1
|
Hashimoto Y, Greene C, Hanley N, Hudson N, Henshall D, Sweeney KJ, O'Brien DF, Campbell M. Pumilio-1 mediated translational control of claudin-5 at the blood-brain barrier. Fluids Barriers CNS 2024; 21:52. [PMID: 38898501 PMCID: PMC11188261 DOI: 10.1186/s12987-024-00553-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Claudin-5 is one of the most essential tight junction proteins at the blood-brain barrier. A single nucleotide polymorphism rs10314 is located in the 3'-untranslated region of claudin-5 and has been shown to be a risk factor for schizophrenia. Here, we show that the pumilio RNA-binding protein, pumilio-1, is responsible for rs10314-mediated claudin-5 regulation. The RNA sequence surrounding rs10314 is highly homologous to the canonical pumilio-binding sequence and claudin-5 mRNA with rs10314 produces 25% less protein due to its inability to bind to pumilio-1. Pumilio-1 formed cytosolic granules under stress conditions and claudin-5 mRNA appeared to preferentially accumulate in these granules. Added to this, we observed granular pumilio-1 in endothelial cells in human brain tissues from patients with psychiatric disorders or epilepsy with increased/accumulated claudin-5 mRNA levels, suggesting translational claudin-5 suppression may occur in a brain-region specific manner. These findings identify a key regulator of claudin-5 translational processing and how its dysregulation may be associated with neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yosuke Hashimoto
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland.
| | - Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Nicole Hanley
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Natalie Hudson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - David Henshall
- Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, FutureNeuro, Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Ireland
- Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | | | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland.
- Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, FutureNeuro, Royal College of Surgeons in Ireland (RCSI), University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
2
|
Dithmer S, Blasig IE, Fraser PA, Qin Z, Haseloff RF. The Basic Requirement of Tight Junction Proteins in Blood-Brain Barrier Function and Their Role in Pathologies. Int J Mol Sci 2024; 25:5601. [PMID: 38891789 PMCID: PMC11172262 DOI: 10.3390/ijms25115601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/10/2024] [Accepted: 03/28/2024] [Indexed: 06/21/2024] Open
Abstract
This review addresses the role of tight junction proteins at the blood-brain barrier (BBB). Their expression is described, and their role in physiological and pathological processes at the BBB is discussed. Based on this, new approaches are depicted for paracellular drug delivery and diagnostics in the treatment of cerebral diseases. Recent data provide convincing evidence that, in addition to its impairment in the course of diseases, the BBB could be involved in the aetiology of CNS disorders. Further progress will be expected based on new insights in tight junction protein structure and in their involvement in signalling pathways.
Collapse
Affiliation(s)
- Sophie Dithmer
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| | - Ingolf E. Blasig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| | | | - Zhihai Qin
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100049, China
| | - Reiner F. Haseloff
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Robert-Rössle-Str. 10, 13125 Berlin, Germany (I.E.B.)
| |
Collapse
|
3
|
Vázquez-Liébanas E, Mocci G, Li W, Laviña B, Reddy A, O'Connor C, Hudson N, Elbeck Z, Nikoloudis I, Gaengel K, Vanlandewijck M, Campbell M, Betsholtz C, Mäe MA. Mosaic deletion of claudin-5 reveals rapid non-cell-autonomous consequences of blood-brain barrier leakage. Cell Rep 2024; 43:113911. [PMID: 38446668 DOI: 10.1016/j.celrep.2024.113911] [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: 07/14/2023] [Revised: 12/19/2023] [Accepted: 02/16/2024] [Indexed: 03/08/2024] Open
Abstract
Claudin-5 (CLDN5) is an endothelial tight junction protein essential for blood-brain barrier (BBB) formation. Abnormal CLDN5 expression is common in brain disease, and knockdown of Cldn5 at the BBB has been proposed to facilitate drug delivery to the brain. To study the consequences of CLDN5 loss in the mature brain, we induced mosaic endothelial-specific Cldn5 gene ablation in adult mice (Cldn5iECKO). These mice displayed increased BBB permeability to tracers up to 10 kDa in size from 6 days post induction (dpi) and ensuing lethality from 10 dpi. Single-cell RNA sequencing at 11 dpi revealed profound transcriptomic differences in brain endothelial cells regardless of their Cldn5 status in mosaic mice, suggesting major non-cell-autonomous responses. Reactive microglia and astrocytes suggested rapid cellular responses to BBB leakage. Our study demonstrates a critical role for CLDN5 in the adult BBB and provides molecular insight into the consequences and risks associated with CLDN5 inhibition.
Collapse
Affiliation(s)
- Elisa Vázquez-Liébanas
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Giuseppe Mocci
- Single Cell Core Facility of Flemingsberg Campus (SICOF), Karolinska Institute, 14157 Huddinge, Sweden
| | - Weihan Li
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Bàrbara Laviña
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Avril Reddy
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Claire O'Connor
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Natalie Hudson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Zaher Elbeck
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Ioannis Nikoloudis
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Konstantin Gaengel
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Michael Vanlandewijck
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden; Single Cell Core Facility of Flemingsberg Campus (SICOF), Karolinska Institute, 14157 Huddinge, Sweden; Department of Medicine, Karolinska Institute, 14157 Huddinge, Sweden
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Christer Betsholtz
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden; Department of Medicine, Karolinska Institute, 14157 Huddinge, Sweden
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| |
Collapse
|
4
|
Markov AG, Bikmurzina AE, Fedorova AA, Vinogradova EP, Kruglova NM, Krivoi II, Amasheh S. Prednisolone Targets Claudins in Mouse Brain Blood Vessels. Int J Mol Sci 2023; 25:276. [PMID: 38203447 PMCID: PMC10779016 DOI: 10.3390/ijms25010276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Endothelial cells in brain capillaries are crucial for the function of the blood-brain barrier (BBB), and members of the tight junction protein family of claudins are regarded to be primarily responsible for barrier properties. Thus, the analysis of bioactive substances that can affect the BBB's permeability is of great importance and may be useful for the development of new therapeutic strategies for brain pathologies. In our study, we tested the hypothesis that the application of the glucocorticoid prednisolone affects the murine blood-brain barrier in vivo. Isolated brain tissue of control and prednisolone-injected mice was examined by employing immunoblotting and confocal laser scanning immunofluorescence microscopy, and the physiological and behavioral effects were analyzed. The control tissue samples revealed the expression of barrier-forming tight junction proteins claudin-1, -3, and -5 and of the paracellular cation and water-channel-forming protein claudin-2. Prednisolone administration for 7 days at doses of 70 mg/kg caused physiological and behavioral effects and downregulated claudin-1 and -3 and the channel-forming claudin-2 without altering their localization in cerebral blood vessels. Changes in the expression of these claudins might have effects on the ionic and acid-base balance in brain tissue, suggesting the relevance of our findings for therapeutic options in disorders such as cerebral edema and psychiatric failure.
Collapse
Affiliation(s)
- Alexander G. Markov
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.G.M.); (A.E.B.); (A.A.F.); (N.M.K.); (I.I.K.)
- Interoception Laboratory, Pavlov Institute of Physiology RAS, 199034 St. Petersburg, Russia
| | - Anastasia E. Bikmurzina
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.G.M.); (A.E.B.); (A.A.F.); (N.M.K.); (I.I.K.)
| | - Arina A. Fedorova
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.G.M.); (A.E.B.); (A.A.F.); (N.M.K.); (I.I.K.)
| | - Ekaterina P. Vinogradova
- Department of Higher Nervous Activity and Psychophysiology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Natalia M. Kruglova
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.G.M.); (A.E.B.); (A.A.F.); (N.M.K.); (I.I.K.)
| | - Igor I. Krivoi
- Department of General Physiology, St. Petersburg State University, 199034 St. Petersburg, Russia; (A.G.M.); (A.E.B.); (A.A.F.); (N.M.K.); (I.I.K.)
| | - Salah Amasheh
- Institute of Veterinary Physiology, Freie Universität Berlin, 14163 Berlin, Germany
| |
Collapse
|
5
|
Bhunia S, Kolishetti N, Vashist A, Yndart Arias A, Brooks D, Nair M. Drug Delivery to the Brain: Recent Advances and Unmet Challenges. Pharmaceutics 2023; 15:2658. [PMID: 38139999 PMCID: PMC10747851 DOI: 10.3390/pharmaceutics15122658] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/02/2023] [Accepted: 11/08/2023] [Indexed: 12/24/2023] Open
Abstract
Brain cancers and neurodegenerative diseases are on the rise, treatments for central nervous system (CNS) diseases remain limited. Despite the significant advancement in drug development technology with emerging biopharmaceuticals like gene therapy or recombinant protein, the clinical translational rate of such biopharmaceuticals to treat CNS disease is extremely poor. The blood-brain barrier (BBB), which separates the brain from blood and protects the CNS microenvironment to maintain essential neuronal functions, poses the greatest challenge for CNS drug delivery. Many strategies have been developed over the years which include local disruption of BBB via physical and chemical methods, and drug transport across BBB via transcytosis by targeting some endogenous proteins expressed on brain-capillary. Drug delivery to brain is an ever-evolving topic, although there were multiple review articles in literature, an update is warranted due to continued growth and new innovations of research on this topic. Thus, this review is an attempt to highlight the recent strategies employed to overcome challenges of CNS drug delivery while emphasizing the necessity of investing more efforts in CNS drug delivery technologies parallel to drug development.
Collapse
Affiliation(s)
- Sukanya Bhunia
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Nagesh Kolishetti
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Arti Vashist
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Adriana Yndart Arias
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Deborah Brooks
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Madhavan Nair
- Department of Immunology and Nano-Medicine, Herbert Wertheim, College of Medicine, Florida International University, Miami, FL 33199, USA
- Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
6
|
O'Brown NM, Patel NB, Hartmann U, Klein AM, Gu C, Megason SG. The secreted neuronal signal Spock1 promotes blood-brain barrier development. Dev Cell 2023; 58:1534-1547.e6. [PMID: 37437574 PMCID: PMC10525910 DOI: 10.1016/j.devcel.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/07/2023] [Accepted: 06/16/2023] [Indexed: 07/14/2023]
Abstract
The blood-brain barrier (BBB) is a unique set of properties of the brain vasculature which severely restrict its permeability to proteins and small molecules. Classic chick-quail chimera studies have shown that these properties are not intrinsic to the brain vasculature but rather are induced by surrounding neural tissue. Here, we identify Spock1 as a candidate neuronal signal for regulating BBB permeability in zebrafish and mice. Mosaic genetic analysis shows that neuronally expressed Spock1 is cell non-autonomously required for a functional BBB. Leakage in spock1 mutants is associated with altered extracellular matrix (ECM), increased endothelial transcytosis, and altered pericyte-endothelial interactions. Furthermore, a single dose of recombinant SPOCK1 partially restores BBB function in spock1 mutants by quenching gelatinase activity and restoring vascular expression of BBB genes including mcamb. These analyses support a model in which neuronally secreted Spock1 initiates BBB properties by altering the ECM, thereby regulating pericyte-endothelial interactions and downstream vascular gene expression.
Collapse
Affiliation(s)
- Natasha M O'Brown
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA.
| | - Nikit B Patel
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | - Ursula Hartmann
- Center for Biochemistry, Medical Faculty, University of Cologne, Joseph-Stelzmann-Str. 52, 50931 Cologne, Germany
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | - Chenghua Gu
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, 220 Longwood Ave, Boston, MA 02115, USA
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Cai Q, Li X, Xiong H, Fan H, Gao X, Vemireddy V, Margolis R, Li J, Ge X, Giannotta M, Hoyt K, Maher E, Bachoo R, Qin Z. Optical blood-brain-tumor barrier modulation expands therapeutic options for glioblastoma treatment. Nat Commun 2023; 14:4934. [PMID: 37582846 PMCID: PMC10427669 DOI: 10.1038/s41467-023-40579-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 07/31/2023] [Indexed: 08/17/2023] Open
Abstract
The treatment of glioblastoma has limited clinical progress over the past decade, partly due to the lack of effective drug delivery strategies across the blood-brain-tumor barrier. Moreover, discrepancies between preclinical and clinical outcomes demand a reliable translational platform that can precisely recapitulate the characteristics of human glioblastoma. Here we analyze the intratumoral blood-brain-tumor barrier heterogeneity in human glioblastoma and characterize two genetically engineered models in female mice that recapitulate two important glioma phenotypes, including the diffusely infiltrative tumor margin and angiogenic core. We show that pulsed laser excitation of vascular-targeted gold nanoparticles non-invasively and reversibly modulates the blood-brain-tumor barrier permeability (optoBBTB) and enhances the delivery of paclitaxel in these two models. The treatment reduces the tumor volume by 6 and 2.4-fold and prolongs the survival by 50% and 33%, respectively. Since paclitaxel does not penetrate the blood-brain-tumor barrier and is abandoned for glioblastoma treatment following its failure in early-phase clinical trials, our results raise the possibility of reevaluating a number of potent anticancer drugs by combining them with strategies to increase blood-brain-tumor barrier permeability. Our study reveals that optoBBTB significantly improves therapeutic delivery and has the potential to facilitate future drug evaluation for cancers in the central nervous system.
Collapse
Affiliation(s)
- Qi Cai
- Department of Mechanical Engineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Xiaoqing Li
- Department of Bioengineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Hejian Xiong
- Department of Mechanical Engineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Hanwen Fan
- Department of Mechanical Engineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Xiaofei Gao
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Vamsidhara Vemireddy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ryan Margolis
- Department of Bioengineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Junjie Li
- Department of Bioengineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Xiaoqian Ge
- Department of Mechanical Engineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Monica Giannotta
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Kenneth Hoyt
- Department of Bioengineering, the University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Elizabeth Maher
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Robert Bachoo
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Zhenpeng Qin
- Department of Mechanical Engineering, the University of Texas at Dallas, Richardson, TX, 75080, USA.
- Department of Bioengineering, the University of Texas at Dallas, Richardson, TX, 75080, USA.
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Center for Advanced Pain Studies, the University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
8
|
Hashimoto Y, Greene C, Munnich A, Campbell M. The CLDN5 gene at the blood-brain barrier in health and disease. Fluids Barriers CNS 2023; 20:22. [PMID: 36978081 PMCID: PMC10044825 DOI: 10.1186/s12987-023-00424-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
The CLDN5 gene encodes claudin-5 (CLDN-5) that is expressed in endothelial cells and forms tight junctions which limit the passive diffusions of ions and solutes. The blood-brain barrier (BBB), composed of brain microvascular endothelial cells and associated pericytes and end-feet of astrocytes, is a physical and biological barrier to maintain the brain microenvironment. The expression of CLDN-5 is tightly regulated in the BBB by other junctional proteins in endothelial cells and by supports from pericytes and astrocytes. The most recent literature clearly shows a compromised BBB with a decline in CLDN-5 expression increasing the risks of developing neuropsychiatric disorders, epilepsy, brain calcification and dementia. The purpose of this review is to summarize the known diseases associated with CLDN-5 expression and function. In the first part of this review, we highlight the recent understanding of how other junctional proteins as well as pericytes and astrocytes maintain CLDN-5 expression in brain endothelial cells. We detail some drugs that can enhance these supports and are being developed or currently in use to treat diseases associated with CLDN-5 decline. We then summarise mutagenesis-based studies which have facilitated a better understanding of the physiological role of the CLDN-5 protein at the BBB and have demonstrated the functional consequences of a recently identified pathogenic CLDN-5 missense mutation from patients with alternating hemiplegia of childhood. This mutation is the first gain-of-function mutation identified in the CLDN gene family with all others representing loss-of-function mutations resulting in mis-localization of CLDN protein and/or attenuated barrier function. Finally, we summarize recent reports about the dosage-dependent effect of CLDN-5 expression on the development of neurological diseases in mice and discuss what cellular supports for CLDN-5 regulation are compromised in the BBB in human diseases.
Collapse
Affiliation(s)
- Yosuke Hashimoto
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| | - Chris Greene
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland
| | - Arnold Munnich
- Institut Imagine, INSERM UMR1163, Université Paris Cité, Paris, F-75015, France
- Departments of Pediatric Neurology and Medical Genetics, Hospital Necker Enfants Malades, Université Paris Cité, Paris, F-75015, France
| | - Matthew Campbell
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin, D02 VF25, Ireland.
| |
Collapse
|
9
|
Rada CC, Yuki K, Ding J, Kuo CJ. Regulation of the Blood-Brain Barrier in Health and Disease. Cold Spring Harb Perspect Med 2023; 13:a041191. [PMID: 36987582 PMCID: PMC10691497 DOI: 10.1101/cshperspect.a041191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
The neurovascular unit is a dynamic microenvironment with tightly controlled signaling and transport coordinated by the blood-brain barrier (BBB). A properly functioning BBB allows sufficient movement of ions and macromolecules to meet the high metabolic demand of the central nervous system (CNS), while protecting the brain from pathogenic and noxious insults. This review describes the main cell types comprising the BBB and unique molecular signatures of these cells. Additionally, major signaling pathways for BBB development and maintenance are highlighted. Finally, we describe the pathophysiology of BBB diseases, their relationship to barrier dysfunction, and identify avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Cara C Rada
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Kanako Yuki
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Jie Ding
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
10
|
Opportunities and challenges in delivering biologics for Alzheimer's disease by low-intensity ultrasound. Adv Drug Deliv Rev 2022; 189:114517. [PMID: 36030018 DOI: 10.1016/j.addr.2022.114517] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/24/2023]
Abstract
Low-intensity ultrasound combined with intravenously injected microbubbles (US+MB) is a novel treatment modality for brain disorders, including Alzheimer's disease (AD), safely and transiently allowing therapeutic agents to overcome the blood-brain barrier (BBB) that constitutes a major barrier for therapeutic agents. Here, we first provide an update on immunotherapies in AD and how US+MB has been applied to AD mouse models and in clinical trials, considering the ultrasound and microbubble parameter space. In the second half of the review, we compare different in vitro BBB models and discuss strategies for combining US+MB with BBB modulators (targeting molecules such as claudin-5), and highlight the insight provided by super-resolution microscopy. Finally, we conclude with a short discussion on how in vitro findings can inform the design of animal studies, and how the insight gained may aid treatment optimization in the clinical ultrasound space.
Collapse
|
11
|
Zhang S, Gan L, Cao F, Wang H, Gong P, Ma C, Ren L, Lin Y, Lin X. The barrier and interface mechanisms of the brain barrier, and brain drug delivery. Brain Res Bull 2022; 190:69-83. [PMID: 36162603 DOI: 10.1016/j.brainresbull.2022.09.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/25/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Three different barriers are formed between the cerebrovascular and the brain parenchyma: the blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the cerebrospinal fluid-brain barrier (CBB). The BBB is the main regulator of blood and central nervous system (CNS) material exchange. The semipermeable nature of the BBB limits the passage of larger molecules and hydrophilic small molecules, Food and Drug Administration (FDA)-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Although the complexity of the BBB affects CNS drug delivery, understanding the composition and function of the BBB can provide a platform for the development of new methods for CNS drug delivery. This review summarizes the classification of the brain barrier, the composition and role of the basic structures of the BBB, and the transport, barrier, and destruction mechanisms of the BBB; discusses the advantages and disadvantages of different drug delivery methods and prospects for future drug delivery strategies.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| | - Lin Gan
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Fengye Cao
- Yiyang The First Hospital of Traditional Chinese Medicine, Yiyang, Hunan Province, 413000, China
| | - Hao Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Peng Gong
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Congcong Ma
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Li Ren
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Xianming Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China.
| |
Collapse
|
12
|
Wakayama E, Kuzu T, Tachibana K, Hirayama R, Okada Y, Kondoh M. Modifying the blood-brain barrier by targeting claudin-5: Safety and risks. Ann N Y Acad Sci 2022; 1514:62-69. [PMID: 35508916 DOI: 10.1111/nyas.14787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The blood-brain barrier is a major obstacle to the delivery of drugs to the central nervous system. In the blood-brain barrier, the spaces between adjacent brain microvascular endothelial cells are sealed by multiprotein complexes known as tight junctions. Among the many components of the tight junction, claudin-5 has received the most attention as a target for loosening the tight-junction seal and allowing drugs to be delivered to the brain. In mice, transient knockdown of claudin-5 and the use of claudin-5 binders have been shown to enhance the permeation of small molecules from the blood into the brain without apparent adverse effects. However, sustained knockdown of claudin-5 in mice is lethal within 40 days, and administration of an anti-claudin-5 antibody induced convulsions in a nonhuman primate. Here, we review the safety concerns of claudin-5-targeted technologies with respect to their clinical application.
Collapse
Affiliation(s)
- Erika Wakayama
- Faculty of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Taiki Kuzu
- College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Japan.,Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Keisuke Tachibana
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | | | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
13
|
Modulation of the Blood-Brain Barrier for Drug Delivery to Brain. Pharmaceutics 2021; 13:pharmaceutics13122024. [PMID: 34959306 PMCID: PMC8708282 DOI: 10.3390/pharmaceutics13122024] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/15/2021] [Accepted: 11/25/2021] [Indexed: 12/26/2022] Open
Abstract
The blood-brain barrier (BBB) precisely controls brain microenvironment and neural activity by regulating substance transport into and out of the brain. However, it severely hinders drug entry into the brain, and the efficiency of various systemic therapies against brain diseases. Modulation of the BBB via opening tight junctions, inhibiting active efflux and/or enhancing transcytosis, possesses the potential to increase BBB permeability and improve intracranial drug concentrations and systemic therapeutic efficiency. Various strategies of BBB modulation have been reported and investigated preclinically and/or clinically. This review describes conventional and emerging BBB modulation strategies and related mechanisms, and safety issues according to BBB structures and functions, to try to give more promising directions for designing more reasonable preclinical and clinical studies.
Collapse
|
14
|
Targeting tight junctions to fight against viral neuroinvasion. Trends Mol Med 2021; 28:12-24. [PMID: 34810086 DOI: 10.1016/j.molmed.2021.10.007] [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: 09/10/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 11/23/2022]
Abstract
The clinical impact of viral neuroinvasion on the central nervous system (CNS) ranges from barely detectable to deadly, including acute and chronic outcomes. Developing innovative therapeutic strategies is important to mitigate virus-induced neurological and psychiatric disorders. A key gatekeeper to the CNS is the neurovascular unit (NVU), a major obstacle to viral neuroinvasion and antiviral therapies. The NVU isolates the brain from the blood through firm sealing operated by the tight junctions (TJs) of endothelial cells. Here, we make the thought-provoking assumption that TJs can be targets to prevent or treat viral neuroinvasion and resulting disorders. This review aims at defining the conceptual diverse mode of actions of such approaches, evaluates their feasibility, and discusses future challenges in the field.
Collapse
|
15
|
Whelan R, Hargaden GC, Knox AJS. Modulating the Blood-Brain Barrier: A Comprehensive Review. Pharmaceutics 2021; 13:1980. [PMID: 34834395 PMCID: PMC8618722 DOI: 10.3390/pharmaceutics13111980] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 12/23/2022] Open
Abstract
The highly secure blood-brain barrier (BBB) restricts drug access to the brain, limiting the molecular toolkit for treating central nervous system (CNS) diseases to small, lipophilic drugs. Development of a safe and effective BBB modulator would revolutionise the treatment of CNS diseases and future drug development in the area. Naturally, the field has garnered a great deal of attention, leading to a vast and diverse range of BBB modulators. In this review, we summarise and compare the various classes of BBB modulators developed over the last five decades-their recent advancements, advantages and disadvantages, while providing some insight into their future as BBB modulators.
Collapse
Affiliation(s)
- Rory Whelan
- School of Biological and Health Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
- Chemical and Structural Biology, Environmental Sustainability and Health Institute, Technological University Dublin, D07 H6K8 Dublin, Ireland
| | - Grainne C. Hargaden
- School of Chemical and Pharmaceutical Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
| | - Andrew J. S. Knox
- School of Biological and Health Sciences, Technological University Dublin, Central Quad, Grangegorman, D07 XT95 Dublin, Ireland;
- Chemical and Structural Biology, Environmental Sustainability and Health Institute, Technological University Dublin, D07 H6K8 Dublin, Ireland
| |
Collapse
|
16
|
Hashimoto Y, Campbell M, Tachibana K, Okada Y, Kondoh M. Claudin-5: A Pharmacological Target to Modify the Permeability of the Blood-Brain Barrier. Biol Pharm Bull 2021; 44:1380-1390. [PMID: 34602546 DOI: 10.1248/bpb.b21-00408] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Claudin-5 is the dominant tight junction protein in brain endothelial cells and exclusively limits the paracellular permeability of molecules larger than 400 Da across the blood-brain barrier (BBB). Its pathological impairment or sustained down-regulation has been shown to lead to the progression of psychiatric and neurological disorders, whereas its expression under physiological conditions prevents the passage of drugs across the BBB. While claudin-5 enhancers could potentially act as vascular stabilizers to treat neurological diseases, claudin-5 inhibitors could function as delivery systems to enhance the brain uptake of hydrophilic small-molecular-weight drugs. Therefore, the effects of claudin-5 manipulation on modulating the BBB in different neurological diseases requires further examination. To manipulate claudin-5 expression levels and function, several claudin-5 modulating molecules have been developed. In this review, we first describe the molecular, cellular and pathological aspects of claudin-5 to highlight the mechanisms of claudin-5 enhancers/inhibitors. We then discuss recently developed claudin-5 enhancers/inhibitors and new methods to discover these molecules.
Collapse
Affiliation(s)
| | | | | | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
17
|
Breitkreuz-Korff O, Tscheik C, Del Vecchio G, Dithmer S, Walther W, Orthmann A, Wolburg H, Haseloff RF, Schröder L, Blasig IE, Winkler L. M01 as a novel drug enhancer for specifically targeting the blood-brain barrier. J Control Release 2021; 338:137-148. [PMID: 34384796 DOI: 10.1016/j.jconrel.2021.08.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 01/17/2023]
Abstract
Drug delivery to the brain is limited for most pharmaceuticals by the blood-brain barrier (BBB) where claudin-5 dominates the paraendothelial tightening. For circumventing the BBB, we identified the compound M01 as a claudin-5 interaction inhibitor. M01 causes transient permeabilisation of the BBB depending on the concentration of small molecules in different cell culture models within 3 to 48 h. In mice, brain uptake of fluorescein peaked within the first 3 h after M01 injection and normalised within 48 h. Compared to the cytostatic paclitaxel alone, M01 improved delivery of paclitaxel to mouse brain and reduced orthotopic glioblastoma growth. Results on interactions of M01 with claudin-5 were incorporated into a binding model which suggests association of its aromatic parts with highly conserved residues of the extracellular domain of claudin-5 and adjacent transmembrane segments. Our results indicate the following mode of action: M01 preferentially binds to the extracellular claudin-5 domain, which weakens trans-interactions between adhering cells. Further decrease in membranous claudin-5 levels due to internalization and transcriptional downregulation enables the paracellular passage of small molecules. In summary, the first small molecule is introduced here as a drug enhancer, which specifically permeabilises the BBB for a sufficient interval for allowing neuropharmaceuticals to enter the brain.
Collapse
Affiliation(s)
| | - Christian Tscheik
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Sophie Dithmer
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Wolfgang Walther
- Experimental and Clinical Research Center, Charité Universitätsmedizin, Berlin, Germany
| | - Andrea Orthmann
- Experimentelle Pharmakologie und Onkologie Berlin-Buch GmbH, Germany
| | | | - Reiner F Haseloff
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Leif Schröder
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ingolf E Blasig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany.
| | - Lars Winkler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany; Experimentelle Pharmakologie und Onkologie Berlin-Buch GmbH, Germany.
| |
Collapse
|
18
|
Scalise AA, Kakogiannos N, Zanardi F, Iannelli F, Giannotta M. The blood-brain and gut-vascular barriers: from the perspective of claudins. Tissue Barriers 2021; 9:1926190. [PMID: 34152937 PMCID: PMC8489939 DOI: 10.1080/21688370.2021.1926190] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In some organs, such as the brain, endothelial cells form a robust and highly selective blood-to-tissue barrier. However, in other organs, such as the intestine, endothelial cells provide less stringent permeability, to allow rapid exchange of solutes and nutrients where needed. To maintain the structural and functional integrity of the highly dynamic blood–brain and gut–vascular barriers, endothelial cells form highly specialized cell-cell junctions, known as adherens junctions and tight junctions. Claudins are a family of four-membrane-spanning proteins at tight junctions and they have both barrier-forming and pore-forming properties. Tissue-specific expression of claudins has been linked to different diseases that are characterized by barrier impairment. In this review, we summarize the more recent progress in the field of the claudins, with particular attention to their expression and function in the blood–brain barrier and the recently described gut–vascular barrier, under physiological and pathological conditions. Abbreviations: 22q11DS 22q11 deletion syndrome; ACKR1 atypical chemokine receptor 1; AD Alzheimer disease; AQP aquaporin; ATP adenosine triphosphate; Aβ amyloid β; BAC bacterial artificial chromosome; BBB blood-brain barrier; C/EBP-α CCAAT/enhancer-binding protein α; cAMP cyclic adenosine monophosphate (or 3ʹ,5ʹ-cyclic adenosine monophosphate); CD cluster of differentiation; CNS central nervous system; DSRED discosoma red; EAE experimental autoimmune encephalomyelitis; ECV304 immortalized endothelial cell line established from the vein of an apparently normal human umbilical cord; EGFP enhanced green fluorescent protein; ESAM endothelial cell-selective adhesion molecule; GLUT-1 glucose transporter 1; GVB gut-vascular barrier; H2B histone H2B; HAPP human amyloid precursor protein; HEK human embryonic kidney; JACOP junction-associated coiled coil protein; JAM junctional adhesion molecules; LYVE1 lymphatic vessel endothelial hyaluronan receptor 1; MADCAM1 mucosal vascular addressin cell adhesion molecule 1; MAPK mitogen-activated protein kinase; MCAO middle cerebral artery occlusion; MMP metalloprotease; MS multiple sclerosis; MUPP multi-PDZ domain protein; PATJ PALS-1-associated tight junction protein; PDGFR-α platelet-derived growth factor receptor α polypeptide; PDGFR-β platelet-derived growth factor receptor β polypeptide; RHO rho-associated protein kinase; ROCK rho-associated, coiled-coil-containing protein kinase; RT-qPCR real time quantitative polymerase chain reactions; PDGFR-β soluble platelet-derived growth factor receptor, β polypeptide; T24 human urinary bladder carcinoma cells; TG2576 transgenic mice expressing the human amyloid precursor protein; TNF-α tumor necrosis factor α; WTwild-type; ZO zonula occludens.
Collapse
|
19
|
Tachibana K, Hashimoto Y, Shirakura K, Okada Y, Hirayama R, Iwashita Y, Nishino I, Ago Y, Takeda H, Kuniyasu H, Kondoh M. Safety and efficacy of an anti-claudin-5 monoclonal antibody to increase blood-brain barrier permeability for drug delivery to the brain in a non-human primate. J Control Release 2021; 336:105-111. [PMID: 34118338 DOI: 10.1016/j.jconrel.2021.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/26/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022]
Abstract
Claudin-5 (CLDN-5) is an essential component of the tight junction seal in the blood-brain barrier. Previously, we showed that CLDN-5 modulation in vitro via an anti-CLDN-5 monoclonal antibody (mAb) may be useful for increasing the permeability of the blood-brain barrier for drug delivery to the brain. Based on these findings, here we examined the safety and efficacy of the anti-CLDN-5 mAb in a non-human primate. Cynomolgus monkeys were intravenously administered the anti-CLDN-5 mAb followed by fluorescein dye (376 Da), and the concentrations of the dye in the cerebrospinal fluid was examined. When the mAb was administered at 3.0 mg/kg, the concentration of dye in the cerebrospinal fluid was increased, and no behavioral changes or changes in plasma biomarkers for inflammation or liver or kidney injury were observed. However, a monkey that received the mAb at 6 mg/kg experienced convulsions, and subsequent histopathological examination of this animal revealed vasodilation in the liver, lung, and kidney; hemorrhage in the lung; and edema in the brain. Together, our data indicate that CLDN-5 might be a potential target for enhancing drug delivery to the brain, but also that the therapeutic window of the anti-CLDN-5 mAb may be narrow for separating efficacy and toxicity.
Collapse
Affiliation(s)
- Keisuke Tachibana
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.
| | - Yosuke Hashimoto
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Keisuke Shirakura
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Ryuichi Hirayama
- Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yumi Iwashita
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Itsuki Nishino
- Faculty of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
| | - Yukio Ago
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Hiroyuki Takeda
- Proteo-Science Center, Ehime University, Ehime 790-8577, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, Nara Medical University, Nara 634-8521, Japan
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
20
|
Brunner J, Ragupathy S, Borchard G. Target specific tight junction modulators. Adv Drug Deliv Rev 2021; 171:266-288. [PMID: 33617902 DOI: 10.1016/j.addr.2021.02.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Intercellular tight junctions represent a formidable barrier against paracellular drug absorption at epithelia (e.g., nasal, intestinal) and the endothelium (e.g., blood-brain barrier). In order to enhance paracellular transport of drugs and increase their bioavailability and organ deposition, active excipients modulating tight junctions have been applied. First-generation of permeation enhancers (PEs) acted by unspecific interactions, while recently developed PEs address specific physiological mechanisms. Such target specific tight junction modulators (TJMs) have the advantage of a defined specific mechanism of action. To date, merely a few of these novel active excipients has entered into clinical trials, as their lack in safety and efficiency in vivo often impedes their commercialisation. A stronger focus on the development of such active excipients would result in an economic and therapeutic improvement of current and future drugs.
Collapse
Affiliation(s)
- Joël Brunner
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Sakthikumar Ragupathy
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Gerrit Borchard
- Section of Pharmaceutical Sciences, Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
21
|
Tight Junction Modulating Bioprobes for Drug Delivery System to the Brain: A Review. Pharmaceutics 2020; 12:pharmaceutics12121236. [PMID: 33352631 PMCID: PMC7767277 DOI: 10.3390/pharmaceutics12121236] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
The blood-brain barrier (BBB), which is composed of endothelial cells, pericytes, astrocytes, and neurons, separates the brain extracellular fluid from the circulating blood, and maintains the homeostasis of the central nervous system (CNS). The BBB endothelial cells have well-developed tight junctions (TJs) and express specific polarized transport systems to tightly control the paracellular movements of solutes, ions, and water. There are two types of TJs: bicellular TJs (bTJs), which is a structure at the contact of two cells, and tricellular TJs (tTJs), which is a structure at the contact of three cells. Claudin-5 and angulin-1 are important components of bTJs and tTJs in the brain, respectively. Here, we review TJ-modulating bioprobes that enable drug delivery to the brain across the BBB, focusing on claudin-5 and angulin-1.
Collapse
|
22
|
Castañeda-Cabral JL, Colunga-Durán A, Ureña-Guerrero ME, Beas-Zárate C, Nuñez-Lumbreras MDLA, Orozco-Suárez S, Alonso-Vanegas M, Guevara-Guzmán R, Deli MA, Valle-Dorado MG, Sánchez-Valle V, Rocha L. Expression of VEGF- and tight junction-related proteins in the neocortical microvasculature of patients with drug-resistant temporal lobe epilepsy. Microvasc Res 2020; 132:104059. [PMID: 32798551 DOI: 10.1016/j.mvr.2020.104059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/30/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) maintains the optimal microenvironment for brain function. Tight junctions (TJs) allow endothelial cells to adhere to each other, leading to the formation of a barrier that prevents the penetration of most molecules via transcellular routes. Evidence has indicated that seizure-induced vascular endothelial growth factor (VEGF) type 2 receptor (VEGFR-2) pathway activation weakens TJs, inducing vasodilatation and increasing vascular permeability and subsequent brain injury. The present study focused on investigating the expression levels of VEGF-related (VEGF-A and VEGFR-2) and TJ-related proteins (claudin-5, occludin and ZO-1) in the neocortical microvasculature of patients with drug-resistant temporal lobe epilepsy (TLE). The results obtained from hippocampal sclerosis TLE (HS-TLE) patients were compared with those obtained from patients with TLE secondary to lesions (lesion-TLE) and autopsy samples. The Western blotting and immunofluorescence results showed that VEGF-A and VEGFR-2 protein expression levels were increased in HS-TLE and lesion-TLE patients compared to autopsy group. On the other hand, claudin-5 expression was higher in HS-TLE patients and lesion-TLE patients than autopsies. The expression level of occludin and ZO-1 was decreased in HS-TLE patients. Our study described modifications to the integrity of the BBB that may contribute to the pathogenesis of TLE, in which the VEGF system may play an important role. We demonstrated that the same modifications were present in both HS-TLE and lesion-TLE patients, which suggests that seizures modify these systems and that they are not associated with the establishment of epilepsy.
Collapse
Affiliation(s)
- José Luis Castañeda-Cabral
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav) Sede Sur, Ciudad de México, Mexico; Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Mexico.
| | - Adacrid Colunga-Durán
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav) Sede Sur, Ciudad de México, Mexico
| | - Mónica E Ureña-Guerrero
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Mexico
| | - Carlos Beas-Zárate
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Mexico
| | - Maria de Los Angeles Nuñez-Lumbreras
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav) Sede Sur, Ciudad de México, Mexico
| | - Sandra Orozco-Suárez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, IMSS, Ciudad de México, Mexico
| | - Mario Alonso-Vanegas
- Servicio de Neurocirugía, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez" (INNNMVS), Ciudad de México, Mexico
| | - Rosalinda Guevara-Guzmán
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - María Guadalupe Valle-Dorado
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav) Sede Sur, Ciudad de México, Mexico
| | | | - Luisa Rocha
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav) Sede Sur, Ciudad de México, Mexico
| |
Collapse
|
23
|
Lochhead JJ, Yang J, Ronaldson PT, Davis TP. Structure, Function, and Regulation of the Blood-Brain Barrier Tight Junction in Central Nervous System Disorders. Front Physiol 2020; 11:914. [PMID: 32848858 PMCID: PMC7424030 DOI: 10.3389/fphys.2020.00914] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
The blood-brain barrier (BBB) allows the brain to selectively import nutrients and energy critical to neuronal function while simultaneously excluding neurotoxic substances from the peripheral circulation. In contrast to the highly permeable vasculature present in most organs that reside outside of the central nervous system (CNS), the BBB exhibits a high transendothelial electrical resistance (TEER) along with a low rate of transcytosis and greatly restricted paracellular permeability. The property of low paracellular permeability is controlled by tight junction (TJ) protein complexes that seal the paracellular route between apposing brain microvascular endothelial cells. Although tight junction protein complexes are principal contributors to physical barrier properties, they are not static in nature. Rather, tight junction protein complexes are highly dynamic structures, where expression and/or localization of individual constituent proteins can be modified in response to pathophysiological stressors. These stressors induce modifications to tight junction protein complexes that involve de novo synthesis of new protein or discrete trafficking mechanisms. Such responsiveness of BBB tight junctions to diseases indicates that these protein complexes are critical for maintenance of CNS homeostasis. In fulfillment of this vital role, BBB tight junctions are also a major obstacle to therapeutic drug delivery to the brain. There is an opportunity to overcome this substantial obstacle and optimize neuropharmacology via acquisition of a detailed understanding of BBB tight junction structure, function, and regulation. In this review, we discuss physiological characteristics of tight junction protein complexes and how these properties regulate delivery of therapeutics to the CNS for treatment of neurological diseases. Specifically, we will discuss modulation of tight junction structure, function, and regulation both in the context of disease states and in the setting of pharmacotherapy. In particular, we will highlight how these properties can be potentially manipulated at the molecular level to increase CNS drug levels via paracellular transport to the brain.
Collapse
|
24
|
Hashimoto Y, Tachibana K, Kondoh M. Tight junction modulators for drug delivery to the central nervous system. Drug Discov Today 2020; 25:1477-1486. [DOI: 10.1016/j.drudis.2020.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/30/2020] [Accepted: 05/10/2020] [Indexed: 12/21/2022]
|
25
|
Hashimoto Y, Campbell M. Tight junction modulation at the blood-brain barrier: Current and future perspectives. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183298. [PMID: 32353377 DOI: 10.1016/j.bbamem.2020.183298] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/09/2020] [Accepted: 03/28/2020] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) is the one of the most robust physical barriers in the body, comprised of tight junction (TJ) proteins in brain microvascular endothelial cells. The need for drugs to treat central nervous systems diseases is ever increasing, however the presence of the BBB significantly hampers the uptake of drugs into the brain. To overcome or circumvent the barrier, many kinds of techniques are being developed. Modulating the paracellular route by disruption of the TJ complex has been proposed as a potential drug delivery system to treat brain diseases, however, it has several limitations and is still in a developmental stage. However, recent significant advance in medical equipment /tools such as targeted ultra-sound technologies may resolve these limitations. In this review, we introduce recent advances in site- or molecular size-selective BBB disruption/modulation technologies and we include details on pharmacological inhibitory molecules against intercellular TJ proteins to modulate the BBB.
Collapse
Affiliation(s)
- Yosuke Hashimoto
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin 2, Ireland.
| | - Matthew Campbell
- Trinity College Dublin, Smurfit Institute of Genetics, Dublin 2, Ireland.
| |
Collapse
|
26
|
O'Brown NM, Megason SG, Gu C. Suppression of transcytosis regulates zebrafish blood-brain barrier function. eLife 2019; 8:e47326. [PMID: 31429822 PMCID: PMC6726461 DOI: 10.7554/elife.47326] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
As an optically transparent model organism with an endothelial blood-brain barrier (BBB), zebrafish offer a powerful tool to study the vertebrate BBB. However, the precise developmental profile of functional zebrafish BBB acquisition and the subcellular and molecular mechanisms governing the zebrafish BBB remain poorly characterized. Here, we capture the dynamics of developmental BBB leakage using live imaging, revealing a combination of steady accumulation in the parenchyma and sporadic bursts of tracer leakage. Electron microscopy studies further reveal high levels of transcytosis in brain endothelium early in development that are suppressed later. The timing of this suppression of transcytosis coincides with the establishment of BBB function. Finally, we demonstrate a key mammalian BBB regulator Mfsd2a, which inhibits transcytosis, plays a conserved role in zebrafish, as mfsd2aa mutants display increased BBB permeability due to increased transcytosis. Our findings indicate a conserved developmental program of barrier acquisition between zebrafish and mice.
Collapse
Affiliation(s)
| | - Sean G Megason
- Department of Systems BiologyHarvard Medical SchoolBostonUnited States
| | - Chenghua Gu
- Department of NeurobiologyHarvard Medical SchoolBostonUnited States
| |
Collapse
|
27
|
Hashimoto Y, Okada Y, Shirakura K, Tachibana K, Sawada M, Yagi K, Doi T, Kondoh M. Anti-Claudin Antibodies as a Concept for Development of Claudin-Directed Drugs. J Pharmacol Exp Ther 2018; 368:179-186. [DOI: 10.1124/jpet.118.252361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/06/2018] [Indexed: 01/17/2023] Open
|
28
|
Keep RF, Andjelkovic AV, Xiang J, Stamatovic SM, Antonetti DA, Hua Y, Xi G. Brain endothelial cell junctions after cerebral hemorrhage: Changes, mechanisms and therapeutic targets. J Cereb Blood Flow Metab 2018; 38:1255-1275. [PMID: 29737222 PMCID: PMC6092767 DOI: 10.1177/0271678x18774666] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/09/2018] [Indexed: 11/15/2022]
Abstract
Vascular disruption is the underlying cause of cerebral hemorrhage, including intracerebral, subarachnoid and intraventricular hemorrhage. The disease etiology also involves cerebral hemorrhage-induced blood-brain barrier (BBB) disruption, which contributes an important component to brain injury after the initial cerebral hemorrhage. BBB loss drives vasogenic edema, allows leukocyte extravasation and may lead to the entry of potentially neurotoxic and vasoactive compounds into brain. This review summarizes current information on changes in brain endothelial junction proteins in response to cerebral hemorrhage (and clot-related factors), the mechanisms underlying junction modification and potential therapeutic targets to limit BBB disruption and, potentially, hemorrhage occurrence. It also addresses advances in the tools that are now available for assessing changes in junctions after cerebral hemorrhage and the potential importance of such junction changes. Recent studies suggest post-translational modification, conformational change and intracellular trafficking of junctional proteins may alter barrier properties. Understanding how cerebral hemorrhage alters BBB properties beyond changes in tight junction protein loss may provide important therapeutic insights to prevent BBB dysfunction and restore normal function.
Collapse
Affiliation(s)
- Richard F Keep
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, MI, USA
| | - Anuska V Andjelkovic
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Ann Arbor, MI, USA
| | - Jianming Xiang
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| | | | - David A Antonetti
- Department of Molecular and Integrative Physiology, University of Michigan Ann Arbor, MI, USA
- Department of Ophthalmology & Visual Science Medical School, University of Michigan Ann Arbor, MI, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan Ann Arbor, MI, USA
| |
Collapse
|
29
|
Engineered membrane protein antigens successfully induce antibodies against extracellular regions of claudin-5. Sci Rep 2018; 8:8383. [PMID: 29849184 PMCID: PMC5976803 DOI: 10.1038/s41598-018-26560-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/15/2018] [Indexed: 01/09/2023] Open
Abstract
The production of antibodies against the extracellular regions (ECR) of multispanning membrane proteins is notoriously difficult because of the low productivity and immunogenicity of membrane proteins due to their complex structure and highly conserved sequences among species. Here, we introduce a new method to generate ECR-binding antibodies utilizing engineered liposomal immunogen prepared using a wheat cell-free protein synthesis system. We used claudin-5 (CLDN-5) as the target antigen, which is a notoriously difficult to produce and poorly immunogenic membrane protein with two highly conserved extracellular loops. We drastically improved the productivity of CLDN-5 in the cell-free system after suppressing and normalizing mRNA GC content. To overcome its low immunogenicity, two engineered antigens were designed and synthesized as proteoliposomes: a human/mouse chimeric CLDN-5, and a CLDN-5-based artificial membrane protein consisting of symmetrically arranged ECRs. Intraperitoneal immunization of both engineered CLDN-5 ECR antigens induced ECR-binding antibodies in mice with a high success rate. We isolated five monoclonal antibodies that specifically recognized CLDN-5 ECR. Antibody clone 2B12 showed high affinity (<10 nM) and inhibited CLDN-5-containing tight junctions. These results demonstrate the effectiveness of the methods for monoclonal antibody development targeting difficult-to-produce membrane proteins such as CLDNs.
Collapse
|
30
|
Wang J, Garancher A, Ramaswamy V, Wechsler-Reya RJ. Medulloblastoma: From Molecular Subgroups to Molecular Targeted Therapies. Annu Rev Neurosci 2018; 41:207-232. [PMID: 29641939 DOI: 10.1146/annurev-neuro-070815-013838] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Brain tumors are the leading cause of cancer-related death in children, and medulloblastoma (MB) is the most common malignant pediatric brain tumor. Advances in surgery, radiation, and chemotherapy have improved the survival of MB patients. But despite these advances, 25-30% of patients still die from the disease, and survivors suffer severe long-term side effects from the aggressive therapies they receive. Although MB is often considered a single disease, molecular profiling has revealed a significant degree of heterogeneity, and there is a growing consensus that MB consists of multiple subgroups with distinct driver mutations, cells of origin, and prognosis. Here, we review recent progress in MB research, with a focus on the genes and pathways that drive tumorigenesis, the animal models that have been developed to study tumor biology, and the advances in conventional and targeted therapy.
Collapse
Affiliation(s)
- Jun Wang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| | - Alexandra Garancher
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| | - Vijay Ramaswamy
- Division of Haematology/Oncology and Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, USA;
| |
Collapse
|
31
|
Neuhaus W, Piontek A, Protze J, Eichner M, Mahringer A, Subileau EA, Lee IFM, Schulzke JD, Krause G, Piontek J. Reversible opening of the blood-brain barrier by claudin-5-binding variants of Clostridium perfringens enterotoxin's claudin-binding domain. Biomaterials 2018; 161:129-143. [DOI: 10.1016/j.biomaterials.2018.01.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
|
32
|
Abstract
The blood-brain barrier (BBB) is increasingly regarded as a dynamic interface that adapts to the needs of the brain, responds to physiological changes, and gets affected by and can even promote diseases. Modulation of BBB function at the molecular level in vivo is beneficial for a variety of basic and clinical studies. Here we show that our heteroduplex oligonucleotide (HDO), composed of an antisense oligonucleotide and its complementary RNA, conjugated to α-tocopherol as a delivery ligand, efficiently reduced the expression of organic anion transporter 3 (OAT3) gene in brain microvascular endothelial cells in mice. This proof-of-concept study demonstrates that intravenous administration of chemically synthesized HDO can remarkably silence OAT3 at the mRNA and protein levels. We also demonstrated modulation of the efflux transport function of OAT3 at the BBB in vivo. HDO will serve as a novel platform technology to advance the biology and pathophysiology of the BBB in vivo, and will also open a new therapeutic field of gene silencing at the BBB for the treatment of various intractable neurological disorders.
Collapse
|
33
|
Menard C, Pfau ML, Hodes GE, Kana V, Wang VX, Bouchard S, Takahashi A, Flanigan ME, Aleyasin H, LeClair KB, Janssen WG, Labonté B, Parise EM, Lorsch ZS, Golden SA, Heshmati M, Tamminga C, Turecki G, Campbell M, Fayad ZA, Tang CY, Merad M, Russo SJ. Social stress induces neurovascular pathology promoting depression. Nat Neurosci 2017; 20:1752-1760. [PMID: 29184215 PMCID: PMC5726568 DOI: 10.1038/s41593-017-0010-3] [Citation(s) in RCA: 579] [Impact Index Per Article: 82.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/27/2017] [Indexed: 02/07/2023]
Abstract
Studies suggest that heightened peripheral inflammation contributes to the pathogenesis of major depressive disorder. We investigated the effect of chronic social defeat stress, a mouse model of depression, on blood-brain barrier (BBB) permeability and infiltration of peripheral immune signals. We found reduced expression of the endothelial cell tight junction protein claudin-5 (Cldn5) and abnormal blood vessel morphology in nucleus accumbens (NAc) of stress-susceptible but not resilient mice. CLDN5 expression was also decreased in NAc of depressed patients. Cldn5 downregulation was sufficient to induce depression-like behaviors following subthreshold social stress whereas chronic antidepressant treatment rescued Cldn5 loss and promoted resilience. Reduced BBB integrity in NAc of stress-susceptible or mice injected with adeno-associated virus expressing shRNA against Cldn5 caused infiltration of the peripheral cytokine interleukin-6 (IL-6) into brain parenchyma and subsequent expression of depression-like behaviors. These findings suggest that chronic social stress alters BBB integrity through loss of tight junction protein Cldn5, promoting peripheral IL-6 passage across the BBB and depression.
Collapse
Affiliation(s)
- Caroline Menard
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Département de psychiatrie et neurosciences, Faculté de médecine and CERVO Brain Research Centre, Université Laval, Quebec City, QC, Canada
| | - Madeline L Pfau
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgia E Hodes
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Veronika Kana
- Department of Oncological Sciences, Tisch Cancer Institute and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Victoria X Wang
- Department of Radiology, Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Sylvain Bouchard
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aki Takahashi
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- University of Tsukuba, Tsukuba, Japan
| | - Meghan E Flanigan
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hossein Aleyasin
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine B LeClair
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William G Janssen
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benoit Labonté
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zachary S Lorsch
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sam A Golden
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mitra Heshmati
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carol Tamminga
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gustavo Turecki
- Douglas Mental Health University Institute and McGill University, Montreal, QC, Canada
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Zahi A Fayad
- Department of Radiology, Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Cheuk Ying Tang
- Department of Radiology, Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Tisch Cancer Institute and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
34
|
Aguiar S, van der Gaag B, Cortese FAB. RNAi mechanisms in Huntington's disease therapy: siRNA versus shRNA. Transl Neurodegener 2017; 6:30. [PMID: 29209494 PMCID: PMC5702971 DOI: 10.1186/s40035-017-0101-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/30/2017] [Indexed: 12/19/2022] Open
Abstract
Huntington's Disease (HD) is a genetically dominant trinucleotide repeat disorder resulting from CAG repeats within the Huntingtin (HTT) gene exceeding a normal range (> 36 CAGs). Symptoms of the disease manifest in middle age and include chorea, dystonia, and cognitive decline. Typical latency from diagnosis to death is 20 years. There are currently no disease-modifying therapies available to HD patients. RNAi is a potentially curative therapy for HD. A popular line of research employs siRNA or antisense oligonucleotides (ASO) to knock down mutant Huntingtin mRNA (mHTT). Unfortunately, this modality requires repeated dosing, commonly exhibit off target effects (OTEs), and exert renal and hepatic toxicity. In contrast, a single AAV-mediated short-hairpin RNA (shRNA) dose can last years with low toxicity. In addition, we highlight research indicating that shRNA elicits fewer OTEs than siRNA when tested head-to-head. Despite this promise, shRNA therapy has been held back by difficulties controlling expression (oversaturating cells with toxic levels of RNA construct). In this review, we compare RNAi modalities for HD and propose novel methods of optimizing shRNA expression and on-target fidelity.
Collapse
Affiliation(s)
- Sebastian Aguiar
- Molecular Neuroscience Laboratory, Swammerdam Institute for Life Sciences (SILS-CNS), University of Amsterdam, Amsterdam, Netherlands
- Fulbright Program, US Department of State (IIE), New York City, NY USA
| | - Bram van der Gaag
- Molecular Neuroscience Laboratory, Swammerdam Institute for Life Sciences (SILS-CNS), University of Amsterdam, Amsterdam, Netherlands
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Francesco Albert Bosco Cortese
- Biogerontology Research Foundation (BGRF), Oxford, UK
- Department of Biomedical and Molecular Sciences, Queen’s University School of Medicine, Queen’s University, Kingston, Canada
| |
Collapse
|
35
|
Campbell M, Cassidy PS, O'Callaghan J, Crosbie DE, Humphries P. Manipulating ocular endothelial tight junctions: Applications in treatment of retinal disease pathology and ocular hypertension. Prog Retin Eye Res 2017; 62:120-133. [PMID: 28951125 DOI: 10.1016/j.preteyeres.2017.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/01/2017] [Accepted: 09/20/2017] [Indexed: 11/25/2022]
Abstract
Protein levels of endothelial tight-junctions of the inner retinal microvasculature, together with those of Schlemm's canal, can be readily manipulated by RNA interference (RNAi), resulting in the paracellular clefts between such cells to be reversibly modulated. This facilitates access to the retina of systemically-deliverable low molecular weight, potentially therapeutic compounds, while also allowing potentially toxic material, for example, soluble Amyloid-β1-40, to be removed from the retina into the peripheral circulation. The technique has also been shown to be highly effective in alleviation of pathological cerebral oedema and we speculate that it may therefore have similar utility in the oedematous retina. Additionally, by manipulating endothelial tight-junctions of Schlemm's canal, inflow of aqueous humour from the trabecular meshwork into the Canal can be radically enhanced, suggesting a novel avenue for control of intraocular pressure. Here, we review the technology underlying this approach together with specific examples of clinical targets that are, or could be, amenable to this novel form of genetic intervention.
Collapse
Affiliation(s)
- Matthew Campbell
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland.
| | - Paul S Cassidy
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Jeffrey O'Callaghan
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Darragh E Crosbie
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Pete Humphries
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
36
|
Hashimoto Y, Shirakura K, Okada Y, Takeda H, Endo K, Tamura M, Watari A, Sadamura Y, Sawasaki T, Doi T, Yagi K, Kondoh M. Claudin-5-Binders Enhance Permeation of Solutes across the Blood-Brain Barrier in a Mammalian Model. J Pharmacol Exp Ther 2017; 363:275-283. [DOI: 10.1124/jpet.117.243014] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 08/07/2017] [Indexed: 12/17/2022] Open
|
37
|
Dithmer S, Staat C, Müller C, Ku MC, Pohlmann A, Niendorf T, Gehne N, Fallier-Becker P, Kittel Á, Walter FR, Veszelka S, Deli MA, Blasig R, Haseloff RF, Blasig IE, Winkler L. Claudin peptidomimetics modulate tissue barriers for enhanced drug delivery. Ann N Y Acad Sci 2017; 1397:169-184. [DOI: 10.1111/nyas.13359] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/20/2017] [Accepted: 03/23/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Sophie Dithmer
- Leibniz Institut für Molekulare Pharmakologie; Berlin Germany
| | - Christian Staat
- Leibniz Institut für Molekulare Pharmakologie; Berlin Germany
| | - Carolin Müller
- Leibniz Institut für Molekulare Pharmakologie; Berlin Germany
| | - Min-Chi Ku
- Berlin Ultrahigh Field Facility; Max Delbrück Center for Molecular Medicine in the Helmholtz Association; Berlin Germany
| | - Andreas Pohlmann
- Berlin Ultrahigh Field Facility; Max Delbrück Center for Molecular Medicine in the Helmholtz Association; Berlin Germany
| | - Thoralf Niendorf
- Berlin Ultrahigh Field Facility; Max Delbrück Center for Molecular Medicine in the Helmholtz Association; Berlin Germany
- Experimental and Clinical Research Center; Charite and Max Delbrück Center for Molecular Medicine in the Helmholtz Association; Berlin Germany
| | - Nora Gehne
- Leibniz Institut für Molekulare Pharmakologie; Berlin Germany
| | - Petra Fallier-Becker
- Institute of Pathology and Neuropathology; University of Tuebingen; Tuebingen Germany
| | - Ágnes Kittel
- Institute of Experimental Medicine; Hungarian Academy of Sciences; Budapest Hungary
| | - Fruzsina R. Walter
- Institute of Biophysics, Biological Research Centre; Hungarian Academy of Sciences; Szeged Hungary
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre; Hungarian Academy of Sciences; Szeged Hungary
| | - Maria A. Deli
- Institute of Biophysics, Biological Research Centre; Hungarian Academy of Sciences; Szeged Hungary
| | - Rosel Blasig
- Leibniz Institut für Molekulare Pharmakologie; Berlin Germany
| | | | | | - Lars Winkler
- Leibniz Institut für Molekulare Pharmakologie; Berlin Germany
| |
Collapse
|
38
|
Size-selective opening of the blood-brain barrier by targeting endothelial sphingosine 1-phosphate receptor 1. Proc Natl Acad Sci U S A 2017; 114:4531-4536. [PMID: 28396408 DOI: 10.1073/pnas.1618659114] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The vasculature of the central nervous system (CNS) forms a selective barrier termed the blood-brain barrier (BBB). Disruption of the BBB may contribute to various CNS diseases. Conversely, the intact BBB restricts efficient penetration of CNS-targeted drugs. Here, we report the BBB-regulatory role of endothelial sphingosine 1-phosphate (S1P) receptor-1, a G protein-coupled receptor known to promote the barrier function in peripheral vessels. Endothelial-specific S1pr1 knockout mice (S1pr1iECKO ) showed BBB breach for small-molecular-mass fluorescence tracers (<3 kDa), but not larger tracers (>10 kDa). Chronic BBB leakiness was associated with cognitive impairment, as assessed by the novel object recognition test, but not signs of brain inflammation. Brain microvessels of S1pr1iECKO mice showed altered subcellular distribution of tight junctional proteins. Pharmacological inhibition of S1P1 function led to transient BBB breach. These data suggest that brain endothelial S1P1 maintain the BBB by regulating the proper localization of tight junction proteins and raise the possibility that endothelial S1P1 inhibition may be a strategy for transient BBB opening and delivery of small molecules into the CNS.
Collapse
|
39
|
Modulating the paracellular pathway at the blood-brain barrier: current and future approaches for drug delivery to the CNS. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 20:35-39. [PMID: 27986221 DOI: 10.1016/j.ddtec.2016.07.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/13/2016] [Indexed: 11/20/2022]
Abstract
The blood-brain barrier (BBB) is the tightly regulated point of entry by which any neuro-targeting therapy must pass through. BBB modulation is a means to loosen the size exclusion properties of the barrier by temporarily interfering with the formation of intercellular tight junction (TJ) or adheren junction (AJ) complexes, allowing for diffusion of small molecule therapeutics from blood to brain. Several technologies, such as RNAi, peptidomimetics, high frequency ultrasound and nanoparticles, have been developed and refined over the years, paving the way for barrier modulation to become an effective part of conventional central nervous system therapies. Here, we review the current and future approaches aimed at facilitating enhanced drug delivery to the central nervous system (CNS).
Collapse
|
40
|
Greene C, Campbell M. Tight junction modulation of the blood brain barrier: CNS delivery of small molecules. Tissue Barriers 2016; 4:e1138017. [PMID: 27141420 DOI: 10.1080/21688370.2015.1138017] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/22/2015] [Accepted: 12/24/2015] [Indexed: 01/06/2023] Open
Abstract
The blood brain barrier (BBB) represents a major obstacle for targeted drug delivery to the brain for the treatment of central nervous system (CNS) disorders. Significant advances in barrier research over the past decade has led to the discovery of an increasing number of structural and regulatory proteins in tight junctions (TJ) and adherens junctions (AJ). These discoveries are providing the framework for the development of novel TJ modulators which can act specifically and temporarily to alter BBB function and regulate paracellular uptake of molecules. TJ modulators that have shown therapeutic potential in preclinical models include claudin-5 and occludin siRNAs, peptides derived from zonula occludens toxin as well as synthetic peptides targeting the extracellular loops of TJs. Adding to the array of modulating agents are novel mechanisms of BBB regulation such as focused ultrasound (FUS). This review will give a succinct overview of BBB biology and TJ modulation in general. Novel insights into BBB regulation in health and disease will also be summarized.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin ; Dublin 2, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin ; Dublin 2, Ireland
| |
Collapse
|
41
|
Kealy J, Campbell M. The Blood-Brain Barrier in Glioblastoma: Pathology and Therapeutic Implications. RESISTANCE TO TARGETED ANTI-CANCER THERAPEUTICS 2016. [DOI: 10.1007/978-3-319-46505-0_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Chen H, Mruk DD, Xia W, Bonanomi M, Silvestrini B, Cheng CY. Effective Delivery of Male Contraceptives Behind the Blood-Testis Barrier (BTB) - Lesson from Adjudin. Curr Med Chem 2016; 23:701-13. [PMID: 26758796 PMCID: PMC4845722 DOI: 10.2174/0929867323666160112122724] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 12/18/2014] [Accepted: 01/11/2016] [Indexed: 12/15/2022]
Abstract
The blood-testis barrier (BTB) is one of the tightest blood-tissue barriers in the mammalian body. It divides the seminiferous epithelium of the seminiferous tubule, the functional unit of the testis, where spermatogenesis takes place, into the basal and the adluminal (apical) compartments. Functionally, the BTB provides a unique microenvironment for meiosis I/II and post-meiotic spermatid development which take place exclusively in the apical compartment, away from the host immune system, and it contributes to the immune privilege status of testis. However, the BTB also poses major obstacles in developing male contraceptives (e.g., adjudin) that exert their effects on germ cells in the apical compartment, such as by disrupting spermatid adhesion to the Sertoli cell, causing germ cell exfoliation from the testis. Besides the tight junction (TJ) between adjacent Sertoli cells at the BTB that restricts the entry of contraceptives from the microvessels in the interstitium to the adluminal compartment, drug transporters, such as P-glycoprotein and multidrug resistance-associated protein 1 (MRP1), are also present that actively pump drugs out of the testis, limiting drug bioavailability. Recent advances in drug formulations, such as drug particle micronization (<50 μm) and co-grinding of drug particles with ß-cyclodextrin have improved bioavailability of contraceptives via considerable increase in solubility. Herein, we discuss development in drug formulations using adjudin as an example. We also put emphasis on the possible use of nanotechnology to deliver adjudin to the apical compartment with multidrug magnetic mesoporous silica nanoparticles. These advances in technology will significantly enhance our ability to develop effective non-hormonal male contraceptives for men.
Collapse
Affiliation(s)
| | | | | | | | | | - Chuen-Yan Cheng
- Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York 10065, USA..
| |
Collapse
|
43
|
Assmann JC, Körbelin J, Schwaninger M. Genetic manipulation of brain endothelial cells in vivo. Biochim Biophys Acta Mol Basis Dis 2015; 1862:381-94. [PMID: 26454206 DOI: 10.1016/j.bbadis.2015.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/30/2015] [Accepted: 10/05/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Julian C Assmann
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Jakob Körbelin
- University Medical Center Hamburg-Eppendorf, Hubertus Wald Cancer Center, Department of Oncology and Hematology, Martinistr. 52, 20246 Hamburg, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, 23562 Lübeck, Germany.
| |
Collapse
|
44
|
Keaney J, Walsh DM, O’Malley T, Hudson N, Crosbie DE, Loftus T, Sheehan F, McDaid J, Humphries MM, Callanan JJ, Brett FM, Farrell MA, Humphries P, Campbell M. Autoregulated paracellular clearance of amyloid-β across the blood-brain barrier. SCIENCE ADVANCES 2015; 1:e1500472. [PMID: 26491725 PMCID: PMC4610013 DOI: 10.1126/sciadv.1500472] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/01/2015] [Indexed: 05/15/2023]
Abstract
The blood-brain barrier (BBB) is essential for maintaining brain homeostasis and protecting neural tissue from damaging blood-borne agents. The barrier is characterized by endothelial tight junctions that limit passive paracellular diffusion of polar solutes and macromolecules from blood to brain. Decreased brain clearance of the neurotoxic amyloid-β (Aβ) peptide is a central event in the pathogenesis of Alzheimer's disease (AD). Whereas transport of Aβ across the BBB can occur via transcellular endothelial receptors, the paracellular movement of Aβ has not been described. We show that soluble human Aβ(1-40) monomers can diffuse across the paracellular pathway of the BBB in tandem with a decrease in the tight junction proteins claudin-5 and occludin in the cerebral vascular endothelium. In a murine model of AD (Tg2576), plasma Aβ(1-40) levels were significantly increased, brain Aβ(1-40) levels were decreased, and cognitive function was enhanced when both claudin-5 and occludin were suppressed. Furthermore, Aβ can cause a transient down-regulation of claudin-5 and occludin, allowing for its own paracellular clearance across the BBB. Our results show, for the first time, the involvement of the paracellular pathway in autoregulated Aβ movement across the BBB and identify both claudin-5 and occludin as potential therapeutic targets for AD. These findings also indicate that controlled modulation of tight junction components at the BBB can enhance the clearance of Aβ from the brain.
Collapse
Affiliation(s)
- James Keaney
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Dominic M. Walsh
- Laboratory for Neurodegenerative Research, Center for Neurological Diseases, Brigham and Women’s Hospital, Harvard Institute of Medicine, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Tiernan O’Malley
- Laboratory for Neurodegenerative Research, Center for Neurological Diseases, Brigham and Women’s Hospital, Harvard Institute of Medicine, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Natalie Hudson
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Darragh E. Crosbie
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Teresa Loftus
- Department of Neuropathology, Beaumont Hospital, Dublin 9, Ireland
| | - Florike Sheehan
- Department of Neuropathology, Beaumont Hospital, Dublin 9, Ireland
| | - Jacqueline McDaid
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Marian M. Humphries
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - John J. Callanan
- UCD School of Veterinary Medicine and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
- Ross University School of Veterinary Medicine, P. O. Box 334, Basseterre, St. Kitts, West Indies
| | | | | | - Peter Humphries
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, Lincoln Place Gate, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
45
|
Tivnan A, Zakaria Z, O'Leary C, Kögel D, Pokorny JL, Sarkaria JN, Prehn JHM. Inhibition of multidrug resistance protein 1 (MRP1) improves chemotherapy drug response in primary and recurrent glioblastoma multiforme. Front Neurosci 2015; 9:218. [PMID: 26136652 PMCID: PMC4468867 DOI: 10.3389/fnins.2015.00218] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 05/31/2015] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain cancer with extremely poor prognostic outcome despite intensive treatment. All chemotherapeutic agents currently used have no greater than 30–40% response rate, many fall into the range of 10–20%, with delivery across the blood brain barrier (BBB) or chemoresistance contributing to the extremely poor outcomes despite treatment. Increased expression of the multidrug resistance protein 1(MRP1) in high grade glioma, and it's role in BBB active transport, highlights this member of the ABC transporter family as a target for improving drug responses in GBM. In this study we show that small molecule inhibitors and gene silencing of MRP1 had a significant effect on GBM cell response to temozolomide (150 μM), vincristine (100 nM), and etoposide (2 μM). Pre-treatment with Reversan (inhibitor of MRP1 and P-glycoprotein) led to a significantly improved response to cell death in the presence of all three chemotherapeutics, in both primary and recurrent GBM cells. The presence of MK571 (inhibitor of MRP1 and multidrug resistance protein 4 (MRP4) led to an enhanced effect of vincristine and etoposide in reducing cell viability over a 72 h period. Specific MRP1 inhibition led to a significant increase in vincristine and etoposide-induced cell death in all three cell lines assessed. Treatment with MK571, or specific MRP1 knockdown, did not have any effect on temozolomide drug response in these cells. These findings have significant implications in providing researchers an opportunity to improve currently used chemotherapeutics for the initial treatment of primary GBM, and improved treatment for recurrent GBM patients.
Collapse
Affiliation(s)
- Amanda Tivnan
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland Dublin, Ireland
| | - Zaitun Zakaria
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland Dublin, Ireland
| | - Caitrín O'Leary
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland Dublin, Ireland
| | - Donat Kögel
- Experimental Neurosurgery, Neuroscience Center, Frankfurt University Hospital Frankfurt am Main, Germany
| | - Jenny L Pokorny
- Department of Radiation Oncology, Mayo Clinic Rochester, MN, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic Rochester, MN, USA
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Centre for Systems Medicine, Royal College of Surgeons in Ireland Dublin, Ireland
| |
Collapse
|
46
|
T Cells-Protective or Pathogenic in Alzheimer's Disease? J Neuroimmune Pharmacol 2015; 10:547-60. [PMID: 25957956 DOI: 10.1007/s11481-015-9612-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/29/2015] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and is characterised by deposits of amyloid β (Aβ), neurofibrillary tangles and neuronal loss. Neuroinflammatory changes have been identified as a feature of the disease, and recent studies have suggested a potential role for the peripheral immune system in driving these changes and, ultimately, the associated neuronal degeneration. A number of reports have detailed changes in the activation state and subtype of T cells in the circulation and CSF of AD patients and there is evidence of T cell infiltration into the brain. In this review, we examine the possible impact of T cell infiltration in the progression of pathology in AD and consider the data obtained from animal models of the disease. We consider how these cells infiltrate the brain, particularly in AD, and discuss whether the presence of T cells in the AD brain is protective or pathogenic. Finally we evaluate the current therapies, particularly those that involve immunization.
Collapse
|
47
|
Gomes MJ, Martins S, Sarmento B. siRNA as a tool to improve the treatment of brain diseases: Mechanism, targets and delivery. Ageing Res Rev 2015; 21:43-54. [PMID: 25796492 DOI: 10.1016/j.arr.2015.03.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/10/2015] [Accepted: 03/16/2015] [Indexed: 10/23/2022]
Abstract
As the population ages, brain pathologies such as neurodegenerative diseases and brain cancer increase their incidence, being the need to find successful treatments of upmost importance. Drug delivery to the central nervous system (CNS) is required in order to reach diseases causes and treat them. However, biological barriers, mainly blood-brain barrier (BBB), are the key obstacles that prevent the effectiveness of possible treatments due to their ability to strongly limit the perfusion of compounds into the brain. Over the past decades, new approaches towards overcoming BBB and its efflux transporters had been proposed. One of these approaches here reviewed is through small interfering RNA (siRNA), which is capable to specifically target one gene and silence it in a post-transcriptional way. There are different possible functional proteins at the BBB, as the ones responsible for transport or just for its tightness, which could be a siRNA target. As important as the effective silence is the way to delivery siRNA to its anatomical site of action. This is where nanotechnology-based systems may help, by protecting siRNA circulation and providing cell/tissue-targeting and intracellular siRNA delivery. After an initial overview on incidence of brain diseases and basic features of the CNS, BBB and its efflux pumps, this review focuses on recent strategies to reach brain based on siRNA, and how to specifically target these approaches in order to treat brain diseases.
Collapse
|
48
|
Rathnasamy G, Sivakumar V, Foulds WS, Ling EA, Kaur C. Vascular changes in the developing rat retina in response to hypoxia. Exp Eye Res 2014; 130:73-86. [PMID: 25433125 DOI: 10.1016/j.exer.2014.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 10/17/2014] [Accepted: 11/26/2014] [Indexed: 12/20/2022]
Abstract
This study was carried out to investigate the roles of tight junction (TJ) proteins and other factors in the increased permeability of the blood retinal barrier (BRB) affecting the immature neonatal retina following a hypoxic insult. The expression of endothelial TJ proteins such as claudin-5, occludin and zonula occludens-1 (ZO-1) and endothelial cell specific molecule-1 (ESM-1), and associated structural changes in the blood vessels were analyzed in the retinas of 1-day-old Wistar rats subjected to hypoxia for 2 h and subsequently sacrificed at different time points ranging from 3 h to 14 d. The mRNA and protein expression of claudin-5, occludin & ZO-1 was found to be reduced in the hypoxic retina, although, at the ultrastructural level, the TJ between the endothelial cells and retinal pigment epithelial cells appeared to be intact. Following the hypoxic insult vascular endothelial cells frequently showed presence of cytoplasmic vacuoles, vacuolated mitochondria and multivesicular aggregations projecting into the lumen of the capillaries. The expression of ESM-1 in the immature retinas was found to be increased following hypoxic exposure. The structural and molecular changes in the hypoxic neonatal retinas were consistent with a hypoxia induced impairment of the BRB. Hypoxia reduced the expression of TJ proteins in the neonatal retina, but the role of increased ESM-1 expression in this process warrants further investigation.
Collapse
Affiliation(s)
- Gurugirijha Rathnasamy
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, Singapore 117594, Singapore
| | - Viswanathan Sivakumar
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, Singapore 117594, Singapore
| | - Wallace S Foulds
- Singapore Eye Research Institute c/o Singapore National Eye Centre, 11 Third Hospital Avenue, Singapore 168751, Singapore; University of Glasgow, Glasgow, Scotland G12 8QQ, UK
| | - Eng Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, Singapore 117594, Singapore
| | - Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, Blk MD10, 4 Medical Drive, National University of Singapore, Singapore 117594, Singapore; Singapore Eye Research Institute c/o Singapore National Eye Centre, 11 Third Hospital Avenue, Singapore 168751, Singapore.
| |
Collapse
|
49
|
Survival from rabies encephalitis. J Neurol Sci 2014; 339:8-14. [PMID: 24582283 DOI: 10.1016/j.jns.2014.02.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 01/29/2014] [Accepted: 02/14/2014] [Indexed: 12/25/2022]
Abstract
Rabies is a major public health problem in Asia and Africa, with nearly 60,000 deaths every year, and represents a substantial economic burden. Neurologists frequently encounter atypical cases, and need to make informed decisions regarding diagnosis and management. No therapy has been shown to unequivocally improve survival in rabies till date. Despite the overwhelmingly fatal nature of this disease, a small number of patients have been reported to survive acute rabies encephalitis with varying degrees of neurological sequelae. This paper presents the eleventh documented case of survival from rabies, which developed after being bitten by a stray dog, albeit with severe neurological residua. Similar to patients in previous reports, this man demonstrated a robust immune response as indicated by peripheral viral clearance and very high serum and cerebrospinal fluid antibody titres. Immunologically-mediated virus clearance therefore appears to be a prerequisite for survival. A detailed review of previously reported survivors, as well as descriptions of the host response and viral clearance in human rabies, current therapy for this disease and future directions in improving the currently dismal prognosis are provided.
Collapse
|
50
|
Zhou L, Yang B, Wang Y, Zhang HL, Chen RW, Wang YB. Bradykinin regulates the expression of claudin-5 in brain microvascular endothelial cells via calcium-induced calcium release. J Neurosci Res 2014; 92:597-606. [PMID: 24464430 DOI: 10.1002/jnr.23350] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Revised: 11/08/2013] [Accepted: 11/25/2013] [Indexed: 11/11/2022]
Affiliation(s)
- Lei Zhou
- Department of Neurosurgery; First Affiliated Hospital of China Medical University; Shenyang People's Republic of China
| | - Bo Yang
- Department of Neurosurgery; General Hospital of Jixi Mining Conglomerate; Jixi People's Republic of China
| | - Yong Wang
- Department of Neurosurgery; First Affiliated Hospital of China Medical University; Shenyang People's Republic of China
| | - Hong-Liang Zhang
- Department of Neurosurgery; First Affiliated Hospital of China Medical University; Shenyang People's Republic of China
| | - Run-Wei Chen
- Department of Neurosurgery; First Affiliated Hospital of China Medical University; Shenyang People's Republic of China
| | - Yi-Bao Wang
- Department of Neurosurgery; First Affiliated Hospital of China Medical University; Shenyang People's Republic of China
| |
Collapse
|