1
|
Rajan A, Fame RM. Brain development and bioenergetic changes. Neurobiol Dis 2024; 199:106550. [PMID: 38849103 PMCID: PMC11495523 DOI: 10.1016/j.nbd.2024.106550] [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] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024] Open
Abstract
Bioenergetics describe the biochemical processes responsible for energy supply in organisms. When these changes become dysregulated in brain development, multiple neurodevelopmental diseases can occur, implicating bioenergetics as key regulators of neural development. Historically, the discovery of disease processes affecting individual stages of brain development has revealed critical roles that bioenergetics play in generating the nervous system. Bioenergetic-dependent neurodevelopmental disorders include neural tube closure defects, microcephaly, intellectual disability, autism spectrum disorders, epilepsy, mTORopathies, and oncogenic processes. Developmental timing and cell-type specificity of these changes determine the long-term effects of bioenergetic disease mechanisms on brain form and function. Here, we discuss key metabolic regulators of neural progenitor specification, neuronal differentiation (neurogenesis), and gliogenesis. In general, transitions between glycolysis and oxidative phosphorylation are regulated in early brain development and in oncogenesis, and reactive oxygen species (ROS) and mitochondrial maturity play key roles later in differentiation. We also discuss how bioenergetics interface with the developmental regulation of other key neural elements, including the cerebrospinal fluid brain environment. While questions remain about the interplay between bioenergetics and brain development, this review integrates the current state of known key intersections between these processes in health and disease.
Collapse
Affiliation(s)
- Arjun Rajan
- Developmental Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ryann M Fame
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
2
|
Pardridge WM. Receptor-mediated drug delivery of bispecific therapeutic antibodies through the blood-brain barrier. FRONTIERS IN DRUG DELIVERY 2023; 3:1227816. [PMID: 37583474 PMCID: PMC10426772 DOI: 10.3389/fddev.2023.1227816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Therapeutic antibody drug development is a rapidly growing sector of the pharmaceutical industry. However, antibody drug development for the brain is a technical challenge, and therapeutic antibodies for the central nervous system account for ~3% of all such agents. The principal obstacle to antibody drug development for brain or spinal cord is the lack of transport of large molecule biologics across the blood-brain barrier (BBB). Therapeutic antibodies can be made transportable through the blood-brain barrier by the re-engineering of the therapeutic antibody as a BBB-penetrating bispecific antibody (BSA). One arm of the BSA is the therapeutic antibody and the other arm of the BSA is a transporting antibody. The transporting antibody targets an exofacial epitope on a BBB receptor, and this enables receptor-mediated transcytosis (RMT) of the BSA across the BBB. Following BBB transport, the therapeutic antibody then engages the target receptor in brain. RMT systems at the BBB that are potential conduits to the brain include the insulin receptor (IR), the transferrin receptor (TfR), the insulin-like growth factor receptor (IGFR) and the leptin receptor. Therapeutic antibodies have been re-engineered as BSAs that target the insulin receptor, TfR, or IGFR RMT systems at the BBB for the treatment of Alzheimer's disease and Parkinson's disease.
Collapse
|
3
|
Korzh V. Development of the brain ventricular system from a comparative perspective. Clin Anat 2023; 36:320-334. [PMID: 36529666 DOI: 10.1002/ca.23994] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
The brain ventricular system (BVS) consists of brain ventricles and channels filled with cerebrospinal fluid (CSF). Disturbance of CSF flow has been linked to scoliosis and neurodegenerative diseases, including hydrocephalus. This could be due to defects of CSF production by the choroid plexus or impaired CSF movement over the ependyma dependent on motile cilia. Most vertebrates have horizontal body posture. They retain additional evolutionary innovations assisting CSF flow, such as the Reissner fiber. The causes of hydrocephalus have been studied using animal models including rodents (mice, rats, hamsters) and zebrafish. However, the horizontal body posture reduces the effect of gravity on CSF flow, which limits the use of mammalian models for scoliosis. In contrast, fish swim against the current and experience a forward-to-backward mechanical force akin to that caused by gravity in humans. This explains the increased popularity of the zebrafish model for studies of scoliosis. "Slit-ventricle" syndrome is another side of the spectrum of BVS anomalies. It develops because of insufficient inflation of the BVS. Recent advances in zebrafish functional genetics have revealed genes that could regulate the development of the BVS and CSF circulation. This review will describe the BVS of zebrafish, a typical teleost, and vertebrates in general, in comparative perspective. It will illustrate the usefulness of the zebrafish model for developmental studies of the choroid plexus (CP), CSF flow and the BVS.
Collapse
Affiliation(s)
- Vladimir Korzh
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
4
|
Saunders NR, Dziegielewska KM, Fame RM, Lehtinen MK, Liddelow SA. The choroid plexus: a missing link in our understanding of brain development and function. Physiol Rev 2023; 103:919-956. [PMID: 36173801 PMCID: PMC9678431 DOI: 10.1152/physrev.00060.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 09/01/2022] [Accepted: 09/17/2022] [Indexed: 11/22/2022] Open
Abstract
Studies of the choroid plexus lag behind those of the more widely known blood-brain barrier, despite a much longer history. This review has two overall aims. The first is to outline long-standing areas of research where there are unanswered questions, such as control of cerebrospinal fluid (CSF) secretion and blood flow. The second aim is to review research over the past 10 years where the focus has shifted to the idea that there are choroid plexuses located in each of the brain's ventricles that make specific contributions to brain development and function through molecules they generate for delivery via the CSF. These factors appear to be particularly important for aspects of normal brain growth. Most research carried out during the twentieth century dealt with the choroid plexus, a brain barrier interface making critical contributions to the composition and stability of the brain's internal environment throughout life. More recent research in the twenty-first century has shown the importance of choroid plexus-generated CSF in neurogenesis, influence of sex and other hormones on choroid plexus function, and choroid plexus involvement in circadian rhythms and sleep. The advancement of technologies to facilitate delivery of brain-specific therapies via the CSF to treat neurological disorders is a rapidly growing area of research. Conversely, understanding the basic mechanisms and implications of how maternal drug exposure during pregnancy impacts the developing brain represents another key area of research.
Collapse
Affiliation(s)
- Norman R Saunders
- Department of Neuroscience, The Alfred Centre, Monash University, Melbourne, Victoria, Australia
| | | | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, New York
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, New York
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, New York
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, New York
| |
Collapse
|
5
|
Pardridge WM. Blood-brain barrier delivery for lysosomal storage disorders with IgG-lysosomal enzyme fusion proteins. Adv Drug Deliv Rev 2022; 184:114234. [PMID: 35307484 DOI: 10.1016/j.addr.2022.114234] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/25/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
The majority of lysosomal storage diseases affect the brain. Treatment of the brain with intravenous enzyme replacement therapy is not successful, because the recombinant lysosomal enzymes do not cross the blood-brain barrier (BBB). Biologic drugs, including lysosomal enzymes, can be re-engineered for BBB delivery as IgG-enzyme fusion proteins. The IgG domain of the fusion protein is a monoclonal antibody directed against an endogenous receptor-mediated transporter at the BBB, such as the insulin receptor or the transferrin receptor. This receptor transports the IgG across the BBB, in parallel with the endogenous receptor ligand, and the IgG acts as a molecular Trojan horse to ferry into brain the lysosomal enzyme genetically fused to the IgG. The IgG-enzyme fusion protein is bi-functional and retains both high affinity binding for the BBB receptor, and high lysosomal enzyme activity. IgG-lysosomal enzymes are presently in clinical trials for treatment of the brain in Mucopolysaccharidosis.
Collapse
|
6
|
Speidel A, Theile M, Pfeiffer L, Herrmann A, Figarella K, Ishikawa H, Schwerk C, Schroten H, Duszenko M, Mogk S. Transmigration of Trypanosoma brucei across an in vitro blood-cerebrospinal fluid barrier. iScience 2022; 25:104014. [PMID: 35313698 PMCID: PMC8933718 DOI: 10.1016/j.isci.2022.104014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/24/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022] Open
Abstract
Trypanosoma brucei is the causative agent of human African trypanosomiasis. The parasite transmigrates from blood vessels across the choroid plexus epithelium to enter the central nervous system, a process that leads to the manifestation of second stage sleeping sickness. Using an in vitro model of the blood-cerebrospinal fluid barrier, we investigated the mechanism of the transmigration process. For this, a monolayer of human choroid plexus papilloma cells was cultivated on a permeable membrane that mimics the basal lamina underlying the choroid plexus epithelial cells. Plexus cells polarize and interconnect forming tight junctions. Deploying different T. brucei brucei strains, we observed that geometry and motility are important for tissue invasion. Using fluorescent microscopy, the parasite's moving was visualized between plexus epithelial cells. The presented model provides a simple tool to screen trypanosome libraries for their ability to infect cerebrospinal fluid or to test the impact of chemical substances on transmigration.
Collapse
Affiliation(s)
- Annika Speidel
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Marianne Theile
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Lena Pfeiffer
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Alexander Herrmann
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | | | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Christian Schwerk
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Horst Schroten
- Pediatric Infectious Diseases, Department of Pediatrics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Duszenko
- Department of Neurophysiology, University of Tübingen, Tübingen, Germany
| | - Stefan Mogk
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Constitutive activation of canonical Wnt signaling disrupts choroid plexus epithelial fate. Nat Commun 2022; 13:633. [PMID: 35110543 PMCID: PMC8810795 DOI: 10.1038/s41467-021-27602-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 11/30/2021] [Indexed: 12/30/2022] Open
Abstract
The choroid plexus secretes cerebrospinal fluid and is critical for the development and function of the brain. In the telencephalon, the choroid plexus epithelium arises from the Wnt- expressing cortical hem. Canonical Wnt signaling pathway molecules such as nuclear β-CATENIN are expressed in the mouse and human embryonic choroid plexus epithelium indicating that this pathway is active. Point mutations in human β-CATENIN are known to result in the constitutive activation of canonical Wnt signaling. In a mouse model that recapitulates this perturbation, we report a loss of choroid plexus epithelial identity and an apparent transformation of this tissue to a neuronal identity. Aspects of this phenomenon are recapitulated in human embryonic stem cell derived organoids. The choroid plexus is also disrupted when β-Catenin is conditionally inactivated. Together, our results indicate that canonical Wnt signaling is required in a precise and regulated manner for normal choroid plexus development in the mammalian brain.
Collapse
|
8
|
Pardridge WM. Kinetics of Blood-Brain Barrier Transport of Monoclonal Antibodies Targeting the Insulin Receptor and the Transferrin Receptor. Pharmaceuticals (Basel) 2021; 15:3. [PMID: 35056060 PMCID: PMC8778919 DOI: 10.3390/ph15010003] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/10/2021] [Accepted: 12/18/2021] [Indexed: 12/21/2022] Open
Abstract
Biologic drugs are large molecule pharmaceuticals that do not cross the blood-brain barrier (BBB), which is formed by the brain capillary endothelium. Biologics can be re-engineered for BBB transport as IgG fusion proteins, where the IgG domain is a monoclonal antibody (MAb) that targets an endogenous BBB transporter, such as the insulin receptor (IR) or transferrin receptor (TfR). The IR and TfR at the BBB transport the receptor-specific MAb in parallel with the transport of the endogenous ligand, insulin or transferrin. The kinetics of BBB transport of insulin or transferrin, or an IRMAb or TfRMAb, can be quantified with separate mathematical models. Mathematical models to estimate the half-time of receptor endocytosis, MAb or ligand exocytosis into brain extracellular space, or receptor recycling back to the endothelial luminal membrane were fit to the brain uptake of a TfRMAb or a IRMAb fusion protein in the Rhesus monkey. Model fits to the data also allow for estimates of the rates of association of the MAb in plasma with the IR or TfR that is embedded within the endothelial luminal membrane in vivo. The parameters generated from the model fits can be used to estimate the brain concentration profile of the MAb over time, and this brain exposure is shown to be a function of the rate of clearance of the antibody fusion protein from the plasma compartment.
Collapse
|
9
|
Alisch JSR, Kiely M, Triebswetter C, Alsameen MH, Gong Z, Khattar N, Egan JM, Bouhrara M. Characterization of Age-Related Differences in the Human Choroid Plexus Volume, Microstructural Integrity, and Blood Perfusion Using Multiparameter Magnetic Resonance Imaging. Front Aging Neurosci 2021; 13:734992. [PMID: 34603011 PMCID: PMC8485051 DOI: 10.3389/fnagi.2021.734992] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
The choroid plexus (CP) is an important cerebral structure involved in cerebrospinal fluid production and transport of solutes into the brain. Recent studies have uncovered the involvement of the CP in neurological disorders such as Alzheimer's disease and multiple sclerosis. However, our understanding of human age-related microstructural and functional changes in the CP with aging and neuropathology is limited. In this cross-sectional study, we investigated age and sex differences in the CP structure and function using advanced quantitative magnetic resonance imaging methodology in a large cohort (n = 155) of cognitively unimpaired individuals over a wide age range between 21 and 94 years. Our analysis included volumetric measurements, relaxometry measures (T 1 and T 2), diffusion tensor imaging (DTI) measures of fractional anisotropy (FA) and mean diffusivity (MD), as well as measures of cerebral blood flow (CBF). Our results revealed that CP volume was increasing with advancing age. We conjecture that this novel observation is likely attributed to alterations in the CP microstructure or function as well as to ventriculomegaly. Indeed, we also found that CBF was lower with advanced age, while, consistent with previous studies, T 1, T 2 and MD were higher, and FA was lower with advanced age. We attribute these functional and microstructural differences to a deteriorated CP structural integrity with aging. Furthermore, our relaxometry and DTI measures were found to be associated with differences in blood perfusion revealing lower microstructural integrity with lower CBF. Finally, in agreement with literature, sex-related differences in MD and CBF were statistically significant. This work lays the foundation for ongoing investigation of the involvement of CP in neurodegeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mustapha Bouhrara
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
10
|
Pardridge WM, Chou T. Mathematical Models of Blood-Brain Barrier Transport of Monoclonal Antibodies Targeting the Transferrin Receptor and the Insulin Receptor. Pharmaceuticals (Basel) 2021; 14:535. [PMID: 34205013 PMCID: PMC8226686 DOI: 10.3390/ph14060535] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
We develop and analyze mathematical models for receptor-mediated transcytosis of monoclonal antibodies (MAb) targeting the transferrin receptor (TfR) or the insulin receptor (IR), which are expressed at the blood-brain barrier (BBB). The mass-action kinetic model for both the TfR and IR antibodies were solved numerically to generate predictions for the concentrations of all species in all compartments considered. Using these models, we estimated the rates of MAb endocytosis into brain capillary endothelium, which forms the BBB in vivo, the rates of MAb exocytosis from the intra-endothelial compartment into brain extracellular space, and the rates of receptor recycling from the endothelial space back to the luminal endothelial plasma membrane. Our analysis highlights the optimal rates of MAb association with the targeted receptor. An important role of the endogenous ligand, transferrin (Tf) or insulin, in receptor-mediated-transport (RMT) of the associated MAb was found and was attributed to the five order magnitude difference between plasma concentrations of Tf (25,000 nM) and insulin (0.3 nM). Our modeling shows that the very high plasma concentration of Tf leads to only 5% of the endothelial TfR expressed on the luminal endothelial membrane.
Collapse
Affiliation(s)
| | - Tom Chou
- Departments of Computational Medicine and Mathematics, UCLA, Los Angeles, CA 90095, USA;
| |
Collapse
|
11
|
Kaiser K, Jang A, Kompanikova P, Lun MP, Prochazka J, Machon O, Dani N, Prochazkova M, Laurent B, Gyllborg D, van Amerongen R, Fame RM, Gupta S, Wu F, Barker RA, Bukova I, Sedlacek R, Kozmik Z, Arenas E, Lehtinen MK, Bryja V. MEIS-WNT5A axis regulates development of fourth ventricle choroid plexus. Development 2021; 148:268365. [PMID: 34032267 DOI: 10.1242/dev.192054] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 04/14/2021] [Indexed: 12/29/2022]
Abstract
The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.
Collapse
Affiliation(s)
- Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 61137, Czech Republic
| | - Ahram Jang
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Petra Kompanikova
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 61137, Czech Republic
| | - Melody P Lun
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jan Prochazka
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Ondrej Machon
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Neil Dani
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Michaela Prochazkova
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Benoit Laurent
- Research Center on Aging, CIUSSS de l'Estrie - CHUS, Sherbrooke, QC 75361, Canada.,Department of Biochemistry and Functional Genomics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC 75281, Canada
| | - Daniel Gyllborg
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna SE-106 91, Sweden
| | - Renee van Amerongen
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, Faculty of Science, University of Amsterdam1098 XH, Netherlands
| | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Suhasini Gupta
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Feizhen Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Roger A Barker
- John van Geest Centre for Brain Repair and WT-MRC Cambridge Stem Cell Centre, University of Cambridge, Cambridge CB2 0PY, UK
| | - Ivana Bukova
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Zbynek Kozmik
- Laboratory of Transcriptional Regulation, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Ernest Arenas
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 61137, Czech Republic
| |
Collapse
|
12
|
Moore SA, Iulianella A. Development of the mammalian cortical hem and its derivatives: the choroid plexus, Cajal-Retzius cells and hippocampus. Open Biol 2021; 11:210042. [PMID: 33947245 PMCID: PMC8097212 DOI: 10.1098/rsob.210042] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/08/2021] [Indexed: 12/14/2022] Open
Abstract
The dorsal medial region of the developing mammalian telencephalon plays a central role in the patterning of the adjacent brain regions. This review describes the development of this specialized region of the vertebrate brain, called the cortical hem, and the formation of the various cells and structures it gives rise to, including the choroid plexus, Cajal-Retzius cells and the hippocampus. We highlight the ontogenic processes that create these different forebrain derivatives from their shared embryonic origin and discuss the key signalling pathways and molecules that influence the patterning of the cortical hem. These include BMP, Wnt, FGF and Shh signalling pathways acting with Homeobox factors to carve the medial telencephalon into district progenitor regions, which in turn give rise to the choroid plexus, dentate gyrus and hippocampus. We then link the formation of the lateral ventricle choroid plexus with embryonic and postnatal neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Samantha A. Moore
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia, Canada, B3H4R2
| | - Angelo Iulianella
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia, Canada, B3H4R2
| |
Collapse
|
13
|
Dani N, Herbst RH, McCabe C, Green GS, Kaiser K, Head JP, Cui J, Shipley FB, Jang A, Dionne D, Nguyen L, Rodman C, Riesenfeld SJ, Prochazka J, Prochazkova M, Sedlacek R, Zhang F, Bryja V, Rozenblatt-Rosen O, Habib N, Regev A, Lehtinen MK. A cellular and spatial map of the choroid plexus across brain ventricles and ages. Cell 2021; 184:3056-3074.e21. [PMID: 33932339 DOI: 10.1016/j.cell.2021.04.003] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 12/15/2020] [Accepted: 04/02/2021] [Indexed: 02/06/2023]
Abstract
The choroid plexus (ChP) in each brain ventricle produces cerebrospinal fluid (CSF) and forms the blood-CSF barrier. Here, we construct a single-cell and spatial atlas of each ChP in the developing, adult, and aged mouse brain. We delineate diverse cell types, subtypes, cell states, and expression programs in epithelial and mesenchymal cells across ages and ventricles. In the developing ChP, we predict a common progenitor pool for epithelial and neuronal cells, validated by lineage tracing. Epithelial and fibroblast cells show regionalized expression by ventricle, starting at embryonic stages and persisting with age, with a dramatic transcriptional shift with maturation, and a smaller shift in each aged cell type. With aging, epithelial cells upregulate host-defense programs, and resident macrophages upregulate interleukin-1β (IL-1β) signaling genes. Our atlas reveals cellular diversity, architecture and signaling across ventricles during development, maturation, and aging of the ChP-brain barrier.
Collapse
Affiliation(s)
- Neil Dani
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Rebecca H Herbst
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Cristin McCabe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gilad S Green
- Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 611 37, Czech Republic
| | - Joshua P Head
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jin Cui
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Frederick B Shipley
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02115, USA
| | - Ahram Jang
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lan Nguyen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christopher Rodman
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Samantha J Riesenfeld
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jan Prochazka
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Michaela Prochazkova
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, Prague 142 20, Czech Republic
| | - Feng Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 611 37, Czech Republic
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Naomi Habib
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Jerusalem 9190401, Israel.
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02115, USA.
| |
Collapse
|
14
|
Fame RM, Lehtinen MK. Emergence and Developmental Roles of the Cerebrospinal Fluid System. Dev Cell 2020; 52:261-275. [PMID: 32049038 DOI: 10.1016/j.devcel.2020.01.027] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/14/2020] [Accepted: 01/24/2020] [Indexed: 12/21/2022]
Abstract
We summarize recent work illuminating how cerebrospinal fluid (CSF) regulates brain function. More than a protective fluid cushion and sink for waste, the CSF is an integral CNS component with dynamic and diverse roles emerging in parallel with the developing CNS. This review examines the current understanding about early CSF and its maturation and roles during CNS development and discusses open questions in the field. We focus on developmental changes in the ventricular system and CSF sources (including neural progenitors and choroid plexus). We also discuss concepts related to the development of fluid dynamics including flow, perivascular transport, drainage, and barriers.
Collapse
Affiliation(s)
- Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
15
|
Kratzer I, Ek J, Stolp H. The molecular anatomy and functions of the choroid plexus in healthy and diseased brain. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183430. [PMID: 32750317 DOI: 10.1016/j.bbamem.2020.183430] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 01/16/2023]
Abstract
The choroid plexus (CP) is located in the ventricular system of the brain (one in each ventricle), and the CP epithelial cells form an important barrier between the blood and the cerebrospinal fluid (CSF). Their main function comprises CSF secretion, maintenance of brain homeostasis, signalling, and forming a neuroprotective barrier against harmful external and internal compounds. The CPs mature early and demonstrate expressional changes of barrier-specific genes and proteins related to location and developmental stage of the CP. Important proteins for the barrier function include tight junction proteins, numerous transporters and enzymes. Natural senescence leads to structural changes in the CP cells and reduced or loss of function, while further loss of CP function and changes in immune status may be relevant in neurodegenerative diseases such as Alzheimer's disease and Multiple Sclerosis. Neuroprotective genes expressed at CPs may be unexplored targets for new therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ingrid Kratzer
- FLUID Team, Lyon Neurosciences Research Center, INSERM U1028 CNRS UMR 5292, University Claude Bernard Lyon 1, 69008 Lyon, France; Friedensgasse 3, 8010 Graz, Austria.
| | - Joakim Ek
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Medicinaregatan 11, Box 432, 40530 Göteborg, Sweden.
| | - Helen Stolp
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW0 1TU, UK.
| |
Collapse
|
16
|
Langford MB, O'Leary CJ, Veeraval L, White A, Lanoue V, Cooper HM. WNT5a Regulates Epithelial Morphogenesis in the Developing Choroid Plexus. Cereb Cortex 2020; 30:3617-3631. [PMID: 31912879 DOI: 10.1093/cercor/bhz330] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 12/13/2022] Open
Abstract
The choroid plexus (CP) is the predominant supplier of cerebral spinal fluid (CSF) and the site of the blood-CSF barrier and is thus essential for brain development and central nervous system homeostasis. Despite these crucial roles, our understanding of the molecular and cellular processes giving rise to the CPs within the ventricles of the mammalian brain is very rudimentary. Here, we identify WNT5a as an important regulator of CP development, where it acts as a pivotal factor driving CP epithelial morphogenesis in all ventricles. We show that WNT5a is essential for the establishment of a cohesive epithelium in the developing CP. We find that in its absence all CPs are substantially reduced in size and complexity and fail to expand into the ventricles. Severe defects were observed in the epithelial cytoarchitecture of all Wnt5a-/- CPs, exemplified by loss of apicobasally polarized morphology and detachment from the ventricular surface and/or basement membrane. We also present evidence that the WNT5a receptor, RYK, and the RHOA kinase, ROCK, are required for normal CP epithelial morphogenesis. Our study, therefore, reveals important insights into the molecular and cellular mechanisms governing CP development.
Collapse
Affiliation(s)
- Michael B Langford
- The University of Queensland, Queensland Brain Institute, Brisbane 4072, Australia and
| | - Conor J O'Leary
- The University of Queensland, Queensland Brain Institute, Brisbane 4072, Australia and
| | - Lenin Veeraval
- The University of Queensland, Queensland Brain Institute, Brisbane 4072, Australia and
| | - Amanda White
- The University of Queensland, Queensland Brain Institute, Brisbane 4072, Australia and
| | - Vanessa Lanoue
- The University of Queensland, Queensland Brain Institute, Brisbane 4072, Australia and.,Victor Chang Cardiac Research Institute, Darlinghurst 2010, Australia
| | - Helen M Cooper
- The University of Queensland, Queensland Brain Institute, Brisbane 4072, Australia and
| |
Collapse
|
17
|
Filatova A, Rey LK, Lechler MB, Schaper J, Hempel M, Posmyk R, Szczaluba K, Santen GWE, Wieczorek D, Nuber UA. Mutations in SMARCB1 and in other Coffin-Siris syndrome genes lead to various brain midline defects. Nat Commun 2019; 10:2966. [PMID: 31273213 PMCID: PMC6609698 DOI: 10.1038/s41467-019-10849-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 06/05/2019] [Indexed: 01/09/2023] Open
Abstract
Mutations in genes encoding components of BAF (BRG1/BRM-associated factor) chromatin remodeling complexes cause neurodevelopmental disorders and tumors. The mechanisms leading to the development of these two disease entities alone or in combination remain unclear. We generated mice with a heterozygous nervous system-specific partial loss-of-function mutation in a BAF core component gene, Smarcb1. These Smarcb1 mutant mice show various brain midline abnormalities that are also found in individuals with Coffin–Siris syndrome (CSS) caused by SMARCB1, SMARCE1, and ARID1B mutations and in SMARCB1-related intellectual disability (ID) with choroid plexus hyperplasia (CPH). Analyses of the Smarcb1 mutant animals indicate that one prominent midline abnormality, corpus callosum agenesis, is due to midline glia aberrations. Our results establish a novel role of Smarcb1 in the development of the brain midline and have important clinical implications for BAF complex-related ID/neurodevelopmental disorders. Why and how mutations in genes encoding BAF complex components lead to distinct disease entitites remains unresolved. In this study, authors establish the first Smarcb1 mutant mouse model with multiple brain abnormalities recapitulating human Coffin–Siris syndrome and show that one prominent midline abnormality, corpus callosum agenesis, is due to midline glia aberrations.
Collapse
Affiliation(s)
- Alina Filatova
- Stem Cell and Developmental Biology, Technical University Darmstadt, Darmstadt, 64287, Germany
| | - Linda K Rey
- Institute of Human Genetics, Medical Faculty, Heinrich Heine University, Düsseldorf, 40225, Germany
| | - Marion B Lechler
- Stem Cell and Developmental Biology, Technical University Darmstadt, Darmstadt, 64287, Germany
| | - Jörg Schaper
- Department of Diagnostic and Interventional Radiology, Medical Faculty, Heinrich Heine University, Düsseldorf, 40225, Germany
| | - Maja Hempel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Renata Posmyk
- Podlaskie Medical Centre "GENETICS" Bialystok and Department of Perinatology and Obstetrics, Medical University of Bialystok, Bialystok, 15-276, Poland
| | - Krzysztof Szczaluba
- Department of Medical Genetics, Medical University Warsaw, Warsaw, 02-106, Poland
| | - Gijs W E Santen
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, 2333 ZA, Netherlands
| | - Dagmar Wieczorek
- Institute of Human Genetics, Medical Faculty, Heinrich Heine University, Düsseldorf, 40225, Germany
| | - Ulrike A Nuber
- Stem Cell and Developmental Biology, Technical University Darmstadt, Darmstadt, 64287, Germany.
| |
Collapse
|
18
|
A Conserved Developmental Mechanism Builds Complex Visual Systems in Insects and Vertebrates. Curr Biol 2017; 26:R1001-R1009. [PMID: 27780043 DOI: 10.1016/j.cub.2016.08.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The visual systems of vertebrates and many other bilaterian clades consist of complex neural structures guiding a wide spectrum of behaviors. Homologies at the level of cell types and even discrete neural circuits have been proposed, but many questions of how the architecture of visual neuropils evolved among different phyla remain open. In this review we argue that the profound conservation of genetic and developmental steps generating the eye and its target neuropils in fish and fruit flies supports a homology between some core elements of bilaterian visual circuitries. Fish retina and tectum, and fly optic lobe, develop from a partitioned, unidirectionally proliferating neurectodermal domain that combines slowly dividing neuroepithelial stem cells and rapidly amplifying progenitors with shared genetic signatures to generate large numbers and different types of neurons in a temporally ordered way. This peculiar 'conveyor belt neurogenesis' could play an essential role in generating the topographically ordered circuitry of the visual system.
Collapse
|
19
|
Campos Y, Qiu X, Gomero E, Wakefield R, Horner L, Brutkowski W, Han YG, Solecki D, Frase S, Bongiovanni A, d'Azzo A. Alix-mediated assembly of the actomyosin-tight junction polarity complex preserves epithelial polarity and epithelial barrier. Nat Commun 2016; 7:11876. [PMID: 27336173 PMCID: PMC4931029 DOI: 10.1038/ncomms11876] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 05/09/2016] [Indexed: 02/07/2023] Open
Abstract
Maintenance of epithelial cell polarity and epithelial barrier relies on the spatial organization of the actin cytoskeleton and proper positioning/assembly of intercellular junctions. However, how these processes are regulated is poorly understood. Here we reveal a key role for the multifunctional protein Alix in both processes. In a knockout mouse model of Alix, we identified overt structural changes in the epithelium of the choroid plexus and in the ependyma, such as asymmetrical cell shape and size, misplacement and abnormal beating of cilia, blebbing of the microvilli. These defects culminate in excessive cell extrusion, enlargement of the lateral ventricles and hydrocephalus. Mechanistically, we find that by interacting with F-actin, the Par complex and ZO-1, Alix ensures the formation and maintenance of the apically restricted actomyosin-tight junction complex. We propose that in this capacity Alix plays a role in the establishment of apical-basal polarity and in the maintenance of the epithelial barrier.
Collapse
Affiliation(s)
- Yvan Campos
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Xiaohui Qiu
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Elida Gomero
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Randall Wakefield
- Cellular Imaging Shared Resource, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Linda Horner
- Cellular Imaging Shared Resource, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Wojciech Brutkowski
- Laboratory of Imaging Tissue Structure and Function, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Young-Goo Han
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - David Solecki
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Sharon Frase
- Cellular Imaging Shared Resource, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| | - Antonella Bongiovanni
- Institute of Biomedicine and Molecular Immunology, National Research Council, 90146 Palermo, Italy
| | - Alessandra d'Azzo
- Department of Genetics, St Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, USA
| |
Collapse
|
20
|
Development and functions of the choroid plexus-cerebrospinal fluid system. Nat Rev Neurosci 2015; 16:445-57. [PMID: 26174708 DOI: 10.1038/nrn3921] [Citation(s) in RCA: 365] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The choroid plexus (ChP) is the principal source of cerebrospinal fluid (CSF), which has accepted roles as a fluid cushion and a sink for nervous system waste in vertebrates. Various animal models have provided insights into how the ChP-CSF system develops and matures. In addition, recent studies have uncovered new, active roles for this dynamic system in the regulation of neural stem cells, critical periods and the overall health of the nervous system. Together, these findings have brought about a paradigm shift in our understanding of brain development and health, and have stimulated new initiatives for the treatment of neurological disease.
Collapse
|
21
|
Liddelow SA. Development of the choroid plexus and blood-CSF barrier. Front Neurosci 2015; 9:32. [PMID: 25784848 PMCID: PMC4347429 DOI: 10.3389/fnins.2015.00032] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/22/2015] [Indexed: 01/17/2023] Open
Abstract
Well-known as one of the main sources of cerebrospinal fluid (CSF), the choroid plexuses have been, and still remain, a relatively understudied tissue in neuroscience. The choroid plexus and CSF (along with the blood-brain barrier proper) are recognized to provide a robust protective effort for the brain: a physical barrier to impede entrance of toxic metabolites to the brain; a “biochemical” barrier that facilitates removal of moieties that circumvent this physical barrier; and buoyant physical protection by CSF itself. In addition, the choroid plexus-CSF system has been shown to be integral for normal brain development, central nervous system (CNS) homeostasis, and repair after disease and trauma. It has been suggested to provide a stem-cell like repository for neuronal and astrocyte glial cell progenitors. By far, the most widely recognized choroid plexus role is as the site of the blood-CSF barrier, controller of the internal CNS microenvironment. Mechanisms involved combine structural diffusion restraint from tight junctions between plexus epithelial cells (physical barrier) and specific exchange mechanisms across the interface (enzymatic barrier). The current hypothesis states that early in development this interface is functional and more specific than in the adult, with differences historically termed as “immaturity” actually correctly reflecting developmental specialization. The advanced knowledge of the choroid plexus-CSF system proves itself imperative to understand a range of neurological diseases, from those caused by plexus or CSF drainage dysfunction (e.g., hydrocephalus) to more complicated late-stage diseases (e.g., Alzheimer's) and failure of CNS regeneration. This review will focus on choroid plexus development, outlining how early specializations may be exploited clinically.
Collapse
Affiliation(s)
- Shane A Liddelow
- Department of Neurobiology, Stanford University CA, USA ; Department of Pharmacology and Therapeutics, The University of Melbourne Parkville, VIC, Australia
| |
Collapse
|
22
|
Bill BR, Korzh V. Choroid plexus in developmental and evolutionary perspective. Front Neurosci 2014; 8:363. [PMID: 25452709 PMCID: PMC4231874 DOI: 10.3389/fnins.2014.00363] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 10/22/2014] [Indexed: 01/17/2023] Open
Abstract
The blood-cerebrospinal fluid boundary is present at the level of epithelial cells of the choroid plexus. As one of the sources of the cerebrospinal fluid (CSF), the choroid plexus (CP) plays an important role during brain development and function. Its formation has been studied largely in mammalian species. Lately, progress in other model animals, in particular the zebrafish, has brought a deeper understanding of CP formation, due in part to the ability to observe CP development in vivo. At the same time, advances in comparative genomics began providing information, which opens a possibility to understand further the molecular mechanisms involved in evolution of the CP and the blood-cerebrospinal fluid boundary formation. Hence this review focuses on analysis of the CP from developmental and evolutionary perspectives.
Collapse
Affiliation(s)
- Brent Roy Bill
- Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles Los Angeles, CA, USA
| | - Vladimir Korzh
- Agency for Science, Technology and Research of Singapore, Institute of Molecular and Cell Biology Singapore, Singapore ; National University of Singapore, Department of Biological Sciences Singapore, Singapore
| |
Collapse
|
23
|
Henson HE, Parupalli C, Ju B, Taylor MR. Functional and genetic analysis of choroid plexus development in zebrafish. Front Neurosci 2014; 8:364. [PMID: 25426018 PMCID: PMC4226144 DOI: 10.3389/fnins.2014.00364] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/22/2014] [Indexed: 01/30/2023] Open
Abstract
The choroid plexus, an epithelial-based structure localized in the brain ventricle, is the major component of the blood-cerebrospinal fluid barrier. The choroid plexus produces the cerebrospinal fluid and regulates the components of the cerebrospinal fluid. Abnormal choroid plexus function is associated with neurodegenerative diseases, tumor formation in the choroid plexus epithelium, and hydrocephaly. In this study, we used zebrafish (Danio rerio) as a model system to understand the genetic components of choroid plexus development. We generated an enhancer trap line, Et(cp:EGFP)sj2, that expresses enhanced green fluorescent protein (EGFP) in the choroid plexus epithelium. Using immunohistochemistry and fluorescent tracers, we demonstrated that the zebrafish choroid plexus possesses brain barrier properties such as tight junctions and transporter activity. Thus, we have established zebrafish as a functionally relevant model to study choroid plexus development. Using an unbiased approach, we performed a forward genetic dissection of the choroid plexus to identify genes essential for its formation and function. Using Et(cp:EGFP)sj2, we isolated 10 recessive mutant lines with choroid plexus abnormalities, which were grouped into five classes based on GFP intensity, epithelial localization, and overall choroid plexus morphology. We also mapped the mutation for two mutant lines to chromosomes 4 and 21, respectively. The mutants generated in this study can be used to elucidate specific genes and signaling pathways essential for choroid plexus development, function, and/or maintenance and will provide important insights into how these genetic mutations contribute to disease.
Collapse
Affiliation(s)
- Hannah E Henson
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital Memphis, TN, USA ; Integrated Program in Biomedical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center Memphis, TN, USA
| | | | - Bensheng Ju
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital Memphis, TN, USA
| | - Michael R Taylor
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital Memphis, TN, USA ; Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison Madison, WI, USA
| |
Collapse
|
24
|
Johansson PA. The choroid plexuses and their impact on developmental neurogenesis. Front Neurosci 2014; 8:340. [PMID: 25386116 PMCID: PMC4208491 DOI: 10.3389/fnins.2014.00340] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 10/07/2014] [Indexed: 11/23/2022] Open
Abstract
During brain development the neural stem cells are regulated by both intrinsic and extrinsic sources. One site of origin of extrinsic regulation is the developing choroid plexuses, primely situated inside the cerebral ventricles. The choroid plexuses are very active in terms of both secretion and barrier function as soon as they appear during development and control the production and contents of cerebrospinal fluid (CSF). This suggests that regulated secretion of signaling molecules from the choroid plexuses into CSF can regulate neural stem cell behavior (as they are in direct contact with CSF) and thereby neurogenesis and brain development. Here, choroid plexus development, particularly with regards to molecular regulation and specification, is reviewed. This is followed by a review and discussion of the role of the developing choroid plexuses in brain development. In particular, recent evidence suggests a region-specific reciprocal regulation between choroid plexuses and the neural stem cells. This is accomplished by site-specific secretion of signaling molecules from the different choroid plexuses into CSF, as well as brain region specific competence of the neural stem cells to respond to the signaling molecules present in CSF. In conclusion, although in its infancy, the field of choroid plexus regulation of neurogenesis has already and will likely continue to shed new light on our understanding of the control and fine-tuning of overall brain development.
Collapse
Affiliation(s)
- Pia A Johansson
- Institute for Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health Munich, Germany
| |
Collapse
|
25
|
Liddelow SA, Dzięgielewska KM, Møllgård K, Whish SC, Noor NM, Wheaton BJ, Gehwolf R, Wagner A, Traweger A, Bauer H, Bauer HC, Saunders NR. Cellular specificity of the blood-CSF barrier for albumin transfer across the choroid plexus epithelium. PLoS One 2014; 9:e106592. [PMID: 25211495 PMCID: PMC4161337 DOI: 10.1371/journal.pone.0106592] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 08/01/2014] [Indexed: 01/29/2023] Open
Abstract
To maintain the precise internal milieu of the mammalian central nervous system, well-controlled transfer of molecules from periphery into brain is required. Recently the soluble and cell-surface albumin-binding glycoprotein SPARC (secreted protein acidic and rich in cysteine) has been implicated in albumin transport into developing brain, however the exact mechanism remains unknown. We postulate that SPARC is a docking site for albumin, mediating its uptake and transfer by choroid plexus epithelial cells from blood into cerebrospinal fluid (CSF). We used in vivo physiological measurements of transfer of endogenous (mouse) and exogenous (human) albumins, in situ Proximity Ligation Assay (in situ PLA), and qRT-PCR experiments to examine the cellular mechanism mediating protein transfer across the blood–CSF interface. We report that at all developmental stages mouse albumin and SPARC gave positive signals with in situ PLAs in plasma, CSF and within individual plexus cells suggesting a possible molecular interaction. In contrast, in situ PLA experiments in brain sections from mice injected with human albumin showed positive signals for human albumin in the vascular compartment that were only rarely identifiable within choroid plexus cells and only at older ages. Concentrations of both endogenous mouse albumin and exogenous (intraperitoneally injected) human albumin were estimated in plasma and CSF and expressed as CSF/plasma concentration ratios. Human albumin was not transferred through the mouse blood–CSF barrier to the same extent as endogenous mouse albumin, confirming results from in situ PLA. During postnatal development Sparc gene expression was higher in early postnatal ages than in the adult and changed in response to altered levels of albumin in blood plasma in a differential and developmentally regulated manner. Here we propose a possible cellular route and mechanism by which albumin is transferred from blood into CSF across a sub-population of specialised choroid plexus epithelial cells.
Collapse
Affiliation(s)
- Shane A. Liddelow
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, Australia
- Department of Neurobiology, Stanford University, Stanford, California, United States of America
| | | | - Kjeld Møllgård
- Institute of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sophie C. Whish
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, Australia
| | - Natassya M. Noor
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, Australia
| | - Benjamin J. Wheaton
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, Australia
| | - Renate Gehwolf
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Andrea Wagner
- Department of Organismic Biology, University of Salzburg, Salzburg, Austria
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Andreas Traweger
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Hannelore Bauer
- Department of Organismic Biology, University of Salzburg, Salzburg, Austria
| | - Hans-Christian Bauer
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University, Salzburg, Austria
| | - Norman R. Saunders
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, Australia
- * E-mail:
| |
Collapse
|
26
|
Liddelow SA, Dziegielewska KM, Ek CJ, Habgood MD, Bauer H, Bauer HC, Lindsay H, Wakefield MJ, Strazielle N, Kratzer I, Møllgård K, Ghersi-Egea JF, Saunders NR. Mechanisms that determine the internal environment of the developing brain: a transcriptomic, functional and ultrastructural approach. PLoS One 2013; 8:e65629. [PMID: 23843944 PMCID: PMC3699566 DOI: 10.1371/journal.pone.0065629] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 04/25/2013] [Indexed: 01/04/2023] Open
Abstract
We provide comprehensive identification of embryonic (E15) and adult rat lateral ventricular choroid plexus transcriptome, with focus on junction-associated proteins, ionic influx transporters and channels. Additionally, these data are related to new structural and previously published permeability studies. Results reveal that most genes associated with intercellular junctions are expressed at similar levels at both ages. In total, 32 molecules known to be associated with brain barrier interfaces were identified. Nine claudins showed unaltered expression, while two claudins (6 and 8) were expressed at higher levels in the embryo. Expression levels for most cytoplasmic/regulatory adaptors (10 of 12) were similar at the two ages. A few junctional genes displayed lower expression in embryos, including 5 claudins, occludin and one junctional adhesion molecule. Three gap junction genes were enriched in the embryo. The functional effectiveness of these junctions was assessed using blood-delivered water-soluble tracers at both the light and electron microscopic level: embryo and adult junctions halted movement of both 286Da and 3kDa molecules into the cerebrospinal fluid (CSF). The molecular identities of many ion channel and transporter genes previously reported as important for CSF formation and secretion in the adult were demonstrated in the embryonic choroid plexus (and validated with immunohistochemistry of protein products), but with some major age-related differences in expression. In addition, a large number of previously unidentified ion channel and transporter genes were identified for the first time in plexus epithelium. These results, in addition to data obtained from electron microscopical and physiological permeability experiments in immature brains, indicate that exchange between blood and CSF is mainly transcellular, as well-formed tight junctions restrict movement of small water-soluble molecules from early in development. These data strongly indicate the brain develops within a well-protected internal environment and the exchange between the blood, brain and CSF is transcellular and not through incomplete barriers.
Collapse
Affiliation(s)
- Shane A Liddelow
- Department of Pharmacology, the University of Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Haddad MR, Donsante A, Zerfas P, Kaler SG. Fetal Brain-directed AAV Gene Therapy Results in Rapid, Robust, and Persistent Transduction of Mouse Choroid Plexus Epithelia. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e101. [PMID: 23799375 PMCID: PMC3696907 DOI: 10.1038/mtna.2013.27] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Fetal brain-directed gene addition represents an under-appreciated tool for investigating novel therapeutic approaches in animal models of central nervous system diseases with early prenatal onset. Choroid plexuses (CPs) are specialized neuroectoderm-derived structures that project into the brain's ventricles, produce cerebrospinal fluid (CSF), and regulate CSF biochemical composition. Targeting the CP may be advantageous for adeno-associated viral (AAV) gene therapy for central nervous system disorders due to its immunoprivileged location and slow rate of epithelial turnover. Yet the capacity of AAV vectors to transduce CP has not been delineated precisely. We performed intracerebroventricular injections of recombinant AAV serotype 5-green fluorescent protein (rAAV5-GFP) or rAAV9-GFP in embryonic day 15 (E15) embryos of CD-1 and C57BL/6 pregnant mice and quantified the percentages of GFP expression in CP epithelia (CPE) from lateral and fourth ventricles on E17, postnatal day 2 (P2), and P22. AAV5 was selective for CPE and showed significantly higher transduction efficiency in C57BL/6 mice (P = 0.0128). AAV9 transduced neurons and glial cells in both the mouse strains, in addition to CPE. We documented GFP expression in CPE on E17, within just 48 hours of rAAV administration to the fetal lateral ventricle, and expression by both the serotypes persisted at P130. Our results indicate that prenatal administration of rAAV5 and rAAV9 enables rapid, robust, and sustained transduction of mouse CPE and buttress the rationale for experimental therapeutics targeting the CP.Molecular Therapy-Nucleic Acids (2013) 2, e101; doi:10.1038/mtna.2013.27; published online 25 June 2013.
Collapse
Affiliation(s)
- Marie Reine Haddad
- Unit on Human Copper Metabolism, Molecular Medicine Program, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|