Skip to content
Publicly Available Published by De Gruyter August 22, 2014

Reproducibility in biological models of the blood-brain barrier

  • Diána Hudecz

    Diána Hudecz started her PhD studies as a Marie Curie Early Stage Researcher within the PathChooser ITN Programme in January 2014. During her work, she is investigating the nanoparticle transport within and through the blood-brain barrier using in vitro models and different imaging techniques. A further important aim of her project is to gain quantitative and reproducible experimental data. Diána holds masters degrees from the Technical University of Denmark and from the University of Copenhagen in Chemical Engineering and Pharmaceutical Sciences, respectively. She completed her BSc studies in Chemical Engineering at the Budapest University of Technology and Economics in Hungary.

    , Louise Rocks

    Louise Rocks joined the Centre for BioNano Interactions in November 2012 as a Postdoctoral Fellow. Her research is focused on the effectiveness of molecular targeting with nanoparticles, specifically, the complications that arise in complex biological fluids. Louise obtained an MSc in chemistry from the University of Strathclyde, Glasgow, prior to undertaking a PhD in the development of a sensitive approach to in vitro/ex vivo biomolecule imaging using surface enhanced Raman spectroscopy.

    , Laurence W. Fitzpatrick

    Laurence W. Fitzpatrick first joined the Centre for BioNano Interactions in February 2011. During which time he worked closely with Dr. Peter Sandin, which resulted in a publication in ACS Nano in January 2012. Laurence returned to the group as a PhD student in September 2012 and is funded by the Irish Research Council under the EMBARK Initiative. His main interests are bio-nanoparticle imaging in cells and tissues and his work focuses on what drives a sub-population of nanoparticles to “unexpected places” within cells, as well as different internalization rates for different sub-populations of particles. He completed his bachelors and graduated with a second class honours BSc in Science with Nanotechnology (Physics Major) in October 2012 from DIT.

    , Luciana-Maria Herda

    Luciana-Maria Herda joined the Centre for BioNano Interactions in January 2014 as a Marie Curie Early Stage Researcher and full-time PhD student, part of the PathChooser ITN programme. Her work focuses on strategies to design functionalised nanoparticles for targeting and controlled negotiation of biological barriers (blood-brain barrier). Luciana graduated from “Carol Davila” University of Medicine and Pharmacy (Bucharest) with a degree in Pharmacy and in September 2013 she completed her MSc in Drug Discovery and Development at University College London School of Pharmacy.

    and Kenneth A. Dawson

    Prof. Kenneth A. Dawson is Director of the Centre for BioNano Interactions (CBNI). He is also a lead investigator of the bionanoscience activities in University College Dublin, and Chair of Physical Chemistry. His professional roles include participation in the EMEA Nanomedicines Expert Group, membership of SCENIHR, participation in the OECD and ISO working groups on standards for Nanotechnology, Chairing the International Alliance for NanoEHS Harmonoisation and co-ordinating several large research projects in the area of nanosafety and nanomedicine. His work has focused on the biomolecule corona that surrounds nanoparticles in biological milieu and how this mediates nanoparticle interactions with living systems. The long-term goal of his research is the development of a rational framework to understand the interactions of nanoparticles with living systems.

    EMAIL logo

Abstract

In the past decade, the importance of quality and reproducibility within research has been re-emphasized, consequently becoming a crucial part of scientific experiments. Their implementation into in vitro and in vivo biological experiments is challenging due to various parameters that can influence the final scientific outcome. In parallel to these activities, there is a huge scientific effort to improve today’s medicines to make them safer and more efficient, and to cure untreatable diseases, such as many neurodegenerative diseases. Nanosized materials have been recognized as potential drug delivery systems in this arena due to their small size and surface properties, which enable the design and synthesis of safe and efficient delivery vehicles that might be able to cross the blood-brain barrier. However, the fundamental understanding behind their uptake mechanism and intracellular trafficking remains unknown. Simple and cost-effective in vitro blood-brain barrier models are widely used to address these questions. This paper aims to critically evaluate the current in vitro models using Transwell systems and to discuss alternative approaches towards more reproducible in vivo features.

Introduction

The importance of reproducibility of scientific experiments has recently been highlighted (1). Assessing the reproducibility of even the most basic techniques and methods, including in vitro biological experiments, has become an essential part of scientific research. Biological experiments can be influenced by various parameters, such as passage-to-passage variation of cell behaviour, receptor expression, batch-to-batch variation of chemical substances, or the interaction between model particle and biological environment (2, 3). Hence, quality and reproducibility must be built into each individual part of the process, as they might have a significant effect on the final outcome and the reproducibility of the final results. Development of detailed protocols and implementation of interlaboratory comparison studies can help to ensure these issues (4).

There is a huge scientific and public interest to find new therapeutic agents for the treatment of neurodegenerative and other diseases localized in the central nervous system (CNS), such as Alzheimer’s, Parkinson’s or Huntington’s diseases, which require the successful crossing of the blood-brain barrier (BBB) of the candidate agents. Due to their size and excellent surface properties, nanoparticles have been recognized as potential drug delivery systems (5), and they can be modified in versatile ways to target the CNS (6, 7).

The BBB strictly controls the molecular trafficking in and out of the brain using its physical, transport, enzymatic, and immune regulatory barrier functions (8, 9) (see Figure 1).

Figure 1 Pathways across the BBB. Grey rectangles represent the brain endothelial cells, which are connected through tight junctions (TJs). Lipophilic compounds are able to path through the barrier through transcellular lipophilic pathway (G), however, most of these compounds are pumped out via efflux transporters (e.g. P-glycoprotein) (C). Although tight junctions acts as a physical barrier and prevent the entrance of many compounds, small water-soluble compounds are able to path them through via the paracellular aqueous pathway (D). Many essential compounds, such as amino acids, peptides, or glucose, enter via transport proteins (solute carriers) (B). Other endogenous macromolecules, such as transferrin and albumin, path through the barrier via receptor-mediated transcytosis (E) and non-specific adsorptive-mediated transcytosis (F), respectively. Furthermore, mononuclear cells, such as monocytes and lymphocytes, can migrate through the barrier (A). Redrawn by D. Hudecz, based on refs (8, 10) with permission of Elsevier and Nature Publishing Group.
Figure 1

Pathways across the BBB. Grey rectangles represent the brain endothelial cells, which are connected through tight junctions (TJs). Lipophilic compounds are able to path through the barrier through transcellular lipophilic pathway (G), however, most of these compounds are pumped out via efflux transporters (e.g. P-glycoprotein) (C). Although tight junctions acts as a physical barrier and prevent the entrance of many compounds, small water-soluble compounds are able to path them through via the paracellular aqueous pathway (D). Many essential compounds, such as amino acids, peptides, or glucose, enter via transport proteins (solute carriers) (B). Other endogenous macromolecules, such as transferrin and albumin, path through the barrier via receptor-mediated transcytosis (E) and non-specific adsorptive-mediated transcytosis (F), respectively. Furthermore, mononuclear cells, such as monocytes and lymphocytes, can migrate through the barrier (A). Redrawn by D. Hudecz, based on refs (8, 10) with permission of Elsevier and Nature Publishing Group.

Physical barriers (formed by tight junctions and adherens junctions between the endothelial cells), transport barriers (composed of adenosine triphosphate binding cassette efflux transporters), and metabolic barriers (formed by enzymes) are responsible for keeping “unwanted objects”, such as toxins, but also most drug candidates, out of the CNS (8–10). Conversely, transport of essential compounds into the brain is possible via paracellular and transcellular pathways, by influx transporters (specific solute carriers), or via receptor- and adsorptive-mediated transcytosis (8, 9).

The most investigated targeting route of the BBB delivery is receptor-mediated transcytosis and it is believed that nanoparticles are transported via this route (7). This targeting strategy is based on the coupling or grafting of receptor substrates that are known to cross the BBB via transcytosis, such as transferrin (11), insulin (12, 13), or apolipoprotein E (ApoE) (14), to the surface of the nanomaterial which trigger receptor-mediated transcytosis and thereby enable them to cross the BBB (6, 7). The success of such modifications using ApoE has been reported (15–17), but in many cases the nanoparticles follow and subsequently accumulate along the endo-lysosomal pathway (18), as is commonly observed with uptake of nanoparticles in other in vitro models (19–22), and never reach the brain. Hence, there is a need for a more specific nanoparticle targeting approach which requires the understanding of the fundamentals behind the transcytosis pathway. This could open a new era of CNS targeting, based on rational, specifically designed carriers.

In vitro BBB models are widely used to study such interactions as they provide a better insight into cellular mechanisms in comparison with in vivo models (23). In the past few decades, various in vitro BBB models have been developed, including cell based, non-cell based (24), and stem cell based models (25–27) [for the most recent reviews about different in vitro models of the BBB see refs (23, 28, 29)]. The cell-based in vitro models include primary, immortalized, and conditionally immortalized models from brain endothelial cells of different species, including rat (30–33), mouse (34, 35), bovine (36), porcine (37, 38), and human (39–41) [the reader can find more examples in a recent review by Bicker et al. (28)]. The endothelial cells in these models can also be co-cultured with astrocytes and/or pericytes (31, 33), which play important roles in the induction, maintenance, and function of the BBB (10, 42).

Investigation of transcytosis and other mechanisms through the BBB using in vitro models

In vitro permeability assays and uptake studies are most often performed in Transwell systems. These multi-well plates comprise an upper (apical- blood side) and a lower (basolateral- brain side) compartment which are separated by a porous membrane. The endothelial cells are seeded onto collagen, or collagen and fibronectin coated membranes, and grown in culture medium until they reach a confluent monolayer. In order to obtain tighter barriers with better barrier function the endothelial cells can also be co-cultured with astrocytes and/or pericytes in mono-, co-, or triple culture models in contact and non-contact format (supporting cells are seeded on the basolateral side of the Transwell insert or to bottom of the basolateral holding plate, respectively) (28, 31) (see ).

Figure 2 In vitro BBB models based on Transwell™ systems. The cerebral endothelial cells (CECs) can be cultured as mono-, co- (A-D), and triple co-cultures (E-F). In the co- and triple co-culture models, CECs are cultured with astrocytes and/or pericytes in contact (B, D) or non-contact (A, C) manner. Reprinted with permission of Bicker et al. (28). Copyright 2014 Elsevier.
Figure 2

In vitro BBB models based on Transwell systems. The cerebral endothelial cells (CECs) can be cultured as mono-, co- (A-D), and triple co-cultures (E-F). In the co- and triple co-culture models, CECs are cultured with astrocytes and/or pericytes in contact (B, D) or non-contact (A, C) manner. Reprinted with permission of Bicker et al. (28). Copyright 2014 Elsevier.

Due to their simplicity, cost-effectiveness, high- to moderate throughput screening properties, and versatility, these Transwell systems are widely used both in academia and in the pharmaceutical industry. They can be applied for rapid measurement of the transendothelial permeability of substances through the BBB, and for the investigation of receptor expression and pathways of the BBB. These systems are also able to measure the transendothelial electrical resistance (TEER), which gives information about the tightness and paracellular permeability of the barrier/cell monolayer (higher TEER values indicate tighter barriers, hence, better mimicking of an in vivo model). The common drawbacks of these systems are the presence of a stagnant liquid layer as a result of their steady condition, and the paracellular leak pathway through the filter pore, which may influence the measured TEER values (23). However, it is possible to correct for these components and calculate the “real” transcellular endothelial permeability (43).

The combination of transmission electron microscopy (TEM) with traditional Transwell systems has revealed some crucial limitations of these models (18, 44, 45), which bring into question the reliability and reproducibility of these systems. These limitations include barrier imperfections, such as formation of multiple layers of endothelial cells on the filter and holes (see Figure 3); nanoparticle adhesion to the microporous filter pore; nanoparticle agglomeration in the culture medium and inside the cell; and leakage of the fluorescent dye. As a result of these limitations, the measured permeability might be an overestimation (due to holes, dye leakage) or underestimation (due to multilayer formation, nanoparticle adhesion and agglomeration) of the true value.

Figure 3 Transmission electron microscope images of the immortalized human brain capillary endothelial cells (hCMEC/D3) grown on collagen/fibronectin-coated 0.4-μm membrane. The endothelial cells can be grown as monolayer (A), multilayer (B), or as imperfect monolayer (C). In subfigure C, the arrows indicate 50 nm SiO2 nanoparticles, which are approaching the filter in the area where cells are not in contact. Adapted from Ye et al. (18) with permission of The Royal Society of Chemistry.
Figure 3

Transmission electron microscope images of the immortalized human brain capillary endothelial cells (hCMEC/D3) grown on collagen/fibronectin-coated 0.4-μm membrane. The endothelial cells can be grown as monolayer (A), multilayer (B), or as imperfect monolayer (C). In subfigure C, the arrows indicate 50 nm SiO2 nanoparticles, which are approaching the filter in the area where cells are not in contact. Adapted from Ye et al. (18) with permission of The Royal Society of Chemistry.

The observed limitations of Transwell systems confirmed that there is a need for more reliable and reproducible measurement techniques which could lead to an improved understanding of nanoparticle/drug candidate-cell behaviour, an insight of the detailed uptake and intracellular trafficking mechanisms, and reproducible quantification of nanoparticle crossing. Utilization of TEM gives better insights into the localization and trafficking routes of nanoparticles (18, 46), however, due to the nature of these techniques (imaging fixed samples), it does not yield a dynamic picture. Immunohistochemistry in conjunction with confocal fluorescent microscopy has yielded further insights into these dynamics (16, 42, 47, 48). However, all of these techniques are based on fixed samples, thereby provide information as a snap shot and limited by the possibility of real time monitoring of the uptake mechanisms and intracellular fate of the nanoparticle.

Development of live-cell imaging techniques (19, 49) and fluorescent probes (50, 51) has enabled the study of dynamic events in a time resolved manner. These live cell imaging techniques include laser scanning- and spinning disc confocal microscopy, widefield fluorescence microscopy, total internal reflection fluorescence microscopy (TIRFM) (for monitoring events that take place on or near the basolateral side of plasma membrane) (52–54), Förster resonance energy transfer (for the measurement of intracellular molecular interactions) (55), fluorescence life-time imaging microscopy (for visualizing the life-time of a molecule’s excitation state) (55). The reader can read more about these and other advanced live cell imaging techniques in refs (56, 57).

Most recently (45), nanoparticle translocation across the BBB was studied using live cell imaging techniques (spinning disc confocal microscopy and TIRFM) coupled with computational analysis. This study revealed the possibility to follow the spatiotemporal movement of nanoparticles quantifying transcytosis/translocation rate, lysosomal accumulation, or other localisation within the barrier. Barrier imperfections (barrier region with holes and/or multiple cell layers) were also observed with the applied live cell imaging techniques, and it was shown that significant amount of nanoparticles could rapidly traverse through the hole (see Figure 4). These events can only be seen using advanced live cell imaging technique, such as TIRFM.

Figure 4 Imperfections of hCMEC/D3 cells grown on collagen-coated glass chamber. The images represent real time events, they were taken using spinning disc confocal microscopy (A and B) and TIRFM (C). Cell membrane was stained with CellMask™ Orange (red). Barrier cross-section with holes (A) (white rectangles) and multiple cell layers (B). Subfigure C) represents the transport of 100 nm carboxylated polystyrene nanoparticles (green) transport through barrier hole over time. Adapted with permission of Bramini et al. (45). Copyright 2014 American Chemical Society.
Figure 4

Imperfections of hCMEC/D3 cells grown on collagen-coated glass chamber. The images represent real time events, they were taken using spinning disc confocal microscopy (A and B) and TIRFM (C). Cell membrane was stained with CellMask Orange (red). Barrier cross-section with holes (A) (white rectangles) and multiple cell layers (B). Subfigure C) represents the transport of 100 nm carboxylated polystyrene nanoparticles (green) transport through barrier hole over time. Adapted with permission of Bramini et al. (45). Copyright 2014 American Chemical Society.

Steps towards a more realistic and reproducible in vitro model

Based on previous observations (18, 44, 45), imperfections exist in all in vitro barriers and monolayers models. Furthermore, the traditional Transwell systems suffer from numerous limitations, preventing their application for accurate and reproducible nanoparticle-BBB studies. Although, TEM imaging enables a more realistic study of such interactions, it does not provide time resolved information, for instance, an endocytotic event from the basolateral site might be mistaken with exocytosis, and events which might be identified as transcytosis might only be due to nanoparticle crossing through a hole located nearby the cell. Alternatively, live cell imaging techniques, including TIRFM, may overcome these issues. Furthermore, regions with a perfect cell monolayer should be investigated, rather than the whole cell population, which might contain holes or multiple cell layers.

Additionally, to have a more comprehensive picture towards the in vivo situations, it is important to apply several models, since every model has their limitations. For instance, different in vitro BBB models have slightly or radically different levels of protein, receptor and transporter expression depending on the type of the model (29, 58). It is also known that primary models have higher TEER values than immortalized models, which means that their junctional protein level expression is higher (23).

Moreover, before the actual nanoparticle-BBB studies, it is necessary to understand the transcytosis mechanism through the BBB. The assumed mechanism of transcytosis is: cell entry mainly via clathrin- or caveolea-mediated endocytosis, or via phagocytosis or macropinocytosis (59), followed by vesicular trafficking, by the avoidance of endo-lysosomal degradation pathway or recycling mechanism, and finally exocytosis on the basolateral side (60). Use of positive controls of transcytosis, such as insulin (12, 13), ApoE (14), transferrin (11), and in house verification of reproducible results are critical and essential part of the above studies.

In addition, application of shear stress in the in vitro BBB models and the use of co-cultures can further improve the models by mimicking the blood flow of in vivo situations, as demonstrated by dynamic in vitro models (61) and microfluidic platforms (62–65) which incorporate these concepts. Microfluidic systems provide the possibility to mimic the anatomical size and hemodynamic shear stress; they are cost effective, require fewer samples than traditional Transwell systems, and give the opportunity of high throughput screening (62–64). These devices are based on a similar concept as Transwell systems, i.e. they consist of an apical and a basolateral compartments, which are separated by a porous membrane (62, 63, 65) or small gaps (64). Most of these systems are based on a so-called sandwich concept; however, Prabhakarpandian et al. (64) have recently presented a different design, synthetic microvasculature model of the BBB (SyM-BBB), in which the position of apical compartment is side-by side with the basolateral compartment, enabling real-time optical monitoring. Figure 5 illustrates the concept behind microfluidic BBB (μBBB) (basolateral side is below the apical) and SyM-BBB (apical side is next to the basolateral side).

Figure 5 Design of μBBB (A) and SyM-BBB (B) models. Adapted from Booth and Kim (62) and Prabhakarpandian et al. (64) with permission of The Royal Society of Chemistry.
Figure 5

Design of μBBB (A) and SyM-BBB (B) models. Adapted from Booth and Kim (62) and Prabhakarpandian et al. (64) with permission of The Royal Society of Chemistry.

There is a fast development in the area of microfluidics, for example Booth et al. (65) have recently reported a multi-channel, multi-assay platform, which enables the study of wide range of wall shear stress conditions and multiple types of assays in a reproducible and controllable manner. A potential drawback to these models is that they are based on the same concept as Transwell systems, whose reliability and reproducibility have been questioned within this current review. However, the design of SyM-BBB and its future derivatives provide the possibility of high throughput pharmaceutical screening and real-time imaging, which enables closer monitoring of holes, cell morphology, and cytotoxicity.

Conclusion and outlook

Although live cell imaging techniques and their combinations with microfluidic systems seems to provide a better understanding and real time monitoring of the intracellular fate of nanoparticles within the BBB, they are not ideal from an industrial point of view due to their current lack of high throughput screening capabilities. On the other hand, these techniques offer the possibility of more reproducible experiments compared to the traditional high throughput Transwell systems, as all processes can be followed visually in a time resolved manner. Regions with imperfect barrier properties can be deselected, and thereby the influence of uptake or retention of nanoparticles through these regions can be excluded from the final estimated nanoparticle uptake. Furthermore, as the accurate investigation of intracellular trafficking and other mechanism through the BBB is only possible using advanced live cell imaging techniques, these techniques will become an essential part of nanomedical and biological barrier research.


Corresponding author: Prof. Kenneth A. Dawson, Director of Centre for BioNano Interactions, School of Chemistry and Chemical Biology, University College Dublin, Belfield, Dublin 4, Ireland, E-mail:

About the authors

Diána Hudecz

Diána Hudecz started her PhD studies as a Marie Curie Early Stage Researcher within the PathChooser ITN Programme in January 2014. During her work, she is investigating the nanoparticle transport within and through the blood-brain barrier using in vitro models and different imaging techniques. A further important aim of her project is to gain quantitative and reproducible experimental data. Diána holds masters degrees from the Technical University of Denmark and from the University of Copenhagen in Chemical Engineering and Pharmaceutical Sciences, respectively. She completed her BSc studies in Chemical Engineering at the Budapest University of Technology and Economics in Hungary.

Louise Rocks

Louise Rocks joined the Centre for BioNano Interactions in November 2012 as a Postdoctoral Fellow. Her research is focused on the effectiveness of molecular targeting with nanoparticles, specifically, the complications that arise in complex biological fluids. Louise obtained an MSc in chemistry from the University of Strathclyde, Glasgow, prior to undertaking a PhD in the development of a sensitive approach to in vitro/ex vivo biomolecule imaging using surface enhanced Raman spectroscopy.

Laurence W. Fitzpatrick

Laurence W. Fitzpatrick first joined the Centre for BioNano Interactions in February 2011. During which time he worked closely with Dr. Peter Sandin, which resulted in a publication in ACS Nano in January 2012. Laurence returned to the group as a PhD student in September 2012 and is funded by the Irish Research Council under the EMBARK Initiative. His main interests are bio-nanoparticle imaging in cells and tissues and his work focuses on what drives a sub-population of nanoparticles to “unexpected places” within cells, as well as different internalization rates for different sub-populations of particles. He completed his bachelors and graduated with a second class honours BSc in Science with Nanotechnology (Physics Major) in October 2012 from DIT.

Luciana-Maria Herda

Luciana-Maria Herda joined the Centre for BioNano Interactions in January 2014 as a Marie Curie Early Stage Researcher and full-time PhD student, part of the PathChooser ITN programme. Her work focuses on strategies to design functionalised nanoparticles for targeting and controlled negotiation of biological barriers (blood-brain barrier). Luciana graduated from “Carol Davila” University of Medicine and Pharmacy (Bucharest) with a degree in Pharmacy and in September 2013 she completed her MSc in Drug Discovery and Development at University College London School of Pharmacy.

Kenneth A. Dawson

Prof. Kenneth A. Dawson is Director of the Centre for BioNano Interactions (CBNI). He is also a lead investigator of the bionanoscience activities in University College Dublin, and Chair of Physical Chemistry. His professional roles include participation in the EMEA Nanomedicines Expert Group, membership of SCENIHR, participation in the OECD and ISO working groups on standards for Nanotechnology, Chairing the International Alliance for NanoEHS Harmonoisation and co-ordinating several large research projects in the area of nanosafety and nanomedicine. His work has focused on the biomolecule corona that surrounds nanoparticles in biological milieu and how this mediates nanoparticle interactions with living systems. The long-term goal of his research is the development of a rational framework to understand the interactions of nanoparticles with living systems.

Acknowledgments

Funding has been generously provided by EU FP7 via the Marie-Curie Initial Training Network PathChooser (PITN-GA-2013-608373).

References

1. Challenges in Irreproducible Research. Nature Special. [cited 2014 May 1]. Available from: http://www.nature.com/nature/focus/reproducibility/.Search in Google Scholar

2. Lundqvist M, Stigler J, Elia G, Lynch I, Cedervall T, Dawson KA. Nanoparticle size and surface properties determine the protein corona with possible implications for biological impacts. Proc Natl Acad Sci USA 2008;105:14265–70.10.1073/pnas.0805135105Search in Google Scholar

3. Monopoli MP, Aberg C, Salvati A, Dawson KA. Biomolecular coronas provide the biological identity of nanosized materials. Nat Nanotechnol. Nature Publishing Group; 2012;7:779–86.Search in Google Scholar

4. Hole P, Sillence K, Hannell C, Maguire CM, Roesslein M, Suarez G, et al. Interlaboratory comparison of size measurements on nanoparticles using nanoparticle tracking analysis (NTA). J Nanopart Res 2013;15:2101.10.1007/s11051-013-2101-8Search in Google Scholar

5. Moghimi SM, Hunter AC, Murray JC. Nanomedicine: current status and future prospects. FASEB J 2005;19:311–30.10.1096/fj.04-2747revSearch in Google Scholar

6. Masserini M. Nanoparticles for Brain Drug Delivery. ISRN Biochem 2013;2013:1–18.10.1155/2013/238428Search in Google Scholar

7. Kreuter J. Drug delivery to the central nervous system by polymeric nanoparticles: what do we know? Adv Drug Deliv Rev 2014;71:2–14.10.1016/j.addr.2013.08.008Search in Google Scholar

8. Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis 2010;37:13–25.10.1016/j.nbd.2009.07.030Search in Google Scholar

9. Abbott NJ. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J Inherit Metab Dis 2013;36:437–49.10.1007/s10545-013-9608-0Search in Google Scholar

10. Abbott NJ, Rönnbäck L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci 2006;7:41–53.10.1038/nrn1824Search in Google Scholar

11. Fishman JB, Rubin JB, Handrahan J V, Connor JR, Fine RE. Receptor-mediated transcytosis of transferrin across the blood-brain barrier. J Neurosci Res 1987;18:299–304.10.1002/jnr.490180206Search in Google Scholar

12. Banks WA, Jaspan JB, Kastin AJ. Selective, physiological transport of insulin across the blood-brain barrier: novel demonstration by species-specific radioimmunoassays. Peptides 1997;18:1257–62.10.1016/S0196-9781(97)00198-8Search in Google Scholar

13. Banks WA, Jaspan JB, Huang W, Kastin AJ. Transport of insulin across the blood-brain barrier: saturability at euglycemic doses of insulin. Peptides 1997;18:1423–9.10.1016/S0196-9781(97)00231-3Search in Google Scholar

14. Dehouck B, Fenart L, Dehouck MP, Pierce A, Torpier G, Cecchelli R. A new function for the LDL receptor: transcytosis of LDL across the blood-brain barrier. J Cell Biol 1997;138:877–89.10.1083/jcb.138.4.877Search in Google Scholar PubMed PubMed Central

15. Zensi A, Begley D, Pontikis C, Legros C, Mihoreanu L, Wagner S, et al. Albumin nanoparticles targeted with Apo E enter the CNS by transcytosis and are delivered to neurones. J Control Release 2009;137:78–86.10.1016/j.jconrel.2009.03.002Search in Google Scholar PubMed

16. Zensi A, Begley D, Pontikis C, Legros C, Mihoreanu L, Büchel C, et al. Human serum albumin nanoparticles modified with apolipoprotein A-I cross the blood-brain barrier and enter the rodent brain. J Drug Target 2010;18:842–8.10.3109/1061186X.2010.513712Search in Google Scholar PubMed

17. Wagner S, Zensi A, Wien SL, Tschickardt SE, Maier W, Vogel T, et al. Uptake Mechanism of ApoE-modified nanoparticles on brain capillary endothelial cells as a blood-brain barrier model. PLoS One. Public Library of Science; 2012;7:e32568.10.1371/journal.pone.0032568Search in Google Scholar PubMed PubMed Central

18. Ye D, Raghnaill MN, Bramini M, Mahon E, Åberg C, Salvati A, et al. Nanoparticle accumulation and transcytosis in brain endothelial cell layers. Nanoscale 2013;5:11153–65.10.1039/c3nr02905kSearch in Google Scholar PubMed

19. Sandin P, Fitzpatrick LW, Simpson JC, Dawson KA. High-speed imaging of Rab family small GTPases reveals rare events in nanoparticle trafficking in living cells. ACS Nano 2012;6:1513–21.10.1021/nn204448xSearch in Google Scholar PubMed

20. Bertoli F, Davies G-L, Monopoli MP, Moloney M, Gun’ko YK, Salvati A, et al. Magnetic nanoparticles to recover cellular organelles and study the time resolved nanoparticle-cell interactome throughout uptake. Small 2014; Available from: http://onlinelibrary.wiley.com/doi/10.1002/smll.201303841/abstract; doi: 10.1002/smll.201303841.10.1002/smll.201303841Search in Google Scholar PubMed

21. Ma D. Enhancing endosomal escape for nanoparticle mediated siRNA delivery. Nanoscale 2014;6:6415–25.10.1039/c4nr00018hSearch in Google Scholar PubMed

22. Nowak JS, Mehn D, Nativo P, García CP, Gioria S, Ojea-Jiménez I, et al. Silica nanoparticle uptake induces survival mechanism in A549 cells by the activation of autophagy but not apoptosis. Toxicol Lett 2014;224:84–92.10.1016/j.toxlet.2013.10.003Search in Google Scholar PubMed

23. Abbott NJ, Dolman DE, Yusof SR, Reichel A. In Vitro models of CNS barriers. In: Hammarlund-Udenaes M, de Lange ECM, Thorne RG, editors. Drug Delivery to the Brain, AAPS Advances in the Pharmaceutical Sciences Series. New York: Springer; 2014 p. 163–97.10.1007/978-1-4614-9105-7_6Search in Google Scholar

24. Di L, Kerns EH, Fan K, McConnell OJ, Carter GT. High throughput artificial membrane permeability assay for blood–brain barrier. Eur J Med Chem 2003;38:223–32.10.1016/S0223-5234(03)00012-6Search in Google Scholar

25. Lippmann ES, Azarin SM, Kay JE, Nessler RA, Wilson HK, Al-Ahmad A, et al. Derivation of blood-brain barrier endothelial cells from human pluripotent stem cells. Nat Biotechnol. Nature Publishing Group; 2012;30:783–91.Search in Google Scholar

26. Lippmann ES, Al-Ahmad A, Palecek SP, Shusta EV. Modeling the blood-brain barrier using stem cell sources. Fluids Barriers CNS. Fluids and Barriers of the CNS; 2013;10:2.10.1186/2045-8118-10-2Search in Google Scholar PubMed PubMed Central

27. Lippmann ES, Al-Ahmad A, Azarin SM, Palecek SP, Shusta EV. A retinoic acid-enhanced, multicellular human blood-brain barrier model derived from stem cell sources. Sci Rep 2014;4:4160.10.1038/srep04160Search in Google Scholar PubMed PubMed Central

28. Bicker J, Alves G, Fortuna A, Falcão A. Blood-brain barrier models and their relevance for a successful development of CNS drug delivery systems: a review. Eur J Pharm Piopharmaceutics 2014;87:409–32.10.1016/j.ejpb.2014.03.012Search in Google Scholar PubMed

29. Wilhelm I, Krizbai IA. In vitro models of the blood–brain barrier for the study of drug delivery to the brain. Mol Pharm 2014;11:1949–63.10.1021/mp500046fSearch in Google Scholar PubMed

30. Roux F, Durieu-Trautmann O, Chaverot N, Claire M, Mailly P, Bourre J-M, et al. Regulation of gamma-glutamyl transpeptidase and alkaline phosphatase activities in immortalized rat brain microvessel endothelial cells. J Cell Physiol 1994;159:101–13.10.1002/jcp.1041590114Search in Google Scholar PubMed

31. Nakagawa S, Deli MA, Kawaguchi H, Shimizudani T, Shimono T, Kittel Á, et al. A new blood–brain barrier model using primary rat brain endothelial cells, pericytes and astrocytes. Neurochem Int 2009;54:253–63.10.1016/j.neuint.2008.12.002Search in Google Scholar PubMed

32. Terasaki T, Hosoya K. Conditionally immortalized cell lines as a new in vitro model for the study of barrier functions. Biol Pharm Bull 2001;24:111–8.10.1248/bpb.24.111Search in Google Scholar PubMed

33. Abbott NJ, Dolman DM, Drndarski S, Fredriksson S. An improved in vitro blood–brain barrier model: rat brain endothelial cells co-cultured with astrocytes. Methods Mol Biol 2012;814:415–30.10.1007/978-1-61779-452-0_28Search in Google Scholar PubMed

34. Williams RL, Courtneidge SA, Wagner EF. Embryonic lethalities and endothelial tumors in chimeric mice expressing polyoma virus middle T oncogene. Cell 1988;52:121–31.10.1016/0092-8674(88)90536-3Search in Google Scholar

35. Williams RL, Risau W, Zerwes H-G, Drexler H, Aguzzi A, Wagner EF. Endothelioma cells expressing the polyoma middle T oncogene induce hemangiomas by host cell recruitment. Cell 1989;57:1053–63.10.1016/0092-8674(89)90343-7Search in Google Scholar

36. Bowman PD, Ennis SR, Rarey KE, Lorris Betz A, Goldstein GW. Brain microvessel endothelial cells in tissue culture: a model for study of blood-brain barrier permeability. Ann Neurol. Little, Brown and Company; 1983;14:396–402.Search in Google Scholar

37. Rubin H, Xu K. Evidence for the progressive and adaptive nature of spontaneous transformation in the NIH 3T3 cell line. Proc Natl Acad Sci 1989;86:1860–4.10.1073/pnas.86.6.1860Search in Google Scholar PubMed PubMed Central

38. Patabendige A, Skinner RA, Abbott NJ. Establishment of a simplified in vitro porcine blood-brain barrier model with high transendothelial electrical resistance. Brain Res 2013;1521:1–15.10.1016/j.brainres.2012.06.057Search in Google Scholar PubMed PubMed Central

39. Weksler BB, Subileau EA, Perrière N, Charneau P, Holloway K, Leveque M, et al. Blood-brain barrier-specific properties of a human adult brain endothelial cell line. FASEB J 2005;19:1872–4.10.1096/fj.04-3458fjeSearch in Google Scholar PubMed

40. Stins MF, Badger J, Sik Kim K. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb Pathog 2001;30:19–28.10.1006/mpat.2000.0406Search in Google Scholar PubMed

41. Sano Y, Shimizu F, Abe M, Maeda T, Kashiwamura Y, Ohtsuki S, et al. Establishment of a new conditionally immortalized human brain microvascular endothelial cell line retaining an in vivo blood–brain barrier function. J Cell Physiol 2010;225:519–28.10.1002/jcp.22232Search in Google Scholar PubMed

42. Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, et al. Pericytes regulate the blood-brain barrier. Nature 2010;468:557–61.10.1038/nature09522Search in Google Scholar PubMed

43. Avdeef A. How well can in vitro brain microcapillary endothelial cell models predict rodent in vivo blood-brain barrier permeability? Eur J Pharm Sci 2011;43:109–24.10.1016/j.ejps.2011.04.001Search in Google Scholar PubMed

44. Ragnaill MN, Brown M, Ye D, Bramini M, Callanan S, Lynch I, et al. Internal benchmarking of a human blood-brain barrier cell model for screening of nanoparticle uptake and transcytosis. Eur J Pharm Biopharm 2011;77:360–7.10.1016/j.ejpb.2010.12.024Search in Google Scholar PubMed

45. Bramini M, Ye D, Hallerbach A, Nic Raghnaill M, Salvati A, Åberg C, et al. Imaging approach to mechanistic study of nanoparticle interactions. ACS Nano 2014;8:4304–12.10.1021/nn5018523Search in Google Scholar PubMed

46. Gromnicova R, Davies HA, Sreekanthreddy P, Romero IA, Lund T, Roitt IM, et al. Glucose-coated gold nanoparticles transfer across human brain endothelium and enter astrocytes in vitro. PLoS One 2013;8:e81043.10.1371/journal.pone.0081043Search in Google Scholar PubMed PubMed Central

47. Eugenin EA, Clements JE, Zink MC, Berman JW. Human immunodeficiency virus infection of human astrocytes disrupts blood-brain barrier integrity by a gap junction-dependent mechanism. J Neurosci 2011;31:9456–65.10.1523/JNEUROSCI.1460-11.2011Search in Google Scholar PubMed PubMed Central

48. Kebir H, Kreymborg K, Ifergan I, Dodelet-Devillers A, Cayrol R, Bernard M, et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med 2007;13:1173–5.10.1038/nm1651Search in Google Scholar PubMed PubMed Central

49. Welsher K, Yang H. Multi-resolution 3D visualization of the early stages of cellular uptake of peptide-coated nanoparticles. Nat Nanotechnol. Nature Publishing Group; 2014;9:198–203.Search in Google Scholar

50. Nienhaus K, Ulrich Nienhaus G. Fluorescent proteins for live-cell imaging with super-resolution. Chem Soc Rev 2014;43:1088–106.10.1039/C3CS60171DSearch in Google Scholar PubMed

51. Yun S-W, Kang N-Y, Park S-J, Ha H-H, Kim YK, Lee J-S, et al. Diversity oriented fluorescence library approach (DOFLA) for live cell imaging probe development. Acc Chem Res. American Chemical Society; 2014;47:1277–86.Search in Google Scholar

52. Burchfield JG, Lopez JA, Mele K, Vallotton P, Hughes WE. Exocytotic vesicle behaviour assessed by total internal reflection fluorescence microscopy. Traffic 2010;11:429–39.10.1111/j.1600-0854.2010.01039.xSearch in Google Scholar PubMed

53. Millis BA. Evanescent-wave field imaging: an introduction to total internal reflection fluorescence microscopy. Methods Mol Biol 2012;823:295–309.10.1007/978-1-60327-216-2_19Search in Google Scholar PubMed

54. Mattheyses AL, Simon SM, Rappoport JZ. Imaging with total internal reflection fluorescence microscopy for the cell biologist. J Cell Sci 2010;123:3621–8.10.1242/jcs.056218Search in Google Scholar PubMed PubMed Central

55. Wallrabe H, Periasamy A. Imaging protein molecules using FRET and FLIM microscopy. Curr Opin Biotechnol 2005;16:19–27.10.1016/j.copbio.2004.12.002Search in Google Scholar PubMed

56. Walker-Daniels J. Live cell imaging methods review. Mater Methods 2012;2:124.10.13070/mm.en.2.124Search in Google Scholar

57. Parhamifar L, Wu L, Andersen H, Moghimi SM. Live-cell fluorescent microscopy platforms for real-time monitoring of polyplex-cell interaction: basic guidelines. Methods 2014;68:300–7.10.1016/j.ymeth.2014.02.004Search in Google Scholar PubMed

58. Hoshi Y, Uchida Y, Tachikawa M, Inoue T, Ohtsuki S, Terasaki T. Quantitative atlas of blood–brain barrier transporters, receptors, and tight junction proteins in rats and common marmoset. J Pharm Sci 2013;102:3343–55.10.1002/jps.23575Search in Google Scholar PubMed

59. Iversen T-G, Skotland T, Sandvig K. Endocytosis and intracellular transport of nanoparticles: present knowledge and need for future studies. Nano Today 2011;6:176–85.10.1016/j.nantod.2011.02.003Search in Google Scholar

60. Tuma PL, Hubbard AL. Transcytosis: crossing cellular barriers. Physiol Rev 2003;83:871–932.10.1152/physrev.00001.2003Search in Google Scholar PubMed

61. Cucullo L, Couraud P-O, Weksler B, Romero I-A, Hossain M, Rapp E, et al. Immortalized human brain endothelial cells and flow-based vascular modeling: a marriage of convenience for rational neurovascular studies. J Cereb Blood Flow Metab 2008;28:312–28.10.1038/sj.jcbfm.9600525Search in Google Scholar PubMed

62. Booth R, Kim H. Characterization of a microfluidic in vitro model of the blood-brain barrier (μBBB). Lab Chip 2012;12:1784–92.10.1039/c2lc40094dSearch in Google Scholar PubMed

63. Griep LM, Wolbers F, de Wagenaar B, ter Braak PM, Weksler BB, Romero IA, et al. BBB on chip: microfluidic platform to mechanically and biochemically modulate blood-brain barrier function. Biomed Microdevices 2013;15:145–50.10.1007/s10544-012-9699-7Search in Google Scholar PubMed

64. Prabhakarpandian B, Shen M-C, Nichols JB, Mills IR, Sidoryk-Wegrzynowicz M, Aschner M, et al. SyM-BBB: a microfluidic blood brain barrier model. Lab Chip 2013;13:1093–101.10.1039/c2lc41208jSearch in Google Scholar PubMed PubMed Central

65. Booth R, Noh S, Kim H. A multiple-channel, multiple-assay platform for characterization of full-range shear stress effects on vascular endothelial cells. Lab Chip 2014;14:1880–90.10.1039/C3LC51304ASearch in Google Scholar PubMed

Received: 2014-5-26
Accepted: 2014-7-18
Published Online: 2014-8-22
Published in Print: 2014-9-1

©2014 by De Gruyter

Downloaded on 15.5.2024 from https://www.degruyter.com/document/doi/10.1515/ejnm-2014-0021/html
Scroll to top button