1
|
Jensen DB, Toft-Bertelsen TL, Barbuskaite D, Stubbe J, Nietzsche S, Capion T, Norager NH, Olsen MH, Sørensen AT, Dimke H, Hübner CA, Juhler M, MacAulay N. The Na +,K +,2Cl - Cotransporter, Not Aquaporin 1, Sustains Cerebrospinal Fluid Secretion While Controlling Brain K + Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2409120. [PMID: 39692709 DOI: 10.1002/advs.202409120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/11/2024] [Indexed: 12/19/2024]
Abstract
Disturbances in the brain fluid balance can lead to life-threatening elevation in intracranial pressure (ICP), which represents a vast clinical challenge. Targeted and efficient pharmaceutical therapy of elevated ICP is not currently available, as the molecular mechanisms governing cerebrospinal fluid (CSF) secretion are largely unresolved. To resolve the quantitative contribution of key choroid plexus transport proteins, this study employs mice with genetic knockout and/or viral choroid plexus-specific knockdown of aquaporin 1 (AQP1) and the Na+, K+, 2Cl- cotransporter 1 (NKCC1) for in vivo determinations of CSF dynamics, ex vivo choroid plexus for transporter-mediated clearance of a CSF K+ load, and patient CSF for [K+] quantification. CSF secretion and ICP management occur independently of choroid plexus AQP1 expression, whereas both parameters are reduced by 40% upon choroid plexus NKCC1 knockdown. Elevation of [K+]CSF increases the choroid plexus Na+/K+-ATPase activity, and favors inwardly-directed net NKCC1 transport, which, together, promote CSF K+ clearance, while maintaining undisturbed CSF secretion rates. CSF from patients with post-hemorrhagic hydrocephalus does not display elevated [K+]CSF, suggesting that NKCC1 maintains net outward transport direction during post-hemorrhagic hydrocephalus formation. Direct or indirect therapeutic modulation of choroid plexus NKCC1 can thus be a potential promising pharmacological approach against brain pathologies associated with elevated ICP.
Collapse
Affiliation(s)
- Dennis Bo Jensen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Dagne Barbuskaite
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Jane Stubbe
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, Odense, 5230, Denmark
| | - Sandor Nietzsche
- Center for Electron Microscopy, Jena University Hospital, Ziegelmühlenweg 1, 07743, Jena, Germany
| | - Tenna Capion
- Department of Neurosurgery, University Hospital of Copenhagen - Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark
| | - Nicolas H Norager
- Department of Neurosurgery, University Hospital of Copenhagen - Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark
| | - Markus H Olsen
- Department of Neuroanaesthesiology, University Hospital of Copenhagen - Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark
| | - Andreas T Sørensen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| | - Henrik Dimke
- Department of Nephrology, Odense University Hospital, J.B. Winsløws Vej 4, Odense, 5000, Denmark
| | - Christian A Hübner
- Hübner, Institute of Human Genetics, Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Marianne Juhler
- Department of Neurosurgery, University Hospital of Copenhagen - Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3, Copenhagen, 2200, Denmark
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, Copenhagen N, 2200, Denmark
| |
Collapse
|
2
|
Marottoli FM, Balu D, Chaudhary R, Lutz SE, Tai LM. Evaluation of BR1 and BI30 AAVs for Brain Endothelial Tropism. ASN Neuro 2024; 16:2427953. [PMID: 39621720 DOI: 10.1080/17590914.2024.2427953] [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: 07/30/2024] [Revised: 10/04/2024] [Accepted: 11/03/2024] [Indexed: 12/06/2024] Open
Abstract
Brain endothelial cells are critical for homeostasis of the central nervous system. Novel adeno-associated viruses (AAV) with brain endothelial cell tropism have been developed and are beginning to be employed in mechanistic and therapeutic research. Studies using AAVs can be involved in terms of cost, time and personnel, and many groups, including our own, are not experts on the technology. Therefore, it is important to report data using AAVs with the research community as a guide for ongoing and future studies. Here, we detail our initial experience with the two most prevalent AAVs with tropism for brain endothelial cells, AAV-BR1 and AAV-BI30. One of our long-term goals is to express key proteins in brain endothelial cells and determine the impact on brain function. For method development, we administered AAV-BR1 and AAV-BI30 with a CMV-driven fluorescent reporter (CMV-P2A-mCherry) to wild-type mice intravenously (retro-orbital) and measured expression in brain and peripheral tissues by RT-PCR and immunostaining. We found that AAV-BR1 transduces neurons and endothelial cells in the brain, and the lung and liver, whereas AAV-BI30 transduces brain endothelial cells and peripheral tissue. Our data highlights the importance of using the AAV best suited to the scientific question.
Collapse
Affiliation(s)
- Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Rohan Chaudhary
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
3
|
Xie S, Li F. Ependymal cells: roles in central nervous system infections and therapeutic application. J Neuroinflammation 2024; 21:255. [PMID: 39385253 PMCID: PMC11465851 DOI: 10.1186/s12974-024-03240-2] [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: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
Ependymal cells are arranged along the inner surfaces of the ventricles and the central canal of the spinal cord, providing anatomical, physiological and immunological barriers that maintain cerebrospinal fluid (CSF) homeostasis. Based on this, studies have found that alterations in gene expression, cell junctions, cytokine secretion and metabolic disturbances can lead to dysfunction of ependymal cells, thereby participating in the onset and progression of central nervous system (CNS) infections. Additionally, ependymal cells can exhibit proliferative and regenerative potential as well as secretory functions during CNS injury, contributing to neuroprotection and post-injury recovery. Currently, studies on ependymal cell primarily focus on the basic investigations of their morphology, function and gene expression; however, there is a notable lack of clinical translational studies examining the molecular mechanisms by which ependymal cells are involved in disease onset and progression. This limits our understanding of ependymal cells in CNS infections and the development of therapeutic applications. Therefore, this review will discuss the molecular mechanism underlying the involvement of ependymal cells in CNS infections, and explore their potential for application in clinical treatment modalities.
Collapse
Affiliation(s)
- Shiqi Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
- Shanghai Institute of Infectious Disease and Biosecurity, 130 Dong An Road, Xuhui District, Shanghai, China.
- Tuberculosis Research Center, Shanghai Public Health Clinical Center, Fudan University, 2901 Cao Lang Road, Jinshan District, Shanghai, China.
| |
Collapse
|
4
|
Wang Q, Liu F, Li Y, Zhang H, Qi X, Wu K, Zhang Y, You S, Liu W, Hui X, Li H, Zhu L, Gao H, Cheng J. Choroid plexus CCL2‒CCR2 signaling orchestrates macrophage recruitment and cerebrospinal fluid hypersecretion in hydrocephalus. Acta Pharm Sin B 2024; 14:4544-4559. [PMID: 39525574 PMCID: PMC11544184 DOI: 10.1016/j.apsb.2024.06.020] [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: 03/15/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 11/16/2024] Open
Abstract
The choroid plexus (ChP) serves as the principal origin of cerebrospinal fluid (CSF). CSF hypersecretion due to ChP inflammation has emerged as an important pathogenesis of hydrocephalus recently. Nevertheless, the precise mechanisms of ChP inflammation and the ensuing CSF hypersecretion in hydrocephalus remain ill-defined. In the present study, we elucidate the critical role of macrophages in the pathogenesis of ChP inflammation. Specifically, we identify the chemokine CCL2, released by ChP epithelial cells, recruits CCR2+ monocytes to the ChP thereby inciting hydrocephalus pathogenesis. The accumulated ChP macrophages increase the inflammation in ChP epithelial cells through TNF-α/TNFR1/NF-κB signaling cascade, thereby leading to CSF hypersecretion. Strikingly, augmentation of ChP‒CCL2 using an adeno-associated viral approach (AAV) exacerbates macrophage recruitment, activation, and ventriculomegaly in rat PHH models. Systemic application of Bindarit, a specific CCL2 inhibitor, significantly inhibits ChP macrophage infiltration and activation and reduces CSF secretion rate. Furthermore, the administration of CCR2 antagonist (INCB 3284) reduces ChP macrophage accumulation and ventriculomegaly. This study not only unveils the ChP CCL2‒CCR2 signaling in the pathophysiology of hydrocephalus but also unveils Bindarit as a promising therapeutic choice for the management of posthemorrhagic hydrocephalus.
Collapse
Affiliation(s)
- Qiguang Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fei Liu
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yue Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huan Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin Qi
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Wu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yi Zhang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shenglan You
- Research Core Facility of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenke Liu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuhui Hui
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Li
- Key Laboratory of Coarse Cereal Processing, School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Lei Zhu
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jian Cheng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
van de Vlekkert D, Hu H, Weesner JA, Fremuth LE, Brown SA, Lu M, Gomero E, Campos Y, Sheppard H, d'Azzo A. AAV-mediated gene therapy for sialidosis. Mol Ther 2024; 32:2094-2112. [PMID: 38796704 PMCID: PMC11287007 DOI: 10.1016/j.ymthe.2024.05.029] [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: 11/21/2023] [Revised: 04/01/2024] [Accepted: 05/23/2024] [Indexed: 05/28/2024] Open
Abstract
Sialidosis (mucolipidosis I) is a glycoprotein storage disease, clinically characterized by a spectrum of systemic and neurological phenotypes. The primary cause of the disease is deficiency of the lysosomal sialidase NEU1, resulting in accumulation of sialylated glycoproteins/oligosaccharides in tissues and body fluids. Neu1-/- mice recapitulate the severe, early-onset forms of the disease, affecting visceral organs, muscles, and the nervous system, with widespread lysosomal vacuolization evident in most cell types. Sialidosis is considered an orphan disorder with no therapy currently available. Here, we assessed the therapeutic potential of AAV-mediated gene therapy for the treatment of sialidosis. Neu1-/- mice were co-injected with two scAAV2/8 vectors, expressing human NEU1 and its chaperone PPCA. Treated mice were phenotypically indistinguishable from their WT controls. NEU1 activity was restored to different extent in most tissues, including the brain, heart, muscle, and visceral organs. This resulted in diminished/absent lysosomal vacuolization in multiple cell types and reversal of sialyl-oligosacchariduria. Lastly, normalization of lysosomal exocytosis in the cerebrospinal fluids and serum of treated mice, coupled to diminished neuroinflammation, were measures of therapeutic efficacy. These findings point to AAV-mediated gene therapy as a suitable treatment for sialidosis and possibly other diseases, associated with low NEU1 expression.
Collapse
Affiliation(s)
| | - Huimin Hu
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jason A Weesner
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Leigh E Fremuth
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Scott A Brown
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Meifen Lu
- Devision of Comparative Pathology, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Elida Gomero
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yvan Campos
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather Sheppard
- Devision of Comparative Pathology, Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Anatomy and Neurobiology, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
6
|
Jeong I, Andreassen SN, Hoang L, Poulain M, Seo Y, Park HC, Fürthauer M, MacAulay N, Jurisch-Yaksi N. The evolutionarily conserved choroid plexus contributes to the homeostasis of brain ventricles in zebrafish. Cell Rep 2024; 43:114331. [PMID: 38843394 DOI: 10.1016/j.celrep.2024.114331] [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: 11/14/2023] [Revised: 04/24/2024] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
The choroid plexus (ChP) produces cerebrospinal fluid (CSF). It also contributes to brain development and serves as the CSF-blood barrier. Prior studies have identified transporters on the epithelial cells that transport water and ions from the blood vasculature to the ventricles and tight junctions involved in the CSF-blood barrier. Yet, how the ChP epithelial cells control brain physiology remains unresolved. We use zebrafish to provide insights into the physiological roles of the ChP. Upon histological and transcriptomic analyses, we identify that the zebrafish ChP is conserved with mammals and expresses transporters involved in CSF secretion. Next, we show that the ChP epithelial cells secrete proteins into CSF. By ablating the ChP epithelial cells, we identify a reduction of the ventricular sizes without alterations of the CSF-blood barrier. Altogether, our findings reveal that the zebrafish ChP is conserved and contributes to the size and homeostasis of the brain ventricles.
Collapse
Affiliation(s)
- Inyoung Jeong
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgsons Gate 1, 7491 Trondheim, Norway
| | - Søren N Andreassen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Linh Hoang
- Cellular and Molecular Imaging Core Facility (CMIC), Norwegian University of Science and Technology, Erling Skjalgsons Gate 1, 7491 Trondheim, Norway
| | - Morgane Poulain
- Université Côte d'Azur, CNRS, Inserm, iBV, 28 Avenue Valrose, 06108 Nice cedex 2, France
| | - Yongbo Seo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Maximilian Fürthauer
- Université Côte d'Azur, CNRS, Inserm, iBV, 28 Avenue Valrose, 06108 Nice cedex 2, France
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgsons Gate 1, 7491 Trondheim, Norway.
| |
Collapse
|
7
|
Tsitsou-Kampeli A, Suzzi S, Kenigsbuch M, Satomi A, Strobelt R, Singer O, Feldmesser E, Purnapatre M, Colaiuta SP, David E, Cahalon L, Hahn O, Wyss-Coray T, Shaul Y, Amit I, Schwartz M. Cholesterol 24-hydroxylase at the choroid plexus contributes to brain immune homeostasis. Cell Rep Med 2023; 4:101278. [PMID: 37944529 PMCID: PMC10694665 DOI: 10.1016/j.xcrm.2023.101278] [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: 11/15/2022] [Revised: 05/26/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
The choroid plexus (CP) plays a key role in remotely controlling brain function in health, aging, and disease. Here, we report that CP epithelial cells express the brain-specific cholesterol 24-hydroxylase (CYP46A1) and that its levels are decreased under different mouse and human brain conditions, including amyloidosis, aging, and SARS-CoV-2 infection. Using primary mouse CP cell cultures, we demonstrate that the enzymatic product of CYP46A1, 24(S)-hydroxycholesterol, downregulates inflammatory transcriptomic signatures within the CP, found here to be elevated across multiple neurological conditions. In vitro, the pro-inflammatory cytokine tumor necrosis factor α (TNF-α) downregulates CYP46A1 expression, while overexpression of CYP46A1 or its pharmacological activation in mouse CP organ cultures increases resilience to TNF-α. In vivo, overexpression of CYP46A1 in the CP in transgenic mice with amyloidosis is associated with better cognitive performance and decreased brain inflammation. Our findings suggest that CYP46A1 expression in the CP impacts the role of this niche as a guardian of brain immune homeostasis.
Collapse
Affiliation(s)
| | - Stefano Suzzi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| | - Mor Kenigsbuch
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Akisawa Satomi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel; Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Romano Strobelt
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Oded Singer
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ester Feldmesser
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liora Cahalon
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Oliver Hahn
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
8
|
van de Vlekkert D, Hu H, Fremuth LE, Brown SA, Weesner JA, Gomero E, Campos Y, d'Azzo A. AAV-mediated gene therapy for Sialidosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566667. [PMID: 38014061 PMCID: PMC10680618 DOI: 10.1101/2023.11.10.566667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Sialidosis is a glycoprotein storage disease caused by deficiency of the lysosomal sialidase NEU1, which leads to pathogenic accumulation of sialylated glycoproteins and oligosaccharides in tissues and body fluids. The disease belongs to the group of orphan disorders with no therapy currently available. Here, we have tested the therapeutic potential of AAV-mediated gene therapy for the treatment of sialidosis in a mouse model of the disease. One-month-old Neu1 -/- mice were co-injected with two scAAV2/8 vectors, expressing NEU1 and its chaperone PPCA, and sacrificed at 3 months post-injection. Treated mice were phenotypically indistinguishable from their WT controls. Histopathologically, they showed diminished or absent vacuolization in cells of visceral organs, including the kidney, as well as the choroid plexus and other areas of the brain. This was accompanied by restoration of NEU1 activity in most tissues, reversal of sialyl-oligosacchariduria, and normalization of lysosomal exocytosis in the CSF and serum of treated mice. AAV injection prevented the occurrence of generalized fibrosis, which is a prominent contributor of disease pathogenesis in Neu1 -/- mice and likely in patients. Overall, this therapeutic strategy holds promise for the treatment of sialidosis and may be applicable to adult forms of human idiopathic fibrosis with low NEU1 expression.
Collapse
|
9
|
Mazucanti CH, Kennedy V, Premathilake HU, Doyle ME, Tian J, Liu QR, O'Connell J, Camandola S, Egan JM. AAV5-mediated manipulation of insulin expression in choroid plexus has long-term metabolic and behavioral consequences. Cell Rep 2023; 42:112903. [PMID: 37515772 PMCID: PMC10529429 DOI: 10.1016/j.celrep.2023.112903] [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: 02/07/2023] [Revised: 06/06/2023] [Accepted: 07/13/2023] [Indexed: 07/31/2023] Open
Abstract
The choroid plexus (CP) is a source of trophic factors for the developing and mature brain. Insulin is produced in epithelial cells of the CP (EChPs), and its secretion is stimulated by Htr2c-mediated signaling. We modulated insulin expression in EChPs with intracerebroventricular injections of AAV5. Insulin overexpression in CP decelerates food intake, whereas its knockdown has the opposite effect. Insulin overexpression also results in reduced anxious behavior. Transcriptomic changes in the hypothalamus, especially in synapse-related processes, are also seen in mice overexpressing insulin in CP. Last, activation of Gq signaling in CP leads to acute Akt phosphorylation in neurons of the arcuate nucleus, indicating a direct action of CP-derived insulin on the hypothalamus. Taken together, our findings signify that CP is a relevant source of insulin in the central nervous system and that CP-derived insulin should be taken into consideration in future work pertaining to insulin actions in the brain.
Collapse
Affiliation(s)
- Caio Henrique Mazucanti
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Vernon Kennedy
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Hasitha U Premathilake
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Maire E Doyle
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Jane Tian
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Qing-Rong Liu
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Jennifer O'Connell
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Simonetta Camandola
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA
| | - Josephine M Egan
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
10
|
Ledri M, Andersson M, Wickham J, Kokaia M. Optogenetics for controlling seizure circuits for translational approaches. Neurobiol Dis 2023:106234. [PMID: 37479090 DOI: 10.1016/j.nbd.2023.106234] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/02/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023] Open
Abstract
The advent of optogenetic tools has had a profound impact on modern neuroscience research, revolutionizing our understanding of the brain. These tools offer a remarkable ability to precisely manipulate specific groups of neurons with an unprecedented level of temporal precision, on the order of milliseconds. This breakthrough has significantly advanced our knowledge of various physiological and pathophysiological processes in the brain. Within the realm of epilepsy research, optogenetic tools have played a crucial role in investigating the contributions of different neuronal populations to the generation of seizures and hyperexcitability. By selectively activating or inhibiting specific neurons using optogenetics, researchers have been able to elucidate the underlying mechanisms and identify key players involved in epileptic activity. Moreover, optogenetic techniques have also been explored as innovative therapeutic strategies for treating epilepsy. These strategies aim to halt seizure progression and alleviate symptoms by utilizing the precise control offered by optogenetics. The application of optogenetic tools has provided valuable insights into the intricate workings of the brain during epileptic episodes. For instance, researchers have discovered how distinct interneuron populations contribute to the initiation of seizures (ictogenesis). They have also revealed how remote circuits in regions such as the cerebellum, septum, or raphe nuclei can interact with hyperexcitable networks in the hippocampus. Additionally, studies have demonstrated the potential of closed-loop systems, where optogenetics is combined with real-time monitoring, to enable precise, on-demand control of seizure activity. Despite the immense promise demonstrated by optogenetic approaches, it is important to acknowledge that many of these techniques are still in the early stages of development and have yet to reach potential clinical applications. The transition from experimental research to practical clinical use poses numerous challenges. In this review, we aim to introduce optogenetic tools, provide a comprehensive survey of their application in epilepsy research, and critically discuss their current potential and limitations in achieving successful clinical implementation for the treatment of human epilepsy. By addressing these crucial aspects, we hope to foster a deeper understanding of the current state and future prospects of optogenetics in epilepsy treatment.
Collapse
Affiliation(s)
- Marco Ledri
- Epilepsy Center, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sölvegatan 17, 223 62 Lund, Sweden
| | - My Andersson
- Epilepsy Center, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sölvegatan 17, 223 62 Lund, Sweden
| | - Jenny Wickham
- Epilepsy Center, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sölvegatan 17, 223 62 Lund, Sweden
| | - Merab Kokaia
- Epilepsy Center, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sölvegatan 17, 223 62 Lund, Sweden.
| |
Collapse
|
11
|
Cheng K, Wang Y, He Y, Tian Y, Li J, Chen C, Xu X, Wu Z, Yu H, Chen X, Wu Y, Song W, Dong Z, Xu H, Xie P. Upregulation of carbonic anhydrase 1 beneficial for depressive disorder. Acta Neuropathol Commun 2023; 11:59. [PMID: 37013604 PMCID: PMC10071615 DOI: 10.1186/s40478-023-01545-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 03/06/2023] [Indexed: 04/05/2023] Open
Abstract
Carbonic Anhydrase 1 (CAR1) is a zinc-metalloenzyme that catalyzes the hydration of carbon dioxide, and the alteration of CAR1 has been implicated in neuropsychiatric disorders. However, the mechanism underlying the role of CAR1 in major depressive disorder (MDD) remains largely unknown. In this study, we report the decreased level of CAR1 in MDD patients and depression-like model rodents. We found that CAR1 is expressed in hippocampal astrocytes and CAR1 regulates extracellular bicarbonate concentration and pH value in the partial hilus. Ablation of the CAR1 gene increased the activity of granule cells via decreasing their miniature inhibitory postsynaptic currents (mIPSC), and caused depression-like behaviors in CAR1-knockout mice. Astrocytic CAR1 expression rescued the deficits in mIPSCs of granule cells and reduced depression-like behaviors in CAR1 deficient mice. Furthermore, pharmacological activation of CAR1 and overexpression of CAR1 in the ventral hippocampus of mice improved depressive behaviors. These findings uncover a critical role of CAR1 in the MDD pathogenesis and its therapeutic potential.
Collapse
Affiliation(s)
- Ke Cheng
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yong He
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Tian
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Junjie Li
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, China
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Chong Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xingzhe Xu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhonghao Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Heming Yu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiangyu Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yili Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Zhejiang Provincial Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Zhifang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing, 400014, China.
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Huatai Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
12
|
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 DOI: 10.1096/fj.202201767r] [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: 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 Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, China
| | - Lanxin Hu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuwen Yang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Cheng Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wenzhan Tu
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, China
| | - Haiyan Lin
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, China
| | - Haotian Wang
- Alberta Institute, Wenzhou Medical University, Wenzhou, China
| | - Yiwei Jiang
- Alberta Institute, Wenzhou Medical University, Wenzhou, China
| | - Songhe Jiang
- Rehabilitation Medicine Center, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Integrative & Optimized Medicine Research center, China-USA Institute for Acupuncture and Rehabilitation, Wenzhou Medical University, Wenzhou, China
| | - Wu Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, China.,The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Liu LL, Shannahan J, Zheng W. Choroid Plexus Modulates Subventricular Zone Adult Neurogenesis and Olfaction Through Secretion of Small Extracellular Vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532966. [PMID: 36993578 PMCID: PMC10055063 DOI: 10.1101/2023.03.16.532966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
UNLABELLED The choroid plexus (CP) in brain ventricles secrete cerebrospinal fluid (CSF) that bathes the adjacent subventricular zone (SVZ); the latter is the largest neurogenic region in adult brain harboring neural stem/progenitor cells (NSPCs) and supplies newborn neurons to the olfactory bulb (OB) for normal olfaction. We discovered the presence of a CP-SVZ regulatory (CSR) axis in which the CP, by secreting small extracellular vesicles (sEVs), regulated adult neurogenesis in the SVZ and maintained olfaction. The proposed CSR axis was supported by 1) differential neurogenesis outcomes in the OB when animals treated with intracerebroventricular (ICV) infusion of sEVs collected from the CP of normal or manganese (Mn)-poisoned mice, 2) progressively diminished SVZ adult neurogenesis in mice following CP-targeted knockdown of SMPD3 to suppress CP sEV secretion, and 3) compromised olfactory performance in these CP-SMPD3-knockdown mice. Collectively, our findings demonstrate the biological and physiological presence of this sEV-dependent CSR axis in adult brains. HIGHLIGHTS CP-secreted sEVs regulate adult neurogenesis in the SVZ.CP-secreted sEVs modulate newborn neurons in the OB.Suppression of sEV secretion from the CP deteriorates olfactory performance.
Collapse
|
14
|
Mani S, Jindal D, Singh M. Gene Therapy, A Potential Therapeutic Tool for Neurological and Neuropsychiatric Disorders: Applications, Challenges and Future Perspective. Curr Gene Ther 2023; 23:20-40. [PMID: 35345999 DOI: 10.2174/1566523222666220328142427] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/18/2022] [Accepted: 02/02/2022] [Indexed: 02/08/2023]
Abstract
Neurological and neuropsychiatric disorders are the main risks for the health care system, exhibiting a huge socioeconomic load. The available range of pharmacotherapeutics mostly provides palliative consequences and fails to treat such conditions. The molecular etiology of various neurological and neuropsychiatric disorders is mostly associated with a change in genetic background, which can be inherited/triggered by other environmental factors. To address such conditions, gene therapy is considered a potential approach claiming a permanent cure of the disease primarily by deletion, silencing, or edition of faulty genes and by insertion of healthier genes. In gene therapy, vectors (viral/nonvial) play an important role in delivering the desired gene to a specific region of the brain. Targeted gene therapy has unraveled opportunities for the treatment of many neurological and neuropsychiatric disorders. For improved gene delivery, the current techniques mainly focus on designing a precise viral vector, plasmid transfection, nanotechnology, microRNA, and in vivo clustered regulatory interspaced short palindromic repeats (CRISPR)-based therapy. These latest techniques have great benefits in treating predominant neurological and neurodevelopmental disorders, including Parkinson's disease, Alzheimer's disease, and autism spectrum disorder, as well as rarer diseases. Nevertheless, all these delivery methods have their limitations, including immunogenic reactions, off-target effects, and a deficiency of effective biomarkers to appreciate the effectiveness of therapy. In this review, we present a summary of the current methods in targeted gene delivery, followed by the limitations and future direction of gene therapy for the cure of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Shalini Mani
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| | - Divya Jindal
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| | - Manisha Singh
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| |
Collapse
|
15
|
Rasmussen CLM, Hede E, Routhe LJ, Körbelin J, Helgudottir SS, Thomsen LB, Schwaninger M, Burkhart A, Moos T. A novel strategy for delivering Niemann-Pick type C2 proteins across the blood-brain barrier using the brain endothelial-specific AAV-BR1 virus. J Neurochem 2023; 164:6-28. [PMID: 35554935 PMCID: PMC10084444 DOI: 10.1111/jnc.15621] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 04/07/2022] [Accepted: 05/03/2022] [Indexed: 02/04/2023]
Abstract
Treating central nervous system (CNS) diseases is complicated by the incapability of numerous therapeutics to cross the blood-brain barrier (BBB), mainly composed of brain endothelial cells (BECs). Genetically modifying BECs into protein factories that supply the CNS with recombinant proteins is a promising approach to overcome this hindrance, especially in genetic diseases, like Niemann Pick disease type C2 (NPC2), where both CNS and peripheral cells are affected. Here, we investigated the potential of the BEC-specific adeno-associated viral vector (AAV-BR1) encoding NPC2 for expression and secretion from primary BECs cultured in an in vitro BBB model with mixed glial cells, and in healthy BALB/c mice. Transduced primary BECs had significantly increased NPC2 gene expression and secreted NPC2 after viral transduction, which significantly reversed cholesterol deposition in NPC2 deficient fibroblasts. Mice receiving an intravenous injection with AAV-BR1-NCP2-eGFP were sacrificed 8 weeks later and examined for its biodistribution and transgene expression of eGFP and NPC2. AAV-BR1-NPC2-eGFP was distributed mainly to the brain and lightly to the heart and lung, but did not label other organs including the liver. eGFP expression was primarily found in BECs throughout the brain but occasionally also in neurons suggesting transport of the vector across the BBB, a phenomenon also confirmed in vitro. NPC2 gene expression was up-regulated in the brain, and recombinant NPC2 protein expression was observed in both transduced brain capillaries and neurons. Our findings show that AAV-BR1 transduction of BECs is possible and that it may denote a promising strategy for future treatment of NPC2.
Collapse
Affiliation(s)
| | - Eva Hede
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Lisa Juul Routhe
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Jakob Körbelin
- Department of Oncology, Hematology and Bone Marrow Transplantation, University Medical Center, Hamburg, Germany
| | - Steinunn Sara Helgudottir
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Louiza Bohn Thomsen
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Annette Burkhart
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Torben Moos
- Neurobiology Research and Drug Delivery, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
16
|
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
|
17
|
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] [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
|
18
|
Jang A, Lehtinen MK. Experimental approaches for manipulating choroid plexus epithelial cells. Fluids Barriers CNS 2022; 19:36. [PMID: 35619113 PMCID: PMC9134666 DOI: 10.1186/s12987-022-00330-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/14/2022] [Indexed: 12/26/2022] Open
Abstract
Choroid plexus (ChP) epithelial cells are crucial for the function of the blood-cerebrospinal fluid barrier (BCSFB) in the developing and mature brain. The ChP is considered the primary source and regulator of CSF, secreting many important factors that nourish the brain. It also performs CSF clearance functions including removing Amyloid beta and potassium. As such, the ChP is a promising target for gene and drug therapy for neurodevelopmental and neurological disorders in the central nervous system (CNS). This review describes the current successful and emerging experimental approaches for targeting ChP epithelial cells. We highlight methodological strategies to specifically target these cells for gain or loss of function in vivo. We cover both genetic models and viral gene delivery systems. Additionally, several lines of reporters to access the ChP epithelia are reviewed. Finally, we discuss exciting new approaches, such as chemical activation and transplantation of engineered ChP epithelial cells. We elaborate on fundamental functions of the ChP in secretion and clearance and outline experimental approaches paving the way to clinical applications.
Collapse
Affiliation(s)
- Ahram Jang
- 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
|
19
|
Fajardo-Serrano A, Rico AJ, Roda E, Honrubia A, Arrieta S, Ariznabarreta G, Chocarro J, Lorenzo-Ramos E, Pejenaute A, Vázquez A, Lanciego JL. Adeno-Associated Viral Vectors as Versatile Tools for Neurological Disorders: Focus on Delivery Routes and Therapeutic Perspectives. Biomedicines 2022; 10:biomedicines10040746. [PMID: 35453499 PMCID: PMC9025350 DOI: 10.3390/biomedicines10040746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/10/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022] Open
Abstract
It is without doubt that the gene therapy field is currently in the spotlight for the development of new therapeutics targeting unmet medical needs. Thus, considering the gene therapy scenario, neurological diseases in general and neurodegenerative disorders in particular are emerging as the most appealing choices for new therapeutic arrivals intended to slow down, stop, or even revert the natural progressive course that characterizes most of these devastating neurodegenerative processes. Since an extensive coverage of all available literature is not feasible in practical terms, here emphasis was made in providing some advice to beginners in the field with a narrow focus on elucidating the best delivery route available for fulfilling any given AAV-based therapeutic approach. Furthermore, it is worth nothing that the number of ongoing clinical trials is increasing at a breath-taking speed. Accordingly, a landscape view of preclinical and clinical initiatives is also provided here in an attempt to best illustrate what is ongoing in this quickly expanding field.
Collapse
Affiliation(s)
- Ana Fajardo-Serrano
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Correspondence: (A.F.-S.); (J.L.L.)
| | - Alberto J. Rico
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Elvira Roda
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Adriana Honrubia
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Sandra Arrieta
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Goiaz Ariznabarreta
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Julia Chocarro
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Elena Lorenzo-Ramos
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Alvaro Pejenaute
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Alfonso Vázquez
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Department of Neurosurgery, Servicio Navarro de Salud, Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
| | - José Luis Lanciego
- Centro de Investigación Médica Aplicada (CIMA), Department of Neuroscience, Universidad de Navarra, 31008 Pamplona, Spain; (A.J.R.); (E.R.); (A.H.); (S.A.); (G.A.); (J.C.); (E.L.-R.); (A.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), 23038 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Correspondence: (A.F.-S.); (J.L.L.)
| |
Collapse
|
20
|
TRAP1 inhibits MIC60 ubiquitination to mitigate the injury of cardiomyocytes and protect mitochondria in extracellular acidosis. Cell Death Dis 2021; 7:389. [PMID: 34907169 PMCID: PMC8671480 DOI: 10.1038/s41420-021-00786-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022]
Abstract
Extracellular acidosis-induced mitochondrial damage of cardiomyocytes leads to cardiac dysfunction, but no detailed mechanism or efficient therapeutic target has been reported. Here we found that the protein levels of MIC60 were decreased in H9C2 cells and heart tissues in extracellular acidosis, which caused mitochondrial damage and cardiac dysfunction. Overexpression of MIC60 maintains H9C2 cells viability, increases ATP production and mitochondrial membrane potential, mitigates the disruptions of mitochondrial structure and cardiac injury. Mechanistically, extracellular acidosis excessively promoted MIC60 ubiquitin-dependent degradation. TRAP1 mitigated acidosis-induced mitochondrial impairments and cardiac injury by directly interacting with MIC60 to decrease its ubiquitin-dependent degradation in extracellular acidosis.
Collapse
|
21
|
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: 3.0] [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
|
22
|
Cui J, Shipley FB, Shannon ML, Alturkistani O, Dani N, Webb MD, Sugden AU, Andermann ML, Lehtinen MK. Inflammation of the Embryonic Choroid Plexus Barrier following Maternal Immune Activation. Dev Cell 2020; 55:617-628.e6. [PMID: 33038331 PMCID: PMC7725967 DOI: 10.1016/j.devcel.2020.09.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/05/2020] [Accepted: 09/18/2020] [Indexed: 01/14/2023]
Abstract
The choroid plexus (ChP) regulates brain development by secreting instructive cues and providing a protective brain barrier. Here, we show that polyI:C-mediated maternal immune activation leads to an inflammatory response in the developing embryonic mouse brain that manifests as pro-inflammatory cerebrospinal fluid (CSF) and accumulation of ChP macrophages. Elevation of CSF-CCL2 was sufficient to drive ChP immune cell recruitment, activation, and proliferation. In addition, ChP macrophages abandoned their regular tiling pattern and relocated to the ChP-free margin where they breached the weakened epithelial barrier. We further found that these immune cells entered from the ChP into the brain via anatomically specialized "hotspots" at the distal tips of ChP villi. In vivo two-photon imaging demonstrated that surveillance behaviors in ChP macrophages had already emerged at this early stage of embryogenesis. Thus, the embryonic ChP forms a functional brain barrier that can mount an inflammatory response to external insults.
Collapse
Affiliation(s)
- Jin Cui
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Frederick B Shipley
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
| | - Morgan L Shannon
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Osama Alturkistani
- IDDRC Cellular Imaging Core, F.M. Kirby Neurobiology Center and Department of Neurology, Boston Children's Hospital, MA, USA
| | - Neil Dani
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Mya D Webb
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
| | - Arthur U Sugden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mark L Andermann
- Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA; Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
23
|
Kaiser K, Bryja V. Choroid Plexus: The Orchestrator of Long-Range Signalling Within the CNS. Int J Mol Sci 2020; 21:E4760. [PMID: 32635478 PMCID: PMC7369786 DOI: 10.3390/ijms21134760] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 06/26/2020] [Accepted: 07/02/2020] [Indexed: 01/24/2023] Open
Abstract
Cerebrospinal fluid (CSF) is the liquid that fills the brain ventricles. CSF represents not only a mechanical brain protection but also a rich source of signalling factors modulating diverse processes during brain development and adulthood. The choroid plexus (CP) is a major source of CSF and as such it has recently emerged as an important mediator of extracellular signalling within the brain. Growing interest in the CP revealed its capacity to release a broad variety of bioactive molecules that, via CSF, regulate processes across the whole central nervous system (CNS). Moreover, CP has been also recognized as a sensor, responding to altered composition of CSF associated with changes in the patterns of CNS activity. In this review, we summarize the recent advances in our understanding of the CP as a signalling centre that mediates long-range communication in the CNS. By providing a detailed account of the CP secretory repertoire, we describe how the CP contributes to the regulation of the extracellular environment-in the context of both the embryonal as well as the adult CNS. We highlight the role of the CP as an important regulator of CNS function that acts via CSF-mediated signalling. Further studies of CP-CSF signalling hold the potential to provide key insights into the biology of the CNS, with implications for better understanding and treatment of neuropathological conditions.
Collapse
Affiliation(s)
- Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| | - Vitezslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, 625 00 Brno, Czech Republic
| |
Collapse
|