Glia-Neuronal and Glia-Vascular Interrelations in Blood-Brain Barrier Formation and Axon Regeneration in Vertebrates

Author(s):  
Hartwig Wolburg
Author(s):  
V. A. Ruzaeva ◽  
A. V. Morgun ◽  
E. D. Khilazheva ◽  
N. V. Kuvacheva ◽  
E. A. Pozhilenkova ◽  
...  

2009 ◽  
Vol 29 (9) ◽  
pp. 1491-1502 ◽  
Author(s):  
Ruth Lyck ◽  
Nadine Ruderisch ◽  
Anton G Moll ◽  
Oliver Steiner ◽  
Clemens D Cohen ◽  
...  

Tight homeostatic control of brain amino acids (AA) depends on transport by solute carrier family proteins expressed by the blood—brain barrier (BBB) microvascular endothelial cells (BMEC). To characterize the mouse BMEC transcriptome and probe culture-induced changes, microarray analyses of platelet endothelial cell adhesion molecule-1-positive (PECAM1+) endothelial cells (ppMBMECs) were compared with primary MBMECs (pMBMEC) cultured in the presence or absence of glial cells and with b.End5 endothelioma cell line. Selected cell marker and AA transporter mRNA levels were further verified by reverse transcription real-time PCR. Regardless of glial coculture, expression of a large subset of genes was strongly altered by a brief culture step. This is consistent with the known dependence of BMECs on in vivo interactions to maintain physiologic functions, for example, tight barrier formation, and their consequent dedifferentiation in culture. Seven ( 4F2hc, Lat1, Taut, Snat3, Snat5, Xpct, and Cat1) of nine AA transporter mRNAs highly expressed in freshly isolated ppMBMECs were strongly downregulated for all cultures and two ( Snat2 and Eaat3) were variably regulated. In contrast, five AA transporter mRNAs with low expression in ppMBMECs, including y+ Lat2, xCT, and Snat1, were upregulated by culture. We hypothesized that the AA transporters highly expressed in ppMBMECs and downregulated in culture have a major in vivo function for BBB transendothelial transport.


2019 ◽  
Vol 35 (1) ◽  
pp. 591-613 ◽  
Author(s):  
Urs H. Langen ◽  
Swathi Ayloo ◽  
Chenghua Gu

The vertebrate vasculature displays high organotypic specialization, with the structure and function of blood vessels catering to the specific needs of each tissue. A unique feature of the central nervous system (CNS) vasculature is the blood-brain barrier (BBB). The BBB regulates substance influx and efflux to maintain a homeostatic environment for proper brain function. Here, we review the development and cell biology of the BBB, focusing on the cellular and molecular regulation of barrier formation and the maintenance of the BBB through adulthood. We summarize unique features of CNS endothelial cells and highlight recent progress in and general principles of barrier regulation. Finally, we illustrate why a mechanistic understanding of the development and maintenance of the BBB could provide novel therapeutic opportunities for CNS drug delivery.


2000 ◽  
Vol 858 (1) ◽  
pp. 172-176 ◽  
Author(s):  
Hideyuki Akiyama ◽  
Takeshi Kondoh ◽  
Takashi Kokunai ◽  
Tatsuya Nagashima ◽  
Naoaki Saito ◽  
...  

Cell ◽  
2005 ◽  
Vol 123 (1) ◽  
pp. 133-144 ◽  
Author(s):  
Tina Schwabe ◽  
Roland J. Bainton ◽  
Richard D. Fetter ◽  
Ulrike Heberlein ◽  
Ulrike Gaul

Stroke ◽  
2020 ◽  
Vol 51 (Suppl_1) ◽  
Author(s):  
Peeyush Thankamani Pandit ◽  
Tanuja Kulkarni ◽  
Devin McBride ◽  
Iny Elizebeth Mathew ◽  
Spiros Blackburn

The blood brain barrier (BBB), unique to the central nervous system (CNS) vessels, tightly controls the passage of molecules between the blood and the brain. BBB breakdown is documented in many pathological conditions including, stroke, and contributes to the severity of disease and poor outcome. Currently, there is no efficient treatment strategy to prevent BBB breakdown or restore disrupted BBB . Despite BBB’s importance in the CNS function, t he genetic program and mechanism, that regulates BBB formation and maintenance is poorly characterized. To understand the molecular features underlying BBB gene regulation during development we utilize endothelial cell (EC) cultures derived from brain of both embryonic (E-13.5) and adult mice and investigated differences in BBB gene expression and epigenetic status using real time PCR analysis and ChIP-qPCR. We found that a distinct BBB profile exist in the CNS ECs during BBB formation and maintenance . When compared to embryonic ECs, adult ECs showed a significant down regulation in expression of tight junction (TJ) genes such as CLDN-1 , CLDN -11 , ZO-1 and OCLN . Conversely, another important TJ gene CLDN- 5 shows a significant increase in adult ECs. Further, we found that epigenetic-histone modifications are involved in the repression in the TJ gene in adult ECs. To more fully understand the role of Wnt/β-catenin in BBB formation and maintenance we block the downstream of the Wnt/β-catenin signaling pathway using LF3 (block the interaction of β-catenin and transcription factor TCF4) in developing ECs. This results in a adult BBB phenotype in developing ECs indicating, silencing of Wnt/β-catenin pathway is required for BBB maturation . Further, epigenetic component HDAC2 was increased significantly in developing ECs when Wnt/β-catenin signaling is blocked demonstrating this pathway can modulate the epigenetics of CNS ECs . In summary, we describe that BBB genes have distinguished expression pattern during BBB formation and maintenance, guided by the Wnt/β-catenin pathway through epigenetic modifications. Understanding the fundamental epigenetic and regulatory mechanisms of BBB formation and maintenance is critical for developing therapeutic interventions against BBB breakdown.


2015 ◽  
Vol 208 (6) ◽  
pp. 703-711 ◽  
Author(s):  
Fabien Sohet ◽  
Christina Lin ◽  
Roeben N. Munji ◽  
Seo Yeon Lee ◽  
Nadine Ruderisch ◽  
...  

The blood–brain barrier (BBB) is a term used to describe the unique properties of central nervous system (CNS) blood vessels. One important BBB property is the formation of a paracellular barrier made by tight junctions (TJs) between CNS endothelial cells (ECs). Here, we show that Lipolysis-stimulated lipoprotein receptor (LSR), a component of paracellular junctions at points in which three cell membranes meet, is greatly enriched in CNS ECs compared with ECs in other nonneural tissues. We demonstrate that LSR is specifically expressed at tricellular junctions and that its expression correlates with the onset of BBB formation during embryogenesis. We further demonstrate that the BBB does not seal during embryogenesis in Lsr knockout mice with a leakage to small molecules. Finally, in mouse models in which BBB was disrupted, including an experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis and a middle cerebral artery occlusion (MCAO) model of stroke, LSR was down-regulated, linking loss of LSR and pathological BBB leakage.


Sign in / Sign up

Export Citation Format

Share Document