51
|
Zhou L, Sohet F, Daneman R. Purification and culture of central nervous system endothelial cells. Cold Spring Harb Protoc 2014; 2014:44-6. [PMID: 24371313 DOI: 10.1101/pdb.top070987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Blood vessels are critical for delivering oxygen and nutrients to all tissues in the body. This is especially important in the central nervous system, which is extremely sensitive to hypoxia and ischemia. Blood vessels are made of two main cell types: endothelial cells and mural cells. Endothelial cells form the walls of the blood vessels that generate a lumen through which blood flows. Mural cells are support cells thought to be involved in vessel contractility, vascular remodeling, and regulation of endothelial permeability. On large vessels, including arteries and veins, mural cells are termed vascular smooth muscle cells. On the small vessels of the capillary bed, they are called pericytes. Here, we provide a brief introduction to the methods for purification of endothelial cells, including an immunopanning method that we developed for isolating these cells from the rodent brain and optic nerve.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305-5125
| | | | | |
Collapse
|
52
|
Maeda T, Sano Y, Abe M, Shimizu F, Kashiwamura Y, Ohtsuki S, Terasaki T, Obinata M, Ueda M, Kanda T. Establishment and characterization of spinal cord microvascular endothelial cell lines. ACTA ACUST UNITED AC 2013. [DOI: 10.1111/cen3.12045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Toshihiko Maeda
- Department of Neurology and Clinical Neuroscience; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - Masaaki Abe
- Department of Neurology and Clinical Neuroscience; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - Yoko Kashiwamura
- Department of Neurology and Clinical Neuroscience; Graduate School of Medicine; Yamaguchi University; Ube Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology; Graduate School of Pharmaceutical Sciences; Kumamoto University; Kumamoto Japan
| | - Tetsuya Terasaki
- Department of Molecular Biopharmacy and Genetics; Graduate School of Pharmaceutical Sciences; Tohoku University; Sendai Japan
| | - Masuo Obinata
- Department of Cell Biology; Institute of Development, Aging and Cancer; Tohoku University; Sendai Japan
| | | | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience; Graduate School of Medicine; Yamaguchi University; Ube Japan
| |
Collapse
|
53
|
Siegenthaler JA, Sohet F, Daneman R. 'Sealing off the CNS': cellular and molecular regulation of blood-brain barriergenesis. Curr Opin Neurobiol 2013; 23:1057-64. [PMID: 23867075 DOI: 10.1016/j.conb.2013.06.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 06/11/2013] [Indexed: 01/24/2023]
Abstract
From their initial ingression into the neural tube to the established, adult vascular plexus, blood vessels within the CNS are truly unique. Covered by a virtually continuous layer of perivascular cells and astrocytic endfeet and connected by specialized cell-cell junctional contacts, mature CNS blood vessels simultaneously provide nutritive blood flow and protect the neural milieu from potentially disruptive or harmful molecules and cells flowing through the vessel lumen. In this review we will discuss how the CNS vasculature acquires blood-brain barrier (BBB) properties with a specific focus on recent work identifying the cell types and molecular pathways that orchestrate barriergenesis.
Collapse
Affiliation(s)
- Julie A Siegenthaler
- Department of Pediatrics, Section of Developmental Biology, University of Colorado, Denver, Aurora, CO 80045, United States.
| | | | | |
Collapse
|
54
|
Watson PMD, Paterson JC, Thom G, Ginman U, Lundquist S, Webster CI. Modelling the endothelial blood-CNS barriers: a method for the production of robust in vitro models of the rat blood-brain barrier and blood-spinal cord barrier. BMC Neurosci 2013; 14:59. [PMID: 23773766 PMCID: PMC3694476 DOI: 10.1186/1471-2202-14-59] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 06/05/2013] [Indexed: 12/16/2022] Open
Abstract
Background Modelling the blood-CNS barriers of the brain and spinal cord in vitro continues to provide a considerable challenge for research studying the passage of large and small molecules in and out of the central nervous system, both within the context of basic biology and for pharmaceutical drug discovery. Although there has been considerable success over the previous two decades in establishing useful in vitro primary endothelial cell cultures from the blood-CNS barriers, no model fully mimics the high electrical resistance, low paracellular permeability and selective influx/efflux characteristics of the in vivo situation. Furthermore, such primary-derived cultures are typically labour-intensive and generate low yields of cells, limiting scope for experimental work. We thus aimed to establish protocols for the high yield isolation and culture of endothelial cells from both rat brain and spinal cord. Our aim was to optimise in vitro conditions for inducing phenotypic characteristics in these cells that were reminiscent of the in vivo situation, such that they developed into tight endothelial barriers suitable for performing investigative biology and permeability studies. Methods Brain and spinal cord tissue was taken from the same rats and used to specifically isolate endothelial cells to reconstitute as in vitro blood-CNS barrier models. Isolated endothelial cells were cultured to expand the cellular yield and then passaged onto cell culture inserts for further investigation. Cell culture conditions were optimised using commercially available reagents and the resulting barrier-forming endothelial monolayers were characterised by functional permeability experiments and in vitro phenotyping by immunocytochemistry and western blotting. Results Using a combination of modified handling techniques and cell culture conditions, we have established and optimised a protocol for the in vitro culture of brain and, for the first time in rat, spinal cord endothelial cells. High yields of both CNS endothelial cell types can be obtained, and these can be passaged onto large numbers of cell culture inserts for in vitro permeability studies. The passaged brain and spinal cord endothelial cells are pure and express endothelial markers, tight junction proteins and intracellular transport machinery. Further, both models exhibit tight, functional barrier characteristics that are discriminating against large and small molecules in permeability assays and show functional expression of the pharmaceutically important P-gp efflux transporter. Conclusions Our techniques allow the provision of high yields of robust sister cultures of endothelial cells that accurately model the blood-CNS barriers in vitro. These models are ideally suited for use in studying the biology of the blood-brain barrier and blood-spinal cord barrier in vitro and for pre-clinical drug discovery.
Collapse
|
55
|
Lippmann ES, Azarin SM, Kay JE, Nessler RA, Wilson HK, Al-Ahmad A, Palecek SP, Shusta EV. Derivation of blood-brain barrier endothelial cells from human pluripotent stem cells. Nat Biotechnol 2013; 30:783-91. [PMID: 22729031 PMCID: PMC3467331 DOI: 10.1038/nbt.2247] [Citation(s) in RCA: 530] [Impact Index Per Article: 48.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 04/30/2012] [Indexed: 11/09/2022]
Abstract
The blood-brain barrier (BBB) is crucial to the health of the brain and is often compromised in neurological disease. Moreover, because of its barrier properties, this endothelial interface restricts uptake of neurotherapeutics. Thus, a renewable source of human BBB endothelium could spur brain research and pharmaceutical development. Here we show that endothelial cells derived from human pluripotent stem cells (hPSCs) acquire BBB properties when co-differentiated with neural cells that provide relevant cues, including those involved in Wnt/β-catenin signaling. The resulting endothelial cells have many BBB attributes, including well-organized tight junctions, appropriate expression of nutrient transporters and polarized efflux transporter activity. Notably, they respond to astrocytes, acquiring substantial barrier properties as measured by transendothelial electrical resistance (1,450 ± 140 Ω cm2), and they possess molecular permeability that correlates well with in vivo rodent blood-brain transfer coefficients.
Collapse
Affiliation(s)
- Ethan S Lippmann
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | | | | | | | | | | | | |
Collapse
|
56
|
Modeling the blood-brain barrier using stem cell sources. Fluids Barriers CNS 2013; 10:2. [PMID: 23305164 PMCID: PMC3564868 DOI: 10.1186/2045-8118-10-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/13/2012] [Indexed: 12/18/2022] Open
Abstract
The blood–brain barrier (BBB) is a selective endothelial interface that controls trafficking between the bloodstream and brain interstitial space. During development, the BBB arises as a result of complex multicellular interactions between immature endothelial cells and neural progenitors, neurons, radial glia, and pericytes. As the brain develops, astrocytes and pericytes further contribute to BBB induction and maintenance of the BBB phenotype. Because BBB development, maintenance, and disease states are difficult and time-consuming to study in vivo, researchers often utilize in vitro models for simplified analyses and higher throughput. The in vitro format also provides a platform for screening brain-penetrating therapeutics. However, BBB models derived from adult tissue, especially human sources, have been hampered by limited cell availability and model fidelity. Furthermore, BBB endothelium is very difficult if not impossible to isolate from embryonic animal or human brain, restricting capabilities to model BBB development in vitro. In an effort to address some of these shortcomings, advances in stem cell research have recently been leveraged for improving our understanding of BBB development and function. Stem cells, which are defined by their capacity to expand by self-renewal, can be coaxed to form various somatic cell types and could in principle be very attractive for BBB modeling applications. In this review, we will describe how neural progenitor cells (NPCs), the in vitro precursors to neurons, astrocytes, and oligodendrocytes, can be used to study BBB induction. Next, we will detail how these same NPCs can be differentiated to more mature populations of neurons and astrocytes and profile their use in co-culture modeling of the adult BBB. Finally, we will describe our recent efforts in differentiating human pluripotent stem cells (hPSCs) to endothelial cells with robust BBB characteristics and detail how these cells could ultimately be used to study BBB development and maintenance, to model neurological disease, and to screen neuropharmaceuticals.
Collapse
|
57
|
Takeshita Y, Ransohoff RM. Inflammatory cell trafficking across the blood-brain barrier: chemokine regulation and in vitro models. Immunol Rev 2012; 248:228-39. [PMID: 22725965 DOI: 10.1111/j.1600-065x.2012.01127.x] [Citation(s) in RCA: 224] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The blood-brain barrier (BBB) is the brain-specific capillary barrier that is critical for preventing toxic substances from entering the central nervous system (CNS). In contrast to vessels of peripheral organs, the BBB limits the exchange of inflammatory cells and mediators under physiological and pathological conditions. Clarifying these limitations and the role of chemokines in regulating the BBB would provide new insights into neuroprotective strategies in neuroinflammatory diseases. Because there is a paucity of in vitro BBB models, however, some mechanistic aspects of transmigration across the BBB still remain largely unknown. In this review, we summarize current knowledge of BBB cellular components, the multistep process of inflammatory cells crossing the BBB, functions of CNS-derived chemokines, and in vitro BBB models for transmigration, with a particular focus on new and recent findings.
Collapse
Affiliation(s)
- Yukio Takeshita
- Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
58
|
Li J, Ye L, Wang X, Liu J, Wang Y, Zhou Y, Ho W. (-)-Epigallocatechin gallate inhibits endotoxin-induced expression of inflammatory cytokines in human cerebral microvascular endothelial cells. J Neuroinflammation 2012; 9:161. [PMID: 22768975 PMCID: PMC3408337 DOI: 10.1186/1742-2094-9-161] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 07/06/2012] [Indexed: 12/18/2022] Open
Abstract
Background (−)-Epigallocatechin gallate (EGCG) is a major polyphenol component of green tea that has antioxidant activities. Lipopolysaccharide (LPS) induces inflammatory cytokine production and impairs blood–brain barrier (BBB) integrity. We examined the effect of EGCG on LPS-induced expression of the inflammatory cytokines in human cerebral microvascular endothelial cells (hCMECs) and BBB permeability. Methods The expression of TNF-α, IL-1β and monocyte chemotactic protein-1 (MCP-1/CCL2) was determined by quantitative real time PCR (qRT-PCR) and ELISA. Intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule (VCAM) in hCMECs were examined by qRT-PCR and Western blotting. Monocytes that adhered to LPS-stimulated endothelial cells were measured by monocyte adhesion assay. Tight junctional factors were detected by qRT-PCR (Claudin 5 and Occludin) and immunofluorescence staining (Claudin 5 and ZO-1). The permeability of the hCMEC monolayer was determined by fluorescence spectrophotometry of transmembrane fluorescin and transendothelial electrical resistance (TEER). NF-kB activation was measured by luciferase assay. Results EGCG significantly suppressed the LPS-induced expression of IL-1β and TNF-α in hCMECs. EGCG also inhibited the expression of MCP-1/CCL2, VCAM-1 and ICAM-1. Functional analysis showed that EGCG induced the expression of tight junction proteins (Occludin and Claudin-5) in hCMECs. Investigation of the mechanism showed that EGCG had the ability to inhibit LPS-mediated NF-κB activation. In addition, 67-kD laminin receptor was involved in the anti-inflammatory effect of EGCG. Conclusions Our results demonstrated that LPS induced inflammatory cytokine production in hCMECs, which could be attenuated by EGCG. These data indicate that EGCG has a therapeutic potential for endotoxin-mediated endothelial inflammation.
Collapse
Affiliation(s)
- Jieliang Li
- The Center for Animal Experiment/Animal Biosafety Level III Laboratory, Wuhan University Wuhan, Hubei 430071, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
59
|
Muffley LA, Pan SC, Smith AN, Ga M, Hocking AM, Gibran NS. Differentiation state determines neural effects on microvascular endothelial cells. Exp Cell Res 2012; 318:2085-93. [PMID: 22683922 DOI: 10.1016/j.yexcr.2012.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/17/2012] [Accepted: 06/01/2012] [Indexed: 12/25/2022]
Abstract
Growing evidence indicates that nerves and capillaries interact paracrinely in uninjured skin and cutaneous wounds. Although mature neurons are the predominant neural cell in the skin, neural progenitor cells have also been detected in uninjured adult skin. The aim of this study was to characterize differential paracrine effects of neural progenitor cells and mature sensory neurons on dermal microvascular endothelial cells. Our results suggest that neural progenitor cells and mature sensory neurons have unique secretory profiles and distinct effects on dermal microvascular endothelial cell proliferation, migration, and nitric oxide production. Neural progenitor cells and dorsal root ganglion neurons secrete different proteins related to angiogenesis. Specific to neural progenitor cells were dipeptidyl peptidase-4, IGFBP-2, pentraxin-3, serpin f1, TIMP-1, TIMP-4 and VEGF. In contrast, endostatin, FGF-1, MCP-1 and thrombospondin-2 were specific to dorsal root ganglion neurons. Microvascular endothelial cell proliferation was inhibited by dorsal root ganglion neurons but unaffected by neural progenitor cells. In contrast, microvascular endothelial cell migration in a scratch wound assay was inhibited by neural progenitor cells and unaffected by dorsal root ganglion neurons. In addition, nitric oxide production by microvascular endothelial cells was increased by dorsal root ganglion neurons but unaffected by neural progenitor cells.
Collapse
Affiliation(s)
- Lara A Muffley
- University of Washington, Campus Box 359796, 300 9th Avenue, Seattle, WA 98104, USA.
| | | | | | | | | | | |
Collapse
|
60
|
Booth R, Kim H. Characterization of a microfluidic in vitro model of the blood-brain barrier (μBBB). LAB ON A CHIP 2012; 12:1784-92. [PMID: 22422217 DOI: 10.1039/c2lc40094d] [Citation(s) in RCA: 500] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The blood-brain barrier (BBB), a unique selective barrier for the central nervous system (CNS), hinders the passage of most compounds to the CNS, complicating drug development. Innovative in vitro models of the BBB can provide useful insights into its role in CNS disease progression and drug delivery. Static transwell models lack fluidic shear stress, while the conventional dynamic in vitro BBB lacks a thin dual cell layer interface. To address both limitations, we developed a microfluidic blood-brain barrier (μBBB) which closely mimics the in vivo BBB with a dynamic environment and a comparatively thin culture membrane (10 μm). To test validity of the fabricated BBB model, μBBBs were cultured with b.End3 endothelial cells, both with and without co-cultured C8-D1A astrocytes, and their key properties were tested with optical imaging, trans-endothelial electrical resistance (TEER), and permeability assays. The resultant imaging of ZO-1 revealed clearly expressed tight junctions in b.End3 cells, Live/Dead assays indicated high cell viability, and astrocytic morphology of C8-D1A cells were confirmed by ESEM and GFAP immunostains. By day 3 of endothelial culture, TEER levels typically exceeded 250 Ω cm(2) in μBBB co-cultures, and 25 Ω cm(2) for transwell co-cultures. Instantaneous transient drop in TEER in response to histamine exposure was observed in real-time, followed by recovery, implying stability of the fabricated μBBB model. Resultant permeability coefficients were comparable to previous BBB models, and were significantly increased at higher pH (>10). These results demonstrate that the developed μBBB system is a valid model for some studies of BBB function and drug delivery.
Collapse
Affiliation(s)
- Ross Booth
- Department of Bioengineering, University of Utah, MEB-1445, 50 S Central Campus Dr, Salt Lake City, UT 84112, USA.
| | | |
Collapse
|
61
|
Cao Q, Whittemore SR. Cell transplantation: stem cells and precursor cells. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:551-61. [PMID: 23098736 DOI: 10.1016/b978-0-444-52137-8.00034-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stem cells have been used to approach four different therapeutic repair strategies in spinal cord injury (SCI): (1) replacement of lost neurons, (2) replacement of oligodendrocytes to promote remyelination of demyelinated and/or regenerated axons, (3) providing a permissive substrate for axonal regeneration to overcome the intrinsic inhibition of surface molecules, and (4) engendering host repair. The first two strategies involve cell-specific differentiation of engrafted neural cells and the latter two may involve grafted neural or non-neural cells. The preclinical data for all of these approaches is at times contradictory and there is no consensus as to what type of stem cell is optimal to facilitate repair in specific injuries. Remyelination has been the most successful stem cell replacement strategy. Partial lineage restriction and pharmacological and/or genetic manipulation to express additional trophic support or restrict responses to host signals appears necessary for optimal neuronal and oligodendrocytic differentiation. However, these modifications will make their clinical application exceedingly difficult. Effects of grafted stem cells on abrogating host immune responses and engendering intrinsic repair is also a mechanism through which stem cells are likely therapeutically beneficial. While clinical trials with stem cell grafting into the injured spinal cord are ongoing, preclinical studies have yet to define mechanisms of action that can be definitively translated to those clinical approaches.
Collapse
Affiliation(s)
- Qilin Cao
- Department of Neurosurgery, University of Texas Medical School, Houston, TX, USA
| | | |
Collapse
|
62
|
Wuest DM, Lee KH. Optimization of endothelial cell growth in a murine in vitro blood-brain barrier model. Biotechnol J 2011; 7:409-17. [PMID: 22095877 PMCID: PMC3488296 DOI: 10.1002/biot.201100189] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 09/29/2011] [Accepted: 11/11/2011] [Indexed: 11/06/2022]
Abstract
In vitro cell culture models of the blood-brain barrier (BBB) are important tools used to study cellular physiology and brain disease therapeutics. Although the number of model configurations is expanding across neuroscience laboratories, it is not clear that any have been effectively optimized. A sequential screening study to identify optimal primary mouse endothelial cell parameter set points, grown alone and in combination with common model enhancements, including co-culturing with primary mouse or rat astrocytes and addition of biochemical agents in the media, was performed. A range of endothelial cell-seeding densities (1-8 × 10(5) cells/cm(2) ) and astrocyte-seeding densities (2-8 × 10(4) cells/cm(2) ) were studied over seven days in the system, and three distinct media-feeding strategies were compared to optimize biochemical agent exposure time. Implementation of all optimal set points increased transendothelial electrical resistance by over 200% compared to an initial model and established a suitable in vitro model for brain disease application studies. These results demonstrate the importance of optimizing cell culture growth, which is the most important parameter in creating an in vitro BBB model as it directly relates the model to the in vivo arrangement.
Collapse
Affiliation(s)
- Diane M Wuest
- Chemical Engineering and Delaware Biotechnology Institute, University of Delaware, Newark, DE 19711, USA
| | | |
Collapse
|
63
|
Wisniewska-Kruk J, Hoeben KA, Vogels IMC, Gaillard PJ, Van Noorden CJF, Schlingemann RO, Klaassen I. A novel co-culture model of the blood-retinal barrier based on primary retinal endothelial cells, pericytes and astrocytes. Exp Eye Res 2011; 96:181-90. [PMID: 22200486 DOI: 10.1016/j.exer.2011.12.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Revised: 11/11/2011] [Accepted: 12/03/2011] [Indexed: 12/13/2022]
Abstract
Loss of blood-retinal barrier (BRB) properties is an important feature in the pathology of diabetic macular edema (DME), but cellular mechanisms underlying BRB dysfunction are poorly understood. Therefore, we developed and characterized a novel in vitro BRB model, based on primary bovine retinal endothelial cells (BRECs). These cells were shown to maintain specific in vivo BRB properties by expressing high levels of the endothelial junction proteins occludin, claudin-5, VE-cadherin and ZO-1 at cell borders, and the specific pumps glucose transporter-1 (GLUT1) and efflux transporter P-glycoprotein (MDR1). To investigate the influence of pericytes and astrocytes on BRB maintenance in vitro, we compared five different co-culture BRB models, based on BRECs, bovine retinal pericytes (BRPCs) and rat glial cells. Co-cultures of BRECs with BRPCs and glial cells showed the highest trans-endothelial resistance (TEER) as well as decreased permeability of tracers after vascular endothelial growth factor (VEGF) stimulation, suggesting a major role for these cell types in maintaining barrier properties. To mimic the in vivo situation of DME, we stimulated BRECs with VEGF, which downregulated MDR1 and GLUT1 mRNA levels, transiently reduced expression levels of endothelial junctional proteins and altered their organization, increased the number of intercellular gaps in BRECs monolayers and influence the permeability of the model to differently-sized molecular tracers. Moreover, as has been shown in vivo, expression of plasmalemma vesicle-associated protein (PLVAP) was increased in endothelial cells in the presence of VEGF. This in vitro model is the first co-culture model of the BRB that mimicks in vivo VEGF-dependent changes occurring in DME.
Collapse
|
64
|
Lippmann ES, Weidenfeller C, Svendsen CN, Shusta EV. Blood-brain barrier modeling with co-cultured neural progenitor cell-derived astrocytes and neurons. J Neurochem 2011; 119:507-20. [PMID: 21854389 DOI: 10.1111/j.1471-4159.2011.07434.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
In vitro blood-brain barrier (BBB) models often consist of brain microvascular endothelial cells (BMECs) that are co-cultured with other cells of the neurovascular unit, such as astrocytes and neurons, to enhance BBB properties. Obtaining primary astrocytes and neurons for co-culture models can be laborious, while yield and heterogeneity of primary isolations can also be limiting. Neural progenitor cells (NPCs), because of their self-renewal capacity and ability to reproducibly differentiate into tunable mixtures of neurons and astrocytes, represent a facile, readily scalable alternative. To this end, differentiated rat NPCs were co-cultured with rat BMECs and shown to induce BBB properties such as elevated trans-endothelial electrical resistance, improved tight junction continuity, polarized p-glycoprotein efflux, and low passive permeability at levels indistinguishable from those induced by primary rat astrocyte co-culture. An NPC differentiation time of 12 days, with the presence of 10% fetal bovine serum, was found to be crucial for generating NPC-derived progeny capable of inducing the optimal response. This approach could also be extended to human NPC-derived astrocytes and neurons which similarly regulated BBB induction. The distribution of rat or human NPC-derived progeny under these conditions was found to be a roughly 3 : 1 mixture of astrocytes to neurons with varying degrees of cellular maturity. BMEC gene expression analysis was conducted using a BBB gene panel, and it was determined that 23 of 26 genes were similarly regulated by either differentiated rat NPC or rat astrocyte co-culture while three genes were differentially altered by the rat NPC-derived progeny. Taken together, these results demonstrate that NPCs are an attractive alternative to primary neural cells for use in BBB co-culture models.
Collapse
Affiliation(s)
- Ethan S Lippmann
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | | | | | | |
Collapse
|
65
|
Shayan G, Shuler ML, Lee KH. The effect of astrocytes on the induction of barrier properties in aortic endothelial cells. Biotechnol Prog 2011; 27:1137-45. [PMID: 21626719 DOI: 10.1002/btpr.620] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 04/08/2011] [Indexed: 11/06/2022]
Abstract
Construction of in vitro models of the blood-brain barrier (BBB) using primary brain microvascular endothelial cells (BMEC) is time intensive and not high throughput, in part due to a lack of culture purity, low yields, and cellular dedifferentiation after the first passage. This problem has created interest in the substitution of BMEC with immortalized brain endothelial cells (EC), or peripheral EC such as bovine aortic EC (BAEC). Many BBB models have focused on further inducing the brain and peripheral ECs by incorporating astrocyte back-to-back or nonback-to-back cocultures. However, previous studies demonstrating induction effects of astrocytes on BAEC in back-to-back cocultures failed to recognize the extensive barrier properties of astrocytes alone, which can have a significant effect on interpreting the results. This manuscript reports the establishment of back-to-back and nonback-to-back cocultures between astrocytes and BAEC or BMEC (as a control) with primary focus on the properties of astrocytes alone and with a linear contrast statistical methodology to interpret the results. Transendothelial electrical resistance and permeability studies revealed that astrocytes can significantly increase the barrier tightening of BMEC by 167%, while having no effect on BAEC. Immunocytochemical studies also revealed the reorganization of BMEC occludin junctions in the presence of astrocytes, while indicating the absence of this junctional protein in BAEC. In contrast to a previous report, here the linear contrast statistical analysis revealed that observed decreases in permeability of BAEC in back-to-back cocultures is due to the addition of astrocytes' properties in series and not due to induction.
Collapse
Affiliation(s)
- Gilda Shayan
- Dept. of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | | |
Collapse
|
66
|
Murine in vitro model of the blood-brain barrier for evaluating drug transport. Eur J Pharm Sci 2010; 42:148-55. [PMID: 21078386 DOI: 10.1016/j.ejps.2010.11.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Revised: 11/02/2010] [Accepted: 11/06/2010] [Indexed: 11/23/2022]
Abstract
In vitro blood-brain barrier (BBB) models help predict brain uptake of potential central nervous system drug candidates. Current in vitro models are composed of brain microvascular endothelial cells (BMEC) that are isolated from rat, bovine, or porcine. However, most in vivo studies on drug transport through the BBB are performed in small laboratory animals, specially mouse and thus murine in vitro BBB models serve as better surrogates to correlate with these studies. Here we describe the functional characterization of a reproducible in vitro model composed of murine BMEC co-cultured with rat primary astrocytes in the presence of biochemical inducing agents. The co-cultures presented high TEER and low sodium fluorescein permeability. Expression of specific BBB tight junction proteins (occludin, claudin-5, ZO-1) and the functionality of transporters (Pgp, GLUT1) were detected by immunocytochemistry and Western blotting. These results indicated a 2.5-fold increase in the expression levels of these proteins in the presence of astrocytes. In addition, a high correlation coefficient (0.98) was obtained between the permeability of a series of hydrophobic and hydrophilic drugs and their corresponding in vivo values. These results together establish the utility of this murine model for future drug transport, pathological, and pharmacological characterizations of the BBB.
Collapse
|
67
|
Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, Barres BA. The mouse blood-brain barrier transcriptome: a new resource for understanding the development and function of brain endothelial cells. PLoS One 2010; 5:e13741. [PMID: 21060791 PMCID: PMC2966423 DOI: 10.1371/journal.pone.0013741] [Citation(s) in RCA: 421] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Accepted: 08/24/2010] [Indexed: 01/21/2023] Open
Abstract
The blood-brain barrier (BBB) maintains brain homeostasis and limits the entry of toxins and pathogens into the brain. Despite its importance, little is known about the molecular mechanisms regulating the development and function of this crucial barrier. In this study we have developed methods to highly purify and gene profile endothelial cells from different tissues, and by comparing the transcriptional profile of brain endothelial cells with those purified from the liver and lung, we have generated a comprehensive resource of transcripts that are enriched in the BBB forming endothelial cells of the brain. Through this comparison we have identified novel tight junction proteins, transporters, metabolic enzymes, signaling components, and unknown transcripts whose expression is enriched in central nervous system (CNS) endothelial cells. This analysis has identified that RXRalpha signaling cascade is specifically enriched at the BBB, implicating this pathway in regulating this vital barrier. This dataset provides a resource for understanding CNS endothelial cells and their interaction with neural and hematogenous cells.
Collapse
Affiliation(s)
- Richard Daneman
- Department of Anatomy, University of California San Francisco, San Francisco, California, United States of America.
| | | | | | | | | | | |
Collapse
|
68
|
Pericytes are required for blood-brain barrier integrity during embryogenesis. Nature 2010; 468:562-6. [PMID: 20944625 DOI: 10.1038/nature09513] [Citation(s) in RCA: 1474] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 09/23/2010] [Indexed: 12/14/2022]
Abstract
Vascular endothelial cells in the central nervous system (CNS) form a barrier that restricts the movement of molecules and ions between the blood and the brain. This blood-brain barrier (BBB) is crucial to ensure proper neuronal function and protect the CNS from injury and disease. Transplantation studies have demonstrated that the BBB is not intrinsic to the endothelial cells, but is induced by interactions with the neural cells. Owing to the close spatial relationship between astrocytes and endothelial cells, it has been hypothesized that astrocytes induce this critical barrier postnatally, but the timing of BBB formation has been controversial. Here we demonstrate that the barrier is formed during embryogenesis as endothelial cells invade the CNS and pericytes are recruited to the nascent vessels, over a week before astrocyte generation. Analysing mice with null and hypomorphic alleles of Pdgfrb, which have defects in pericyte generation, we demonstrate that pericytes are necessary for the formation of the BBB, and that absolute pericyte coverage determines relative vascular permeability. We demonstrate that pericytes regulate functional aspects of the BBB, including the formation of tight junctions and vesicle trafficking in CNS endothelial cells. Pericytes do not induce BBB-specific gene expression in CNS endothelial cells, but inhibit the expression of molecules that increase vascular permeability and CNS immune cell infiltration. These data indicate that pericyte-endothelial cell interactions are critical to regulate the BBB during development, and disruption of these interactions may lead to BBB dysfunction and neuroinflammation during CNS injury and disease.
Collapse
|
69
|
Veiga DD, Antunes JC, Gómez RG, Mano JF, Ribelles JLG, Soria JM. Three-Dimensional Scaffolds as a Model System for Neural and Endothelial ‘In Vitro’ Culture. J Biomater Appl 2010; 26:293-310. [DOI: 10.1177/0885328210365005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Biomaterials based on the hydrophobic homopolymer poly(ethyl acrylate), PEA, and its copolymers with hydroxyethyl acrylate, p(EA-co-HEA) and methacrylic acid, p(EA-co-MAAc) were prepared as polymeric scaffolds with interconnected pores of 90 microns and tested in vitro as culture substrates and compared for their impact on the differentiation of neural stem cells (NSC) obtained from the subventricular zone (SVZ) of postnatal rats and human endothelial cells (HUVEC). Immunocytochemical staining assay for specific markers show that p(EA-co-MAAc) scaffolds were suitable substrates to promote cell attachment and differentiation of adult NSC and HUVEC cells.
Collapse
Affiliation(s)
- D. Dias Veiga
- Center for Biomaterials and Tissue Engineering, Universidad Politécnica de Valencia, Camino de Vera s/n E -46022 Valencia, Spain, 3B's Research Group - Biomaterials, Biodegradables and Biomimetics Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio do Barco 4806-909 Taipas, Guimarães, Portugal, IBB - Institute for Biotechnology and Bioengineering, PT Government Associated Laboratory, Guimarães, Portugal
| | - Joana Costa Antunes
- Center for Biomaterials and Tissue Engineering, Universidad Politécnica de Valencia, Camino de Vera s/n E -46022 Valencia, Spain, 3B's Research Group - Biomaterials, Biodegradables and Biomimetics Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio do Barco 4806-909 Taipas, Guimarães, Portugal, IBB - Institute for Biotechnology and Bioengineering, PT Government Associated Laboratory, Guimarães, Portugal
| | - Roberto García Gómez
- Centro de Investigación Príncipe Felipe, Regenerative Medicine Unit Autopista del Saler 16, E -46013 Valencia, Spain
| | - João F. Mano
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio do Barco 4806-909 Taipas, Guimarães, Portugal, IBB - Institute for Biotechnology and Bioengineering, PT Government Associated Laboratory, Guimarães, Portugal
| | - Jose Luis Gómez Ribelles
- Center for Biomaterials and Tissue Engineering, Universidad Politécnica de Valencia, Camino de Vera s/n E -46022 Valencia, Spain, Centro de Investigación Príncipe Felipe, Regenerative Medicine Unit Autopista del Saler 16, E -46013 Valencia, Spain, CIBER en Bioingeniería, Biomateriales y Nanomedicina, Valencia, Spain
| | - Jose Miguel Soria
- Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera Avda Seminario s/n, 46113 Moncada, Valencia, Spain, CIBER en enfermedades Neurodegenerativas, Valencia, Spain,
| |
Collapse
|
70
|
Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol 2010; 119:7-35. [PMID: 20012068 PMCID: PMC2799634 DOI: 10.1007/s00401-009-0619-8] [Citation(s) in RCA: 3515] [Impact Index Per Article: 251.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 11/23/2009] [Accepted: 11/24/2009] [Indexed: 02/06/2023]
Abstract
Astrocytes are specialized glial cells that outnumber neurons by over fivefold. They contiguously tile the entire central nervous system (CNS) and exert many essential complex functions in the healthy CNS. Astrocytes respond to all forms of CNS insults through a process referred to as reactive astrogliosis, which has become a pathological hallmark of CNS structural lesions. Substantial progress has been made recently in determining functions and mechanisms of reactive astrogliosis and in identifying roles of astrocytes in CNS disorders and pathologies. A vast molecular arsenal at the disposal of reactive astrocytes is being defined. Transgenic mouse models are dissecting specific aspects of reactive astrocytosis and glial scar formation in vivo. Astrocyte involvement in specific clinicopathological entities is being defined. It is now clear that reactive astrogliosis is not a simple all-or-none phenomenon but is a finely gradated continuum of changes that occur in context-dependent manners regulated by specific signaling events. These changes range from reversible alterations in gene expression and cell hypertrophy with preservation of cellular domains and tissue structure, to long-lasting scar formation with rearrangement of tissue structure. Increasing evidence points towards the potential of reactive astrogliosis to play either primary or contributing roles in CNS disorders via loss of normal astrocyte functions or gain of abnormal effects. This article reviews (1) astrocyte functions in healthy CNS, (2) mechanisms and functions of reactive astrogliosis and glial scar formation, and (3) ways in which reactive astrocytes may cause or contribute to specific CNS disorders and lesions.
Collapse
|
71
|
Daneman R, Rescigno M. The gut immune barrier and the blood-brain barrier: are they so different? Immunity 2009; 31:722-35. [PMID: 19836264 DOI: 10.1016/j.immuni.2009.09.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Accepted: 09/29/2009] [Indexed: 12/18/2022]
Abstract
In order to protect itself from a diverse set of environmental pathogens and toxins, the body has developed a number of barrier mechanisms to limit the entry of potential hazards. Here, we compare two such barriers: the gut immune barrier, which is the primary barrier against pathogens and toxins ingested in food, and the blood-brain barrier, which protects the central nervous system from pathogens and toxins in the blood. Although each barrier provides defense in very different environments, there are many similarities in their mechanisms of action. In both cases, there is a physical barrier formed by a cellular layer that tightly regulates the movement of ions, molecules, and cells between two tissue spaces. These barrier cells interact with different cell types, which dynamically regulate their function, and with a different array of immune cells that survey the physical barrier and provide innate and adaptive immunity.
Collapse
Affiliation(s)
- Richard Daneman
- University of California, San Francisco, Department of Anatomy, San Francisco, CA 94143-0452, USA.
| | | |
Collapse
|
72
|
Plane JM, Andjelkovic AV, Keep RF, Parent JM. Intact and injured endothelial cells differentially modulate postnatal murine forebrain neural stem cells. Neurobiol Dis 2009; 37:218-27. [PMID: 19837162 DOI: 10.1016/j.nbd.2009.10.008] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 09/04/2009] [Accepted: 10/09/2009] [Indexed: 11/20/2022] Open
Abstract
Neural stem cells (NSCs) persist in the forebrain subventricular zone (SVZ) within a niche containing endothelial cells. Evidence suggests that endothelial cells stimulate NSC expansion and neurogenesis. Experimental stroke increases neurogenesis and angiogenesis, but how endothelial cells influence stroke-induced neurogenesis is unknown. We hypothesized intact or oxygen-glucose deprived (OGD) endothelial cells secrete factors that enhance neurogenesis. We co-cultured mouse SVZ neurospheres (NS) with endothelial cells, or differentiated NS in endothelial cell-conditioned medium (ECCM). NS also were expanded in ECCM from OGD-exposed (OGD-ECCM) endothelial cells to assess injury effects. ECCM significantly increased NS production. NS co-cultured with endothelial cells or ECCM generated more immature-appearing neurons and oligodendrocytes, and astrocytes with radial glial-like/reactive morphology than controls. OGD-ECCM stimulated neuroblast migration and yielded neurons with longer processes and more branching. These data indicate that intact and injured endothelial cells exert differing effects on NSCs, and suggest targets for stimulating regeneration after brain insults.
Collapse
Affiliation(s)
- Jennifer M Plane
- Department of Neurology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| | | | | | | |
Collapse
|
73
|
Alcock J, Sottile V. Dynamic distribution and stem cell characteristics of Sox1-expressing cells in the cerebellar cortex. Cell Res 2009; 19:1324-33. [PMID: 19823196 DOI: 10.1038/cr.2009.119] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Bergmann glia cells are a discrete radial glia population surrounding Purkinje cells in the cerebellar cortex. Although Bergmann glia are essential for the development and correct arborization of Purkinje cells, little is known about the regulation of this cell population after the developmental phase. In an effort to characterize this population at the molecular level, we have analyzed marker expression and established that adult Bergmann glia express Sox1, Sox2 and Sox9, a feature otherwise associated with neural stem cells (NSCs). In the present study, we have further analyzed the developmental pattern of Sox1-expressing cells in the developing cerebellum. We report that before becoming restricted to the Purkinje cell layer, Sox1-positive cells are present throughout the immature tissue, and that these cells show characteristics of Bergmann glia progenitors. Our study shows that these progenitors express Sox1, Sox2 and Sox9, a signature maintained throughout cerebellar maturation into adulthood. When isolated in culture, the Sox1-expressing cerebellar population exhibited neurosphere-forming ability, NSC-marker characteristics, and demonstrated multipotency at the clonal level. Our results show that the Bergmann glia population expresses Sox1 during cerebellar development, and that these cells can be isolated and show stem cell characteristics in vitro, suggesting that they could hold a broader potential than previously thought.
Collapse
Affiliation(s)
- Joelle Alcock
- Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), University Park - CBS building, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | | |
Collapse
|
74
|
Ivey NS, MacLean AG, Lackner AA. Acquired immunodeficiency syndrome and the blood-brain barrier. J Neurovirol 2009; 15:111-22. [PMID: 19306229 DOI: 10.1080/13550280902769764] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier (BBB) plays a critical role in normal physiology of the central nervous system by regulating what reaches the brain from the periphery. The BBB also plays a major role in neurologic disease including neuropathologic sequelae associated with infection by human immunodeficiency virus (HIV) in humans and the closely related simian immunodeficiency virus (SIV) in macaques. In this review, we provide an overview of the function, structure, and components of the BBB, followed by a more detailed discussion of the subcellular structures and regulation of the tight junction. We then discuss the ways in which HIV/SIV affects the BBB, largely through infection of monocytes/macrophages, and how infected macrophages crossing the BBB ultimately results in breakdown of the barrier.
Collapse
Affiliation(s)
- Nathan S Ivey
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | | | | |
Collapse
|
75
|
Abstract
Cell therapy plays an important role in multidisciplinary management of the two major forms of central nervous system (CNS) injury, traumatic brain injury and spinal cord injury, which are caused by external physical trauma. Cell therapy for CNS disorders involves the use of cells of neural or non-neural origin to replace, repair, or enhance the function of the damaged nervous system and is usually achieved by transplantation of the cells, which are isolated and may be modified, e.g., by genetic engineering, when it may be referred to as gene therapy. Because the adult brain cells have a limited capacity to migrate to and regenerate at sites of injury, the use of embryonic stem cells that can be differentiated into various cell types as well as the use of neural stem cells has been explored. Preclinical studies and clinical trials are reviewed. Advantages as well as limitations are discussed. Cell therapy is promising for the treatment of CNS injury because it targets multiple mechanisms in a sustained manner. It can provide repair and regeneration of damaged tissues as well as prolonged release of neuroprotective and other therapeutic substances.
Collapse
|
76
|
Wnt/beta-catenin signaling is required for CNS, but not non-CNS, angiogenesis. Proc Natl Acad Sci U S A 2009; 106:641-6. [PMID: 19129494 DOI: 10.1073/pnas.0805165106] [Citation(s) in RCA: 562] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite the importance of CNS blood vessels, the molecular mechanisms that regulate CNS angiogenesis and blood-brain barrier (BBB) formation are largely unknown. Here we analyze the role of Wnt/beta-catenin signaling in regulating the formation of CNS blood vessels. First, through the analysis of TOP-Gal Wnt reporter mice, we identify that canonical Wnt/beta-catenin signaling is specifically activated in CNS, but not non-CNS, blood vessels during development. This activation correlates with the expression of different Wnt ligands by neural progenitor cells in distinct locations throughout the CNS, including Wnt7a and Wnt7b in ventral regions and Wnt1, Wnt3, Wnt3a, and Wnt4 in dorsal regions. Blockade of Wnt/beta-catenin signaling in vivo specifically disrupts CNS, but not non-CNS, angiogenesis. These defects include reduction in vessel number, loss of capillary beds, and the formation of hemorrhagic vascular malformations that remain adherent to the meninges. Furthermore, we demonstrate that Wnt/beta-catenin signaling regulates the expression of the BBB-specific glucose transporter glut-1. Taken together these experiments reveal an essential role for Wnt/beta-catenin signaling in driving CNS-specific angiogenesis and provide molecular evidence that angiogenesis and BBB formation are in part linked.
Collapse
|
77
|
Cecchelli R, Berezowski V, Lundquist S, Culot M, Renftel M, Dehouck MP, Fenart L. Modelling of the blood–brain barrier in drug discovery and development. Nat Rev Drug Discov 2007; 6:650-61. [PMID: 17667956 DOI: 10.1038/nrd2368] [Citation(s) in RCA: 435] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The market for neuropharmaceuticals is potentially one of the largest sectors of the global pharmaceutical market owing to the increase in average life expectancy and the fact that many neurological disorders have been largely refractory to pharmacotherapy. The brain is a delicate organ that can efficiently protect itself from harmful compounds and precisely regulate its microenvironment. Unfortunately, the same mechanisms can also prove to be formidable hurdles in drug development. An improved understanding of the regulatory interfaces that exist between blood and brain may provide novel and more effective strategies to treat neurological disorders.
Collapse
Affiliation(s)
- Romeo Cecchelli
- Laboratoire de physiopathologie de la barrière hémato-encéphalique E.A.2465, IMPRT IFR-114, Université d'Artois, Faculté Jean Perrin, 62307 Lens, France.
| | | | | | | | | | | | | |
Collapse
|