251
|
Immortalized endothelial cell lines for in vitro blood–brain barrier models: A systematic review. Brain Res 2016; 1642:532-545. [DOI: 10.1016/j.brainres.2016.04.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/05/2016] [Accepted: 04/12/2016] [Indexed: 12/18/2022]
|
252
|
Aparicio-Blanco J, Martín-Sabroso C, Torres-Suárez AI. In vitro screening of nanomedicines through the blood brain barrier: A critical review. Biomaterials 2016; 103:229-255. [PMID: 27392291 DOI: 10.1016/j.biomaterials.2016.06.051] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/14/2016] [Accepted: 06/20/2016] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier accounts for the high attrition rate of the treatments of most brain disorders, which therefore remain one of the greatest health-care challenges of the twenty first century. Against this background of hindrance to brain delivery, nanomedicine takes advantage of the assembly at the nanoscale of available biomaterials to provide a delivery platform with potential to raising brain levels of either imaging or therapeutic agents. Nevertheless, to prevent later failure due to ineffective drug levels at the target site, researchers have been endeavoring to develop a battery of in vitro screening procedures that can predict earlier in the drug discovery process the ability of these cutting-edge drug delivery platforms to cross the blood-brain barrier for biomedical purposes. This review provides an in-depth analysis of the currently available in vitro blood-brain barrier models (both cell-based and non-cell-based) with the focus on their suitability for understanding the biological brain distribution of forthcoming nanomedicines. The relationship between experimental factors and underlying physiological assumptions that would ultimately lead to a more predictive capacity of their in vivo performance, and those methods already assayed for the evaluation of the brain distribution of nanomedicines are comprehensively discussed.
Collapse
Affiliation(s)
- Juan Aparicio-Blanco
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Cristina Martín-Sabroso
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain
| | - Ana-Isabel Torres-Suárez
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Complutense University, 28040, Madrid, Spain; University Institute of Industrial Pharmacy, Complutense University, 28040, Madrid, Spain.
| |
Collapse
|
253
|
Chatard M, Puech C, Roche F, Perek N. Hypoxic Stress Induced by Hydralazine Leads to a Loss of Blood-Brain Barrier Integrity and an Increase in Efflux Transporter Activity. PLoS One 2016; 11:e0158010. [PMID: 27337093 PMCID: PMC4919080 DOI: 10.1371/journal.pone.0158010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/08/2016] [Indexed: 11/18/2022] Open
Abstract
Understanding cellular and molecular mechanisms induced by hypoxic stress is crucial to reduce blood-brain barrier (BBB) disruption in some neurological diseases. Since the brain is a complex organ, it makes the interpretation of in vivo data difficult, so BBB studies are often investigated using in vitro models. However, the investigation of hypoxia in cellular pathways is complex with physical hypoxia because HIF-1α (factor induced by hypoxia) has a short half-life. We had set up an innovative and original method of induction of hypoxic stress by hydralazine that was more reproducible, which allowed us to study its impact on an in vitro BBB model. Our results showed that hydralazine, a mimetic agent of the hypoxia pathway, had the same effect as physical hypoxia, with few cytotoxicity effects on our cells. Hypoxic stress led to an increase of BBB permeability which corresponded to an opening of our BBB model. Study of tight junction proteins revealed that this hypoxic stress decreased ZO-1 but not occludin expression. In contrast, cells established a defence mechanism by increasing expression and activity of their efflux transporters (Pgp and MRP-1). This induction method of hypoxic stress by hydralazine is simple, reproducible, controllable and suitable to understand the cellular and molecular mechanisms involved by hypoxia on the BBB.
Collapse
Affiliation(s)
- Morgane Chatard
- Université de Lyon, UJM-Saint-Etienne, SNA-EPIS, EA4607, F-42023, Saint-Etienne, France
- Université de Lyon, UJM-Saint-Etienne, INSERM, SAINBIOSE U1089 Team DVH, F-42023, Saint-Etienne, France
| | - Clémentine Puech
- Université de Lyon, UJM-Saint-Etienne, INSERM, SAINBIOSE U1089 Team DVH, F-42023, Saint-Etienne, France
| | - Frederic Roche
- Université de Lyon, UJM-Saint-Etienne, SNA-EPIS, EA4607, F-42023, Saint-Etienne, France
- * E-mail:
| | - Nathalie Perek
- Université de Lyon, UJM-Saint-Etienne, INSERM, SAINBIOSE U1089 Team DVH, F-42023, Saint-Etienne, France
| |
Collapse
|
254
|
Neuroinvasion and Inflammation in Viral Central Nervous System Infections. Mediators Inflamm 2016; 2016:8562805. [PMID: 27313404 PMCID: PMC4897715 DOI: 10.1155/2016/8562805] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/16/2016] [Accepted: 04/12/2016] [Indexed: 12/31/2022] Open
Abstract
Neurotropic viruses can cause devastating central nervous system (CNS) infections, especially in young children and the elderly. The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) have been described as relevant sites of entry for specific viruses as well as for leukocytes, which are recruited during the proinflammatory response in the course of CNS infection. In this review, we illustrate examples of established brain barrier models, in which the specific reaction patterns of different viral families can be analyzed. Furthermore, we highlight the pathogen specific array of cytokines and chemokines involved in immunological responses in viral CNS infections. We discuss in detail the link between specific cytokines and chemokines and leukocyte migration profiles. The thorough understanding of the complex and interrelated inflammatory mechanisms as well as identifying universal mediators promoting CNS inflammation is essential for the development of new diagnostic and treatment strategies.
Collapse
|
255
|
Strazza M, Maubert ME, Pirrone V, Wigdahl B, Nonnemacher MR. Co-culture model consisting of human brain microvascular endothelial and peripheral blood mononuclear cells. J Neurosci Methods 2016; 269:39-45. [PMID: 27216631 DOI: 10.1016/j.jneumeth.2016.05.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 04/29/2016] [Accepted: 05/17/2016] [Indexed: 01/25/2023]
Abstract
BACKGROUND Numerous systems exist to model the blood-brain barrier (BBB) with the goal of understanding the regulation of passage into the central nervous system (CNS) and the potential impact of selected insults on BBB function. These models typically focus on the intrinsic cellular properties of the BBB, yet studies of peripheral cell migration are often excluded due to technical restraints. NEW METHOD This method allows for the study of in vitro cellular transmigration following exposure to any treatment of interest through optimization of co-culture conditions for the human brain microvascular endothelial cells (BMEC) cell line, hCMEC/D3, and primary human peripheral blood mononuclear cells (PBMCs). RESULTS hCMEC/D3 cells form functionally confluent monolayers on collagen coated polytetrafluoroethylene (PTFE) transwell inserts, as assessed by microscopy and tracer molecule (FITC-dextran (FITC-D)) exclusion. Two components of complete hCMEC/D3 media, EBM-2 base-media and hydrocortisone (HC), were determined to be cytotoxic to PBMCs. By combining the remaining components of complete hCMEC/D3 media with complete PBMC media a resulting co-culture media was established for use in hCMEC/D3-PBMC co-culture functional assays. COMPARISON WITH EXISTING METHODS Through this method, issues of extensive differences in culture media conditions are resolved allowing for treatments and functional assays to be conducted on the two cell populations co-cultured simultaneously. CONCLUSION Described here is an in vitro co-culture model of the BBB, consisting of the hCMEC/D3 cell line and primary human PBMCs. The co-culture media will now allow for the study of exposure to potential insults to BBB function over prolonged time courses.
Collapse
Affiliation(s)
- Marianne Strazza
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA.
| | - Monique E Maubert
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA.
| | - Vanessa Pirrone
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA.
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Bluemle Life Sciences Building, 233 South 10th Street Suite 1050, Philadelphia, PA 19107, USA.
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, 245 N. 15th Street, MS #1013A, Philadelphia, PA 19102, USA.
| |
Collapse
|
256
|
Gräfe C, Slabu I, Wiekhorst F, Bergemann C, von Eggeling F, Hochhaus A, Trahms L, Clement JH. Magnetic particle spectroscopy allows precise quantification of nanoparticles after passage through human brain microvascular endothelial cells. Phys Med Biol 2016; 61:3986-4000. [PMID: 27163489 DOI: 10.1088/0031-9155/61/11/3986] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Crossing the blood-brain barrier is an urgent requirement for the treatment of brain disorders. Superparamagnetic iron oxide nanoparticles (SPIONs) are a promising tool as carriers for therapeutics because of their physical properties, biocompatibility, and their biodegradability. In order to investigate the interaction of nanoparticles with endothelial cell layers in detail, in vitro systems are of great importance. Human brain microvascular endothelial cells are a well-suited blood-brain barrier model. Apart from generating optimal conditions for the barrier-forming cell units, the accurate detection and quantification of SPIONs is a major challenge. For that purpose we use magnetic particle spectroscopy to sensitively and directly quantify the SPION-specific iron content. We could show that SPION concentration depends on incubation time, nanoparticle concentration and location. This model system allows for further investigations on particle uptake and transport at cellular barriers with regard to parameters including particles' shape, material, size, and coating.
Collapse
Affiliation(s)
- C Gräfe
- Abteilung Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Erlanger Allee 101, D-07747 Jena, Germany
| | | | | | | | | | | | | | | |
Collapse
|
257
|
Helms HC, Abbott NJ, Burek M, Cecchelli R, Couraud PO, Deli MA, Förster C, Galla HJ, Romero IA, Shusta EV, Stebbins MJ, Vandenhaute E, Weksler B, Brodin B. In vitro models of the blood-brain barrier: An overview of commonly used brain endothelial cell culture models and guidelines for their use. J Cereb Blood Flow Metab 2016; 36:862-90. [PMID: 26868179 PMCID: PMC4853841 DOI: 10.1177/0271678x16630991] [Citation(s) in RCA: 541] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
The endothelial cells lining the brain capillaries separate the blood from the brain parenchyma. The endothelial monolayer of the brain capillaries serves both as a crucial interface for exchange of nutrients, gases, and metabolites between blood and brain, and as a barrier for neurotoxic components of plasma and xenobiotics. This "blood-brain barrier" function is a major hindrance for drug uptake into the brain parenchyma. Cell culture models, based on either primary cells or immortalized brain endothelial cell lines, have been developed, in order to facilitate in vitro studies of drug transport to the brain and studies of endothelial cell biology and pathophysiology. In this review, we aim to give an overview of established in vitro blood-brain barrier models with a focus on their validation regarding a set of well-established blood-brain barrier characteristics. As an ideal cell culture model of the blood-brain barrier is yet to be developed, we also aim to give an overview of the advantages and drawbacks of the different models described.
Collapse
Affiliation(s)
- Hans C Helms
- Department of Pharmacy, University of Copenhagen, Denmark
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, UK
| | - Malgorzata Burek
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | | | - Pierre-Olivier Couraud
- Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Maria A Deli
- Institute of Biophysics, Biological Research Centre, HAS, Szeged, Hungary
| | - Carola Förster
- Klinik und Poliklinik für Anästhesiologie, University of Wurzburg, Germany
| | - Hans J Galla
- Institute of Biochemistry, University of Muenster, Germany
| | - Ignacio A Romero
- Department of Biological Sciences, The Open University, Walton Hall, Milton Keynes, UK
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | - Matthew J Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, WI, USA
| | | | - Babette Weksler
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, NY, USA
| | - Birger Brodin
- Department of Pharmacy, University of Copenhagen, Denmark
| |
Collapse
|
258
|
Vandenhaute E, Drolez A, Sevin E, Gosselet F, Mysiorek C, Dehouck MP. Adapting coculture in vitro models of the blood-brain barrier for use in cancer research: maintaining an appropriate endothelial monolayer for the assessment of transendothelial migration. J Transl Med 2016; 96:588-98. [PMID: 26901835 DOI: 10.1038/labinvest.2016.35] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/06/2016] [Accepted: 01/14/2016] [Indexed: 12/14/2022] Open
Abstract
Although brain metastases are the most common brain tumors in adults, there are few treatment options in this setting. To colonize the brain, circulating tumor cells must cross the blood-brain barrier (BBB), which is situated within specialized, restrictive microvascular endothelium. Understanding how cancer cells manage to transmigrate through the BBB might enable this process to be prevented. In vitro models are dedicated tools for characterizing the cellular and molecular mechanisms that underlie transendothelial migration process, as long as they accurately mimic the brain endothelium's in vivo characteristics. The objective of the present study was to adapt an existing in vitro model of the human BBB for use in studying cancer cell transmigration. The model is based on the coculture of endothelial cells (ECs, derived from cord blood hematopoietic stem cells) and brain pericytes. To allow the migration of cancer cells into the lower compartment, our model had to be transposed onto inserts with a larger pore size. However, we encountered a problem when culturing ECs on large (3-μm)-pore inserts: the cells crossed the membrane and formed a non-physiological second layer on the lower face of the insert. Using 3-μm-pore inserts (in a 12-well plate format), we report here on a method that enables the maintenance of a single monolayer of ECs on the insert's upper face only. Under these chosen conditions, the ECs exhibited typical BBB properties found in the original model (including restricted paracellular permeability and the expression of continuous tight junctions). This modified in vitro model of the human BBB enabled us to investigate the migratory potential of the MDA-MB-231 cell line (derived from highly metastatic human breast cancer cells). Last, the results obtained were compared with the rate of transmigration through endothelia with no BBB features.
Collapse
Affiliation(s)
- Elodie Vandenhaute
- Laboratoire de la Barrière Hémato-Encéphalique-EA 2465, Faculté des Sciences Jean Perrin, Université d'Artois, LBHE, Lens, France
| | - Aurore Drolez
- Laboratoire de la Barrière Hémato-Encéphalique-EA 2465, Faculté des Sciences Jean Perrin, Université d'Artois, LBHE, Lens, France
| | - Emmanuel Sevin
- Laboratoire de la Barrière Hémato-Encéphalique-EA 2465, Faculté des Sciences Jean Perrin, Université d'Artois, LBHE, Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique-EA 2465, Faculté des Sciences Jean Perrin, Université d'Artois, LBHE, Lens, France
| | - Caroline Mysiorek
- Laboratoire de la Barrière Hémato-Encéphalique-EA 2465, Faculté des Sciences Jean Perrin, Université d'Artois, LBHE, Lens, France
| | - Marie-Pierre Dehouck
- Laboratoire de la Barrière Hémato-Encéphalique-EA 2465, Faculté des Sciences Jean Perrin, Université d'Artois, LBHE, Lens, France
| |
Collapse
|
259
|
Mu R, Cutting AS, Del Rosario Y, Villarino N, Stewart L, Weston TA, Patras KA, Doran KS. Identification of CiaR Regulated Genes That Promote Group B Streptococcal Virulence and Interaction with Brain Endothelial Cells. PLoS One 2016; 11:e0153891. [PMID: 27100296 PMCID: PMC4839699 DOI: 10.1371/journal.pone.0153891] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/05/2016] [Indexed: 11/18/2022] Open
Abstract
Group B Streptococcus (GBS) is a major causative agent of neonatal meningitis due to its ability to efficiently cross the blood-brain barrier (BBB) and enter the central nervous system (CNS). It has been demonstrated that GBS can invade human brain microvascular endothelial cells (hBMEC), a primary component of the BBB; however, the mechanism of intracellular survival and trafficking is unclear. We previously identified a two component regulatory system, CiaR/H, which promotes GBS intracellular survival in hBMEC. Here we show that a GBS strain deficient in the response regulator, CiaR, localized more frequently with Rab5, Rab7 and LAMP1 positive vesicles. Further, lysosomes isolated from hBMEC contained fewer viable bacteria following initial infection with the ΔciaR mutant compared to the WT strain. To characterize the contribution of CiaR-regulated genes, we constructed isogenic mutant strains lacking the two most down-regulated genes in the CiaR-deficient mutant, SAN_2180 and SAN_0039. These genes contributed to bacterial uptake and intracellular survival. Furthermore, competition experiments in mice showed that WT GBS had a significant survival advantage over the Δ2180 and Δ0039 mutants in the bloodstream and brain.
Collapse
Affiliation(s)
- Rong Mu
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
| | - Andrew S. Cutting
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
| | - Yvette Del Rosario
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
| | - Nicholas Villarino
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
| | - Lara Stewart
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
| | - Thomas A. Weston
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
| | - Kathryn A. Patras
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
| | - Kelly S. Doran
- Department of Biology and Center for Microbial Sciences, San Diego State University, San Diego, California, 92182, United States of America
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, 92093, United States of America
- * E-mail:
| |
Collapse
|
260
|
Katt ME, Xu ZS, Gerecht S, Searson PC. Human Brain Microvascular Endothelial Cells Derived from the BC1 iPS Cell Line Exhibit a Blood-Brain Barrier Phenotype. PLoS One 2016; 11:e0152105. [PMID: 27070801 PMCID: PMC4829259 DOI: 10.1371/journal.pone.0152105] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/08/2016] [Indexed: 01/08/2023] Open
Abstract
The endothelial cells that form capillaries in the brain are highly specialized, with tight junctions that minimize paracellular transport and an array of broad-spectrum efflux pumps that make drug delivery to the brain extremely challenging. One of the major limitations in blood-brain barrier research and the development of drugs to treat central nervous system diseases is the lack of appropriate cell lines. Recent reports indicate that the derivation of human brain microvascular endothelial cells (hBMECs) from human induced pluripotent stem cells (iPSCs) may provide a solution to this problem. Here we demonstrate the derivation of hBMECs extended to two new human iPSC lines: BC1 and GFP-labeled BC1. These hBMECs highly express adherens and tight junction proteins VE-cadherin, ZO-1, occludin, and claudin-5. The addition of retinoic acid upregulates VE-cadherin expression, and results in a significant increase in transendothelial electrical resistance to physiological values. The permeabilities of tacrine, rhodamine 123, and Lucifer yellow are similar to values obtained for MDCK cells. The efflux ratio for rhodamine 123 across hBMECs is in the range 2-4 indicating polarization of efflux transporters. Using the rod assay to assess cell organization in small vessels and capillaries, we show that hBMECs resist elongation with decreasing diameter but show progressive axial alignment. The derivation of hBMECs with a blood-brain barrier phenotype from the BC1 cell line highlights that the protocol is robust. The expression of GFP in hBMECs derived from the BC1-GFP cell line provides an important new resource for BBB research.
Collapse
Affiliation(s)
- Moriah E. Katt
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
- Institute of Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
| | - Zinnia S. Xu
- Institute of Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD, 21205, United States of America
| | - Sharon Gerecht
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
- Institute of Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
| | - Peter C. Searson
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
- Institute of Nanobiotechnology, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD, 21218, United States of America
| |
Collapse
|
261
|
Eigenmann DE, Dürig C, Jähne EA, Smieško M, Culot M, Gosselet F, Cecchelli R, Helms HCC, Brodin B, Wimmer L, Mihovilovic MD, Hamburger M, Oufir M. In vitro blood-brain barrier permeability predictions for GABAA receptor modulating piperine analogs. Eur J Pharm Biopharm 2016; 103:118-126. [PMID: 27018328 DOI: 10.1016/j.ejpb.2016.03.029] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/14/2016] [Accepted: 03/23/2016] [Indexed: 11/19/2022]
Abstract
The alkaloid piperine from black pepper (Piper nigrum L.) and several synthetic piperine analogs were recently identified as positive allosteric modulators of γ-aminobutyric acid type A (GABAA) receptors. In order to reach their target sites of action, these compounds need to enter the brain by crossing the blood-brain barrier (BBB). We here evaluated piperine and five selected analogs (SCT-66, SCT-64, SCT-29, LAU397, and LAU399) regarding their BBB permeability. Data were obtained in three in vitro BBB models, namely a recently established human model with immortalized hBMEC cells, a human brain-like endothelial cells (BLEC) model, and a primary animal (bovine endothelial/rat astrocytes co-culture) model. For each compound, quantitative UHPLC-MS/MS methods in the range of 5.00-500ng/mL in the corresponding matrix were developed, and permeability coefficients in the three BBB models were determined. In vitro predictions from the two human BBB models were in good agreement, while permeability data from the animal model differed to some extent, possibly due to protein binding of the screened compounds. In all three BBB models, piperine and SCT-64 displayed the highest BBB permeation potential. This was corroborated by data from in silico prediction. For the other piperine analogs (SCT-66, SCT-29, LAU397, and LAU399), BBB permeability was low to moderate in the two human BBB models, and moderate to high in the animal BBB model. Efflux ratios (ER) calculated from bidirectional permeability experiments indicated that the compounds were likely not substrates of active efflux transporters.
Collapse
Affiliation(s)
- Daniela Elisabeth Eigenmann
- Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Carmen Dürig
- Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Evelyn Andrea Jähne
- Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Martin Smieško
- Molecular Modeling, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Maxime Culot
- Univ. Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300 Lens Cedex, France
| | - Fabien Gosselet
- Univ. Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300 Lens Cedex, France
| | - Romeo Cecchelli
- Univ. Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), F-62300 Lens Cedex, France
| | - Hans Christian Cederberg Helms
- Drug Transporters in ADME, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Birger Brodin
- Drug Transporters in ADME, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Laurin Wimmer
- Institute of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, A-1060 Vienna, Austria
| | - Marko D Mihovilovic
- Institute of Applied Synthetic Chemistry, TU Wien, Getreidemarkt 9, A-1060 Vienna, Austria
| | - Matthias Hamburger
- Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Mouhssin Oufir
- Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland.
| |
Collapse
|
262
|
Glucose Transporters at the Blood-Brain Barrier: Function, Regulation and Gateways for Drug Delivery. Mol Neurobiol 2016; 54:1046-1077. [PMID: 26801191 DOI: 10.1007/s12035-015-9672-6] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/17/2015] [Indexed: 12/31/2022]
Abstract
Glucose transporters (GLUTs) at the blood-brain barrier maintain the continuous high glucose and energy demands of the brain. They also act as therapeutic targets and provide routes of entry for drug delivery to the brain and central nervous system for treatment of neurological and neurovascular conditions and brain tumours. This article first describes the distribution, function and regulation of glucose transporters at the blood-brain barrier, the major ones being the sodium-independent facilitative transporters GLUT1 and GLUT3. Other GLUTs and sodium-dependent transporters (SGLTs) have also been identified at lower levels and under various physiological conditions. It then considers the effects on glucose transporter expression and distribution of hypoglycemia and hyperglycemia associated with diabetes and oxygen/glucose deprivation associated with cerebral ischemia. A reduction in glucose transporters at the blood-brain barrier that occurs before the onset of the main pathophysiological changes and symptoms of Alzheimer's disease is a potential causative effect in the vascular hypothesis of the disease. Mutations in glucose transporters, notably those identified in GLUT1 deficiency syndrome, and some recreational drug compounds also alter the expression and/or activity of glucose transporters at the blood-brain barrier. Approaches for drug delivery across the blood-brain barrier include the pro-drug strategy whereby drug molecules are conjugated to glucose transporter substrates or encapsulated in nano-enabled delivery systems (e.g. liposomes, micelles, nanoparticles) that are functionalised to target glucose transporters. Finally, the continuous development of blood-brain barrier in vitro models is important for studying glucose transporter function, effects of disease conditions and interactions with drugs and xenobiotics.
Collapse
|
263
|
Validation of an immortalized human (hBMEC) in vitro blood-brain barrier model. Anal Bioanal Chem 2016; 408:2095-107. [PMID: 26790872 DOI: 10.1007/s00216-016-9313-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/12/2015] [Accepted: 01/05/2016] [Indexed: 12/28/2022]
Abstract
We recently established and optimized an immortalized human in vitro blood-brain barrier (BBB) model based on the hBMEC cell line. In the present work, we validated this mono-culture 24-well model with a representative series of drug substances which are known to cross or not to cross the BBB. For each individual compound, a quantitative UHPLC-MS/MS method in Ringer HEPES buffer was developed and validated according to current regulatory guidelines, with respect to selectivity, precision, and reliability. Various biological and analytical challenges were met during method validation, highlighting the importance of careful method development. The positive controls antipyrine, caffeine, diazepam, and propranolol showed mean endothelial permeability coefficients (P e) in the range of 17-70 × 10(-6) cm/s, indicating moderate to high BBB permeability when compared to the barrier integrity marker sodium fluorescein (mean P e 3-5 × 10(-6) cm/s). The negative controls atenolol, cimetidine, and vinblastine showed mean P e values < 10 × 10(-6) cm/s, suggesting low permeability. In silico calculations were in agreement with in vitro data. With the exception of quinidine (P-glycoprotein inhibitor and substrate), BBB permeability of all control compounds was correctly predicted by this new, easy, and fast to set up human in vitro BBB model. Addition of retinoic acid and puromycin did not increase transendothelial electrical resistance (TEER) values of the BBB model.
Collapse
|
264
|
Hayes M, Moen LF, Auty MAE, Lea TE. Transport of a Prolyl Endopeptidase Inhibitory Peptide across the Blood-Brain Barrier Demonstrated Using the hCMEC/D3 Cell Line Transcytosis Assay. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:146-150. [PMID: 26716467 DOI: 10.1021/acs.jafc.5b04696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The blood-brain barrier (BBB) remains a significant hurdle for treatment of central nervous system (CNS) and mental health disorders. A prolyl endopeptidase (PEP) inhibitory peptide with the amino acid sequence proline-proline-leucine (PPL) was chemically synthesized labeled with 5-FAM and assessed using a transcytosis assay for its ability to cross the BBB. Transport of this peptide across the BBB was determined using an in vitro model of the human BBB, which utilizes the human cerebral microvascular endothelial cell line (hCMEC/D3). Uptake and transport of 5-FAM-PPL across the hCMEC/D3 cell model was determined using confocal microscopy and mass spectrometry. This is an important parameter in determining whether peptides may reach the target organ (i.e., the brain and central nervous system).This work assessed, for the first time, the ability of a food-derived PEP inhibitory peptide to cross the BBB without the use of animal models.
Collapse
Affiliation(s)
- Maria Hayes
- Food BioSciences Department, Teagasc, The Irish Agricultural and Food Development Authority , Ashtown, Dublin 15, Ireland
| | - Lars Fredrik Moen
- Department of Chemistry, Biotechnology and Food Science, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences (NMBU) , 1432 Ås, Oslo area, Norway
| | - Mark A E Auty
- Food Chemistry and Technology Department, National Food Imaging Centre , Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Tor Erling Lea
- Department of Chemistry, Biotechnology and Food Science, Faculty of Veterinary Medicine and Biosciences, Norwegian University of Life Sciences (NMBU) , 1432 Ås, Oslo area, Norway
| |
Collapse
|
265
|
Holloway PM, Gavins FNE. Modeling Ischemic Stroke In Vitro: Status Quo and Future Perspectives. Stroke 2016; 47:561-9. [PMID: 26742797 DOI: 10.1161/strokeaha.115.011932] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 12/04/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Paul M Holloway
- From the Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA
| | - Felicity N E Gavins
- From the Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA.
| |
Collapse
|
266
|
Bischel LL, Coneski PN, Lundin JG, Wu PK, Giller CB, Wynne J, Ringeisen BR, Pirlo RK. Electrospun gelatin biopapers as substrate for in vitro bilayer models of blood-brain barrier tissue. J Biomed Mater Res A 2016; 104:901-9. [PMID: 26650896 DOI: 10.1002/jbm.a.35624] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/03/2015] [Accepted: 12/03/2015] [Indexed: 12/16/2022]
Abstract
Gaining a greater understanding of the blood-brain barrier (BBB) is critical for improvement in drug delivery, understanding pathologies that compromise the BBB, and developing therapies to protect the BBB. In vitro human tissue models are valuable tools for studying these issues. The standard in vitro BBB models use commercially available cell culture inserts to generate bilayer co-cultures of astrocytes and endothelial cells (EC). Electrospinning can be used to produce customized cell culture substrates with optimized material composition and mechanical properties with advantages over off-the-shelf materials. Electrospun gelatin is an ideal cell culture substrate because it is a natural polymer that can aid cell attachment and be modified and degraded by cells. Here, we have developed a method to produce cell culture inserts with electrospun gelatin "biopaper" membranes. The electrospun fiber diameter and cross-linking method were optimized for the growth of primary human endothelial cell and primary human astrocyte bilayer co-cultures to model human BBB tissue. BBB co-cultures on biopaper were characterized via cell morphology, trans-endothelial electrical resistance (TEER), and permeability to FITC-labeled dextran and compared to BBB co-cultures on standard cell culture inserts. Over longer culture periods (up to 21 days), cultures on the optimized electrospun gelatin biopapers were found to have improved TEER, decreased permeability, and permitted a smaller separation between co-cultured cells when compared to standard PET inserts.
Collapse
Affiliation(s)
- Lauren L Bischel
- American Society for Engineering Education Postdoctoral Fellow at the U.S. Naval Research Laboratory, Washington, DC
| | - Peter N Coneski
- American Society for Engineering Education Postdoctoral Fellow at the U.S. Naval Research Laboratory, Washington, DC
| | - Jeffrey G Lundin
- U.S. Naval Research Laboratory, Chemistry Division, Washington, DC
| | - Peter K Wu
- Department of Physics, Southern Oregon University, Ashland, Oregon
| | - Carl B Giller
- Contractor at the U.S. Naval Research Laboratory, Leidos, Washington, DC
| | - James Wynne
- U.S. Naval Research Laboratory, Chemistry Division, Washington, DC
| | - Brad R Ringeisen
- U.S. Naval Research Laboratory, Chemistry Division, Washington, DC
| | - Russell K Pirlo
- U.S. Naval Research Laboratory, Chemistry Division, Washington, DC
| |
Collapse
|
267
|
Behrens M, Hüwel S, Galla HJ, Humpf HU. Blood-Brain Barrier Effects of the Fusarium Mycotoxins Deoxynivalenol, 3 Acetyldeoxynivalenol, and Moniliformin and Their Transfer to the Brain. PLoS One 2015; 10:e0143640. [PMID: 26600019 PMCID: PMC4658139 DOI: 10.1371/journal.pone.0143640] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/07/2015] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Secondary metabolites produced by Fusarium fungi frequently contaminate food and feed and have adverse effects on human and animal health. Fusarium mycotoxins exhibit a wide structural and biosynthetic diversity leading to different toxicokinetics and toxicodynamics. Several studies investigated the toxicity of mycotoxins, focusing on very specific targets, like the brain. However, it still remains unclear how fast mycotoxins reach the brain and if they impair the integrity of the blood-brain barrier. This study investigated and compared the effects of the Fusarium mycotoxins deoxynivalenol, 3-acetyldeoxynivalenol and moniliformin on the blood-brain barrier. Furthermore, the transfer properties to the brain were analyzed, which are required for risk assessment, including potential neurotoxic effects. METHODS Primary porcine brain capillary endothelial cells were cultivated to study the effects of the examined mycotoxins on the blood-brain barrier in vitro. The barrier integrity was monitored by cellular impedance spectroscopy and 14C radiolabeled sucrose permeability measurements. The distribution of the applied toxins between blood and brain compartments of the cell monolayer was analyzed by high performance liquid chromatography-mass spectrometry to calculate transfer rates and permeability coefficients. RESULTS Deoxynivalenol reduced the barrier integrity and caused cytotoxic effects at 10 μM concentrations. Slight alterations of the barrier integrity were also detected for 3-acetyldeoxynivalenol. The latter was transferred very quickly across the barrier and additionally cleaved to deoxynivalenol. The transfer of deoxynivalenol and moniliformin was slower, but clearly exceeded the permeability of the negative control. None of the compounds was enriched in one of the compartments, indicating that no efflux transport protein is involved in their transport.
Collapse
Affiliation(s)
- Matthias Behrens
- Institute of Food Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 45, 48149, Münster, Germany
| | - Sabine Hüwel
- Institute of Biochemistry, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Str. 2, 48149, Münster, Germany
| | - Hans-Joachim Galla
- Institute of Biochemistry, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Str. 2, 48149, Münster, Germany
| | - Hans-Ulrich Humpf
- Institute of Food Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstr. 45, 48149, Münster, Germany
- * E-mail:
| |
Collapse
|
268
|
Loureiro JA, Gomes B, Coelho MAN, do Carmo Pereira M, Rocha S. Immunoliposomes doubly targeted to transferrin receptor and to α-synuclein. Future Sci OA 2015; 1:FSO71. [PMID: 28031922 PMCID: PMC5137902 DOI: 10.4155/fso.15.71] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/10/2015] [Indexed: 01/06/2023] Open
Abstract
AIM The present study was designed to test the cellular uptake of PEGylated liposomes targeted to transferrin receptor and to α-synuclein by a cell model of the blood-brain barrier (BBB). MATERIALS & METHODS PEGylated immunoliposomes were prepared with anti-transferrin receptor OX26 and anti-α-synuclein LB509 antibodies to overcome the BBB in Parkinson's disease. RESULTS The doubly targeted immunoliposomes bind to transferrin receptor and to α-synuclein protein, as assessed by ELISA assays. We establish that 40% of an encapsulated tested drug (epigallocatechin-3-gallate) is released in a time frame of 44 h, which is reasonable for sustained release. The cellular uptake of doubly targeted immunoliposomes in cultured brain endothelial cells hCMEC/D3 was two-times more efficient than that of PEGylated liposomes. CONCLUSION Immunoliposomes targeted to BBB receptors and to α-synuclein could potentially enable the transport of drugs across the BBB and reach one of the drug targets in Parkinson's disease.
Collapse
Affiliation(s)
- Joana A Loureiro
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Bárbara Gomes
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Manuel AN Coelho
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Maria do Carmo Pereira
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal
| | - Sandra Rocha
- Department of Biology & Biological Engineering, Chalmers University of Technology, Gothenburg SE-41296, Sweden
| |
Collapse
|
269
|
Alexandrov PN, Kruck TPA, Lukiw WJ. Nanomolar aluminum induces expression of the inflammatory systemic biomarker C-reactive protein (CRP) in human brain microvessel endothelial cells (hBMECs). J Inorg Biochem 2015; 152:210-3. [PMID: 26265215 DOI: 10.1016/j.jinorgbio.2015.07.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/07/2015] [Accepted: 07/15/2015] [Indexed: 12/16/2022]
Abstract
C-reactive protein (CRP; also known as pentraxin 1, PTX1), a 224 amino acid soluble serum protein organized into a novel pentameric ring-shaped structure, is a highly sensitive pathogenic biomarker for systemic inflammation. High CRP levels are found in practically every known inflammatory state, and elevated CRP levels indicate an increased risk for several common age-related human degenerative disorders, including cardiovascular disease, cancer, diabetes, and Alzheimer's disease (AD). While the majority of CRP is synthesized in the liver for secretion into the systemic circulation, it has recently been discovered that an appreciable amount of CRP is synthesized in highly specialized endothelial cells that line the vasculature of the brain and central nervous system (CNS). These highly specialized cells, the major cell type lining the human CNS vasculature, are known as human brain microvessel endothelial cells (hBMECs). In the current pilot study we examined (i) CRP levels in human serum obtained from AD and age-matched control patients; and (ii) analyzed the effects of nanomolar aluminum sulfate on CRP expression in primary hBMECs. The three major findings in this short communication are: (i) that CRP is up-regulated in AD serum; (ii) that CRP serum levels increased in parallel with AD progression; and (iii) for the first time show that nanomolar aluminum potently up-regulates CRP expression in hBMECs to many times its 'basal abundance'. The results suggest that aluminum-induced CRP may in part contribute to a pathophysiological state associated with a chronic systemic inflammation of the human vasculature.
Collapse
Affiliation(s)
| | - Theodore P A Kruck
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA; Department of Neurology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA.
| |
Collapse
|
270
|
Lénárt N, Walter FR, Bocsik A, Sántha P, Tóth ME, Harazin A, Tóth AE, Vizler C, Török Z, Pilbat AM, Vígh L, Puskás LG, Sántha M, Deli MA. Cultured cells of the blood-brain barrier from apolipoprotein B-100 transgenic mice: effects of oxidized low-density lipoprotein treatment. Fluids Barriers CNS 2015; 12:17. [PMID: 26184769 PMCID: PMC4504453 DOI: 10.1186/s12987-015-0013-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 07/01/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The apolipoprotein B-100 (ApoB-100) transgenic mouse line is a model of human atherosclerosis. Latest findings suggest the importance of ApoB-100 in the development of neurodegenerative diseases and microvascular/perivascular localization of ApoB-100 protein was demonstrated in the cerebral cortex of ApoB-100 transgenic mice. The aim of the study was to characterize cultured brain endothelial cells, pericytes and glial cells from wild-type and ApoB-100 transgenic mice and to study the effect of oxidized low-density lipoprotein (oxLDL) on these cells. METHODS Morphology of cells isolated from brains of wild type and ApoB-100 transgenic mice was characterized by immunohistochemistry and the intensity of immunolabeling was quantified by image analysis. Toxicity of oxLDL treatment was monitored by real-time impedance measurement and lactate dehydrogenase release. Reactive oxygen species and nitric oxide production, barrier permeability in triple co-culture blood-brain barrier model and membrane fluidity were also determined after low-density lipoprotein (LDL) or oxLDL treatment. RESULTS The presence of ApoB-100 was confirmed in brain endothelial cells, while no morphological change was observed between wild type and transgenic cells. Oxidized but not native LDL exerted dose-dependent toxicity in all three cell types, induced barrier dysfunction and increased reactive oxygen species (ROS) production in both genotypes. A partial protection from oxLDL toxicity was seen in brain endothelial and glial cells from ApoB-100 transgenic mice. Increased membrane rigidity was measured in brain endothelial cells from ApoB-100 transgenic mice and in LDL or oxLDL treated wild type cells. CONCLUSION The morphological and functional properties of cultured brain endothelial cells, pericytes and glial cells from ApoB-100 transgenic mice were characterized and compared to wild type cells for the first time. The membrane fluidity changes in ApoB-100 transgenic cells related to brain microvasculature indicate alterations in lipid composition which may be linked to the partial protection against oxLDL toxicity.
Collapse
Affiliation(s)
- Nikolett Lénárt
- Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6720, Szeged, Hungary. .,Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Fruzsina R Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Alexandra Bocsik
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Petra Sántha
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Melinda E Tóth
- Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6720, Szeged, Hungary.
| | - András Harazin
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Andrea E Tóth
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Csaba Vizler
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Zsolt Török
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Ana-Maria Pilbat
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - László Vígh
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - László G Puskás
- Laboratory of Functional Genomics, Laboratories of Core Facilities, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Miklós Sántha
- Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6720, Szeged, Hungary.
| | - Mária A Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| |
Collapse
|
271
|
Dexamethasone potentiates in vitro blood-brain barrier recovery after primary blast injury by glucocorticoid receptor-mediated upregulation of ZO-1 tight junction protein. J Cereb Blood Flow Metab 2015; 35:1191-8. [PMID: 25757751 PMCID: PMC4640274 DOI: 10.1038/jcbfm.2015.38] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/05/2015] [Accepted: 02/09/2015] [Indexed: 11/08/2022]
Abstract
Owing to the frequent incidence of blast-induced traumatic brain injury (bTBI) in recent military conflicts, there is an urgent need to develop effective therapies for bTBI-related pathologies. Blood-brain barrier (BBB) breakdown has been reported to occur after primary blast exposure, making restoration of BBB function and integrity a promising therapeutic target. We tested the hypothesis that treatment with dexamethasone (DEX) after primary blast injury potentiates recovery of an in vitro BBB model consisting of mouse brain endothelial cells (bEnd.3). DEX treatment resulted in complete recovery of transendothelial electrical resistance and hydraulic conductivity 1 day after injury, compared with 3 days for vehicle-treated injured cultures. Administration of RU486 (mifepristone) inhibited effects of DEX, confirming that barrier restoration was mediated by glucocorticoid receptor signaling. Potentiated recovery with DEX treatment was accompanied by stronger zonula occludens (ZO)-1 tight junction immunostaining and expression, suggesting that increased ZO-1 expression was a structural correlate to BBB recovery after blast. Interestingly, augmented ZO-1 protein expression was associated with specific upregulation of the α(+) isoform but not the α(-) isoform. This is the first study to provide a mechanistic basis for potentiated functional recovery of an in vitro BBB model because of glucocorticoid treatment after primary blast injury.
Collapse
|
272
|
Xiao X, Zhang C, Ma X, Miao H, Wang J, Liu L, Chen S, Zeng R, Chen Y, Bihl JC. Angiotensin-(1-7) counteracts angiotensin II-induced dysfunction in cerebral endothelial cells via modulating Nox2/ROS and PI3K/NO pathways. Exp Cell Res 2015; 336:58-65. [PMID: 26101159 DOI: 10.1016/j.yexcr.2015.06.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 11/29/2022]
Abstract
Angiotensin (Ang) II, the main effector of the renin-angiotensin system, has been implicated in the pathogenesis of vascular diseases. Ang-(1-7) binds to the G protein-coupled Mas receptor (MasR) and can exert vasoprotective effects. We investigated the effects and underlying mechanisms of Ang-(1-7) on Ang II-induced dysfunction and oxidative stress in human brain microvascular endothelial cells (HbmECs). The pro-apoptotic activity, reactive oxygen species (ROS) and nitric oxide (NO) productions in HbmECs were measured. The protein expressions of nicotinamide adenine dinucleotide phosphate oxidase 2 (Nox2), serine/threonine kinase (Akt), endothelial nitric oxide synthase (eNOS) and their phosphorylated forms (p-Akt and p-eNOS) were examined by western blot. MasR antagonist and phosphatidylinositol-3-kinase (PI3K) inhibitor were used for receptor/pathway verification. We found that Ang-(1-7) suppressed Ang II-induced pro-apoptotic activity, ROS over-production and NO reduction in HbmECs, which were abolished by MasR antagonist. In addition, Ang-(1-7) down-regulated the expression of Nox2, and up-regulated the ratios of p-Akt/Akt and its downstream p-eNOS/eNOS in HbmECs. Exposure to PI3K inhibitor partially abrogated Ang-(1-7)-mediated protective effects in HbmECs. Our data suggests that Ang-(1-7)/MasR axis protects HbmECs from Ang II-induced dysfunction and oxidative stress via inhibition of Nox2/ROS and activation of PI3K/NO pathways.
Collapse
Affiliation(s)
- Xiang Xiao
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Cheng Zhang
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Xiaotang Ma
- Institute of Neurology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Huilai Miao
- Department of Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Jinju Wang
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Langni Liu
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Shuzhen Chen
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Rong Zeng
- Department of Surgery, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Yanfang Chen
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA.
| | - Ji C Bihl
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA; Institute of Neurology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China.
| |
Collapse
|
273
|
Wilson HK, Canfield SG, Hjortness MK, Palecek SP, Shusta EV. Exploring the effects of cell seeding density on the differentiation of human pluripotent stem cells to brain microvascular endothelial cells. Fluids Barriers CNS 2015; 12:13. [PMID: 25994964 PMCID: PMC4455681 DOI: 10.1186/s12987-015-0007-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/24/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Brain microvascular-like endothelial cells (BMECs) derived from human pluripotent stem cells (hPSCs) have significant promise as tools for drug screening and studying the structure and function of the BBB in health and disease. The density of hPSCs is a key factor in regulating cell fate and yield during differentiation. Prior reports of hPSC differentiation to BMECs have seeded hPSCs in aggregates, leading to non-uniform cell densities that may result in differentiation heterogeneity. Here we report a singularized-cell seeding approach compatible with hPSC-derived BMEC differentiation protocols and evaluate the effects of initial hPSC seeding density on the subsequent differentiation, yield, and blood-brain barrier (BBB) phenotype. METHODS A range of densities of hPSCs was seeded and differentiated, with the resultant endothelial cell yield quantified via VE-cadherin flow cytometry. Barrier phenotype of purified hPSC-derived BMECs was measured via transendothelial electrical resistance (TEER), and purification protocols were subsequently optimized to maximize TEER. Expression of characteristic vascular markers, tight junction proteins, and transporters was confirmed by immunocytochemistry and quantified by flow cytometry. P-glycoprotein and MRP-family transporter activity was assessed by intracellular accumulation assay. RESULTS The initial hPSC seeding density of approximately 30,000 cells/cm(2) served to maximize the yield of VE-cadherin+ BMECs per input hPSC. BMECs displayed the highest TEER (>2,000 Ω × cm(2)) within this same range of initial seeding densities, although optimization of the BMEC purification method could minimize the seeding density dependence for some lines. Localization and expression levels of tight junction proteins as well as efflux transporter activity were largely independent of hPSC seeding density. Finally, the utility of the singularized-cell seeding approach was demonstrated by scaling the differentiation and purification process down from 6-well to 96-well culture without impacting BBB phenotype. CONCLUSIONS Given the yield and barrier dependence on initial seeding density, the singularized-cell seeding approach reported here should enhance the reproducibility and scalability of hPSC-derived BBB models, particularly for the application to new pluripotent stem cell lines.
Collapse
Affiliation(s)
- Hannah K Wilson
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Scott G Canfield
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Michael K Hjortness
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| |
Collapse
|
274
|
Palmela I, Correia L, Silva RFM, Sasaki H, Kim KS, Brites D, Brito MA. Hydrophilic bile acids protect human blood-brain barrier endothelial cells from disruption by unconjugated bilirubin: an in vitro study. Front Neurosci 2015; 9:80. [PMID: 25821432 PMCID: PMC4358072 DOI: 10.3389/fnins.2015.00080] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Accepted: 02/24/2015] [Indexed: 12/13/2022] Open
Abstract
Ursodeoxycholic acid and its main conjugate glycoursodeoxycholic acid are bile acids with neuroprotective properties. Our previous studies demonstrated their anti-apoptotic, anti-inflammatory, and antioxidant properties in neural cells exposed to elevated levels of unconjugated bilirubin (UCB) as in severe jaundice. In a simplified model of the blood-brain barrier, formed by confluent monolayers of a cell line of human brain microvascular endothelial cells, UCB has shown to induce caspase-3 activation and cell death, as well as interleukin-6 release and a loss of blood-brain barrier integrity. Here, we tested the preventive and restorative effects of these bile acids regarding the disruption of blood-brain barrier properties by UCB in in vitro conditions mimicking severe neonatal hyperbilirubinemia and using the same experimental blood-brain barrier model. Both bile acids reduced the apoptotic cell death induced by UCB, but only glycoursodeoxycholic acid significantly counteracted caspase-3 activation. Bile acids also prevented the upregulation of interleukin-6 mRNA, whereas only ursodeoxycholic acid abrogated cytokine release. Regarding barrier integrity, only ursodeoxycholic acid abrogated UCB-induced barrier permeability. Better protective effects were obtained by bile acid pre-treatment, but a strong efficacy was still observed by their addition after UCB treatment. Finally, both bile acids showed ability to cross confluent monolayers of human brain microvascular endothelial cells in a time-dependent manner. Collectively, data disclose a therapeutic time-window for preventive and restorative effects of ursodeoxycholic acid and glycoursodeoxycholic acid against UCB-induced blood-brain barrier disruption and damage to human brain microvascular endothelial cells.
Collapse
Affiliation(s)
- Inês Palmela
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| | - Leonor Correia
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| | - Rui F M Silva
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| | - Hiroyuki Sasaki
- Division of Fine Morphology, Core Research Facilities, The Jikei University School of Medicine Tokyo Japan
| | - Kwang S Kim
- Division of Infectious Diseases, Johns Hopkins University School of Medicine Baltimore, MA, USA
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| | - Maria A Brito
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisbon, Portugal
| |
Collapse
|
275
|
Hydrocortisone enhances the barrier properties of HBMEC/ciβ, a brain microvascular endothelial cell line, through mesenchymal-to-endothelial transition-like effects. Fluids Barriers CNS 2015; 12:7. [PMID: 25763180 PMCID: PMC4355132 DOI: 10.1186/s12987-015-0003-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 02/22/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Because in vitro blood-brain barrier (BBB) models are important tools for studying brain diseases and drug development, we recently established a new line of conditionally immortalized human brain microvascular endothelial cells (HBMEC/ciβ) for use in such models. Since one of the most important functional features of the BBB is its strong intercellular adhesion, in this study, we aimed at improving HBMEC/ciβ barrier properties by means of culture media modifications, thus enhancing their use for future BBB studies. In addition, we simultaneously attempted to obtain insights on related mechanistic properties. METHODS Several types of culture media were prepared in an effort to identify the medium most suitable for culturing HBMEC/ciβ. The barrier properties of HBMEC/ciβ were examined by determining Na(+)-fluorescein permeability and transendothelial electric resistance (TEER). Endothelial marker mRNA expression levels were determined by quantitative real-time polymerase chain reaction. Adherens junction (AJ) formation was examined by immunocytochemistry. Cell migration ability was analyzed by scratch assay. Furthermore, cellular lipid composition was examined by liquid chromatography-time-of-flight mass spectrometry. RESULTS Our initial screening tests showed that addition of hydrocortisone (HC) to the basal medium significantly reduced the Na(+)-fluorescein permeability and increased the TEER of HBMEC/ciβ monolayers. It was also found that, while AJ proteins were diffused in the cytoplasm of HBMEC/ciβ cultured without HC, those expressed in cells cultured with HC were primarily localized at the cell border. Furthermore, this facilitation of AJ formation by HC was in concert with increased endothelial marker mRNA levels and increased ether-type phosphatidylethanolamine levels, while cell migration was retarded in the presence of HC. CONCLUSIONS Our results show that HC supplementation to the basal medium significantly enhances the barrier properties of HBMEC/ciβ. This was associated with a marked phenotypic alteration in HBMEC/ciβ through orchestration of various signaling pathways. Taken together, it appears that overall effects of HC on HBMEC/ciβ could be summarized as facilitating endothelial differentiation characteristics while concurrently retarding mesenchymal characteristics.
Collapse
|
276
|
Reinitz A, DeStefano J, Ye M, Wong AD, Searson PC. Human brain microvascular endothelial cells resist elongation due to shear stress. Microvasc Res 2015; 99:8-18. [PMID: 25725258 DOI: 10.1016/j.mvr.2015.02.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 02/02/2023]
Abstract
Endothelial cells in straight sections of vessels are known to elongate and align in the direction of flow. This phenotype has been replicated in confluent monolayers of bovine aortic endothelial cells and human umbilical vein endothelial cells (HUVECs) in cell culture under physiological shear stress. Here we report on the morphological response of human brain microvascular endothelial cells (HBMECs) in confluent monolayers in response to shear stress. Using a microfluidic platform we image confluent monolayers of HBMECs and HUVECs under shear stresses up to 16 dyne cm(-2). From live-cell imaging we quantitatively analyze the cell morphology and cell speed as a function of time. We show that HBMECs do not undergo a classical transition from cobblestone to spindle-like morphology in response to shear stress. We further show that under shear stress, actin fibers are randomly oriented in the cells indicating that there is no cytoskeletal remodeling. These results suggest that HBMECs are programmed to resist elongation and alignment under shear stress, a phenotype that may be associated with the unique properties of the blood-brain barrier.
Collapse
Affiliation(s)
- Adam Reinitz
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Jackson DeStefano
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Mao Ye
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Andrew D Wong
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Peter C Searson
- Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
277
|
Hopkins AM, DeSimone E, Chwalek K, Kaplan DL. 3D in vitro modeling of the central nervous system. Prog Neurobiol 2015; 125:1-25. [PMID: 25461688 PMCID: PMC4324093 DOI: 10.1016/j.pneurobio.2014.11.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 10/12/2014] [Accepted: 11/15/2014] [Indexed: 12/15/2022]
Abstract
There are currently more than 600 diseases characterized as affecting the central nervous system (CNS) which inflict neural damage. Unfortunately, few of these conditions have effective treatments available. Although significant efforts have been put into developing new therapeutics, drugs which were promising in the developmental phase have high attrition rates in late stage clinical trials. These failures could be circumvented if current 2D in vitro and in vivo models were improved. 3D, tissue-engineered in vitro systems can address this need and enhance clinical translation through two approaches: (1) bottom-up, and (2) top-down (developmental/regenerative) strategies to reproduce the structure and function of human tissues. Critical challenges remain including biomaterials capable of matching the mechanical properties and extracellular matrix (ECM) composition of neural tissues, compartmentalized scaffolds that support heterogeneous tissue architectures reflective of brain organization and structure, and robust functional assays for in vitro tissue validation. The unique design parameters defined by the complex physiology of the CNS for construction and validation of 3D in vitro neural systems are reviewed here.
Collapse
Affiliation(s)
- Amy M Hopkins
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Elise DeSimone
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - Karolina Chwalek
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Science & Technology Center, 4 Colby Street, Medford, MA 02155, USA.
| |
Collapse
|
278
|
Wolff A, Antfolk M, Brodin B, Tenje M. In Vitro Blood-Brain Barrier Models-An Overview of Established Models and New Microfluidic Approaches. J Pharm Sci 2015; 104:2727-46. [PMID: 25630899 DOI: 10.1002/jps.24329] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 12/25/2022]
Abstract
The societal need for new central nervous system (CNS) medicines is substantial, because of the global increase in life expectancy and the accompanying increase in age-related CNS diseases. Low blood-brain barrier (BBB) permeability has been one of the major causes of failure for new CNS drug candidates. There has therefore been a great interest in cell models, which mimic BBB permeation properties. In this review, we present an overview of the performance of monocultured, cocultured, and triple-cultured primary cells and immortalized cell lines, including key parameters such as transendothelial electrical resistance values, permeabilities of paracellular flux markers, and expression of BBB-specific marker proteins. Microfluidic systems are gaining ground as a new automated technical platform for cell culture and systematic analysis. The performance of these systems was compared with current state-of-the-art models and it was noted that, although they show great promise, these systems have not yet reached beyond the proof-of-concept stage. In general, it was found that there were large variations in experimental protocols, BBB phenotype markers, and paracellular flux markers used. It is the author's opinion that the field may benefit greatly from developing standardized methodologies and initiating collaborative efforts on optimizing culture protocols.
Collapse
Affiliation(s)
- Anette Wolff
- Lund University, Department of Biomedical Engineering, Lund, Sweden
| | - Maria Antfolk
- Lund University, Department of Biomedical Engineering, Lund, Sweden
| | - Birger Brodin
- University of Copenhagen, Department of Pharmacy, Copenhagen, Denmark
| | - Maria Tenje
- Lund University, Department of Biomedical Engineering, Lund, Sweden.,Uppsala University, Department of Engineering Sciences, Uppsala, Sweden
| |
Collapse
|
279
|
Kim JH, Cho MH, Choi KC, Lee K, Kim KS, Shim SM. Oxidative Stress Induced by Cigarette Smoke Extracts in Human Brain Cells (T98G) and Human Brain Microvascular Endothelial Cells (HBMEC) in Mono- and Co-Culture. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2015; 78:1019-27. [PMID: 26262444 DOI: 10.1080/15287394.2015.1043607] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The objective of the current study was to examine oxidative stress induced by cigarette smoke extract (CSE) or cigarette smoke condensate (CSC) in human brain cells (T98G) and human brain microvascular endothelial cells (HBMEC) in mono- and co-culture systems. Cell viability of T98G cells exposed to CSC (0.05-4 mg/ml) was significantly decreased compared to CSE (0.025-20%). There were no marked differences between quantities of reactive oxygen species (ROS) generation by either CSE (2, 4, and 10%) or CSC (0.2, 0.4, and 0.8 mg/ml) treatment compared to control. However, a significant effect was noted in ROS generation following CSC incubation at 4mg/ml. Cellular integrity of HBMEC decreased to 74 and 64% within 120 h of exposure at the IC50 value of CSE and CSC, respectively. This study suggests that chronic exposure to cigarette smoking might initiate damage to the blood-brain barrier.
Collapse
Affiliation(s)
- Ju-Hyeong Kim
- a Department of Food Science and Technology , Sejong University , Seoul , Republic of Korea
| | | | | | | | | | | |
Collapse
|
280
|
Sándor N, Walter FR, Bocsik A, Sántha P, Schilling-Tóth B, Léner V, Varga Z, Kahán Z, Deli MA, Sáfrány G, Hegyesi H. Low dose cranial irradiation-induced cerebrovascular damage is reversible in mice. PLoS One 2014; 9:e112397. [PMID: 25393626 PMCID: PMC4231057 DOI: 10.1371/journal.pone.0112397] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/03/2014] [Indexed: 11/21/2022] Open
Abstract
Background High-dose radiation-induced blood-brain barrier breakdown contributes to acute radiation toxicity syndrome and delayed brain injury, but there are few data on the effects of low dose cranial irradiation. Our goal was to measure blood-brain barrier changes after low (0.1 Gy), moderate (2 Gy) and high (10 Gy) dose irradiation under in vivo and in vitro conditions. Methodology Cranial irradiation was performed on 10-day-old and 10-week-old mice. Blood-brain barrier permeability for Evans blue, body weight and number of peripheral mononuclear and circulating endothelial progenitor cells were evaluated 1, 4 and 26 weeks postirradiation. Barrier properties of primary mouse brain endothelial cells co-cultured with glial cells were determined by measurement of resistance and permeability for marker molecules and staining for interendothelial junctions. Endothelial senescence was determined by senescence associated β-galactosidase staining. Principle Findings Extravasation of Evans blue increased in cerebrum and cerebellum in adult mice 1 week and in infant mice 4 weeks postirradiation at all treatment doses. Head irradiation with 10 Gy decreased body weight. The number of circulating endothelial progenitor cells in blood was decreased 1 day after irradiation with 0.1 and 2 Gy. Increase in the permeability of cultured brain endothelial monolayers for fluorescein and albumin was time- and radiation dose dependent and accompanied by changes in junctional immunostaining for claudin-5, ZO-1 and β-catenin. The number of cultured brain endothelial and glial cells decreased from third day of postirradiation and senescence in endothelial cells increased at 2 and 10 Gy. Conclusion Not only high but low and moderate doses of cranial irradiation increase permeability of cerebral vessels in mice, but this effect is reversible by 6 months. In-vitro experiments suggest that irradiation changes junctional morphology, decreases cell number and causes senescence in brain endothelial cells.
Collapse
Affiliation(s)
- Nikolett Sándor
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
- Doctoral Schools of Pathological Sciences, Semmelweis University, Budapest, Hungary
| | - Fruzsina R. Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Alexandra Bocsik
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Petra Sántha
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Boglárka Schilling-Tóth
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Violetta Léner
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
- Department of Morphology and Physiology, Faculty of Health Care, Semmelweis University, Budapest, Hungary
| | - Zoltán Varga
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Kahán
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Mária A. Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Géza Sáfrány
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
| | - Hargita Hegyesi
- Division of Molecular Radiobiology and Biodosimetry, “Frédéric Joliot-Curie” National Research Institute for Radiobiology and Radiohygiene, Budapest, Hungary
- Department of Morphology and Physiology, Faculty of Health Care, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
281
|
Stanimirovic DB, Bani-Yaghoub M, Perkins M, Haqqani AS. Blood-brain barrier models: in vitro to in vivo translation in preclinical development of CNS-targeting biotherapeutics. Expert Opin Drug Discov 2014; 10:141-55. [PMID: 25388782 DOI: 10.1517/17460441.2015.974545] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The majority of therapeutics, small molecule or biologics, developed for the CNS do not penetrate the blood-brain barrier (BBB) sufficiently to induce pharmacologically meaningful effects on CNS targets. To improve the efficiency of CNS drug discovery, several in vitro models of the BBB have been used to aid early selection of molecules with CNS exposure potential. However, correlative studies suggest relatively poor predictability of in vitro BBB models underscoring the need to combine in vitro and in vivo BBB penetration assessment into an integrated preclinical workflow. AREAS COVERED This review gives a brief general overview of in vitro and in vivo BBB models used in the pre-clinical evaluation of CNS-targeting drugs, with particular focus on the recent progress in developing humanized models. The authors discuss the advantages, limitations, in vitro-in vivo correlation, and integration of these models into CNS drug discovery and development with the aim of improving translation. EXPERT OPINION Often, a simplistic rationalization of the CNS drug discovery and development process overlooks or even ignores the need for an early and predictive assessment of the BBB permeability. Indeed, past failures of CNS candidates in clinical trials argue strongly that the early deployment of in vitro and in vivo models for assessing BBB permeability, mechanisms of transport and brain exposure of leads, and the co-development of BBB delivery strategies will improve translation and increase the clinical success of CNS pipelines.
Collapse
Affiliation(s)
- Danica B Stanimirovic
- Human Health Therapeutics Portfolio, National Research Council of Canada , 1200 Montreal Road, Bldg M-54 Ottawa, ON K4P 1R7 , Canada +1 613 993 3730 ; +1 613 941 4475 ;
| | | | | | | |
Collapse
|
282
|
Dando SJ, Mackay-Sim A, Norton R, Currie BJ, St John JA, Ekberg JAK, Batzloff M, Ulett GC, Beacham IR. Pathogens penetrating the central nervous system: infection pathways and the cellular and molecular mechanisms of invasion. Clin Microbiol Rev 2014; 27:691-726. [PMID: 25278572 PMCID: PMC4187632 DOI: 10.1128/cmr.00118-13] [Citation(s) in RCA: 289] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The brain is well protected against microbial invasion by cellular barriers, such as the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB). In addition, cells within the central nervous system (CNS) are capable of producing an immune response against invading pathogens. Nonetheless, a range of pathogenic microbes make their way to the CNS, and the resulting infections can cause significant morbidity and mortality. Bacteria, amoebae, fungi, and viruses are capable of CNS invasion, with the latter using axonal transport as a common route of infection. In this review, we compare the mechanisms by which bacterial pathogens reach the CNS and infect the brain. In particular, we focus on recent data regarding mechanisms of bacterial translocation from the nasal mucosa to the brain, which represents a little explored pathway of bacterial invasion but has been proposed as being particularly important in explaining how infection with Burkholderia pseudomallei can result in melioidosis encephalomyelitis.
Collapse
Affiliation(s)
- Samantha J Dando
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Alan Mackay-Sim
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Robert Norton
- Townsville Hospital, Townsville, Queensland, Australia
| | - Bart J Currie
- Menzies School of Health Research and Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - James A St John
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Jenny A K Ekberg
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Michael Batzloff
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C Ulett
- School of Medical Science and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Ifor R Beacham
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
283
|
Pinzón-Daza ML, Salaroglio IC, Kopecka J, Garzòn R, Couraud PO, Ghigo D, Riganti C. The cross-talk between canonical and non-canonical Wnt-dependent pathways regulates P-glycoprotein expression in human blood-brain barrier cells. J Cereb Blood Flow Metab 2014; 34:1258-69. [PMID: 24896565 PMCID: PMC4126086 DOI: 10.1038/jcbfm.2014.100] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/19/2014] [Accepted: 04/13/2014] [Indexed: 12/11/2022]
Abstract
In this work, we investigate if and how transducers of the 'canonical' Wnt pathway, i.e., Wnt/glycogen synthase kinase 3 (GSK3)/β-catenin, and transducers of the 'non-canonical' Wnt pathway, i.e., Wnt/RhoA/RhoA kinase (RhoAK), cooperate to control the expression of P-glycoprotein (Pgp) in blood-brain barrier (BBB) cells. By analyzing human primary brain microvascular endothelial cells constitutively activated for RhoA, silenced for RhoA or treated with the RhoAK inhibitor Y27632, we found that RhoAK phosphorylated and activated the protein tyrosine phosphatase 1B (PTP1B), which dephosphorylated tyrosine 216 of GSK3, decreasing the GSK3-mediated inhibition of β-catenin. By contrast, the inhibition of RhoA/RhoAK axis prevented the activation of PTP1B, enhanced the GSK3-induced phosphorylation and ubiquitination of β-catenin, and reduced the β-catenin-driven transcription of Pgp. The RhoAK inhibition increased the delivery of Pgp substrates like doxorubicin across the BBB and improved the doxorubicin efficacy against glioblastoma cells co-cultured under a BBB monolayer. Our data demonstrate that in human BBB cells the expression of Pgp is controlled by a cross-talk between canonical and non-canonical Wnt pathways. The disruption of this cross-talk, e.g., by inhibiting RhoAK, downregulates Pgp and increases the delivery of Pgp substrates across the BBB.
Collapse
Affiliation(s)
- Martha L Pinzón-Daza
- 1] Department of Oncology, School of Medicine, University of Turin, Turin, Italy [2] Unidad de Bioquímica, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Iris C Salaroglio
- Department of Oncology, School of Medicine, University of Turin, Turin, Italy
| | - Joanna Kopecka
- Department of Oncology, School of Medicine, University of Turin, Turin, Italy
| | - Ruth Garzòn
- Unidad de Bioquímica, Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Pierre-Olivier Couraud
- Institut Cochin, Centre National de la Recherche Scientifique UMR 8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U567, Université René Descartes, Paris, France
| | - Dario Ghigo
- 1] Department of Oncology, School of Medicine, University of Turin, Turin, Italy [2] Center for Experimental Research and Medical Studies, University of Turin, Turin, Italy
| | - Chiara Riganti
- 1] Department of Oncology, School of Medicine, University of Turin, Turin, Italy [2] Center for Experimental Research and Medical Studies, University of Turin, Turin, Italy
| |
Collapse
|
284
|
Winger RC, Koblinski JE, Kanda T, Ransohoff RM, Muller WA. Rapid remodeling of tight junctions during paracellular diapedesis in a human model of the blood-brain barrier. THE JOURNAL OF IMMUNOLOGY 2014; 193:2427-37. [PMID: 25063869 DOI: 10.4049/jimmunol.1400700] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Leukocyte transendothelial migration (TEM; diapedesis) is a critical event in immune surveillance and inflammation. Most TEM occurs at endothelial cell borders (paracellular). However, there is indirect evidence to suggest that, at the tight junctions of the blood-brain barrier (BBB), leukocytes migrate directly through the endothelial cell body (transcellular). Why leukocytes migrate through the endothelial cell body rather than the cell borders is unknown. To test the hypothesis that the tightness of endothelial cell junctions influences the pathway of diapedesis, we developed an in vitro model of the BBB that possessed 10-fold higher electrical resistance than standard culture conditions and strongly expressed the BBB tight junction proteins claudin-5 and claudin-3. We found that paracellular TEM was still the predominant pathway (≥98%) and TEM was dependent on PECAM-1 and CD99. We show that endothelial tight junctions expressing claudin-5 are dynamic and undergo rapid remodeling during TEM. Membrane from the endothelial lateral border recycling compartment is mobilized to the exact site of tight junction remodeling. This preserves the endothelial barrier by sealing the intercellular gaps with membrane and engaging the migrating leukocyte with unligated adhesion molecules (PECAM-1 and CD99) as it crosses the cell border. These findings provide new insights into leukocyte-endothelial interactions at the BBB and suggest that tight junctions are more dynamic than previously appreciated.
Collapse
Affiliation(s)
- Ryan C Winger
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Jennifer E Koblinski
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan; and
| | - Richard M Ransohoff
- Neuroinflammation Research Center, Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - William A Muller
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611;
| |
Collapse
|
285
|
Pamies D, Hartung T, Hogberg HT. Biological and medical applications of a brain-on-a-chip. Exp Biol Med (Maywood) 2014; 239:1096-1107. [PMID: 24912505 DOI: 10.1177/1535370214537738] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The desire to develop and evaluate drugs as potential countermeasures for biological and chemical threats requires test systems that can also substitute for the clinical trials normally crucial for drug development. Current animal models have limited predictivity for drug efficacy in humans as the large majority of drugs fails in clinical trials. We have limited understanding of the function of the central nervous system and the complexity of the brain, especially during development and neuronal plasticity. Simple in vitro systems do not represent physiology and function of the brain. Moreover, the difficulty of studying interactions between human genetics and environmental factors leads to lack of knowledge about the events that induce neurological diseases. Microphysiological systems (MPS) promise to generate more complex in vitro human models that better simulate the organ's biology and function. MPS combine different cell types in a specific three-dimensional (3D) configuration to simulate organs with a concrete function. The final aim of these MPS is to combine different "organoids" to generate a human-on-a-chip, an approach that would allow studies of complex physiological organ interactions. The recent discovery of induced pluripotent stem cells (iPSCs) gives a range of possibilities allowing cellular studies of individuals with different genetic backgrounds (e.g., human disease models). Application of iPSCs from different donors in MPS gives the opportunity to better understand mechanisms of the disease and can be a novel tool in drug development, toxicology, and medicine. In order to generate a brain-on-a-chip, we have established a 3D model from human iPSCs based on our experience with a 3D rat primary aggregating brain model. After four weeks of differentiation, human 3D aggregates stain positive for different neuronal markers and show higher gene expression of various neuronal differentiation markers compared to 2D cultures. Here we present the applications and challenges of this emerging technology.
Collapse
Affiliation(s)
- David Pamies
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| | - Thomas Hartung
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| | - Helena T Hogberg
- Centers for Alternatives to Animal Testing (CAAT) at Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD 21205, USA; University of Konstanz, POB 600, Konstanz 78457, Germany
| |
Collapse
|
286
|
Jähne EA, Eigenmann DE, Culot M, Cecchelli R, Walter FR, Deli MA, Tremmel R, Fricker G, Smiesko M, Hamburger M, Oufir M. Development and validation of a LC-MS/MS method for assessment of an anti-inflammatory indolinone derivative by in vitro blood-brain barrier models. J Pharm Biomed Anal 2014; 98:235-46. [PMID: 24949819 DOI: 10.1016/j.jpba.2014.05.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 12/18/2022]
Abstract
The compound (E,Z)-3-(4-hydroxy-3,5-dimethoxybenzylidene)indolin-2-one (indolinone) was identified from lipophilic woad extracts (Isatis tinctoria L., Brassicaceae) as a compound possessing potent histamine release inhibitory and anti-inflammatory properties [1]. To further evaluate the potential of indolinone in terms of crossing the blood-brain barrier (BBB), we screened the compound in several in vitro cell-based human and animal BBB models. Therefore, we developed a quantitative LC-MS/MS method for the compound in modified Ringer HEPES buffer (RHB) and validated it according to FDA and EMA guidelines [2,3]. The calibration curve of indolinone in the range between 30.0 and 3000ng/ml was quadratic, and the limit of quantification was 30.0ng/ml. Dilution of samples up to 100-fold did not affect precision and accuracy. The carry-over was within acceptance criteria. Indolinone proved to be stable in RHB for 3h at room temperature (RT), and for three successive freeze/thaw cycles. The processed samples could be stored in the autosampler at 10°C for at least 28h. Moreover, indolinone was stable for at least 16 days in RHB when stored below -65°C. This validation study demonstrates that our method is specific, selective, precise, accurate, and capable to produce reliable results. In the immortalized human BBB mono-culture model, the apparent permeability coefficient from apical to basolateral (PappA→B), and the Papp from basolateral to apical (PappB→A) were 19.2±0.485×10(-6)cm/s and 21.7±0.326×10(-6)cm/s, respectively. For the primary rat/bovine BBB co-culture model a PappA→B of 27.1±1.67×10(-6)cm/s was determined. In the primary rat BBB triple co-culture model, the PappA→B and the PappB→A were 56.2±3.63×10(-6)cm/s and 34.6±1.41×10(-6)cm/s, respectively. The data obtained with the different models showed good correlation and were indicative of a high BBB permeation potential of indolinone confirmed by in silico prediction calculations. P-glycoprotein (P-gp) interaction for indolinone was studied with the aid of a calcein-AM uptake assay, and by calculation of the efflux ratio (ER) from the bidirectional permeability assays. For both bidirectional BBB models an ER below 2 was calculated, indicating that no active mediated transport mechanism is involved for indolinone. In porcine brain capillary endothelial cells (PBCECs), the calcein-AM uptake assay demonstrated that indolinone is neither a P-gp substrate nor a P-gp inhibitor and is accumulated into cells at high extent.
Collapse
Affiliation(s)
- Evelyn A Jähne
- Institute of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Daniela E Eigenmann
- Institute of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Maxime Culot
- Université Lille Nord de France, UArtois, BBB Laboratory EA 2465, IMPRT: IFR114, 62307 Lens Cedex, France
| | - Roméo Cecchelli
- Université Lille Nord de France, UArtois, BBB Laboratory EA 2465, IMPRT: IFR114, 62307 Lens Cedex, France
| | - Fruzsina R Walter
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvari krt. 62, H-6726 Szeged, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Temesvari krt. 62, H-6726 Szeged, Hungary
| | - Robin Tremmel
- Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 329, D-69120 Heidelberg, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 329, D-69120 Heidelberg, Germany
| | - Martin Smiesko
- Molecular Modeling, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Matthias Hamburger
- Institute of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Mouhssin Oufir
- Institute of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland.
| |
Collapse
|
287
|
Wilhelm I, Krizbai IA. In vitro models of the blood-brain barrier for the study of drug delivery to the brain. Mol Pharm 2014; 11:1949-63. [PMID: 24641309 DOI: 10.1021/mp500046f] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The most important obstacle to the drug delivery into the brain is the presence of the blood-brain barrier, which limits the traffic of substances between the blood and the nervous tissue. Therefore, adequate in vitro models need to be developed in order to characterize the penetration properties of drug candidates into the central nervous system. This review article summarizes the presently used and the most promising in vitro BBB models based on the culture of brain endothelial cells. Robust models can be obtained using primary porcine brain endothelial cells and rodent coculture models, which have low paracellular permeability and express functional efflux transporters, showing good correlation of drug penetration data with in vivo results. Models mimicking the in vivo anatomophysiological complexity of the BBB are also available, including triple coculture (culture of brain endothelial cells in the presence of pericytes and astrocytes), dynamic, and microfluidic models; however, these are not suitable for rapid, high throughput studies. Potent human cell lines would be needed for easily available and reproducible models which avoid interspecies differences.
Collapse
Affiliation(s)
- Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences , Temesvári krt. 62, 6726 Szeged, Hungary
| | | |
Collapse
|
288
|
Brain microvascular endothelial cells resist elongation due to curvature and shear stress. Sci Rep 2014; 4:4681. [PMID: 24732421 PMCID: PMC3986701 DOI: 10.1038/srep04681] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/14/2014] [Indexed: 11/08/2022] Open
Abstract
The highly specialized endothelial cells in brain capillaries are a key component of the blood-brain barrier, forming a network of tight junctions that almost completely block paracellular transport. In contrast to vascular endothelial cells in other organs, we show that brain microvascular endothelial cells resist elongation in response to curvature and shear stress. Since the tight junction network is defined by endothelial cell morphology, these results suggest that there may be an evolutionary advantage to resisting elongation by minimizing the total length of cell-cell junctions per unit length of vessel.
Collapse
|
289
|
Influence of basement membrane proteins and endothelial cell-derived factors on the morphology of human fetal-derived astrocytes in 2D. PLoS One 2014; 9:e92165. [PMID: 24647106 PMCID: PMC3960172 DOI: 10.1371/journal.pone.0092165] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/19/2014] [Indexed: 12/13/2022] Open
Abstract
Astrocytes are the most prevalent type of glial cell in the brain, participating in a variety of diverse functions from regulating cerebral blood flow to controlling synapse formation. Astrocytes and astrocyte-conditioned media are widely used in models of the blood-brain barrier (BBB), however, very little is known about astrocyte culture in 2D. To test the hypothesis that surface coating and soluble factors influence astrocyte morphology in 2D, we quantitatively analyzed the morphology of human fetal derived astrocytes on glass, matrigel, fibronectin, collagen IV, and collagen I, and after the addition soluble factors including platelet-derived growth factor (PDGF), laminin, basic fibroblast growth factor (bFGF), and leukemia inhibitory factor (LIF). Matrigel surface coatings, as well as addition of leukemia inhibitory factor (LIF) to the media, were found to have the strongest effects on 2D astrocyte morphology, and may be important in improving existing BBB models. In addition, the novel set of quantitative parameters proposed in this paper provide a test for determining the influence of compounds on astrocyte morphology, both to screen for new endothelial cell-secreted factors that influence astrocytes, and to determine in a high-throughput way which factors are important for translation to more complex, 3D BBB models.
Collapse
|