsinki Institute of Life Science, University of Helsinki, Tukholmankatu 8, Helsinki, 00290, Finland
| | - Homa Ehsan
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
| | - Eija S Jokitalo
- Electron Microscopy Unit (EMBI), Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Viikinkaari 9, Helsinki, 00790, Finland
| | - Thomas G McWilliams
- Translational Stem Cell Biology and Metabolism Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
| |
Collapse
|
4
|
Schwerk C, Schroten H. In vitro models of the choroid plexus and the blood-cerebrospinal fluid barrier: advances, applications, and perspectives. Hum Cell 2024; 37:1235-1242. [PMID: 39103559 PMCID: PMC11341628 DOI: 10.1007/s13577-024-01115-5] [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: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
The choroid plexus (CP), a highly vascularized endothelial-epithelial convolute, is placed in the ventricular system of the brain and produces a large part of the cerebrospinal fluid (CSF). Additionally, the CP is the location of a blood-CSF barrier (BCSFB) that separates the CSF from the blood stream in the CP endothelium. In vitro models of the CP and the BCSFB are of high importance to investigate the biological functions of the CP and the BCSFB. Since the CP is involved in several serious diseases, these in vitro models promise help in researching the processes contributing to the diseases and during the development of treatment options. In this review, we provide an overview on the available models and the advances that have been made toward more sophisticated and "in vivo near" systems as organoids and microfluidic lab-on-a-chip approaches. We go into the applications and research objectives for which the various modeling systems can be used and discuss the possible future prospects and perspectives.
Collapse
Affiliation(s)
- Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany.
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany.
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| |
Collapse
|
5
|
Delvenne A, Vandendriessche C, Gobom J, Burgelman M, Dujardin P, De Nolf C, Tijms BM, Teunissen CE, Schindler SE, Verhey F, Ramakers I, Martinez-Lage P, Tainta M, Vandenberghe R, Schaeverbeke J, Engelborghs S, De Roeck E, Popp J, Peyratout G, Tsolaki M, Freund-Levi Y, Lovestone S, Streffer J, Bertram L, Blennow K, Zetterberg H, Visser PJ, Vandenbroucke RE, Vos SJB. Involvement of the choroid plexus in Alzheimer's disease pathophysiology: findings from mouse and human proteomic studies. Fluids Barriers CNS 2024; 21:58. [PMID: 39020361 PMCID: PMC11256635 DOI: 10.1186/s12987-024-00555-3] [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: 04/15/2024] [Accepted: 06/03/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Structural and functional changes of the choroid plexus (ChP) have been reported in Alzheimer's disease (AD). Nonetheless, the role of the ChP in the pathogenesis of AD remains largely unknown. We aim to unravel the relation between ChP functioning and core AD pathogenesis using a unique proteomic approach in mice and humans. METHODS We used an APP knock-in mouse model, APPNL-G-F, exhibiting amyloid pathology, to study the association between AD brain pathology and protein changes in mouse ChP tissue and CSF using liquid chromatography mass spectrometry. Mouse proteomes were investigated at the age of 7 weeks (n = 5) and 40 weeks (n = 5). Results were compared with previously published human AD CSF proteomic data (n = 496) to identify key proteins and pathways associated with ChP changes in AD. RESULTS ChP tissue proteome was dysregulated in APPNL-G-F mice relative to wild-type mice at both 7 and 40 weeks. At both ages, ChP tissue proteomic changes were associated with epithelial cells, mitochondria, protein modification, extracellular matrix and lipids. Nonetheless, some ChP tissue proteomic changes were different across the disease trajectory; pathways related to lysosomal function, endocytosis, protein formation, actin and complement were uniquely dysregulated at 7 weeks, while pathways associated with nervous system, immune system, protein degradation and vascular system were uniquely dysregulated at 40 weeks. CSF proteomics in both mice and humans showed similar ChP-related dysregulated pathways. CONCLUSIONS Together, our findings support the hypothesis of ChP dysfunction in AD. These ChP changes were related to amyloid pathology. Therefore, the ChP could become a novel promising therapeutic target for AD.
Collapse
Affiliation(s)
- Aurore Delvenne
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands.
| | - Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Johan Gobom
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Marlies Burgelman
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Pieter Dujardin
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Clint De Nolf
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam University Medical Centers (AUMC), Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, USA
- Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, USA
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | - Inez Ramakers
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | | | - Mikel Tainta
- Fundación CITA-Alzhéimer Fundazioa, San Sebastian, Spain
| | - Rik Vandenberghe
- Neurology Service, University Hospitals Leuven, Louvain, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Louvain, Belgium
| | - Jolien Schaeverbeke
- Neurology Service, University Hospitals Leuven, Louvain, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Louvain, Belgium
| | - Sebastiaan Engelborghs
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Bru-BRAIN, Universitair Ziekenhuis Brussel, Brussels, Belgium
- NEUR Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ellen De Roeck
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, Hospital Network Antwerp (ZNA) Middelheim and Hoge Beuken, Antwerp, Belgium
| | - Julius Popp
- Old Age Psychiatry, University Hospital Lausanne, Lausanne, Switzerland
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatry University Hospital Zürich, Zurich, Switzerland
| | | | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, Makedonia, Thessaloniki, Greece
| | - Yvonne Freund-Levi
- Department of Neurobiology, Caring Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry in Region Örebro County and School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Old Age Psychiatry, Psychology & Neuroscience, King's College, London, UK
| | - Simon Lovestone
- University of Oxford, Oxford, UK
- Johnson and Johnson Medical Ltd., Wokingham, UK
| | - Johannes Streffer
- Reference Center for Biological Markers of Dementia (BIODEM), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- H. Lundbeck A/S, Valby, Denmark
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, University of Lübeck, Lübeck, Germany
| | - Kaj Blennow
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, Alzheimer Centrum Limburg, School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| |
Collapse
|
6
|
Hladky SB, Barrand MA. Regulation of brain fluid volumes and pressures: basic principles, intracranial hypertension, ventriculomegaly and hydrocephalus. Fluids Barriers CNS 2024; 21:57. [PMID: 39020364 PMCID: PMC11253534 DOI: 10.1186/s12987-024-00532-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 03/21/2024] [Indexed: 07/19/2024] Open
Abstract
The principles of cerebrospinal fluid (CSF) production, circulation and outflow and regulation of fluid volumes and pressures in the normal brain are summarised. Abnormalities in these aspects in intracranial hypertension, ventriculomegaly and hydrocephalus are discussed. The brain parenchyma has a cellular framework with interstitial fluid (ISF) in the intervening spaces. Framework stress and interstitial fluid pressure (ISFP) combined provide the total stress which, after allowing for gravity, normally equals intracerebral pressure (ICP) with gradients of total stress too small to measure. Fluid pressure may differ from ICP in the parenchyma and collapsed subarachnoid spaces when the parenchyma presses against the meninges. Fluid pressure gradients determine fluid movements. In adults, restricting CSF outflow from subarachnoid spaces produces intracranial hypertension which, when CSF volumes change very little, is called idiopathic intracranial hypertension (iIH). Raised ICP in iIH is accompanied by increased venous sinus pressure, though which is cause and which effect is unclear. In infants with growing skulls, restriction in outflow leads to increased head and CSF volumes. In adults, ventriculomegaly can arise due to cerebral atrophy or, in hydrocephalus, to obstructions to intracranial CSF flow. In non-communicating hydrocephalus, flow through or out of the ventricles is somehow obstructed, whereas in communicating hydrocephalus, the obstruction is somewhere between the cisterna magna and cranial sites of outflow. When normal outflow routes are obstructed, continued CSF production in the ventricles may be partially balanced by outflow through the parenchyma via an oedematous periventricular layer and perivascular spaces. In adults, secondary hydrocephalus with raised ICP results from obvious obstructions to flow. By contrast, with the more subtly obstructed flow seen in normal pressure hydrocephalus (NPH), fluid pressure must be reduced elsewhere, e.g. in some subarachnoid spaces. In idiopathic NPH, where ventriculomegaly is accompanied by gait disturbance, dementia and/or urinary incontinence, the functional deficits can sometimes be reversed by shunting or third ventriculostomy. Parenchymal shrinkage is irreversible in late stage hydrocephalus with cellular framework loss but may not occur in early stages, whether by exclusion of fluid or otherwise. Further studies that are needed to explain the development of hydrocephalus are outlined.
Collapse
Affiliation(s)
- Stephen B Hladky
- Department of Pharmacology, Tennis Court Rd, Cambridge, CB2 1PD, UK.
| | | |
Collapse
|
7
|
Mack AF, Bihlmaier R, Deffner F. Shifting from ependyma to choroid plexus epithelium and the changing expressions of aquaporin-1 and aquaporin-4. J Physiol 2024; 602:3097-3110. [PMID: 37975746 DOI: 10.1113/jp284196] [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: 06/24/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023] Open
Abstract
The cells of the choroid plexus (CP) epithelium are specialized ependymal cells (ECs) but have distinct properties. The CP cells and ECs form single-cell sheets contiguous to each other at a transitional zone. The CP is underlined by a basal lamina and has barrier properties, whereas the ECs do not. The basal lamina of the CP is continuous with the glia limitans superficialis and, consequently, the CP stroma is continuous with the meninges along entering blood vessels. The CP has previously been reported to express aquaporin-1 (AQP1) mostly apically, and ECs show mostly basolateral aquaporin-4 (AQP4) expression. Recent evidence in various systems has shown that in changing conditions the expression and distribution of AQP4 can be modified, involving phosphorylation and calmodulin-triggered translocation. Studies on the human CP revealed that AQP4 is also expressed in some CP cells, which is likely to be increased during ageing based on mouse data. Moreover, subependymal astrocytic processes in the ependyma-CP transition, forming a glial plate around blood vessels and facing the CP stroma, were strongly positive for AQP4. We propose that the increased AQP4 expression might be a compensatory mechanism for the observed reduction in CSF production in the ageing human brain. The high AQP4 density in the transition zone might facilitate the transport of water into and out of the CP stroma and serve as a drainage and clearing pathway for metabolites in the CNS.
Collapse
Affiliation(s)
- Andreas F Mack
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| | - Ronja Bihlmaier
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
| | - Felix Deffner
- Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, Tübingen, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Freiburg, Freiburg, Germany
| |
Collapse
|
8
|
Ueno M, Chiba Y, Murakami R, Miyai Y, Matsumoto K, Wakamatsu K, Nakagawa T, Takebayashi G, Uemura N, Yanase K, Ogino Y. Transporters, Ion Channels, and Junctional Proteins in Choroid Plexus Epithelial Cells. Biomedicines 2024; 12:708. [PMID: 38672064 PMCID: PMC11048166 DOI: 10.3390/biomedicines12040708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
The choroid plexus (CP) plays significant roles in secreting cerebrospinal fluid (CSF) and forming circadian rhythms. A monolayer of epithelial cells with tight and adherens junctions of CP forms the blood-CSF barrier to control the movement of substances between the blood and ventricles, as microvessels in the stroma of CP have fenestrations in endothelial cells. CP epithelial cells are equipped with several kinds of transporters and ion channels to transport nutrient substances and secrete CSF. In addition, junctional components also contribute to CSF production as well as blood-CSF barrier formation. However, it remains unclear how junctional components as well as transporters and ion channels contribute to the pathogenesis of neurodegenerative disorders. In this manuscript, recent findings regarding the distribution and significance of transporters, ion channels, and junctional proteins in CP epithelial cells are introduced, and how changes in expression of their epithelial proteins contribute to the pathophysiology of brain disorders are reviewed.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yumi Miyai
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Keiji Wakamatsu
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Toshitaka Nakagawa
- Division of Research Instrument and Equipment, Research Facility Center, Kagawa University, Kagawa 761-0793, Japan;
| | - Genta Takebayashi
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Naoya Uemura
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Ken Yanase
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| | - Yuichi Ogino
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan; (G.T.); (N.U.); (K.Y.); (Y.O.)
| |
Collapse
|
9
|
Abstract
The brain is a complex organ, fundamentally changing across the day to perform basic functions like sleep, thought, and regulating whole-body physiology. This requires a complex symphony of nutrients, hormones, ions, neurotransmitters and more to be properly distributed across the brain to maintain homeostasis throughout 24 hours. These solutes are distributed both by the blood and by cerebrospinal fluid. Cerebrospinal fluid contents are distinct from the general circulation because of regulation at brain barriers including the choroid plexus, glymphatic system, and blood-brain barrier. In this review, we discuss the overlapping circadian (≈24-hour) rhythms in brain fluid biology and at the brain barriers. Our goal is for the reader to gain both a fundamental understanding of brain barriers alongside an understanding of the interactions between these fluids and the circadian timing system. Ultimately, this review will provide new insight into how alterations in these finely tuned clocks may lead to pathology.
Collapse
Affiliation(s)
- Velia S Vizcarra
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ryann M Fame
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| |
Collapse
|
10
|
Badaut J, Ghersi-Egea JF, Thorne RG, Konsman JP. Blood-brain borders: a proposal to address limitations of historical blood-brain barrier terminology. Fluids Barriers CNS 2024; 21:3. [PMID: 38183042 PMCID: PMC10770911 DOI: 10.1186/s12987-023-00478-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/11/2023] [Indexed: 01/07/2024] Open
Abstract
Many neuroscientists use the term Blood-Brain Barrier (BBB) to emphasize restrictiveness, often equating or reducing the notion of BBB properties to tight junction molecules physically sealing cerebral endothelial cells, rather than pointing out the complexity of this biological interface with respect to its selectivity and variety of exchange between the general blood circulation and the central nervous tissue. Several authors in the field find it unfortunate that the exquisitely dynamic interfaces between blood and brain continue to be viewed primarily as obstructive barriers to transport. Although the term blood-brain interface is an excellent descriptor that does not convey the idea of a barrier, it is important and preferable for the spreading of an idea beyond specialist communities to try to appeal to well-chosen metaphors. Recent evidence reviewed here indicates that blood-brain interfaces are more than selective semi-permeable membranes in that they display many dynamic processes and complex mechanisms for communication. They are thus more like 'geopolitical borders'. Furthermore, some authors working on blood-brain interface-relevant issues have started to use the word border, for example in border-associated macrophages. Therefore, we suggest adopting the term Blood-Brain Border to better communicate the flexibility of and movement across blood-brain interfaces.
Collapse
Affiliation(s)
- Jerome Badaut
- Brain Molecular Imaging Lab, UMR 5536, CNRS, RMSB, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
- Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Jean-François Ghersi-Egea
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028, CNRS UMR 5292, Lyon-1 University, Bron, France.
| | - Robert G Thorne
- Denali Therapeutics, Inc, 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
- Department of Pharmaceutics, University of Minnesota, 9-177 Weaver-Densford Hall, 308 Harvard St. SE, Minneapolis, MN, 55455, USA.
| | - Jan Pieter Konsman
- UMR 5164, CNRS, ImmunoConcEpT, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux Cedex, France.
| |
Collapse
|
11
|
MacAulay N, Toft-Bertelsen TL. Dual function of the choroid plexus: Cerebrospinal fluid production and control of brain ion homeostasis. Cell Calcium 2023; 116:102797. [PMID: 37801806 DOI: 10.1016/j.ceca.2023.102797] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/08/2023]
Abstract
The choroid plexus is a small monolayered epithelium located in the brain ventricles and serves to secrete the cerebrospinal fluid (CSF) that envelops the brain and fills the central ventricles. The CSF secretion is sustained with a concerted effort of a range of membrane transporters located in a polarized fashion in this tissue. Prominent amongst these are the Na+/K+-ATPase, the Na+,K+,2Cl- cotransporter (NKCC1), and several HCO3- transporters, which together support the net transepithelial transport of the major electrolytes, Na+ and Cl-, and thus drive the CSF secretion. The choroid plexus, in addition, serves an important role in keeping the CSF K+ concentration at a level compatible with normal brain function. The choroid plexus Na+/K+-ATPase represents a key factor in the barrier-mediated control of the CSF K+ homeostasis, as it increases its K+ uptake activity when faced with elevated extracellular K+ ([K+]o). In certain developmental or pathological conditions, the NKCC1 may revert its net transport direction to contribute to CSF K+ homeostasis. The choroid plexus ion transport machinery thus serves dual, yet interconnected, functions with its contribution to electrolyte and fluid secretion in combination with its control of brain K+ levels.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen 2200, Denmark.
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen 2200, Denmark
| |
Collapse
|
12
|
Israelsen IME, Kamp-Jensen C, Westgate CSJ, Styrishave B, Jensen RH, Eftekhari S. Cycle-dependent sex differences in expression of membrane proteins involved in cerebrospinal fluid secretion at rat choroid plexus. BMC Neurosci 2023; 24:60. [PMID: 37946101 PMCID: PMC10633912 DOI: 10.1186/s12868-023-00829-w] [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: 06/15/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Female sex is a known risk factor of brain disorders with raised intracranial pressure (ICP) and sex hormones have been suggested to alter cerebrospinal fluid (CSF) dynamics, thus impairing ICP regulation in CSF disorders such as idiopathic intracranial hypertension (IIH). The choroid plexus (CP) is the tissue producing CSF and it has been hypothesized that altered hormonal composition could affect the activity of transporters involved in CSF secretion, thus affecting ICP. Therefore, we aimed to investigate if expression of various transporters involved in CSF secretion at CP were different between males and females and between females in different estrous cycle states. Steroid levels in serum was also investigated. METHODS Female and male rats were used to determine sex-differences in the genes encoding for the transporters Aqp1 and 4, NKCC1, NBCe2, NCBE; carbonic anhydrase enzymes II and III (CA), subunits of the Na+/K+-ATPase including Atp1a1, Atp1b1 and Fxyd1 at CP. The estrous cycle stage metestrus (MET) and estrous (ES) were determined before euthanasia. Serum and CP were collected and subjected to RT-qPCR analysis and western blots. Serum was used to measure steroid levels using liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS Significant differences in gene expression and steroid levels between males and ES females were found, while no differences were found between male and MET females. During ES, expression of Aqp1 was lower (p < 0.01) and NKCC1 was higher in females compared to males. CAII was lower while CAIII was higher in ES females (p < 0.0001). Gene expression of Atp1a1 was lower in ES compared to male (p = 0.0008). Several of these choroidal genes were also significantly different in MET compared to females in ES. Differences in gene expression during the estrus cycle were correlated to serum level of steroid hormones. Protein expression of AQP1 (p = 0.008) and CAII (p = 0.035) was reduced in ES females compared to males. CONCLUSIONS This study demonstrates for the first time that expression at CP is sex-dependent and markedly affected by the estrous cycle in female rats. Further, expression was related to hormone levels in serum. This opens a completely new avenue for steroid regulation of the expression of CSF transporters and the close link to the understanding of CSF disorders such as IIH.
Collapse
Affiliation(s)
- Ida Marchen Egerod Israelsen
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Christina Kamp-Jensen
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Connar Stanley James Westgate
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Bjarne Styrishave
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rigmor H Jensen
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark
| | - Sajedeh Eftekhari
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600, Glostrup, Denmark.
| |
Collapse
|
13
|
Johnsen LØ, Friis KA, Damkier HH. In vitro investigation of the effect of proinflammatory cytokines on mouse choroid plexus membrane transporters Ncbe and NKCC1. Fluids Barriers CNS 2023; 20:71. [PMID: 37828581 PMCID: PMC10568836 DOI: 10.1186/s12987-023-00474-9] [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: 10/06/2022] [Accepted: 10/04/2023] [Indexed: 10/14/2023] Open
Abstract
Intraventricular hemorrhage is a potentially life-threatening condition. Approximately 20% of patients develop posthemorrhagic hydrocephalus with increased ventricular volume and intracranial pressure. Hydrocephalus develops partially due to increased secretion of cerebrospinal fluid by the choroid plexus. During hemorrhage a multitude of factors are released into the cerebrospinal fluid. Many of these have been implicated in the hypersecretion. In this study, we have investigated the isolated effect of inflammatory components, on the abundance of two membrane transporters involved in cerebrospinal fluid secretion by the choroid plexus: the Na+-dependent Cl-/HCO3- exchanger, Ncbe, and the Na+, K+, 2Cl- cotransporter, NKCC1. We have established a primary choroid plexus epithelial cell culture from 1 to 7 days old mouse pups. Seven days after seeding, the cells formed a monolayer. The cells were treated with either tumor necrosis factor alpha (TNFα), interleukin 1 beta (IL-1β), or interleukin 6 (IL-6) to mimic inflammation. The data show that treatment with TNFα, and IL-1β only transiently increased NKCC1 abundance whereas the effect on Ncbe abundance was a transient decrease. IL-6 however significantly increased NKCC1 (242%), the phosphorylated NKCC1 (147%), as well as pSPAK (406%) abundance, but had no effect on Ncbe. This study suggests that the inflammatory pathway involved in hypersecretion primarily is mediated by activation of basolateral receptors in the choroid plexus, mainly facilitated by IL-6. This study highlights the complexity of the pathophysiological circumstances occurring during intraventricular hemorrhage.
Collapse
|
14
|
Nutter CA, Kidd BM, Carter HA, Hamel JI, Mackie PM, Kumbkarni N, Davenport ML, Tuyn DM, Gopinath A, Creigh PD, Sznajder ŁJ, Wang ET, Ranum LPW, Khoshbouei H, Day JW, Sampson JB, Prokop S, Swanson MS. Choroid plexus mis-splicing and altered cerebrospinal fluid composition in myotonic dystrophy type 1. Brain 2023; 146:4217-4232. [PMID: 37143315 PMCID: PMC10545633 DOI: 10.1093/brain/awad148] [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: 01/23/2023] [Revised: 04/08/2023] [Accepted: 04/18/2023] [Indexed: 05/06/2023] Open
Abstract
Myotonic dystrophy type 1 is a dominantly inherited multisystemic disease caused by CTG tandem repeat expansions in the DMPK 3' untranslated region. These expanded repeats are transcribed and produce toxic CUG RNAs that sequester and inhibit activities of the MBNL family of developmental RNA processing factors. Although myotonic dystrophy is classified as a muscular dystrophy, the brain is also severely affected by an unusual cohort of symptoms, including hypersomnia, executive dysfunction, as well as early onsets of tau/MAPT pathology and cerebral atrophy. To address the molecular and cellular events that lead to these pathological outcomes, we recently generated a mouse Dmpk CTG expansion knock-in model and identified choroid plexus epithelial cells as particularly affected by the expression of toxic CUG expansion RNAs. To determine if toxic CUG RNAs perturb choroid plexus functions, alternative splicing analysis was performed on lateral and hindbrain choroid plexi from Dmpk CTG knock-in mice. Choroid plexus transcriptome-wide changes were evaluated in Mbnl2 knockout mice, a developmental-onset model of myotonic dystrophy brain dysfunction. To determine if transcriptome changes also occurred in the human disease, we obtained post-mortem choroid plexus for RNA-seq from neurologically unaffected (two females, three males; ages 50-70 years) and myotonic dystrophy type 1 (one female, three males; ages 50-70 years) donors. To test that choroid plexus transcriptome alterations resulted in altered CSF composition, we obtained CSF via lumbar puncture from patients with myotonic dystrophy type 1 (five females, five males; ages 35-55 years) and non-myotonic dystrophy patients (three females, four males; ages 26-51 years), and western blot and osmolarity analyses were used to test CSF alterations predicted by choroid plexus transcriptome analysis. We determined that CUG RNA induced toxicity was more robust in the lateral choroid plexus of Dmpk CTG knock-in mice due to comparatively higher Dmpk and lower Mbnl RNA levels. Impaired transitions to adult splicing patterns during choroid plexus development were identified in Mbnl2 knockout mice, including mis-splicing previously found in Dmpk CTG knock-in mice. Whole transcriptome analysis of myotonic dystrophy type 1 choroid plexus revealed disease-associated RNA expression and mis-splicing events. Based on these RNA changes, predicted alterations in ion homeostasis, secretory output and CSF composition were confirmed by analysis of myotonic dystrophy type 1 CSF. Our results implicate choroid plexus spliceopathy and concomitant alterations in CSF homeostasis as an unappreciated contributor to myotonic dystrophy type 1 CNS pathogenesis.
Collapse
Affiliation(s)
- Curtis A Nutter
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Benjamin M Kidd
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Helmut A Carter
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Johanna I Hamel
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA
| | - Philip M Mackie
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Nayha Kumbkarni
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Mackenzie L Davenport
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Dana M Tuyn
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Adithya Gopinath
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Peter D Creigh
- Department of Neurology, University of Rochester, Rochester, NY 14642, USA
| | - Łukasz J Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Eric T Wang
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Laura P W Ranum
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, McKnight Brain Institute and the Fixel Institute for Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - John W Day
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Jacinda B Sampson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Stefan Prokop
- Department of Pathology, Immunology, and Laboratory Medicine, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute and the Fixel Institute for Neurological Diseases, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| |
Collapse
|
15
|
Hochstetler A, Smith H, Reed M, Hulme L, Territo P, Bedwell A, Persohn S, Perrotti N, D'Antona L, Musumeci F, Schenone S, Blazer-Yost BL. Inhibition of serum- and glucocorticoid-induced kinase 1 ameliorates hydrocephalus in preclinical models. Fluids Barriers CNS 2023; 20:61. [PMID: 37596666 PMCID: PMC10439616 DOI: 10.1186/s12987-023-00461-0] [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: 11/22/2022] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Hydrocephalus is a pathological accumulation of cerebrospinal fluid (CSF), leading to ventriculomegaly. Hydrocephalus may be primary or secondary to traumatic brain injury, infection, or intracranial hemorrhage. Regardless of cause, current treatment involves surgery to drain the excess CSF. Importantly, there are no long-term, effective pharmaceutical treatments and this represents a clinically unmet need. Many forms of hydrocephalus involve dysregulation in water and electrolyte homeostasis, making this an attractive, druggable target. METHODS In vitro, a combination of electrophysiological and fluid flux assays was used to elucidate secretory transepithelial electrolyte and fluid flux in a human cell culture model of the choroid plexus epithelium and to determine the involvement of serum-, glucocorticoid-induced kinase 1 (SGK1). In vivo, MRI studies were performed in a genetic rat model of hydrocephalus to determine effects of inhibition of SGK1 with a novel inhibitor, SI113. RESULTS In the cultured cell line, SI113 reduced secretory transepithelial electrolyte and fluid flux. In vivo, SI113 blocks the development of hydrocephalus with no effect on ventricular size of wild-type animals and no overt toxic effects. Mechanistically, the development of hydrocephalus in the rat model involves an increase in activated, phosphorylated SGK1 with no change in the total amount of SGK1. SI113 inhibits phosphorylation with no changes in total SGK1 levels in the choroid plexus epithelium. CONCLUSION These data provide a strong preclinical basis for the use of SGK1 inhibitors in the treatment of hydrocephalus.
Collapse
Affiliation(s)
- Alexandra Hochstetler
- Department of Biology, SL358, Indiana University Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN, 46202, USA
| | - Hillary Smith
- Department of Biology, SL358, Indiana University Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN, 46202, USA
| | - Makenna Reed
- Department of Biology, SL358, Indiana University Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN, 46202, USA
| | - Louise Hulme
- Department of Biology, SL358, Indiana University Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN, 46202, USA
| | - Paul Territo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Amanda Bedwell
- Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Scott Persohn
- Department of Medicine, Indiana University School of Medicine, Indianapolis, USA
| | - Nicola Perrotti
- Dipartimento di Scienze della Salute, Università" Magna Graecia" di Catanzaro, Catanzaro, Italy
| | - Lucia D'Antona
- Dipartimento di Scienze della Salute, Università" Magna Graecia" di Catanzaro, Catanzaro, Italy
| | | | | | - Bonnie L Blazer-Yost
- Department of Biology, SL358, Indiana University Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN, 46202, USA.
| |
Collapse
|
16
|
Mineiro R, Albuquerque T, Neves AR, Santos CRA, Costa D, Quintela T. The Role of Biological Rhythms in New Drug Formulations to Cross the Brain Barriers. Int J Mol Sci 2023; 24:12541. [PMID: 37628722 PMCID: PMC10454916 DOI: 10.3390/ijms241612541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
For brain protection, the blood-brain barrier and blood-cerebrospinal fluid barrier limit the traffic of molecules between blood and brain tissue and between blood and cerebrospinal fluid, respectively. Besides their protective function, brain barriers also limit the passage of therapeutic drugs to the brain, which constitutes a great challenge for the development of therapeutic strategies for brain disorders. This problem has led to the emergence of novel strategies to treat neurological disorders, like the development of nanoformulations to deliver therapeutic agents to the brain. Recently, functional molecular clocks have been identified in the blood-brain barrier and in the blood-cerebrospinal fluid barrier. In fact, circadian rhythms in physiological functions related to drug disposition were also described in brain barriers. This opens the possibility for chronobiological approaches that aim to use time to improve drug efficacy and safety. The conjugation of nanoformulations with chronobiology for neurological disorders is still unexplored. Facing this, here, we reviewed the circadian rhythms in brain barriers, the nanoformulations studied to deliver drugs to the brain, and the nanoformulations with the potential to be conjugated with a chronobiological approach to therapeutic strategies for the brain.
Collapse
Affiliation(s)
- Rafael Mineiro
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Tânia Albuquerque
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Raquel Neves
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Cecília R. A. Santos
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Diana Costa
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI—Health Sciences Research Centre, Universidade da Beira Interior, Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal
- UDI-IPG—Unidade de Investigação para o Desenvolvimento do Interior, Instituto Politécnico da Guarda, 6300-559 Guarda, Portugal
| |
Collapse
|
17
|
Ueno M, Chiba Y, Murakami R, Miyai Y, Matsumoto K, Wakamatsu K, Takebayashi G, Uemura N, Yanase K. Distribution of Monocarboxylate Transporters in Brain and Choroid Plexus Epithelium. Pharmaceutics 2023; 15:2062. [PMID: 37631275 PMCID: PMC10458808 DOI: 10.3390/pharmaceutics15082062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
The choroid plexus (CP) plays central roles in regulating the microenvironment of the central nervous system by secreting the majority of cerebrospinal fluid (CSF) and controlling its composition. A monolayer of epithelial cells of CP plays a significant role in forming the blood-CSF barrier to restrict the movement of substances between the blood and ventricles. CP epithelial cells are equipped with transporters for glucose and lactate that are used as energy sources. There are many review papers on glucose transporters in CP epithelial cells. On the other hand, distribution of monocarboxylate transporters (MCTs) in CP epithelial cells has received less attention compared with glucose transporters. Some MCTs are known to transport lactate, pyruvate, and ketone bodies, whereas others transport thyroid hormones. Since CP epithelial cells have significant carrier functions as well as the barrier function, a decline in the expression and function of these transporters leads to a poor supply of thyroid hormones as well as lactate and can contribute to the process of age-associated brain impairment and pathophysiology of neurodegenerative diseases. In this review paper, recent findings regarding the distribution and significance of MCTs in the brain, especially in CP epithelial cells, are summarized.
Collapse
Affiliation(s)
- Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yoichi Chiba
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Ryuta Murakami
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Yumi Miyai
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Koichi Matsumoto
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Keiji Wakamatsu
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (Y.C.); (R.M.); (Y.M.); (K.M.); (K.W.)
| | - Genta Takebayashi
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (N.U.); (K.Y.)
| | - Naoya Uemura
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (N.U.); (K.Y.)
| | - Ken Yanase
- Department of Anesthesiology, Faculty of Medicine, Kagawa University, Takamatsu 761-0793, Kagawa, Japan; (G.T.); (N.U.); (K.Y.)
| |
Collapse
|
18
|
Sadegh C, Xu H, Sutin J, Fatou B, Gupta S, Pragana A, Taylor M, Kalugin PN, Zawadzki ME, Alturkistani O, Shipley FB, Dani N, Fame RM, Wurie Z, Talati P, Schleicher RL, Klein EM, Zhang Y, Holtzman MJ, Moore CI, Lin PY, Patel AB, Warf BC, Kimberly WT, Steen H, Andermann ML, Lehtinen MK. Choroid plexus-targeted NKCC1 overexpression to treat post-hemorrhagic hydrocephalus. Neuron 2023; 111:1591-1608.e4. [PMID: 36893755 PMCID: PMC10198810 DOI: 10.1016/j.neuron.2023.02.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/17/2023] [Accepted: 02/13/2023] [Indexed: 03/11/2023]
Abstract
Post-hemorrhagic hydrocephalus (PHH) refers to a life-threatening accumulation of cerebrospinal fluid (CSF) that occurs following intraventricular hemorrhage (IVH). An incomplete understanding of this variably progressive condition has hampered the development of new therapies beyond serial neurosurgical interventions. Here, we show a key role for the bidirectional Na-K-Cl cotransporter, NKCC1, in the choroid plexus (ChP) to mitigate PHH. Mimicking IVH with intraventricular blood led to increased CSF [K+] and triggered cytosolic calcium activity in ChP epithelial cells, which was followed by NKCC1 activation. ChP-targeted adeno-associated viral (AAV)-NKCC1 prevented blood-induced ventriculomegaly and led to persistently increased CSF clearance capacity. These data demonstrate that intraventricular blood triggered a trans-choroidal, NKCC1-dependent CSF clearance mechanism. Inactive, phosphodeficient AAV-NKCC1-NT51 failed to mitigate ventriculomegaly. Excessive CSF [K+] fluctuations correlated with permanent shunting outcome in humans following hemorrhagic stroke, suggesting targeted gene therapy as a potential treatment to mitigate intracranial fluid accumulation following hemorrhage.
Collapse
Affiliation(s)
- Cameron Sadegh
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Huixin Xu
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jason Sutin
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Benoit Fatou
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Suhasini Gupta
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Aja Pragana
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Milo Taylor
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard College, Harvard University, Cambridge, MA 02138, USA
| | - Peter N Kalugin
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA
| | - Miriam E Zawadzki
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA
| | - Osama Alturkistani
- Cellular Imaging Core, Boston Children's Hospital, Boston, MA 02115, USA
| | - Frederick B Shipley
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Neil Dani
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Zainab Wurie
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Pratik Talati
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Riana L Schleicher
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Eric M Klein
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Yong Zhang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St. Louis, MO, 63110, USA
| | - Christopher I Moore
- Carney Institute for Brain Science, Brown University, Providence, RI 02912, USA
| | - Pei-Yi Lin
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Benjamin C Warf
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA 02115, USA
| | - W Taylor Kimberly
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hanno Steen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Precision Vaccines Program, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mark L Andermann
- Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
19
|
Zheng Y, Hu L, Yang Y, Zheng C, Tu W, Lin H, Wang H, Jiang Y, Jiang S, Zheng W. Blocking the IFN-gamma signal in the choroid plexus confers resistance to experimental autoimmune encephalomyelitis. FASEB J 2023; 37:e22833. [PMID: 36921064 PMCID: PMC11977598 DOI: 10.1096/fj.202201767r] [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: 10/29/2022] [Revised: 01/09/2023] [Accepted: 02/09/2023] [Indexed: 03/17/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by inflammatory infiltration and demyelination in the central nervous system (CNS). IFN-gamma (IFN-γ), a critically important immunomodulator, has been widely studied in MS pathology. The confusing and complex effects of IFN-γ in MS patients and rodent models, however, cause us to look more closely at its exact role in MS. In this study, we identified the role of the IFN-γ signaling in the choroid plexus (CP) in the experimental autoimmune encephalomyelitis (EAE) model. We found that the IFN-γ signal was rapidly amplified when CNS immune cell infiltration occurred in the CP during the progressive stage. Furthermore, using two CP-specific knockdown strategies, we demonstrated that blocking the IFN-γ signal via knockdown of IFN-γR1 in the CP could protect mice against EAE pathology, as evidenced by improvements in clinical scores and infiltration. Notably, knocking down IFN-γR1 in the CP reduced the local expression of adhesion molecules and chemokines. This finding suggests that IFN-γ signaling in the CP may participate in the pathological process of EAE by preventing pathological T helper (Th) 17+ cells from infiltrating into the CNS. Finally, we showed that the unbalanced state of IFN-γ signaling between peripheral lymphocytes and the choroid plexus may determine whether IFN-γ has a protective or aggravating effect on EAE pathology. Above all, we discovered that IFN-γR1-mediated IFN-γ signaling in the CP was a vital pathway in the pathological process of EAE.
Collapse
Affiliation(s)
- Yuyin Zheng
- Rehabilitation Medicine CenterThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Integrative & Optimized Medicine Research center, China‐USA Institute for Acupuncture and RehabilitationWenzhou Medical UniversityWenzhouChina
| | - Lanxin Hu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- The Molecular Neuropharmacology Laboratory and the Eye‐Brain Research Center, School of Ophthalmology & Optometry and Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Yuwen Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- The Molecular Neuropharmacology Laboratory and the Eye‐Brain Research Center, School of Ophthalmology & Optometry and Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Cheng Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- The Molecular Neuropharmacology Laboratory and the Eye‐Brain Research Center, School of Ophthalmology & Optometry and Eye HospitalWenzhou Medical UniversityWenzhouChina
| | - Wenzhan Tu
- Rehabilitation Medicine CenterThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Integrative & Optimized Medicine Research center, China‐USA Institute for Acupuncture and RehabilitationWenzhou Medical UniversityWenzhouChina
| | - Haiyan Lin
- Rehabilitation Medicine CenterThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Integrative & Optimized Medicine Research center, China‐USA Institute for Acupuncture and RehabilitationWenzhou Medical UniversityWenzhouChina
| | - Haotian Wang
- Alberta InstituteWenzhou Medical UniversityWenzhouChina
| | - Yiwei Jiang
- Alberta InstituteWenzhou Medical UniversityWenzhouChina
| | - Songhe Jiang
- Rehabilitation Medicine CenterThe Second Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Integrative & Optimized Medicine Research center, China‐USA Institute for Acupuncture and RehabilitationWenzhou Medical UniversityWenzhouChina
| | - Wu Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye HospitalWenzhou Medical UniversityWenzhouChina
- The Molecular Neuropharmacology Laboratory and the Eye‐Brain Research Center, School of Ophthalmology & Optometry and Eye HospitalWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
20
|
Karimy JK, Newville JC, Sadegh C, Morris JA, Monuki ES, Limbrick DD, McAllister Ii JP, Koschnitzky JE, Lehtinen MK, Jantzie LL. Outcomes of the 2019 hydrocephalus association workshop, "Driving common pathways: extending insights from posthemorrhagic hydrocephalus". Fluids Barriers CNS 2023; 20:4. [PMID: 36639792 PMCID: PMC9838022 DOI: 10.1186/s12987-023-00406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
The Hydrocephalus Association (HA) workshop, Driving Common Pathways: Extending Insights from Posthemorrhagic Hydrocephalus, was held on November 4 and 5, 2019 at Washington University in St. Louis. The workshop brought together a diverse group of basic, translational, and clinical scientists conducting research on multiple hydrocephalus etiologies with select outside researchers. The main goals of the workshop were to explore areas of potential overlap between hydrocephalus etiologies and identify drug targets that could positively impact various forms of hydrocephalus. This report details the major themes of the workshop and the research presented on three cell types that are targets for new hydrocephalus interventions: choroid plexus epithelial cells, ventricular ependymal cells, and immune cells (macrophages and microglia).
Collapse
Affiliation(s)
- Jason K Karimy
- Department of Family Medicine, Mountain Area Health Education Center - Boone, North Carolina, 28607, USA
| | - Jessie C Newville
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Cameron Sadegh
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, MA, Boston, 02114, USA
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Jill A Morris
- National Institute of Neurological Disorders and Stroke, Neuroscience Center, National Institutes of Health, 6001 Executive Blvd, NSC Rm 2112, Bethesda, MD, 20892, USA
| | - Edwin S Monuki
- Departments of Pathology & Laboratory Medicine and Developmental & Cell Biology, University of California Irvine, Irvine, CA, 92697, USA
| | - David D Limbrick
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - James P McAllister Ii
- Departments of Neurosurgery and Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | | | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, 02115, USA.
| | - Lauren L Jantzie
- Department of Pediatrics and Neurosurgery, Johns Hopkins Children's Center, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
- Kennedy Krieger Institute, Baltimore, MD, 21287, USA.
| |
Collapse
|
21
|
Alvarez MR, Alarcon JM, Roman CA, Lazaro D, Bobrowski-Khoury N, Baena-Caldas GP, Esber GR. Can a basic solution activate the inflammatory reflex? A review of potential mechanisms, opportunities, and challenges. Pharmacol Res 2023; 187:106525. [PMID: 36441036 DOI: 10.1016/j.phrs.2022.106525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/09/2022] [Accepted: 10/25/2022] [Indexed: 11/24/2022]
Abstract
Stimulation of the inflammatory reflex (IR) is a promising strategy to treat systemic inflammatory disorders. However, this strategy is hindered by the cost and side effects of traditional IR activators. Recently, oral intake of sodium bicarbonate (NaHCO3) has been suggested to activate the IR, providing a safe and inexpensive alternative. Critically, the mechanisms whereby NaHCO3 might achieve this effect and more broadly the pathways underlying the IR remain poorly understood. Here, we argue that the recognition of NaHCO3 as a potential IR activator presents exciting clinical and research opportunities. To aid this quest, we provide an integrative review of our current knowledge of the neural and cellular pathways mediating the IR and discuss the status of physiological models of IR activation. From this vantage point, we derive testable hypotheses on potential mechanisms whereby NaHCO3 might stimulate the IR and compare NaHCO3 with classic IR activators. Elucidation of these mechanisms will help determine the therapeutic value of NaHCO3 as an IR activator and provide new insights into the IR circuitry.
Collapse
Affiliation(s)
- Milena Rodriguez Alvarez
- Department of Internal Medicine, Division of Rheumatology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA.
| | - Juan Marcos Alarcon
- Department of Pathology, The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Christopher A Roman
- Department of Cell Biology, State University of New York (SUNY) Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Deana Lazaro
- Division of Rheumatology, Department of Internal Medicine, Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY, USA
| | | | | | | |
Collapse
|
22
|
Kann O, Almouhanna F, Chausse B. Interferon γ: a master cytokine in microglia-mediated neural network dysfunction and neurodegeneration. Trends Neurosci 2022; 45:913-927. [PMID: 36283867 DOI: 10.1016/j.tins.2022.10.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Traditionally, lymphocytic interferon γ (IFN-γ) was considered to be a simple 'booster' of proinflammatory responses by microglia (brain-resident macrophages) during bacterial or viral infection. Recent slice culture (in situ) and in vivo studies suggest, however, that IFN-γ has a unique role in microglial activation. Priming by IFN-γ results in proliferation (microgliosis), enhanced synapse elimination, and moderate nitric oxide release sufficient to impair synaptic transmission, gamma rhythm activity, and cognitive functions. Moreover, IFN-γ is pivotal for driving Toll-like receptor (TLR)-activated microglia into neurotoxic phenotypes that induce energetic and oxidative stress, severe network dysfunction, and neuronal death. Pharmacological targeting of activated microglia could be beneficial during elevated IFN-γ levels, blood-brain barrier leakage, and parenchymal T lymphocyte infiltration associated with, for instance, encephalitis, multiple sclerosis, and Alzheimer's disease.
Collapse
Affiliation(s)
- Oliver Kann
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, D-69120 Heidelberg, Germany.
| | - Fadi Almouhanna
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Bruno Chausse
- Institute of Physiology and Pathophysiology, University of Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
23
|
Hulme L, Hochstetler A, Schwerk C, Schroten H, Ishikawa H, Tung CY, Perrin B, Blazer-Yost B. Characterization of TRPV4-mediated signaling pathways in an optimized human choroid plexus epithelial cell line. Am J Physiol Cell Physiol 2022; 323:C1823-C1842. [PMID: 35938676 PMCID: PMC9744646 DOI: 10.1152/ajpcell.00193.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022]
Abstract
The objectives of these studies were twofold: 1) to characterize the human choroid plexus papilloma (HIBCPP) cell line as a model of the blood-cerebrospinal fluid barrier (BCSFB) via morphology, tightness, and polarization of transporters in choroid plexus epithelia (CPe), and 2) to utilize Ussing-style electrophysiology to elucidate signaling pathways associated with the activation of the transient receptor potential vanilloid 4 (TRPV4) channel involved in cerebrospinal fluid (CSF) secretion. RT-PCR was implemented to determine gene expression of cell fate markers, junctional complex proteins, and transporters of interest. Scanning electron microscopy and confocal three-dimensional renderings of cultures grown on permeable supports were utilized to delineate the morphology of the brush border, junctional complexes, and polarization of key transporters. Electrophysiology was used to understand and explore TRPV4-mediated signaling in the HIBCPP cell line, considering both short-circuit current (Isc) and conductance responses. HIBCPP cells grown under optimized culture conditions exhibited minimal multilayering, developed an intermediate resistance monolayer, retained differentiation properties, and expressed, and correctly localized, junctional proteins and native transporters. We found that activation of TRPV4 resulted in a robust, multiphasic change in electrogenic ion flux and increase in conductance accompanied by substantial fluid secretion. This response appears to be modulated by a number of different effectors, implicating phospholipase C (PLC), protein kinase C (PKC), and phosphoinositide 3-kinase (PI3K) in TRPV4-mediated ion flux. The HIBCPP cell line is a representative model of the human BCSFB, which can be utilized for studies of transporter function, intracellular signaling, and regulation of CSF production.
Collapse
Affiliation(s)
- Louise Hulme
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Alexandra Hochstetler
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Christian Schwerk
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Horst Schroten
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hiroshi Ishikawa
- Department of Neurosurgery, Laboratory of Clinical Regenerative Medicine, University of Tsukuba, Ibaraki, Japan
| | - Chun-Yu Tung
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Benjamin Perrin
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Bonnie Blazer-Yost
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
24
|
Liu R, Zhang Z, Chen Y, Liao J, Wang Y, Liu J, Lin Z, Xiao G. Choroid plexus epithelium and its role in neurological diseases. Front Mol Neurosci 2022; 15:949231. [PMID: 36340696 PMCID: PMC9633854 DOI: 10.3389/fnmol.2022.949231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/15/2022] [Indexed: 02/16/2024] Open
Abstract
Choroid plexus epithelial cells can secrete cerebrospinal fluid into the ventricles, serving as the major structural basis of the selective barrier between the neurological system and blood in the brain. In fact, choroid plexus epithelial cells release the majority of cerebrospinal fluid, which is connected with particular ion channels in choroid plexus epithelial cells. Choroid plexus epithelial cells also produce and secrete a number of essential growth factors and peptides that help the injured cerebrovascular system heal. The pathophysiology of major neurodegenerative disorders like Alzheimer's disease, Parkinson's disease, as well as minor brain damage diseases like hydrocephalus and stroke is still unknown. Few studies have previously connected choroid plexus epithelial cells to the etiology of these serious brain disorders. Therefore, in the hopes of discovering novel treatment options for linked conditions, this review extensively analyzes the association between choroid plexus epithelial cells and the etiology of neurological diseases such as Alzheimer's disease and hydrocephalus. Finally, we review CPE based immunotherapy, choroid plexus cauterization, choroid plexus transplantation, and gene therapy.
Collapse
Affiliation(s)
- Ruizhen Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiping Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yibing Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Junbo Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yuchang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jingping Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Lin
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
Blazer-Yost BL. Following Ussing's legacy: from amphibian models to mammalian kidney and brain. Am J Physiol Cell Physiol 2022; 323:C1061-C1069. [PMID: 36036449 PMCID: PMC9529261 DOI: 10.1152/ajpcell.00303.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022]
Abstract
Professor Hans H. Ussing (1911-2000) was one of the founding members of the field of epithelial cell biology. He is most famous for the electrophysiological technique that he developed to measure electrogenic ion flux across epithelial tissues. Ussing-style electrophysiology has been applied to multiple tissues and has informed fields as diverse as amphibian biology and medicine. In the latter, this technique has contributed to a basic understanding of maladies such as hypertension, polycystic kidney disease, cystic fibrosis, and diarrheal diseases to mention but a few. In addition to this valuable contribution to biological methods, Prof. Ussing also provided strong evidence for the concept of active transport several years before the elucidation of Na+K+ATPase. In addition, he provided cell biologists with the important concept of polarized epithelia with specific and different transporters found in the apical and basolateral membranes, thus providing these cells with the ability to conduct directional, active and passive transepithelial transport. My studies have used Ussing chamber electrophysiology to study the toad urinary bladder, an amphibian cell line, renal cell lines, and, most recently, choroid plexus cell lines. This technique has formed the basis of our in vitro mechanistic studies that are used in an iterative manner with animal models to better understand disease progress and treatment. I was honored to be invited to deliver the 2022 Hans Ussing Lecture sponsored by the Epithelial Transport Group of the American Physiological Society. This manuscript is a version of the material presented in that lecture.
Collapse
Affiliation(s)
- Bonnie L Blazer-Yost
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
26
|
Driedonks T, Jiang L, Carlson B, Han Z, Liu G, Queen SE, Shirk EN, Gololobova O, Liao Z, Nyberg LH, Lima G, Paniushkina L, Garcia‐Contreras M, Schonvisky K, Castell N, Stover M, Guerrero‐Martin S, Richardson R, Smith B, Mahairaki V, Lai CP, Izzi JM, Hutchinson EK, Pate KAM, Witwer KW. Pharmacokinetics and biodistribution of extracellular vesicles administered intravenously and intranasally to Macaca nemestrina. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e59. [PMID: 36591537 PMCID: PMC9799283 DOI: 10.1002/jex2.59] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/23/2022] [Accepted: 08/13/2022] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) have potential in disease treatment since they can be loaded with therapeutic molecules and engineered for retention by specific tissues. However, questions remain on optimal dosing, administration, and pharmacokinetics. Previous studies have addressed biodistribution and pharmacokinetics in rodents, but little evidence is available for larger animals. Here, we investigated the pharmacokinetics and biodistribution of Expi293F-derived EVs labelled with a highly sensitive nanoluciferase reporter (palmGRET) in a non-human primate model (Macaca nemestrina), comparing intravenous (IV) and intranasal (IN) administration over a 125-fold dose range. We report that EVs administered IV had longer circulation times in plasma than previously reported in mice and were detectable in cerebrospinal fluid (CSF) after 30-60 minutes. EV association with PBMCs, especially B-cells, was observed as early as one minute post-administration. EVs were detected in liver and spleen within one hour of IV administration. However, IN delivery was minimal, suggesting that pretreatment approaches may be needed in large animals. Furthermore, EV circulation times strongly decreased after repeated IV administration, possibly due to immune responses and with clear implications for xenogeneic EV-based therapeutics. We hope that our findings from this baseline study in macaques will help to inform future research and therapeutic development of EVs.
Collapse
Affiliation(s)
- Tom Driedonks
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Linglei Jiang
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Bess Carlson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zheng Han
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Guanshu Liu
- Russell H. Morgan Department of RadiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- F.M. Kirby Research CenterKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Suzanne E. Queen
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Erin N. Shirk
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Olesia Gololobova
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Zhaohao Liao
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lyle H. Nyberg
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Gabriela Lima
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Liliia Paniushkina
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Marta Garcia‐Contreras
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kayla Schonvisky
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Natalie Castell
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mitchel Stover
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Selena Guerrero‐Martin
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Riley Richardson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Barbara Smith
- Department of Cell BiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Vasiliki Mahairaki
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Charles P. Lai
- Institute of Atomic and Molecular SciencesAcademia SinicaTaipeiTaiwan
| | - Jessica M. Izzi
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Eric K. Hutchinson
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kelly A. M. Pate
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Division of Comparative Medicine and Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
27
|
Bitanihirwe BKY, Lizano P, Woo TUW. Deconstructing the functional neuroanatomy of the choroid plexus: an ontogenetic perspective for studying neurodevelopmental and neuropsychiatric disorders. Mol Psychiatry 2022; 27:3573-3582. [PMID: 35618887 PMCID: PMC9133821 DOI: 10.1038/s41380-022-01623-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023]
Abstract
The choroid plexus (CP) is a delicate and highly vascularized structure in the brain comprised of a dense network of fenestrated capillary loops that help in the synthesis, secretion and circulation of cerebrospinal fluid (CSF). This unique neuroanatomical structure is comprised of arachnoid villi stemming from frond-like surface projections-that protrude into the lumen of the four cerebral ventricles-providing a key source of nutrients to the brain parenchyma in addition to serving as a 'sink' for central nervous system metabolic waste. In fact, the functions of the CP are often described as being analogous to those of the liver and kidney. Beyond forming a barrier/interface between the blood and CSF compartments, the CP has been identified as a modulator of leukocyte trafficking, inflammation, cognition, circadian rhythm and the gut brain-axis. In recent years, advances in molecular biology techniques and neuroimaging along with the use of sophisticated animal models have played an integral role in shaping our understanding of how the CP-CSF system changes in relation to the maturation of neural circuits during critical periods of brain development. In this article we provide an ontogenetic perspective of the CP and review the experimental evidence implicating this structure in the pathophysiology of neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Byron K Y Bitanihirwe
- Humanitarian and Conflict Response Institute, University of Manchester, Manchester, UK.
| | - Paulo Lizano
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Translational Neuroscience Division, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tsung-Ung W Woo
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Program in Molecular Neuropathology, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
28
|
Oernbo EK, Steffensen AB, Razzaghi Khamesi P, Toft-Bertelsen TL, Barbuskaite D, Vilhardt F, Gerkau NJ, Tritsaris K, Simonsen AH, Lolansen SD, Andreassen SN, Hasselbalch SG, Zeuthen T, Rose CR, Kurtcuoglu V, MacAulay N. Membrane transporters control cerebrospinal fluid formation independently of conventional osmosis to modulate intracranial pressure. Fluids Barriers CNS 2022; 19:65. [PMID: 36038945 PMCID: PMC9422132 DOI: 10.1186/s12987-022-00358-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/13/2022] [Indexed: 11/23/2022] Open
Abstract
Background Disturbances in the brain fluid balance can lead to life-threatening elevation in the intracranial pressure (ICP), which represents a vast clinical challenge. Nevertheless, the details underlying the molecular mechanisms governing cerebrospinal fluid (CSF) secretion are largely unresolved, thus preventing targeted and efficient pharmaceutical therapy of cerebral pathologies involving elevated ICP. Methods Experimental rats were employed for in vivo determinations of CSF secretion rates, ICP, blood pressure and ex vivo excised choroid plexus for morphological analysis and quantification of expression and activity of various transport proteins. CSF and blood extractions from rats, pigs, and humans were employed for osmolality determinations and a mathematical model employed to determine a contribution from potential local gradients at the surface of choroid plexus. Results We demonstrate that CSF secretion can occur independently of conventional osmosis and that local osmotic gradients do not suffice to support CSF secretion. Instead, the CSF secretion across the luminal membrane of choroid plexus relies approximately equally on the Na+/K+/2Cl− cotransporter NKCC1, the Na+/HCO3− cotransporter NBCe2, and the Na+/K+-ATPase, but not on the Na+/H+ exchanger NHE1. We demonstrate that pharmacological modulation of CSF secretion directly affects the ICP. Conclusions CSF secretion appears to not rely on conventional osmosis, but rather occur by a concerted effort of different choroidal transporters, possibly via a molecular mode of water transport inherent in the proteins themselves. Therapeutic modulation of the rate of CSF secretion may be employed as a strategy to modulate ICP. These insights identify new promising therapeutic targets against brain pathologies associated with elevated ICP. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00358-4.
Collapse
Affiliation(s)
- Eva K Oernbo
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Annette B Steffensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Pooya Razzaghi Khamesi
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Dagne Barbuskaite
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Niklas J Gerkau
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Katerina Tritsaris
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Anja H Simonsen
- Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Sara D Lolansen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Søren N Andreassen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Steen G Hasselbalch
- Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Zeuthen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225, Düsseldorf, Germany
| | - Vartan Kurtcuoglu
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
29
|
Dabbagh F, Schroten H, Schwerk C. In Vitro Models of the Blood–Cerebrospinal Fluid Barrier and Their Applications in the Development and Research of (Neuro)Pharmaceuticals. Pharmaceutics 2022; 14:pharmaceutics14081729. [PMID: 36015358 PMCID: PMC9412499 DOI: 10.3390/pharmaceutics14081729] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 11/30/2022] Open
Abstract
The pharmaceutical research sector has been facing the challenge of neurotherapeutics development and its inherited high-risk and high-failure-rate nature for decades. This hurdle is partly attributable to the presence of brain barriers, considered both as obstacles and opportunities for the entry of drug substances. The blood–cerebrospinal fluid (CSF) barrier (BCSFB), an under-studied brain barrier site compared to the blood–brain barrier (BBB), can be considered a potential therapeutic target to improve the delivery of CNS therapeutics and provide brain protection measures. Therefore, leveraging robust and authentic in vitro models of the BCSFB can diminish the time and effort spent on unproductive or redundant development activities by a preliminary assessment of the desired physiochemical behavior of an agent toward this barrier. To this end, the current review summarizes the efforts and progresses made to this research area with a notable focus on the attribution of these models and applied techniques to the pharmaceutical sector and the development of neuropharmacological therapeutics and diagnostics. A survey of available in vitro models, with their advantages and limitations and cell lines in hand will be provided, followed by highlighting the potential applications of such models in the (neuro)therapeutics discovery and development pipelines.
Collapse
|
30
|
de Mélo Silva Júnior ML, Diniz PRB, de Souza Vilanova MV, Basto GPT, Valença MM. Brain ventricles, CSF and cognition: a narrative review. Psychogeriatrics 2022; 22:544-552. [PMID: 35488797 DOI: 10.1111/psyg.12839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/07/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022]
Abstract
The brain ventricles are structures that have been related to cognition since antiquity. They are essential components in the development and maintenance of brain functions. The aging process runs with the enlargement of ventricles and is related to a less selective blood-cerebrospinal fluid barrier and then a more toxic cerebrospinal fluid environment. The study of brain ventricles as a biological marker of aging is promissing because they are structures easily identified in neuroimaging studies, present good inter-rater reliability, and measures of them can identify brain atrophy earlier than cortical structures. The ventricular system also plays roles in the development of dementia, since dysfunction in the clearance of beta-amyloid protein is a key mechanism in sporadic Alzheimer's disease. The morphometric and volumetric studies of the brain ventricles can help to distinguish between healthy elderly and persons with mild cognitive impairment (MCI) and dementia. Brain ventricle data may contribute to the appropriate allocation of individuals in groups at higher risk for MCI-dementia progression in clinical trials and to measuring therapeutic responses in these studies, as well as providing differential diagnosis, such as normal pressure hydrocephalus. Here, we reviewed the pathophysiology of healthy aging and cognitive decline, focusing on the role of the choroid plexus and brain ventricles in this process.
Collapse
Affiliation(s)
- Mário Luciano de Mélo Silva Júnior
- Medical School, Universidade Federal de Pernambuco, Recife, Brazil.,Medical School, Centro Universitário Maurício de Nassau, Recife, Brazil.,Neurology Unit, Hospital da Restauração, Recife, Brazil
| | | | | | | | | |
Collapse
|
31
|
Hochstetler A, Hulme L, Delpire E, Schwerk C, Schroten H, Preston D, Simpson S, Blazer-Yost BL. Porcine choroid plexus-Riems cell line demonstrates altered polarization of transport proteins compared with the native epithelium. Am J Physiol Cell Physiol 2022; 323:C1-C13. [PMID: 35508188 PMCID: PMC9236870 DOI: 10.1152/ajpcell.00374.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023]
Abstract
The choroid plexus epithelium (CPe) forms a barrier between the cerebral blood supply and the cerebrospinal fluid (CSF), establishing the blood-CSF barrier (BCSFB). CSF is actively secreted by the CPe via tightly controlled processes involving multiple channels, transporters, and pumps. The importance of controlling CSF production and composition has been accentuated recently with an appreciation of CSF dysfunction in many pathologies. For mechanistic studies of CSF production, isolated CPe cell lines are valuable for the testing of hypotheses and potential drug targets. Although several continuous CPe cell lines have been described, none appear to have all the characteristics of the native epithelium and each must be used judiciously. The porcine choroid plexus-Riems (PCP-R) cell line forms a high-resistance monolayer characteristic of a barrier epithelium. Conservation of this phenotype is unusual among CPe cell lines, making this model useful for studies of the effects of infection, injury, and drugs on permeability. We have recently discovered that, although this line expresses many of the transporters expressed in the native tissue, some are mispolarized. As a result, inferences regarding fluid/electrolyte flux and the resultant CSF production should be pursued with caution. Furthermore, extended culture periods and changes in media composition result in significant morphological and functional variability. These studies provide a more detailed characterization of the PCP-R cell line concerning transporter expression, polarization, and functionality, as well as plasticity in culture, with the goal to provide the scientific community with information necessary to optimize future experiments with this model.
Collapse
Affiliation(s)
- Alexandra Hochstetler
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Louise Hulme
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| | - Eric Delpire
- Department of Anesthesiology and Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Christian Schwerk
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Horst Schroten
- Department of Pediatrics, Pediatric Infectious Diseases, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Preston
- Indiana Biosciences Research Institute, Indianapolis, Indiana
| | - Stefanie Simpson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Bonnie L Blazer-Yost
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
32
|
Zhang Z, Tan Q, Guo P, Huang S, Jia Z, Liu X, Feng H, Chen Y. NLRP3 inflammasome-mediated choroid plexus hypersecretion contributes to hydrocephalus after intraventricular hemorrhage via phosphorylated NKCC1 channels. J Neuroinflammation 2022; 19:163. [PMID: 35729645 PMCID: PMC9210649 DOI: 10.1186/s12974-022-02530-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 06/14/2022] [Indexed: 12/26/2022] Open
Abstract
Background Hydrocephalus is a severe complication of intracerebral hemorrhage with ventricular extension (ICH-IVH) and causes cerebrospinal fluid (CSF) accumulation. The choroid plexus epithelium plays an important role in CSF secretion and constitutes the blood–CSF barrier within the brain–immune system interface. Although the NLRP3 inflammasome, as a key component of the innate immune system, promotes neuroinflammation, its role in the pathogenesis of hydrocephalus after hemorrhage has not been investigated. Therefore, this study aimed to investigate the potential mechanism of NLRP3 in hydrocephalus to discover a potential marker for targeted therapy. Methods A rat model of hydrocephalus after ICH-IVH was developed through autologous blood infusion in wild-type and Nlrp3−/− rats. By studying the features and processes of the model, we investigated the relationship between the NLRP3 inflammasome and CSF hypersecretion in the choroid plexus. Results The ICH-IVH model rats showed ventricular dilation accompanied by CSF hypersecretion for 3 days. Based on the choroid plexus RNA-seq and proteomics results, we found that an inflammatory response was activated. The NLRP3 inflammasome was investigated, and the expression levels of NLRP3 inflammasome components reached a peak at 3 days after ICH-IVH. Inhibition of NLRP3 by an MCC950 inflammasome inhibitor or Nlrp3 knockout decreased CSF secretion and ventricular dilation and attenuated neurological deficits after ICH-IVH. The mechanism underlying the neuroprotective effects of NLRP3 inhibition involved decreased phosphorylation of NKCC1, which is a major protein that regulates CSF secretion by altering Na+- and K+-coupled water transport, via MCC950 or Nlrp3 knockout. In combination with the in vitro experiments, this experiment confirmed the involvement of the NLRP3/p-NKCC1 pathway and Na+ and K+ flux. Conclusions This study demonstrates that NKCC1 phosphorylation in the choroid plexus epithelium promotes NLRP3 inflammasome-mediated CSF hypersecretion and that NLRP3 plays an important role in the pathogenesis of hydrocephalus after hemorrhage. These findings provide a new therapeutic strategy for treating hydrocephalus. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02530-x.
Collapse
Affiliation(s)
- Zhaoqi Zhang
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qiang Tan
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Suna Huang
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhengcai Jia
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Liu
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| |
Collapse
|
33
|
Histological reinterpretation of paraphysis cerebri in Ambystoma mexicanum. Acta Histochem 2022; 124:151915. [PMID: 35738026 DOI: 10.1016/j.acthis.2022.151915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/02/2022] [Accepted: 06/05/2022] [Indexed: 11/22/2022]
Abstract
Intraventricular and extraventricular choroid plexuses are neuroepithelial folds which arise from the roof of the diencephalon. We describe the circumventricular structure of the diencephalon roof (paraphysis cerebri) during the various development stages of Ambystoma mexicanum. The parasagittal sections of the larvae epithalamus exhibit the presence, in addition to the epiphysis, of two dorsal primordia in nearby areas, which appear to be extraventricular saccular evaginations of different origin that give rise to two structures we define as the anterior extraventricular choroid plexus (AEP) and posterior extraventricular choroid plexus (PEP). During larvae development, the primordia arise perpendicular to each other, grow and show luminal folds and invaginations. Later, the two extraventricular evaginations, which are separate units, become interrelated. As the PEP grows, it covers the AEP dorsally, but it is difficult to define the borders of these organs. AEP is formed by alveolar-acinar epithelial aggregates with evidence of secretion-like content. PEP structure is like a choroid plexus, but its position is extraventricular and dorsal to the AEP. The PEP is always between the AEP and the meninges and can be small or large in size. This means that in A. mexicanum, the paraphysis cerebri is made up of two adjacent organs, which arise almost simultaneously from two different primordia (the AEP and the PEP) and as the posterior one grows, it overlaps the anterior one and masks itself. In conclusion, we suggest that AEP and PEP are homologous to paraphysis cerebri and the dorsal sac, respectively.
Collapse
|
34
|
Targeting choroid plexus epithelium as a novel therapeutic strategy for hydrocephalus. J Neuroinflammation 2022; 19:156. [PMID: 35715859 PMCID: PMC9205094 DOI: 10.1186/s12974-022-02500-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
The choroid plexus is a tissue located in the lateral ventricles of the brain and is composed mainly of choroid plexus epithelium cells. The main function is currently thought to be the secretion of cerebrospinal fluid and the regulation of its pH, and more functions are gradually being demonstrated. Assistance in the removal of metabolic waste and participation in the apoptotic pathway are also the functions of choroid plexus. Besides, it helps to repair the brain by regulating the secretion of neuropeptides and the delivery of drugs. It is involved in the immune response to assist in the clearance of infections in the central nervous system. It is now believed that the choroid plexus is in an inflammatory state after damage to the brain. This state, along with changes in the cilia, is thought to be an abnormal physiological state of the choroid plexus, which in turn leads to abnormal conditions in cerebrospinal fluid and triggers hydrocephalus. This review describes the pathophysiological mechanism of hydrocephalus following choroid plexus epithelium cell abnormalities based on the normal physiological functions of choroid plexus epithelium cells, and analyzes the attempts and future developments of using choroid plexus epithelium cells as a therapeutic target for hydrocephalus.
Collapse
|
35
|
Gibel-Russo R, Benacom D, Di Nardo AA. Non-Cell-Autonomous Factors Implicated in Parvalbumin Interneuron Maturation and Critical Periods. Front Neural Circuits 2022; 16:875873. [PMID: 35601531 PMCID: PMC9115720 DOI: 10.3389/fncir.2022.875873] [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: 02/14/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
From birth to adolescence, the brain adapts to its environmental stimuli through structural and functional remodeling of neural circuits during critical periods of heightened plasticity. They occur across modalities for proper sensory, motor, linguistic, and cognitive development. If they are disrupted by early-life adverse experiences or genetic deficiencies, lasting consequences include behavioral changes, physiological and cognitive deficits, or psychiatric illness. Critical period timing is orchestrated not only by appropriate neural activity but also by a multitude of signals that participate in the maturation of fast-spiking parvalbumin interneurons and the consolidation of neural circuits. In this review, we describe the various signaling factors that initiate critical period onset, such as BDNF, SPARCL1, or OTX2, which originate either from local neurons or glial cells or from extracortical sources such as the choroid plexus. Critical period closure is established by signals that modulate extracellular matrix and myelination, while timing and plasticity can also be influenced by circadian rhythms and by hormones and corticosteroids that affect brain oxidative stress levels or immune response. Molecular outcomes include lasting epigenetic changes which themselves can be considered signals that shape downstream cross-modal critical periods. Comprehensive knowledge of how these signals and signaling factors interplay to influence neural mechanisms will help provide an inclusive perspective on the effects of early adversity and developmental defects that permanently change perception and behavior.
Collapse
|
36
|
Bramall AN, Anton ES, Kahle KT, Fecci PE. Navigating the ventricles: Novel insights into the pathogenesis of hydrocephalus. EBioMedicine 2022; 78:103931. [PMID: 35306341 PMCID: PMC8933686 DOI: 10.1016/j.ebiom.2022.103931] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/16/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Congenital hydrocephalus occurs in one in 500-1000 babies born in the United States and acquired hydrocephalus may occur as the consequence of stroke, intraventricular and subarachnoid hemorrhage, traumatic brain injuries, brain tumors, craniectomy or may be idiopathic, as in the case of normal pressure hydrocephalus. Irrespective of its prevalence and significant impact on quality of life, neurosurgeons still rely on invasive cerebrospinal fluid shunt systems for the treatment of hydrocephalus that are exceptionally prone to failure and/or infection. Further understanding of this process at a molecular level, therefore, may have profound implications for improving treatment and quality of life for millions of individuals worldwide. The purpose of this article is to review the current research landscape on hydrocephalus with a focus on recent advances in our understanding of cerebrospinal fluid pathways from an evolutionary, genetics and molecular perspective.
Collapse
Affiliation(s)
- Alexa N Bramall
- Department of Neurosurgery, Duke University Hospital, 2301 Erwin Rd., Durham, NC 27710, United States.
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, United States
| | - Peter E Fecci
- Department of Neurosurgery, Duke University Hospital, 2301 Erwin Rd., Durham, NC 27710, United States
| |
Collapse
|
37
|
Fang Y, Huang L, Wang X, Si X, Lenahan C, Shi H, Shao A, Tang J, Chen S, Zhang J, Zhang JH. A new perspective on cerebrospinal fluid dynamics after subarachnoid hemorrhage: From normal physiology to pathophysiological changes. J Cereb Blood Flow Metab 2022; 42:543-558. [PMID: 34806932 PMCID: PMC9051143 DOI: 10.1177/0271678x211045748] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 11/17/2022]
Abstract
Knowledge about the dynamic metabolism and function of cerebrospinal fluid (CSF) physiology has rapidly progressed in recent decades. It has traditionally been suggested that CSF is produced by the choroid plexus and drains to the arachnoid villi. However, recent findings have revealed that the brain parenchyma produces a large portion of CSF and drains through the perivascular glymphatic system and meningeal lymphatic vessels into the blood. The primary function of CSF is not limited to maintaining physiological CNS homeostasis but also participates in clearing waste products resulting from neurodegenerative diseases and acute brain injury. Aneurysmal subarachnoid hemorrhage (SAH), a disastrous subtype of acute brain injury, is associated with high mortality and morbidity. Post-SAH complications contribute to the poor outcomes associated with SAH. Recently, abnormal CSF flow was suggested to play an essential role in the post-SAH pathophysiological changes, such as increased intracerebral pressure, brain edema formation, hydrocephalus, and delayed blood clearance. An in-depth understanding of CSF dynamics in post-SAH events would shed light on potential development of SAH treatment options. This review summarizes and updates the latest physiological characteristics of CSF dynamics and discusses potential pathophysiological changes and therapeutic targets after SAH.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Huang
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoli Si
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cameron Lenahan
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
- Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Hui Shi
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
- Department of Neurosurgery, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiping Tang
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - John H Zhang
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
38
|
MacAulay N, Keep RF, Zeuthen T. Cerebrospinal fluid production by the choroid plexus: a century of barrier research revisited. Fluids Barriers CNS 2022; 19:26. [PMID: 35317823 PMCID: PMC8941821 DOI: 10.1186/s12987-022-00323-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/09/2022] [Indexed: 12/20/2022] Open
Abstract
Cerebrospinal fluid (CSF) envelops the brain and fills the central ventricles. This fluid is continuously replenished by net fluid extraction from the vasculature by the secretory action of the choroid plexus epithelium residing in each of the four ventricles. We have known about these processes for more than a century, and yet the molecular mechanisms supporting this fluid secretion remain unresolved. The choroid plexus epithelium secretes its fluid in the absence of a trans-epithelial osmotic gradient, and, in addition, has an inherent ability to secrete CSF against an osmotic gradient. This paradoxical feature is shared with other 'leaky' epithelia. The assumptions underlying the classical standing gradient hypothesis await experimental support and appear to not suffice as an explanation of CSF secretion. Here, we suggest that the elusive local hyperosmotic compartment resides within the membrane transport proteins themselves. In this manner, the battery of plasma membrane transporters expressed in choroid plexus are proposed to sustain the choroidal CSF secretion independently of the prevailing bulk osmotic gradient.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Thomas Zeuthen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| |
Collapse
|
39
|
Munro DAD, Movahedi K, Priller J. Macrophage compartmentalization in the brain and cerebrospinal fluid system. Sci Immunol 2022; 7:eabk0391. [PMID: 35245085 DOI: 10.1126/sciimmunol.abk0391] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Macrophages reside within the diverse anatomical compartments of the central nervous system (CNS). Within each compartment, these phagocytes are exposed to unique combinations of niche signals and mechanical stimuli that instruct their tissue-specific identities. Whereas most CNS macrophages are tissue-embedded, the macrophages of the cerebrospinal fluid (CSF) system are bathed in an oscillating liquid. Studies using multiomics technologies have recently uncovered the transcriptomic and proteomic profiles of CSF macrophages, enhancing our understanding of their cellular characteristics in both rodents and humans. Here, we review the relationships between CNS macrophage populations, with a focus on the origins, phenotypes, and functions of CSF macrophages in health and disease.
Collapse
Affiliation(s)
- David A D Munro
- UK Dementia Research Institute at University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium.,Laboratory of Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Josef Priller
- UK Dementia Research Institute at University of Edinburgh, Edinburgh EH16 4TJ, UK.,Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin and DZNE, 10117 Berlin, Germany.,Technical University of Munich, School of Medicine, Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, 81675 Munich, Germany.,Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London SE5 8AF, UK
| |
Collapse
|
40
|
Severe Acute Hepatic Dysfunction Induced by Ammonium Acetate Treatment Results in Choroid Plexus Swelling and Ventricle Enlargement in the Brain. Int J Mol Sci 2022; 23:ijms23042010. [PMID: 35216129 PMCID: PMC8879736 DOI: 10.3390/ijms23042010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/08/2022] [Accepted: 02/10/2022] [Indexed: 02/04/2023] Open
Abstract
Hepatic encephalopathy is a major cause of liver failure. However, the pathophysiological role of ventricle enlargement in brain edema remains unclear. Here, we used an acute hepatic encephalopathy mouse model to examine the sequential pathological changes in the brain associated with this condition. We collected tissue samples from experimental animals treated with ammonium acetate at 3 and 24 h post-injection. Despite the normalization of the animal’s ammonia levels, samples taken at 24 h after injection exhibited distinct enlargement of lateral ventricles. The choroid plexus samples obtained at 3 h post-ammonium acetate treatment indicated enlargement; however, this swelling was reduced at the later timepoint. The aquaporin-1 proteins that regulate the choroid plexus were localized both in the apical membrane and the cytoplasm of the epithelia in the control; however, they translocated to the apical membranes of the epithelia in response to ammonia treatment. Therefore, severe acute hepatic encephalopathy induced by ammonium acetate administration caused enlargement of the ventricles, through swelling of the choroid plexus and aquaporin-1 transport and aggregation within the apical membranes.
Collapse
|
41
|
Abstract
In the field of neuropsychiatry, neuroinflammation is one of the prevailing hypotheses to explain the pathophysiology of mood and psychotic disorders. Neuroinflammation encompasses an ill-defined set of pathophysiological processes in the central nervous system that cause neuronal or glial atrophy or death and disruptions in neurotransmitter signaling, resulting in cognitive and behavioral changes. Positron emission tomography for the brain-based translocator protein has been shown to be a useful tool to measure glial activation in neuropsychiatric disorders. Recent neuroimaging studies also indicate a potential disruption in the choroid plexus and blood-brain barrier, which modulate the transfer of ions, molecules, toxins, and cells from the periphery into the brain. Simultaneously, peripheral inflammatory markers have consistently been shown to be altered in mood and psychotic disorders. The crosstalk (i.e., the communication between peripheral and central inflammatory pathways) is not well understood in these disorders, however, and neuroimaging studies hold promise to shed light on this complex process. In the current Perspectives article, we discuss the neuroimaging insights into neuroimmune crosstalk offered in selected works. Overall, evidence exists for peripheral immune cell infiltration into the central nervous system in some patients, but the reason for this is unknown. Future neuroimaging studies should aim to extend our knowledge of this system and the role it likely plays in symptom onset and recurrence.
Collapse
|
42
|
Otun A, Morales DM, Garcia-Bonilla M, Goldberg S, Castaneyra-Ruiz L, Yan Y, Isaacs AM, Strahle JM, McAllister JP, Limbrick DD. Biochemical profile of human infant cerebrospinal fluid in intraventricular hemorrhage and post-hemorrhagic hydrocephalus of prematurity. Fluids Barriers CNS 2021; 18:62. [PMID: 34952604 PMCID: PMC8710025 DOI: 10.1186/s12987-021-00295-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/06/2021] [Indexed: 12/05/2022] Open
Abstract
Background Intraventricular hemorrhage (IVH) and post-hemorrhagic hydrocephalus (PHH) have a complex pathophysiology involving inflammatory response, ventricular zone and cell–cell junction disruption, and choroid-plexus (ChP) hypersecretion. Increased cerebrospinal fluid (CSF) cytokines, extracellular matrix proteins, and blood metabolites have been noted in IVH/PHH, but osmolality and electrolyte disturbances have not been evaluated in human infants with these conditions. We hypothesized that CSF total protein, osmolality, electrolytes, and immune cells increase in PHH. Methods CSF samples were obtained from lumbar punctures of control infants and infants with IVH prior to the development of PHH and any neurosurgical intervention. Osmolality, total protein, and electrolytes were measured in 52 infants (18 controls, 10 low grade (LG) IVH, 13 high grade (HG) IVH, and 11 PHH). Serum electrolyte concentrations, and CSF and serum cell counts within 1-day of clinical sampling were obtained from clinical charts. Frontal occipital horn ratio (FOR) was measured for estimating the degree of ventriculomegaly. Dunn or Tukey’s post-test ANOVA analysis were used for pair-wise comparisons. Results CSF osmolality, sodium, potassium, and chloride were elevated in PHH compared to control (p = 0.012 − < 0.0001), LGIVH (p = 0.023 − < 0.0001), and HGIVH (p = 0.015 − 0.0003), while magnesium and calcium levels were higher compared to control (p = 0.031) and LGIVH (p = 0.041). CSF total protein was higher in both HGIVH and PHH compared to control (p = 0.0009 and 0.0006 respectively) and LGIVH (p = 0.034 and 0.028 respectively). These differences were not reflected in serum electrolyte concentrations nor calculated osmolality across the groups. However, quantitatively, CSF sodium and chloride contributed 86% of CSF osmolality change between control and PHH; and CSF osmolality positively correlated with CSF sodium (r, p = 0.55,0.0015), potassium (r, p = 0.51,0.0041), chloride (r, p = 0.60,0.0004), but not total protein across the entire patient cohort. CSF total cells (p = 0.012), total nucleated cells (p = 0.0005), and percent monocyte (p = 0.016) were elevated in PHH compared to control. Serum white blood cell count increased in PHH compared to control (p = 0.042) but there were no differences in serum cell differential across groups. CSF total nucleated cells also positively correlated with CSF osmolality, sodium, potassium, and total protein (p = 0.025 − 0.0008) in the whole cohort. Conclusions CSF osmolality increased in PHH, largely driven by electrolyte changes rather than protein levels. However, serum electrolytes levels were unchanged across groups. CSF osmolality and electrolyte changes were correlated with CSF total nucleated cells which were also increased in PHH, further suggesting PHH is a neuro-inflammatory condition. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00295-8.
Collapse
Affiliation(s)
- Ayodamola Otun
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA.
| | - Diego M Morales
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Maria Garcia-Bonilla
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Seth Goldberg
- Department of Nephrology, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | | | - Yan Yan
- Department of Surgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - Albert M Isaacs
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Calgary, Calgary, AB, T2N 2T9, Canada
| | - Jennifer M Strahle
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - James P McAllister
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| | - David D Limbrick
- Department of Neurosurgery, Washington University in St. Louis School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
43
|
Abstract
Reparative inflammation is an important protective response that eliminates foreign organisms, damaged cells, and physical irritants. However, inappropriately triggered or sustained inflammation can respectively initiate, propagate, or prolong disease. Post-hemorrhagic (PHH) and post-infectious hydrocephalus (PIH) are the most common forms of hydrocephalus worldwide. They are treated using neurosurgical cerebrospinal fluid (CSF) diversion techniques with high complication and failure rates. Despite their distinct etiologies, clinical studies in human patients have shown PHH and PIH share similar CSF cytokine and immune cell profiles. Here, in light of recent work in model systems, we discuss the concept of "inflammatory hydrocephalus" to emphasize potential shared mechanisms and potential therapeutic vulnerabilities of these disorders. We propose that this change of emphasis could shift our thinking of PHH and PIH from a framework of life-long neurosurgical disorders to that of preventable conditions amenable to immunomodulation.
Collapse
|
44
|
Cui J, Xu H, Lehtinen MK. Macrophages on the margin: choroid plexus immune responses. Trends Neurosci 2021; 44:864-875. [PMID: 34312005 PMCID: PMC8551004 DOI: 10.1016/j.tins.2021.07.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022]
Abstract
The choroid plexus (ChP), an epithelial bilayer containing a network of mesenchymal, immune, and neuronal cells, forms the blood-cerebrospinal fluid (CSF) barrier (BCSFB). While best recognized for secreting CSF, the ChP is also a hotbed of immune cell activity and can provide circulating peripheral immune cells with passage into the central nervous system (CNS). Here, we review recent studies on ChP immune cells, with a focus on the ontogeny, development, and behaviors of ChP macrophages, the principal resident immune cells of the ChP. We highlight the implications of immune cells for ChP barrier function, CSF cytokines and volume regulation, and their contribution to neurodevelopmental disorders, with possible age-specific features to be elucidated in the future.
Collapse
Affiliation(s)
- Jin Cui
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Huixin Xu
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
45
|
Francis Stuart SD, Villalobos AR. GSH and Zinc Supplementation Attenuate Cadmium-Induced Cellular Stress and Stimulation of Choline Uptake in Cultured Neonatal Rat Choroid Plexus Epithelia. Int J Mol Sci 2021; 22:ijms22168857. [PMID: 34445563 PMCID: PMC8396310 DOI: 10.3390/ijms22168857] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/01/2021] [Accepted: 08/11/2021] [Indexed: 12/27/2022] Open
Abstract
Choroid plexus (CP) sequesters cadmium and other metals, protecting the brain from these neurotoxins. These metals can induce cellular stress and modulate homeostatic functions of CP, such as solute transport. We previously showed in primary cultured neonatal rat CP epithelial cells (CPECs) that cadmium induced cellular stress and stimulated choline uptake at the apical membrane, which interfaces with cerebrospinal fluid in situ. Here, in CPECs, we characterized the roles of glutathione (GSH) and Zinc supplementation in the adaptive stress response to cadmium. Cadmium increased GSH and decreased the reduced GSH-to-oxidized GSH (GSSG) ratio. Heat shock protein-70 (Hsp70), heme oxygenase (HO-1), and metallothionein (Mt-1) were induced along with the catalytic and modifier subunits of glutamate cysteine ligase (GCL), the rate-limiting enzyme in GSH synthesis. Inhibition of GCL by l-buthionine sulfoximine (BSO) enhanced stress protein induction and stimulation of choline uptake by cadmium. Zinc alone did not induce Hsp70, HO-1, or GCL subunits, or modulate choline uptake. Zinc supplementation during cadmium exposure attenuated stress protein induction and stimulation of choline uptake; this effect persisted despite inhibition of GSH synthesis. These data indicated up-regulation of GSH synthesis promotes adaptation to cadmium-induced cellular stress in CP, but Zinc may confer cytoprotection independent of GSH.
Collapse
Affiliation(s)
- Samantha D. Francis Stuart
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA;
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Alice R. Villalobos
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA;
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
- Correspondence: ; Tel.: +1-806-743-2057
| |
Collapse
|
46
|
Quintela T, Furtado A, Duarte AC, Gonçalves I, Myung J, Santos CRA. The role of circadian rhythm in choroid plexus functions. Prog Neurobiol 2021; 205:102129. [PMID: 34343629 DOI: 10.1016/j.pneurobio.2021.102129] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/17/2022]
Abstract
For several years, a great effort has been devoted to understand how circadian oscillations in physiological processes are determined by the circadian clock system. This system is composed by the master clock at the suprachiasmatic nucleus which sets the pace and tunes peripheral clocks in several organs. It was recently demonstrated that the choroid plexus epithelial cells that compose the blood-cerebrospinal fluid barrier hold a circadian clock which might control their multiple functions with implications for the maintenance of brain homeostasis. However, the choroid plexus activities regulated by its inner clock are still largely unknown. In this review, we propose that several choroid plexus functions might be regulated by the circadian clock, alike in other tissues. We provide evidences that the timing of cerebrospinal fluid secretion, clearance of amyloid-beta peptides and xenobiotics, and the barrier function of the blood-cerebrospinal fluid barrier are regulated by the circadian clock. These data, highlight that the circadian regulation of the blood-cerebrospinal fluid barrier must be taken into consideration for enhancing drug delivery to central nervous system disorders.
Collapse
Affiliation(s)
- Telma Quintela
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - André Furtado
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, No. 172-1 Sec. 2 Keelung Road, Da'an District, Taipei 106, Taiwan; Brain and Consciousness Research Centre, Shuang Ho Hospital, Ministry of Health and Welfare, No. 291 Zhongzheng Road, Zhonghe District, New Taipei City 235, Taiwan
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
47
|
Abstract
Our brains consist of 80% water, which is continuously shifted between different compartments and cell types during physiological and pathophysiological processes. Disturbances in brain water homeostasis occur with pathologies such as brain oedema and hydrocephalus, in which fluid accumulation leads to elevated intracranial pressure. Targeted pharmacological treatments do not exist for these conditions owing to our incomplete understanding of the molecular mechanisms governing brain water transport. Historically, the transmembrane movement of brain water was assumed to occur as passive movement of water along the osmotic gradient, greatly accelerated by water channels termed aquaporins. Although aquaporins govern the majority of fluid handling in the kidney, they do not suffice to explain the overall brain water movement: either they are not present in the membranes across which water flows or they appear not to be required for the observed flow of water. Notably, brain fluid can be secreted against an osmotic gradient, suggesting that conventional osmotic water flow may not describe all transmembrane fluid transport in the brain. The cotransport of water is an unconventional molecular mechanism that is introduced in this Review as a missing link to bridge the gap in our understanding of cellular and barrier brain water transport.
Collapse
Affiliation(s)
- Nanna MacAulay
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
48
|
Tobiansky DJ, Long KM, Hamden JE, Brawn JD, Fuxjager MJ. Cost-reducing traits for agonistic head collisions: a case for neurophysiology. Integr Comp Biol 2021; 61:1394-1405. [PMID: 33885750 DOI: 10.1093/icb/icab034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Many animal species have evolved extreme behaviors requiring them to engage in repeated high-impact collisions. These behaviors include mating displays like headbutting in sheep and drumming in woodpeckers. To our knowledge, these taxa do not experience any notable acute head trauma, even though the deceleration forces would cause traumatic brain injury in most animals. Previous research has focused on skeletomuscular morphology, biomechanics, and material properties in an attempt to explain how animals moderate these high-impact forces. However, many of these behaviors are understudied, and most morphological or computational studies make assumptions about the behavior without accounting for the physiology of an organism. Studying neurophysiological and immune adaptations that co-vary with these behaviors can highlight unique or synergistic solutions to seemingly deleterious behavioral displays. Here, we argue that selection for repeated, high-impact head collisions may rely on a suite of coadaptations in intracranial physiology as a cost-reducing mechanism. We propose that there are three physiological systems that could mitigate the effects of repeated head trauma: (i) the innate neuroimmune response, (ii) the glymphatic system, and (iii) the choroid plexus. These systems are interconnected yet can evolve in an independent manner. We then briefly describe the function of these systems, their role in head trauma, and research that has examined how these systems may evolve to help reduce the cost of repeated, forceful head impacts. Ultimately, we note that little is known about cost-reducing intracranial mechanisms making it a novel field of comparative study that is ripe for exploration.
Collapse
Affiliation(s)
| | - Kira M Long
- The University of Illinois at Urbana-Champaign, Urbana-Champaign, IL USAKML
| | | | - Jeffrey D Brawn
- The University of Illinois at Urbana-Champaign, Urbana-Champaign, IL USAJDB
| | | |
Collapse
|
49
|
Costa-Brito AR, Quintela T, Gonçalves I, Duarte AC, Costa AR, Arosa FA, Cavaco JE, Lemos MC, Santos CRA. The Choroid Plexus Is an Alternative Source of Prolactin to the Rat Brain. Mol Neurobiol 2021; 58:1846-1858. [PMID: 33409838 DOI: 10.1007/s12035-020-02267-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/16/2020] [Indexed: 10/22/2022]
Abstract
Among the more than 300 functions attributed to prolactin (PRL), this hormone has been associated with the induction of neurogenesis and differentiation of olfactory neurons especially during pregnancy, which are essential for maternal behavior. Despite the original hypothesis that PRL enters the central nervous system through a process mediated by PRL receptors (PRLR) at the choroid plexus (CP), recent data suggested that PRL transport into the brain is independent of its receptors. Based on transcriptomic data suggesting that PRL could be expressed in the CP, this work aimed to confirm PRL synthesis and secretion by CP epithelial cells (CPEC). The secretion of PRL and the distribution of PRLR in CPEC were further characterized using an in vitro model of the rat blood-cerebrospinal fluid barrier. RT-PCR analysis of PRL transcripts showed its presence in pregnant rat CP, in CPEC, and in the rat immortalized CP cell line, Z310. These observations were reinforced by immunocytochemistry staining of PRL in CPEC and Z310 cell cytoplasm. A 63-kDa immunoreactive PRL protein was detected by Western blot in CP protein extracts as well as in culture medium incubated with rat pituitary and samples of rat cerebrospinal fluid and serum. Positive immunocytochemistry staining of PRLR was present throughout the CPEC cytoplasm and in the apical and basal membrane of these cells. Altogether, our evidences suggest that CP is an alternative source of PRL to the brain, which might impact neurogenesis of olfactory neurons at the subventricular zone, given its proximity to the CP.
Collapse
Affiliation(s)
- Ana R Costa-Brito
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ana R Costa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Fernando A Arosa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - José E Cavaco
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Manuel C Lemos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cecília R A Santos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| |
Collapse
|
50
|
Bryniarski MA, Ren T, Rizvi AR, Snyder AM, Morris ME. Targeting the Choroid Plexuses for Protein Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12100963. [PMID: 33066423 PMCID: PMC7602164 DOI: 10.3390/pharmaceutics12100963] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/05/2020] [Accepted: 10/10/2020] [Indexed: 12/15/2022] Open
Abstract
Delivery of therapeutic agents to the central nervous system is challenged by the barriers in place to regulate brain homeostasis. This is especially true for protein therapeutics. Targeting the barrier formed by the choroid plexuses at the interfaces of the systemic circulation and ventricular system may be a surrogate brain delivery strategy to circumvent the blood-brain barrier. Heterogenous cell populations located at the choroid plexuses provide diverse functions in regulating the exchange of material within the ventricular space. Receptor-mediated transcytosis may be a promising mechanism to deliver protein therapeutics across the tight junctions formed by choroid plexus epithelial cells. However, cerebrospinal fluid flow and other barriers formed by ependymal cells and perivascular spaces should also be considered for evaluation of protein therapeutic disposition. Various preclinical methods have been applied to delineate protein transport across the choroid plexuses, including imaging strategies, ventriculocisternal perfusions, and primary choroid plexus epithelial cell models. When used in combination with simultaneous measures of cerebrospinal fluid dynamics, they can yield important insight into pharmacokinetic properties within the brain. This review aims to provide an overview of the choroid plexuses and ventricular system to address their function as a barrier to pharmaceutical interventions and relevance for central nervous system drug delivery of protein therapeutics. Protein therapeutics targeting the ventricular system may provide new approaches in treating central nervous system diseases.
Collapse
|