1
|
Fetsko AR, Sebo DJ, Budzynski LB, Scharbarth A, Taylor MR. IL-1β disrupts the initiation of blood-brain barrier development by inhibiting endothelial Wnt/β-catenin signaling. iScience 2024; 27:109651. [PMID: 38638574 PMCID: PMC11025013 DOI: 10.1016/j.isci.2024.109651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/06/2024] [Accepted: 03/29/2024] [Indexed: 04/20/2024] Open
Abstract
During neuroinflammation, the proinflammatory cytokine interleukin-1β (IL-1β) impacts blood-brain barrier (BBB) function by disrupting brain endothelial tight junctions, promoting vascular permeability, and increasing transmigration of immune cells. Here, we examined the effects of Il-1β on the in vivo initiation of BBB development. We generated doxycycline-inducible transgenic zebrafish to secrete Il-1β in the CNS. To validate the utility of our model, we showed Il-1β dose-dependent mortality, recruitment of neutrophils, and expansion of microglia. Using live imaging, we discovered that Il-1β causes a significant reduction in CNS angiogenesis and barriergenesis. To demonstrate specificity, we rescued the Il-1β induced phenotypes by targeting the zebrafish il1r1 gene using CRISPR-Cas9. Mechanistically, we determined that Il-1β disrupts the initiation of BBB development by decreasing Wnt/β-catenin transcriptional activation in brain endothelial cells. Given that several neurodevelopmental disorders are associated with inflammation, our findings support further investigation into the connections between proinflammatory cytokines, neuroinflammation, and neurovascular development.
Collapse
Affiliation(s)
- Audrey R. Fetsko
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dylan J. Sebo
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Lilyana B. Budzynski
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Alli Scharbarth
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michael R. Taylor
- School of Pharmacy, Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
- School of Pharmacy, Pharmacology and Toxicology Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
2
|
Göbel C, Schoof M, Holdhof D, Spohn M, Schüller U. SMARCA4 Loss and Mutated β-Catenin Induce Proliferative Lesions in the Murine Embryonic Cerebellum. J Neurosci 2024; 44:e1605232024. [PMID: 38383496 PMCID: PMC11007475 DOI: 10.1523/jneurosci.1605-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
Almost all medulloblastomas (MB) of the Wingless/Int-1 (WNT) type are characterized by hotspot mutations in CTNNB1, and mouse models have convincingly demonstrated the tumor-initiating role of these mutations. Additional alterations in SMARCA4 are detected in ∼20% of WNT MB, but their functional role is mostly unknown. We, therefore, amended previously described brain lipid binding protein (Blbp)-cre::Ctnnb1(ex3)fl/wt mice by the introduction of floxed Smarca4 alleles. Unexpectedly, mutated and thereby stabilized β-catenin on its own induced severe developmental phenotypes in male and female Blbp-cre::Ctnnb1(ex3)fl/wt mice in our hands, including a thinned cerebral cortex, hydrocephalus, missing cerebellar layering, and cell accumulations in the brainstem and cerebellum. An additional loss of SMARCA4 even resulted in prenatal death for most mice. Respective Blbp-cre::Ctnnb1(ex3)fl/wt::Smarca4fl/rec mutants (male and female) developed large proliferative lesions in the cerebellum evolving from E13.5 to E16.5. Histological and molecular analysis of these lesions by DNA methylation profiling and single-cell RNA sequencing suggested an origin in early undifferentiated SOX2-positive cerebellar progenitors. Furthermore, upregulated WNT signaling, altered actin/cytoskeleton organization, and reduced neuronal differentiation were evident in mutant cells. In vitro, cells harboring alterations in both Ctnnb1 and Smarca4 were negatively selected and did not show tumorigenic potential after transplantation in adult female recipient mice. However, in cerebellar explant cultures, mutant cells displayed significantly increased proliferation, suggesting an important role of the embryonic microenvironment in the development of lesions. Altogether, these results represent an important first step toward the unraveling of tumorigenic mechanisms induced by aberrant WNT signaling and SMARCA4 deficiency.
Collapse
Affiliation(s)
- Carolin Göbel
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg D-20251, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg D-20251, Germany
| | - Melanie Schoof
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg D-20251, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg D-20251, Germany
| | - Dörthe Holdhof
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg D-20251, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg D-20251, Germany
| | - Michael Spohn
- Research Institute Children's Cancer Center Hamburg, Hamburg D-20251, Germany
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg D-20251, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg D-20251, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg D-20251, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg D-20251, Germany
| |
Collapse
|
3
|
Goncalves A, Antonetti DA. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS. Fluids Barriers CNS 2022; 19:86. [PMID: 36320068 PMCID: PMC9628113 DOI: 10.1186/s12987-022-00386-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
The unique environment of the brain and retina is tightly regulated by blood-brain barrier and the blood-retinal barrier, respectively, to ensure proper neuronal function. Endothelial cells within these tissues possess distinct properties that allow for controlled passage of solutes and fluids. Pericytes, glia cells and neurons signal to endothelial cells (ECs) to form and maintain the barriers and control blood flow, helping to create the neurovascular unit. This barrier is lost in a wide range of diseases affecting the central nervous system (CNS) and retina such as brain tumors, stroke, dementia, and in the eye, diabetic retinopathy, retinal vein occlusions and age-related macular degeneration to name prominent examples. Recent studies directly link barrier changes to promotion of disease pathology and degradation of neuronal function. Understanding how these barriers form and how to restore these barriers in disease provides an important point for therapeutic intervention. This review aims to describe the fundamentals of the blood-tissue barriers of the CNS and how the use of transgenic animal models led to our current understanding of the molecular framework of these barriers. The review also highlights examples of targeting barrier properties to protect neuronal function in disease states.
Collapse
Affiliation(s)
- Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA
| | - David A Antonetti
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, 1000 Wall St Rm, Ann Arbor, MI, 7317, USA.
| |
Collapse
|
4
|
Zheng K, Huang H, Yang J, Qiu M. Origin, molecular specification and stemness of astrocytes. Dev Neurobiol 2022; 82:149-159. [DOI: 10.1002/dneu.22863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 10/08/2021] [Accepted: 11/09/2021] [Indexed: 11/08/2022]
Affiliation(s)
- Kang Zheng
- Institute of Developmental and Regenerative Biology, College of Life Sciences Hangzhou Normal University Hangzhou 311121 China
| | - Hao Huang
- Institute of Developmental and Regenerative Biology, College of Life Sciences Hangzhou Normal University Hangzhou 311121 China
| | - Junlin Yang
- Institute of Developmental and Regenerative Biology, College of Life Sciences Hangzhou Normal University Hangzhou 311121 China
| | - Mengsheng Qiu
- Institute of Developmental and Regenerative Biology, College of Life Sciences Hangzhou Normal University Hangzhou 311121 China
| |
Collapse
|
5
|
Menaceur C, Gosselet F, Fenart L, Saint-Pol J. The Blood-Brain Barrier, an Evolving Concept Based on Technological Advances and Cell-Cell Communications. Cells 2021; 11:cells11010133. [PMID: 35011695 PMCID: PMC8750298 DOI: 10.3390/cells11010133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 02/07/2023] Open
Abstract
The construction of the blood–brain barrier (BBB), which is a natural barrier for maintaining brain homeostasis, is the result of a meticulous organisation in space and time of cell–cell communication processes between the endothelial cells that carry the BBB phenotype, the brain pericytes, the glial cells (mainly the astrocytes), and the neurons. The importance of these communications for the establishment, maturation and maintenance of this unique phenotype had already been suggested in the pioneering work to identify and demonstrate the BBB. As for the history of the BBB, the evolution of analytical techniques has allowed knowledge to evolve on the cell–cell communication pathways involved, as well as on the role played by the cells constituting the neurovascular unit in the maintenance of the BBB phenotype, and more particularly the brain pericytes. This review summarises the key points of the history of the BBB, from its origin to the current knowledge of its physiology, as well as the cell–cell communication pathways identified so far during its development, maintenance, and pathophysiological alteration.
Collapse
|
6
|
Yan L, Moriarty RA, Stroka KM. Recent progress and new challenges in modeling of human pluripotent stem cell-derived blood-brain barrier. Theranostics 2021; 11:10148-10170. [PMID: 34815809 PMCID: PMC8581424 DOI: 10.7150/thno.63195] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
The blood-brain barrier (BBB) is a semipermeable unit that serves to vascularize the central nervous system (CNS) while tightly regulating the movement of molecules, ions, and cells between the blood and the brain. The BBB precisely controls brain homeostasis and protects the neural tissue from toxins and pathogens. The BBB is coordinated by a tight monolayer of brain microvascular endothelial cells, which is subsequently supported by mural cells, astrocytes, and surrounding neuronal cells that regulate the barrier function with a series of specialized properties. Dysfunction of barrier properties is an important pathological feature in the progression of various neurological diseases. In vitro BBB models recapitulating the physiological and diseased states are important tools to understand the pathological mechanism and to serve as a platform to screen potential drugs. Recent advances in this field have stemmed from the use of pluripotent stem cells (PSCs). Various cell types of the BBB such as brain microvascular endothelial cells (BMECs), pericytes, and astrocytes have been derived from PSCs and synergistically incorporated to model the complex BBB structure in vitro. In this review, we summarize the most recent protocols and techniques for the differentiation of major cell types of the BBB. We also discuss the progress of BBB modeling by using PSC-derived cells and perspectives on how to reproduce more natural BBBs in vitro.
Collapse
Affiliation(s)
- Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Rebecca A. Moriarty
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Biophysics Program, University of Maryland, College Park, MD 20742, USA
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
7
|
Siqueira M, Araujo APB, Gomes FCA, Stipursky J. Ethanol Gestational Exposure Impairs Vascular Development and Endothelial Potential to Control BBB-Associated Astrocyte Function in the Developing Cerebral Cortex. Mol Neurobiol 2021; 58:1755-1768. [PMID: 33387302 DOI: 10.1007/s12035-020-02214-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 11/16/2020] [Indexed: 10/22/2022]
Abstract
Ethanol consumption during pregnancy or lactation period can induce permanent damage to the development of the central nervous system (CNS), resulting in fetal alcohol spectrum disorders (FASD). CNS development depends on proper neural cells and blood vessel (BV) development and blood-brain barrier (BBB) establishment; however, little is known about how ethanol affects these events. Here, we investigated the impact of ethanol exposure to endothelial cells (ECs) function and to ECs interaction with astrocytes in the context of BBB establishment. Cerebral cortex of newborn mice exposed in utero to ethanol (FASD model) presented a hypervascularized phenotype, revealed by augmented vessel density, length, and branch points. Further, aberrant distribution of the tight junction ZO-1 protein along BVs and increased rates of perivascular astrocytic endfeet around BVs were observed. In vitro exposure of human brain microcapillary ECs (HBMEC) to ethanol significantly disrupted ZO-1 distribution, decreased Claudin-5 and GLUT-1 expression and impaired glucose uptake, and increased nitric oxide secretion. These events were accompanied by upregulation of angiogenesis-related secreted proteins by ECs in response to ethanol exposure. Treatment of cortical astrocytes with conditioned medium (CM) from ethanol exposed ECs, upregulated astrocyte's expression of GFAP, Cx43, and Lipocalin-2 genes, as well as the pro-inflammatory genes, IL-1beta, IL-6, and TNF-alpha, which was accompanied by NF-kappa B protein nuclear accumulation. Our findings suggest that ethanol triggers a dysfunctional phenotype in brain ECs, leading to impairment of cortical vascular network formation, and promotes ECs-induced astrocyte dysfunction, which could dramatically affect BBB establishment in the developing brain.
Collapse
Affiliation(s)
- Michele Siqueira
- Laboratório de Neurobiologia Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Bloco F, Sala F15, Ilha do Fundão, Rio de Janeiro, RJ, 21949-902, Brazil
| | - Ana Paula Bérgamo Araujo
- Laboratório de Neurobiologia Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Bloco F, Sala F15, Ilha do Fundão, Rio de Janeiro, RJ, 21949-902, Brazil
| | - Flávia Carvalho Alcantara Gomes
- Laboratório de Neurobiologia Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Bloco F, Sala F15, Ilha do Fundão, Rio de Janeiro, RJ, 21949-902, Brazil
| | - Joice Stipursky
- Laboratório de Neurobiologia Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal do Rio de Janeiro, Bloco F, Sala F15, Ilha do Fundão, Rio de Janeiro, RJ, 21949-902, Brazil.
| |
Collapse
|
8
|
Abstract
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.
Collapse
Affiliation(s)
- Urs H Langen
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Swathi Ayloo
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Chenghua Gu
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA;
| |
Collapse
|
9
|
Quiñonez-Silvero C, Hübner K, Herzog W. Development of the brain vasculature and the blood-brain barrier in zebrafish. Dev Biol 2019; 457:181-190. [PMID: 30862465 DOI: 10.1016/j.ydbio.2019.03.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022]
Abstract
To ensure tissue homeostasis the brain needs to be protected from blood-derived fluctuations or pathogens that could affect its function. Therefore, the brain capillaries develop tissue-specific properties to form a selective blood-brain barrier (BBB), allowing the passage of essential molecules to the brain and blocking the penetration of potentially harmful compounds or cells. Previous studies reported the presence of this barrier in zebrafish. The intrinsic features of the zebrafish embryos and larvae in combination with optical techniques, make them suitable for the study of barrier establishment and maturation. In this review, we discuss the most recent contributions to the development and formation of a functional zebrafish BBB. Moreover, we compare the molecular and cellular characteristic of the zebrafish and the mammalian BBB.
Collapse
Affiliation(s)
- Claudia Quiñonez-Silvero
- University of Muenster, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Germany
| | - Kathleen Hübner
- University of Muenster, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Germany
| | - Wiebke Herzog
- University of Muenster, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Germany; Max Planck Institute for Molecular Biomedicine, Muenster, Germany.
| |
Collapse
|
10
|
β-Catenin Is Required for Endothelial Cyp1b1 Regulation Influencing Metabolic Barrier Function. J Neurosci 2017; 36:8921-35. [PMID: 27559173 DOI: 10.1523/jneurosci.0148-16.2016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 06/27/2016] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED The canonical Wnt/β-catenin signaling pathway is crucial for blood-brain barrier (BBB) formation in brain endothelial cells. Although glucose transporter 1, claudin-3, and plasmalemma vesicular-associated protein have been identified as Wnt/β-catenin targets in brain endothelial cells, further downstream targets relevant to BBB formation and function are incompletely explored. By Affymetrix expression analysis, we show that the cytochrome P450 enzyme Cyp1b1 was significantly decreased in β-catenin-deficient mouse endothelial cells, whereas its close homolog Cyp1a1 was upregulated in an aryl hydrocarbon receptor-dependent manner, hence indicating that β-catenin is indispensable for Cyp1b1 but not for Cyp1a1 expression. Functionally, Cyp1b1 could generate retinoic acid from retinol leading to cell-autonomous induction of the barrier-related ATP-binding cassette transporter P-glycoprotein. Cyp1b1 could also generate 20-hydroxyeicosatetraenoic acid from arachidonic acid, decreasing endothelial barrier function in vitro In mice in vivo pharmacological inhibition of Cyp1b1 increased BBB permeability for small molecular tracers, and Cyp1b1 was downregulated in glioma vessels in which BBB function is lost. Hence, we propose Cyp1b1 as a target of β-catenin indirectly influencing BBB properties via its metabolic activity, and as a potential target for modulating barrier function in endothelial cells. SIGNIFICANCE STATEMENT Wnt/β-catenin signaling is crucial for blood-brain barrier (BBB) development and maintenance; however, its role in regulating metabolic characteristics of endothelial cells is unclear. We provide evidence that β-catenin influences endothelial metabolism by transcriptionally regulating the cytochrome P450 enzyme Cyp1b1 Furthermore, expression of its close homolog Cyp1a1 was inhibited by β-catenin. Functionally, Cyp1b1 generated retinoic acid as well as 20-hydroxyeicosatetraenoic acid that regulated P-glycoprotein and junction proteins, respectively, thereby modulating BBB properties. Inhibition of Cyp1b1 in vivo increased BBB permeability being in line with its downregulation in glioma endothelia, potentially implicating Cyp1b1 in other brain pathologies. In conclusion, Wnt/β-catenin signaling regulates endothelial metabolic barrier function through Cyp1b1 transcription.
Collapse
|
11
|
Umans RA, Henson HE, Mu F, Parupalli C, Ju B, Peters JL, Lanham KA, Plavicki JS, Taylor MR. CNS angiogenesis and barriergenesis occur simultaneously. Dev Biol 2017; 425:101-108. [PMID: 28365243 DOI: 10.1016/j.ydbio.2017.03.017] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 02/24/2017] [Accepted: 03/18/2017] [Indexed: 01/04/2023]
Abstract
The blood-brain barrier (BBB) plays a vital role in the central nervous system (CNS). A comprehensive understanding of BBB development has been hampered by difficulties in observing the differentiation of brain endothelial cells (BECs) in real-time. Here, we generated two transgenic zebrafish line, Tg(glut1b:mCherry) and Tg(plvap:EGFP), to serve as in vivo reporters of BBB development. We showed that barriergenesis (i.e. the induction of BEC differentiation) occurs immediately as endothelial tips cells migrate into the brain parenchyma. Using the Tg(glut1b:mCherry) transgenic line, we performed a genetic screen and identified a zebrafish mutant with a nonsense mutation in gpr124, a gene known to play a role in CNS angiogenesis and BBB development. We also showed that our transgenic plvap:EGFP line, a reporter of immature brain endothelium, is initially expressed in newly formed brain endothelial cells, but subsides during BBB maturation. Our results demonstrate the ability to visualize the in vivo differentiation of brain endothelial cells into the BBB phenotype and establish that CNS angiogenesis and barriergenesis occur simultaneously.
Collapse
Affiliation(s)
- Robyn A Umans
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA; Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hannah E Henson
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA; Integrated Program in Biomedical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Fangzhou Mu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Chaithanyarani Parupalli
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bensheng Ju
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jennifer L Peters
- Cell and Tissue Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kevin A Lanham
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Jessica S Plavicki
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Michael R Taylor
- Department of Chemical Biology & Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA; Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA.
| |
Collapse
|
12
|
Lange C, Turrero Garcia M, Decimo I, Bifari F, Eelen G, Quaegebeur A, Boon R, Zhao H, Boeckx B, Chang J, Wu C, Le Noble F, Lambrechts D, Dewerchin M, Kuo CJ, Huttner WB, Carmeliet P. Relief of hypoxia by angiogenesis promotes neural stem cell differentiation by targeting glycolysis. EMBO J 2016; 35:924-41. [PMID: 26856890 DOI: 10.15252/embj.201592372] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/05/2016] [Indexed: 12/23/2022] Open
Abstract
Blood vessels are part of the stem cell niche in the developing cerebral cortex, but their in vivo role in controlling the expansion and differentiation of neural stem cells (NSCs) in development has not been studied. Here, we report that relief of hypoxia in the developing cerebral cortex by ingrowth of blood vessels temporo-spatially coincided with NSC differentiation. Selective perturbation of brain angiogenesis in vessel-specific Gpr124 null embryos, which prevented the relief from hypoxia, increased NSC expansion at the expense of differentiation. Conversely, exposure to increased oxygen levels rescued NSC differentiation in Gpr124 null embryos and increased it further in WT embryos, suggesting that niche blood vessels regulate NSC differentiation at least in part by providing oxygen. Consistent herewith, hypoxia-inducible factor (HIF)-1α levels controlled the switch of NSC expansion to differentiation. Finally, we provide evidence that high glycolytic activity of NSCs is required to prevent their precocious differentiation in vivo Thus, blood vessel function is required for efficient NSC differentiation in the developing cerebral cortex by providing oxygen and possibly regulating NSC metabolism.
Collapse
Affiliation(s)
- Christian Lange
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | | | - Ilaria Decimo
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Francesco Bifari
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Annelies Quaegebeur
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Ruben Boon
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Hui Zhao
- Laboratory of Translational Genetics, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Translational Genetics, Department of Oncology, KU Leuven Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Translational Genetics, Department of Oncology, KU Leuven Leuven, Belgium
| | - Junlei Chang
- Department of Medicine, Hematology Division Stanford University, Stanford, CA, USA
| | - Christine Wu
- Department of Medicine, Hematology Division Stanford University, Stanford, CA, USA
| | - Ferdinand Le Noble
- Angiogenesis and Cardiovascular Pathology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany Department of Cell and Developmental Biology, KIT, Karlsruhe, Germany
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Translational Genetics, Department of Oncology, KU Leuven Leuven, Belgium
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| | - Calvin J Kuo
- Department of Medicine, Hematology Division Stanford University, Stanford, CA, USA
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Department of Oncology, KU Leuven Leuven, Belgium
| |
Collapse
|
13
|
Saunders NR, Dreifuss JJ, Dziegielewska KM, Johansson PA, Habgood MD, Møllgård K, Bauer HC. The rights and wrongs of blood-brain barrier permeability studies: a walk through 100 years of history. Front Neurosci 2014; 8:404. [PMID: 25565938 PMCID: PMC4267212 DOI: 10.3389/fnins.2014.00404] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/20/2014] [Indexed: 12/16/2022] Open
Abstract
Careful examination of relevant literature shows that many of the most cherished concepts of the blood-brain barrier are incorrect. These include an almost mythological belief in its immaturity that is unfortunately often equated with absence or at least leakiness in the embryo and fetus. The original concept of a blood-brain barrier is often attributed to Ehrlich; however, he did not accept that permeability of cerebral vessels was different from other organs. Goldmann is often credited with the first experiments showing dye (trypan blue) exclusion from the brain when injected systemically, but not when injected directly into it. Rarely cited are earlier experiments of Bouffard and of Franke who showed methylene blue and trypan red stained all tissues except the brain. The term “blood-brain barrier” “Blut-Hirnschranke” is often attributed to Lewandowsky, but it does not appear in his papers. The first person to use this term seems to be Stern in the early 1920s. Studies in embryos by Stern and colleagues, Weed and Wislocki showed results similar to those in adult animals. These were well-conducted experiments made a century ago, thus the persistence of a belief in barrier immaturity is puzzling. As discussed in this review, evidence for this belief, is of poor experimental quality, often misinterpreted and often not properly cited. The functional state of blood-brain barrier mechanisms in the fetus is an important biological phenomenon with implications for normal brain development. It is also important for clinicians to have proper evidence on which to advise pregnant women who may need to take medications for serious medical conditions. Beliefs in immaturity of the blood-brain barrier have held the field back for decades. Their history illustrates the importance of taking account of all the evidence and assessing its quality, rather than selecting papers that supports a preconceived notion or intuitive belief. This review attempts to right the wrongs. Based on careful translation of original papers, some published a century ago, as well as providing discussion of studies claiming to show barrier immaturity, we hope that readers will have evidence on which to base their own conclusions.
Collapse
Affiliation(s)
- Norman R Saunders
- Department of Pharmacology and Therapeutics, University of Melbourne Parkville, VIC, Australia
| | | | | | - Pia A Johansson
- Institute for Stem Cell Research, Helmholtz Center Munich Munich, Germany
| | - Mark D Habgood
- Department of Pharmacology and Therapeutics, University of Melbourne Parkville, VIC, Australia
| | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, University of Copenhagen Copenhagen, Denmark
| | - Hans-Christian Bauer
- Institute of Tendon and Bone Regeneration, Paracelsus Medical University Salzburg, Austria ; Spinal Cord Injury and Tissue Regeneration Center, Paracelsus Medical University Salzburg, Austria
| |
Collapse
|
14
|
Saunders NR, Liddelow SA, Dziegielewska KM. Barrier mechanisms in the developing brain. Front Pharmacol 2012; 3:46. [PMID: 22479246 PMCID: PMC3314990 DOI: 10.3389/fphar.2012.00046] [Citation(s) in RCA: 344] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 02/29/2012] [Indexed: 01/24/2023] Open
Abstract
The adult brain functions within a well-controlled stable environment, the properties of which are determined by cellular exchange mechanisms superimposed on the diffusion restraint provided by tight junctions at interfaces between blood, brain and cerebrospinal fluid (CSF). These interfaces are referred to as “the” blood–brain barrier. It is widely believed that in embryos and newborns, this barrier is immature or “leaky,” rendering the developing brain more vulnerable to drugs or toxins entering the fetal circulation from the mother. New evidence shows that many adult mechanisms, including functionally effective tight junctions are present in embryonic brain and some transporters are more active during development than in the adult. Additionally, some mechanisms present in embryos are not present in adults, e.g., specific transport of plasma proteins across the blood–CSF barrier and embryo-specific intercellular junctions between neuroependymal cells lining the ventricles. However developing cerebral vessels appear to be more fragile than in the adult. Together these properties may render developing brains more vulnerable to drugs, toxins, and pathological conditions, contributing to cerebral damage and later neurological disorders. In addition, after birth loss of protection by efflux transporters in placenta may also render the neonatal brain more vulnerable than in the fetus.
Collapse
Affiliation(s)
- Norman R Saunders
- Department of Pharmacology, The University of Melbourne Parkville, VIC, Australia
| | | | | |
Collapse
|
15
|
Ek CJ, Dziegielewska KM, Habgood MD, Saunders NR. Barriers in the developing brain and Neurotoxicology. Neurotoxicology 2011; 33:586-604. [PMID: 22198708 DOI: 10.1016/j.neuro.2011.12.009] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 12/11/2011] [Accepted: 12/11/2011] [Indexed: 01/16/2023]
Abstract
The brain develops and grows within a well-controlled internal environment that is provided by cellular exchange mechanisms in the interfaces between blood, cerebrospinal fluid and brain. These are generally referred to by the term "brain barriers": blood-brain barrier across the cerebral endothelial cells and blood-CSF barrier across the choroid plexus epithelial cells. An essential component of barrier mechanisms is the presence of tight junctions between the endothelial and epithelial cells of these interfaces. This review outlines historical evidence for the presence of effective barrier mechanisms in the embryo and newborn and provides an up to date description of recent morphological, biochemical and molecular data for the functional effectiveness of these barriers. Intercellular tight junctions between cerebral endothelial cells and between choroid plexus epithelial cells are functionally effective as soon as they differentiate. Many of the influx and efflux mechanisms are not only present from early in development, but the genes for some are expressed at much higher levels in the embryo than in the adult and there is physiological evidence that these transport systems are functionally more active in the developing brain. This substantial body of evidence supporting the concept of well developed barrier mechanisms in the developing brain is contrasted with the widespread belief amongst neurotoxicologists that "the" blood-brain barrier is immature or even absent in the embryo and newborn. A proper understanding of the functional capacity of the barrier mechanisms to restrict the entry of harmful substances or administered therapeutics into the developing brain is critical. This knowledge would assist the clinical management of pregnant mothers and newborn infants and development of protocols for evaluation of risks of drugs used in pregnancy and the neonatal period prior to their introduction into clinical practice.
Collapse
Affiliation(s)
- C Joakim Ek
- Department of Pharmacology, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | |
Collapse
|
16
|
Saunders N, Habgood M. Understanding barrier mechanisms in the developing brain to aid therapy for the dysfunctional brain. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.10.84] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The brain, both in the adult and during development, is protected by morphological barriers and functional mechanisms that provide a stable internal environment. Understanding these processes in the developing brain may lead to novel therapies for brain disorders, as some transport mechanisms, particularly those in the choroid plexus, may prove more amenable to devising novel delivery systems. Based on results from studies of the transfer of specific proteins across the blood–cerebrospinal fluid interface in the developing brain, the steps required to develop such a delivery system are outlined. Knowledge of barrier mechanisms in the developing brain may be relevant to treating neuropsychiatric conditions in children and adults for whom barrier dysfunction in the fetus, precipitated by adverse factors such as maternal infection, may contribute to the neuropathology underlying disorders such as autism and schizophrenia.
Collapse
Affiliation(s)
- Norman Saunders
- University of Melbourne, Department of Pharmacology, Parkville, Victoria 3010, Australia
| | - Mark Habgood
- University of Melbourne, Department of Pharmacology, Parkville, Victoria 3010, Australia
| |
Collapse
|
17
|
Preservation of transendothelial glucose transporter 1 and P-glycoprotein transporters in a cortical slice culture model of the blood–brain barrier. Neuroscience 2010; 170:361-71. [DOI: 10.1016/j.neuroscience.2010.06.073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 06/13/2010] [Accepted: 06/25/2010] [Indexed: 11/24/2022]
|
18
|
Affiliation(s)
- Eckhard Lammert
- Institute of Animal Physiology, Heinrich-Heine-University, D-40225 Düsseldorf, Germany
| |
Collapse
|
19
|
Lazzari M, Bettini S, Ciani F, Franceschini V. Glucose transporter distribution in the vessels of the central nervous system of the axolotl Ambystoma mexicanum (Urodela: Ambystomatidae). Anat Rec (Hoboken) 2008; 291:1293-300. [PMID: 18727107 DOI: 10.1002/ar.20741] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The GLUT-1 isoform of the glucose transporter is commonly considered a reliable molecular marker of blood-brain barrier endothelia in the neural vasculature organized in a three-dimensional network of single vessels. The central nervous system of the axolotl Ambystoma mexicanum is characterized by a vascular architecture that contains both single and paired vessels. The presence and distribution of the GLUT-1 transporter are studied in this urodele using both immunoperoxidase histochemistry and immunogold technique. Light microscopy reveals immunopositivity in both parenchymal and meningeal vessels. The transverse-sectioned pairs of vessels do not show the same size. Furthermore, in the same pair, the two elements often differ in diameter. The main regions of the central nervous system show a different percentage of the paired structures. Only immunogold cytochemistry reveals different staining intensity in the two adjoined elements of a vascular pair. Colloidal gold particles show an asymmetric distribution in the endothelia of both single and paired vessels. These particles are more numerous on the abluminal surface than on the luminal one. The particle density is calculated in both vascular types. The different values could indicate functional differences between single and paired vessels and between the two adjoined elements of a pair, regarding glucose transport.
Collapse
|
20
|
Kraus J, Oschmann P. The impact of interferon-beta treatment on the blood-brain barrier. Drug Discov Today 2006; 11:755-62. [PMID: 16846804 DOI: 10.1016/j.drudis.2006.06.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 05/24/2006] [Accepted: 06/09/2006] [Indexed: 01/09/2023]
Abstract
Changes in the blood-brain barrier (BBB) are crucial to the pathogenesis of multiple sclerosis (MS). There are currently few established treatments for MS, and interferon-beta (IFN-beta) therapy is one of the most promising - proposed to act as an immunomodulator of the cytokine network reducing inflammatory damage. However, there is increasing evidence that direct effects on the BBB could also be relevant. This review surveys the evidence that IFN-beta stabilizes the BBB, and that this process itself might be the key target. Understanding IFN-beta-derived changes at the BBB will not only provide new insights in the pathogenesis of MS but will also be helpful to develop new, more-specific drugs for MS treatment.
Collapse
Affiliation(s)
- Jörg Kraus
- Paracelsus Private Medical University and Salzburger Landesklinken, Christian-Doppler-Klinik, Department of Neurology, Ignaz-Harrer-Strasse 79, 5020 Salzburg, Austria.
| | | |
Collapse
|
21
|
Lazzari M, Franceschini V. Glucose transporter (GLUT-1) distribution in the brain vessels of the adult Italian wall lizard, Podarcis sicula. Acta Histochem 2006; 108:385-93. [PMID: 16828846 DOI: 10.1016/j.acthis.2006.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2006] [Revised: 04/13/2006] [Accepted: 05/17/2006] [Indexed: 10/24/2022]
Abstract
The GLUT-1 isoform of the glucose transporter is commonly accepted as a reliable molecular marker of blood-brain barrier endothelia in neural vasculature organized in a three-dimensional network of single vessels. The brain of the lizard Podarcis sicula is characterized by a vascular architecture based on a pattern of paired vessels. The presence and distribution of GLUT-1 were studied in adult lizards using both light and transmission electron microscopic techniques. Immunoperoxidase histochemistry was applied to sections from paraffin-embedded brain using gold-conjugated secondary antibodies to localize this antigen on ultrathin sections. The transverse sectioned pairs of vessels did not show the same size and, in particular, the two elements of the same pair often differed in their diameters. Light microscopy revealed immunopositivity in both parenchymal and meningeal vessels. In each transverse-sectioned vascular pair, one element was intensely labelled, and the adjacent one showed only slight or negligible reaction. Colloidal gold particles were restricted to endothelial cells, showing an asymmetric labelling pattern, which was always characterized by markedly higher density of immunolabelling of the abluminal rather than the luminal plasmalemma. Moreover, in every vascular pair, one profile had lower amounts of scantier labelling by gold particles than the adjacent element. This pattern indicates functional differences between the adjacent vascular limbs regarding glucose transport.
Collapse
Affiliation(s)
- Maurizio Lazzari
- Department of Biology, University of Bologna, via F. Selmi 3, 40126 Bologna, Italy.
| | | |
Collapse
|
22
|
Cornford EM, Hyman S. Localization of brain endothelial luminal and abluminal transporters with immunogold electron microscopy. NeuroRx 2005; 2:27-43. [PMID: 15717055 PMCID: PMC539318 DOI: 10.1602/neurorx.2.1.27] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Immunogold electron microscopy has identified a variety of blood-brain barrier (BBB) proteins with transporter and regulatory functions. For example, isoforms of the glucose transporter, protein kinase C (PKC), and caveolin-1 are BBB specific. Isoform 1 of the facilitative glucose transporter family (GLUT1) is expressed solely in endothelial (and pericyte) domains, and approximately 75% of the protein is membrane-localized in humans. Evidence is presented for a water cotransport function of BBB GLUT1. A shift in transporter polarity characterized by increased luminal membrane GLUT1 is seen when BBB glucose transport is upregulated; but a greater abluminal membrane density is seen in the human BBB when GLUT1 is downregulated. PKC colocalizes with GLUT1 within these endothelial domains during up- and downregulation, suggesting that a PKC-mediated mechanism regulates human BBB glucose transporter expression. Occludin and claudin-5 (like other tight-junctional proteins) exhibit a restricted distribution, and are expressed solely within interendothelial clefts of the BBB. GFAP (glial fibrillary acidic protein) is uniformly expressed throughout the foot-processes and the entire astrocyte. But the microvascular-facing membranes of the glial processes that contact the basal laminae are also polarized, and their transporters may also redistribute within the astrocyte. Monocarboxylic acid transporter and water channel (Aquaporin-4) expression are enriched at the glial foot-process, and both undergo physiological modulation. We suggest that as transcytosis and efflux mechanisms generate interest as potential neurotherapeutic targets, electron microscopic confirmation of their site-specific expression patterns will continue to support the CNS drug discovery process.
Collapse
Affiliation(s)
- Eain M Cornford
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California 90073, USA.
| | | |
Collapse
|
23
|
Cornford EM, Hyman S. Localization of brain endothelial luminal and abluminal transporters with immunogold electron microscopy. Neurotherapeutics 2005. [DOI: 10.1007/bf03206640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
24
|
Rajakumar A, Thamotharan S, Raychaudhuri N, Menon RK, Devaskar SU. Trans-activators regulating neuronal glucose transporter isoform-3 gene expression in mammalian neurons. J Biol Chem 2004; 279:26768-79. [PMID: 15054091 DOI: 10.1074/jbc.m402735200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The murine facilitative glucose transporter isoform 3 is developmentally regulated and is predominantly expressed in neurons. By employing the primer extension assay, the transcription start site of the murine Glut 3 gene in the brain was localized to -305 bp 5' to the ATG translation start codon. Transient transfection assays in N2A neuroblasts using murine GLUT3-luciferase reporter constructs mapped enhancer activities to two regions located at -203 to -177 and -104 to -29 bp flanking a previously described repressor element (-137 to -130 bp). Dephosphorylated Sp1 and Sp3 proteins from the 1- and 21-day-old mouse brain nuclear extracts bound the repressor elements, whereas both dephosphorylated and phosphorylated cAMP-response element-binding protein (CREB) in N2A, 1- and 21-day-old mouse brain nuclear extracts bound the 5'-enhancer cis-elements (-187 to -180 bp) of the Glut 3 gene, and the Y box protein MSY-1 bound the sense strand of the -83- to -69-bp region. Sp3, CREB, and MSY-1 binding to the GLUT 3 DNA was confirmed by the chromatin immunoprecipitation assay, whereas CREB and MSY-1 interaction was detected by the co-immunoprecipitation assay. Furthermore, small interference RNA targeted at CREB in N2A cells decreased endogenous CREB concentrations, and CREB mediated GLUT 3 transcription. Thus, in the murine brain similar to the N2A cells, phosphorylated CREB and MSY-1 bound the Glut 3 gene trans-activating the expression in neurons, whereas Sp1/Sp3 bound the repressor elements. We speculate that phosphorylated CREB and Sp3 also interacted to bring about GLUT 3 expression in response to development/cell differentiation and neurotransmission.
Collapse
Affiliation(s)
- Augustine Rajakumar
- Division of Neonatology and Developmental Biology, Department of Pediatrics, David Geffen School of Medicine, UCLA, Los Angeles, California 90095-1752, USA
| | | | | | | | | |
Collapse
|
25
|
Sinha C, Agrawal AK, Islam F, Seth K, Chaturvedi RK, Shukla S, Seth PK. Mosquito repellent (pyrethroid‐based) induced dysfunction of blood–brain barrier permeability in developing brain. Int J Dev Neurosci 2003; 22:31-7. [PMID: 15013076 DOI: 10.1016/j.ijdevneu.2003.10.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2003] [Revised: 09/22/2003] [Accepted: 10/15/2003] [Indexed: 11/17/2022] Open
Abstract
Pyrethroid-based mosquito repellents (MR) are commonly used to protect humans against mosquito vector. New born babies and children are often exposed to pyrethroids for long periods by the use of liquid vaporizers. Occupational and experimental studies indicate that pyrethroids can cause clinical, biochemical and neurological changes, and that exposure to pyrethroids during organogenesis and early developmental period is especially harmful. The neurotoxicity caused by MR has aroused concern among public regarding their use. In the present study, the effect of exposure of rat pups during early developmental stages to a pyrethroid-based MR (allethrin, 3.6% w/v, 8h per day through inhalation) on blood-brain barrier (BBB) permeability was investigated. Sodium fluororescein (SF) and Evan's blue (EB) were used as micromolecular and macromolecular tracers, respectively. Exposure during prenatal (gestation days 1-20), postnatal (PND1-30) and perinatal (gestation days 1-20 + PND1-30) periods showed significant increase in the brain uptake index (BUI) of SF by 54% (P < 0.01), 70% (P < 0.01), 79% (P < 0.01), respectively. This increase persisted (68%, P < 0.01) even 1 week after withdrawal of exposure (as assessed on PND37). EB did not exhibit significant change in BBB permeability in any of the group. The results suggest that MR inhalation during early prenatal/postnatal/perinatal life may have adverse effects on infants leading to central nervous system (CNS) abnormalities, if a mechanism operates in humans similar to that in rat pups.
Collapse
Affiliation(s)
- C Sinha
- Developmental Toxicology Division, Industrial Toxicology Research Centre, P.O. Box 80, Mahatma Gandhi Marg, Lucknow 226001, India
| | | | | | | | | | | | | |
Collapse
|
26
|
Mani N, Khaibullina A, Krum JM, Rosenstein JM. Activation of receptor-mediated angiogenesis and signaling pathways after VEGF administration in fetal rat CNS explants. J Cereb Blood Flow Metab 2003; 23:1420-9. [PMID: 14663337 DOI: 10.1097/01.wcb.0000090620.86921.9c] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The angiogenic role of vascular endothelial growth factor (VEGF) receptors, flk-1 and flt-1, and their downstream signaling pathways, MAPK/ERK and PI-3 kinase, were examined in a fetal rat cortical explant model after exposure to exogenous VEGF. Treatment with VEGF resulted in substantial neovascularization characterized by increased vascular flk-1 receptor expression, whereas flt-1 receptor protein expression was absent. The specific role of flk-1 receptors in the angiogenic process was confirmed by the addition of antisense oligonucleotides (AS-ODNs) to flk-1, which blocked angiogenesis, whereas AS-ODNs to flt-1 had no effect. These results were further supported by the finding that specific chemical inhibition of flk-1 receptors caused disruption of the angiogenic response, whereas inhibition of the flt-1 receptors had no effect. Application of either MAPK/ERK or PI-3 kinase pathway inhibitors disrupted VEGF-induced angiogenesis, thereby indicating that both signaling pathways mediate this process. Thus VEGF binding to the endothelial flk-1 receptor activates the MAPK/ERK and PI-3 kinase pathways, resulting in neoangiogenic events. Of interest is the fact that although VEGF is regarded as a vascular permeability factor, its application to nascent cortical tissue caused an increase in a key physiologic protein of the blood-brain barrier function, glucose transporter-1, suggesting that the cytokine may have a role in blood-brain barrier development.
Collapse
Affiliation(s)
- Nina Mani
- Department of Anatomy and Cell Biology, The George University Medical Center, Washington, DC 20037, USA
| | | | | | | |
Collapse
|
27
|
McAllister MS, Krizanac-Bengez L, Macchia F, Naftalin RJ, Pedley KC, Mayberg MR, Marroni M, Leaman S, Stanness KA, Janigro D. Mechanisms of glucose transport at the blood-brain barrier: an in vitro study. Brain Res 2001; 904:20-30. [PMID: 11516408 DOI: 10.1016/s0006-8993(01)02418-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
How the brain meets its continuous high metabolic demand in light of varying plasma glucose levels and a functional blood-brain barrier (BBB) is poorly understood. GLUT-1, found in high density at the BBB appears to maintain the continuous shuttling of glucose across the blood-brain barrier irrespective of the plasma concentration. We examined the process of glucose transport across a quasi-physiological in vitro blood-brain barrier model. Radiolabeled tracer permeability studies revealed a concentration ratio of abluminal to luminal glucose in this blood-brain barrier model of approximately 0.85. Under conditions where [glucose](lumen) was higher than [glucose](ablumen), influx of radiolabeled 2-deoxyglucose from lumen to the abluminal compartment was approximately 35% higher than efflux from the abluminal side to the lumen. However, when compartmental [glucose] were maintained equal, a reversal of this trend was seen (approximately 19% higher efflux towards the lumen), favoring establishment of a luminal to abluminal concentration gradient. Immunocytochemical experiments revealed that in addition to segregation of GLUT-1 (luminal>abluminal), the intracellular enzyme hexokinase was also asymmetrically distributed (abluminal>luminal). We conclude that glucose transport at the CNS/blood interface appears to be dependent on and regulated by a serial chain of membrane-bound and intracellular transporters and enzymes.
Collapse
Affiliation(s)
- M S McAllister
- Department of Neurological Surgery, Cerebrovascular Research Center, Cleveland Clinic Foundation/NB20, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ghazanfari FA, Stewart RR. Characteristics of endothelial cells derived from the blood-brain barrier and of astrocytes in culture. Brain Res 2001; 890:49-65. [PMID: 11164768 DOI: 10.1016/s0006-8993(00)03053-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, cultures of astrocytes and capillary endothelial cells from the blood-brain barrier (BBB) of the postnatal (P1) mouse cerebral cortex were analyzed with the aim of acquiring information on the distinguishing characteristics of each cell type. For isolation and purification of astrocyte cells, the methods of McCarthy and DeVellis [J. Cell Biol. 85 (1980) 890] were employed. The methods of Chen et al. [Lab. Invest. 78 (1998) 353], Duport et al. [Proc. Natl. Acad. Sci. USA 95 (1998) 1840], Rubin et al. [J Cell Biol. 115 (1991) 1725] and Tontsch and Bauer [Microvasc. Res. 37 (1989) 148] were utilized for culturing of cells from the BBB. A simple protocol was also created for isolating and purifying brain endothelial cells with 10 mM sodium cyanide. The vascular system of the cerebral cortex is derived from the leptomeningeal blood vessels [Qin and Sato, Dev. Dyn. 202 (1995) 172; Risau et al., EMBO J. 5 (1986) 3179]. With this in mind, cultures of the P1 mouse meninges were used as a comparative cell type in order to differentiate between BBB cells and astrocytes. In this regard, the expression of a number of markers were correlated, and an antibody double labeling technique was employed. The staining of these markers was then compared to cells cultured from leptomeninges and to two other types of endothelial cells, human umbilical vein and bovine aortic. Reverse transcription-polymerase chain reaction (RT-PCR) was performed on total RNA isolated from adult mouse brain, cells cultured from P1 mouse cortex or meninges, bovine aortic endothelial cells and human umbilical vein endothelial cells (HUV-EC) to detect the expression of glial fibrillary acidic protein (GFAP), Von Willebrand factor (factor VIII-related antigen) and fibronectin. These analyses revealed the presence of GFAP mRNA in the cultures of cortical and leptomeningeal cells and of protein in all cell types; Von Willebrand factor mRNA was detectable in HUV-EC cells but undetectable in cortical, leptomeningeal and bovine aortic endothelial cells. Fibronectin mRNA and protein were present in all of the cell types. Given the results of our investigations we conclude that in culture, astrocytes are actually brain endothelial cells.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology
- Actins/analysis
- Actins/immunology
- Animals
- Antibodies
- Aorta/cytology
- Astrocytes/chemistry
- Astrocytes/cytology
- Astrocytes/metabolism
- Blood-Brain Barrier/physiology
- Cattle
- Cell Survival/drug effects
- Cerebral Cortex/blood supply
- Cerebral Cortex/cytology
- Coculture Techniques/methods
- DNA-Binding Proteins/analysis
- DNA-Binding Proteins/immunology
- Endothelium, Vascular/chemistry
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Enzyme Inhibitors/pharmacology
- Fibronectins/analysis
- Fibronectins/immunology
- Galactosylceramides/analysis
- Galactosylceramides/immunology
- Gene Expression/physiology
- Glial Fibrillary Acidic Protein/analysis
- Glial Fibrillary Acidic Protein/genetics
- Glial Fibrillary Acidic Protein/immunology
- Glucose Transporter Type 1
- Lectins
- Lipoproteins, LDL/pharmacokinetics
- Meninges/cytology
- Mice
- Mice, Inbred C57BL
- Monosaccharide Transport Proteins/analysis
- Monosaccharide Transport Proteins/immunology
- PAX2 Transcription Factor
- RNA, Messenger/analysis
- Sodium Cyanide/pharmacology
- Transcription Factors/analysis
- Transcription Factors/immunology
- von Willebrand Factor/analysis
- von Willebrand Factor/genetics
- von Willebrand Factor/immunology
Collapse
Affiliation(s)
- F A Ghazanfari
- R.O.W. Sciences, Inc., 1700 Research Boulevard, Rockville, MD 20850, USA
| | | |
Collapse
|
29
|
Virgintino D, Robertson D, Benagiano V, Errede M, Bertossi M, Ambrosi G, Roncali L. Immunogold cytochemistry of the blood-brain barrier glucose transporter GLUT1 and endogenous albumin in the developing human brain. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 2000; 123:95-101. [PMID: 11020555 DOI: 10.1016/s0165-3806(00)00086-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The blood-brain barrier (BBB) glucose transporter, GLUT1, was detected by immunogold electron microscopy on the microvascular compartment of the human foetus telencephalon at the 12th and 18th weeks of gestation. By computerized morphometry, the cellular and subcellular localization of the immunosignal for GLUT1 was quantitatively evaluated. The study showed that the glucose transporter is strongly expressed by endothelial cells while a very low signal is detected on vascular pericytes. The GLUT1 antigenic sites are preferentially associated to the ablumenal and junctional plasma membranes of the endothelial cells and tend to increase significantly with age. A parallel study carried out by the endogenous serum protein albumin demonstrated that already at the 12th week the endothelial routes are hindered to the protein as happens at the blood-endothelium interface of mature brain. The results demonstrate that in the human foetus the brain microvessels express BBB-specific functional activities early.
Collapse
Affiliation(s)
- D Virgintino
- Department of Human Anatomy and Histology, University of Bari School of Medicine, Piazza Giulio Cesare, I-70124, Bari, Italy.
| | | | | | | | | | | | | |
Collapse
|
30
|
Chang S, Lee S, Lee C, Kim JI, Kim Y. Expression of the human erythrocyte glucose transporter in transitional cell carcinoma of the bladder. Urology 2000; 55:448-52. [PMID: 10699635 DOI: 10.1016/s0090-4295(99)00474-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVES It has previously been shown that glucose uptake and use is more prevalent in carcinomas than in normal cells and tissues. We hypothesized that human erythrocyte glucose transporter (Glut-1) expression is increased in bladder transitional cell carcinoma (TCC) and that the grade of expression might correlate with the degree of malignancy. METHODS Immunostaining of Glut-1 protein was studied in normal bladder (5 cases), benign papilloma (10 cases), superficial tumor (48 cases), and invasive tumor (31 cases) tissue. The immunoreactivity grading system used was as follows: absence of immunoreactivity in tumor cell = 0; less than 10% of the tumor cells immunoreactive = 1+; 10% to 50% of the tumor cells immunoreactive = 2+; and greater than 50% of the tumor cells immunoreactive = 3+. RESULTS Immunostaining of Glut-1 protein was not expressed in the normal bladder or benign papilloma samples, but it was expressed in 63.0% (46 of 73) of the TCC samples. In the pattern of expression of Glut-1 protein, superficial TCC was stained focally, but invasive TCC was stained diffusely in the tumor nests. The grade of Glut-1 protein expression increased more significantly in the invasive TCC than in the superficial TCC samples (P = 0.002) and more significantly in the high nuclear grade than in the low nuclear grade samples (P = 0.007). In the superficial TCC samples, the bladder tumor recurrence rate did not significantly correlate with Glut-1 protein expression (P = 0.40). CONCLUSIONS Our results suggest that the Glut-1 protein is not expressed in normal bladder mucosa and benign lesions, that Glut-1 protein expression is strongly associated with neoplastic progression in bladder TCC, and that Glut-1 expression does not correlate with the recurrence rate in superficial bladder TCC.
Collapse
Affiliation(s)
- S Chang
- Department of Urology Kyung Hee University School of Medicine, Seoul, South Korea
| | | | | | | | | |
Collapse
|
31
|
Loda M, Xu X, Pession A, Vortmeyer A, Giangaspero F. Membranous expression of glucose transporter-1 protein (GLUT-1) in embryonal neoplasms of the central nervous system. Neuropathol Appl Neurobiol 2000; 26:91-7. [PMID: 10736070 DOI: 10.1046/j.1365-2990.2000.00225.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The human erythrocyte GLUT-1 is a transmembrane protein which facilitates transport of glucose in the cell in an energy-independent fashion. Neuroectodermal stem cells show strong membrane immunoreactivitry with this marker at early developmental stages in rodents. Membranous expression by undifferentiated neuroectodermal cells gradually decreases while GLUT-1 becomes confined to the endothelial cells, when these acquire blood-brain barrier function. We thus sought to determine whether GLUT-1 expression was limited to embryonal neoplasms of the central nervous system (CNS) which are presumably derived from developmentally arrested neuroectodermal stem cells. Archival material of 40 primary CNS neoplasms were examined for immunoreactivity with anti-GLUT-1. This included both non-embryonal neoplasms (18 astrocytic tumours, one ependymoma and three oligodendroglioma) and embryonal neoplasms (12 cerebellar medulloblastomas, four supratentorial PNETs and two atypical teratoid/rhabdoid tumours (AT/RhT)). In addition, cell lines and nude mice xenografts derived from both undifferentiated and differentiated tumours were assessed for GLUT-1 immunoreactivity by both immunohistochemistry and Western blotting. All embryonal tumours, MBs and PNET xenografts consistently showed GLUT-1 membrane staining. Non-embryonal neoplasms were negative except for vascular staining. Membrane protein fraction of embryonal tumours cell lines immunoreacted by immunoblot with GLUT-1, whereas the glioblastoma cell line was negative. Expression of GLUT-1 supports the stem cell nature of the cells of origin of MBs, supratentorial PNET and AT/RhTs. As a result, GLUT-1 is a useful marker to define the embryonal nature of CNS neoplasms.
Collapse
Affiliation(s)
- M Loda
- Departments of Adult Oncology, Dana Farber Cancer Institute, Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
32
|
Abstract
Neural tissue is entirely dependent on glucose for normal metabolic activity. Since glucose stores in the brain and retina are negligible compared to glucose demand, metabolism in these tissues is dependent upon adequate glucose delivery from the systemic circulation. In the brain, the critical interface for glucose transport is at the brain capillary endothelial cells which comprise the blood-brain barrier (BBB). In the retina, transport occurs across the retinal capillary endothelial cells of the inner blood-retinal barrier (BRB) and the retinal pigment epithelium of the outer BRB. Because glucose transport across these barriers is mediated exclusively by the sodium-independent glucose transporter GLUT1, changes in endothelial glucose transport and GLUT1 abundance in the barriers of the brain and retina may have profound consequences on glucose delivery to these tissues and major implications in the development of two major diabetic complications, namely insulin-induced hypoglycemia and diabetic retinopathy. This review discusses the regulation of brain and retinal glucose transport and glucose transporter expression and considers the role of changes in glucose transporter expression in the development of two of the most devastating complications of long-standing diabetes mellitus and its management.
Collapse
Affiliation(s)
- A K Kumagai
- Department of Internal Medicine, Michigan Diabetes Research and Training Center, University of Michigan Medical School, Ann Arbor, MI 48109-0678, USA.
| |
Collapse
|
33
|
Wolburg H, Liebner S, Reichenbach A, Gerhardt H. The pecten oculi of the chicken: a model system for vascular differentiation and barrier maturation. INTERNATIONAL REVIEW OF CYTOLOGY 1999; 187:111-59. [PMID: 10212979 DOI: 10.1016/s0074-7696(08)62417-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The pecten oculi is a convolute of blood vessels in the vitreous body of the avian eye. This structure is well known for more than a century, but its functions are still a matter of controversies. One of these functions must be the formation of a blood-retina barrier because there is no diffusion barrier for blood-borne compounds available between the pecten and the retina. Surprisingly, the blood-retina barrier characteristics of this organ have not been studied so far, although the pecten oculi may constitute a fascinating model of vascular differentiation and barrier maturation: Pectinate endothelial cells grow by angiogenesis from the ophthalmotemporal artery into the pecten primordium and consecutively gain barrier properties. The pectinate pigmented cells arise during development from retinal pigment epithelial cells and subsequently lose barrier properties. These inverse transdifferentiation processes may be triggered by the peculiar microenvironment in the vitreous body. In addition, the question is discussed whether the avascularity of the avian retina may be due to the specific metabolic activity of the pecten.
Collapse
Affiliation(s)
- H Wolburg
- Institute of Pathology, University of Tübingen, Germany
| | | | | | | |
Collapse
|
34
|
Nualart F, Godoy A, Reinicke K. Expression of the hexose transporters GLUT1 and GLUT2 during the early development of the human brain. Brain Res 1999; 824:97-104. [PMID: 10095047 DOI: 10.1016/s0006-8993(99)01078-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We used immunohistochemistry with anti-glucose transporter antibodies to document the presence of facilitative hexose transporters in the fetal human brain. GLUT1 is expressed in all regions of the fetal brain from ages 10 to 21 weeks. GLUT1 was present in the endothelial cells of the brain capillaries, the epithelial cells of the choroid plexus and neurons. High expression of GLUT2 was observed in the granular layer of the cerebellum in brains 21 weeks old, but GLUT2 immunoreactivity was absent at earlier stages. GLUT3 and GLUT4 immunoreactivities were absent at all stages studied. GLUT5 immunoreactivity was evident only in the cerebellar region of 21-week old fetal brains. We conclude that GLUT1 plays a fundamental role in early human brain development. The data also suggest that the cerebellum of the developing brain has the capacity to transport fructose, a substrate that has not been previously identified as a source of metabolic energy in the adult human brain.
Collapse
Affiliation(s)
- F Nualart
- Departamento de Histología y Embriología, Facultad de Ciencias Biológicas, Casilla 160-C, Universidad de Concepción, Concepción, Chile.
| | | | | |
Collapse
|
35
|
Devaskar SU, Rajakumar PA, Mink RB, McKnight RA, Thamotharan S, Hicks SJ. Effect of development and hypoxic-ischemia upon rabbit brain glucose transporter expression. Brain Res 1999; 823:113-28. [PMID: 10095018 DOI: 10.1016/s0006-8993(99)01143-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have cloned and sequenced a full length rabbit GLUT 1 and partial rabbit GLUT 3 cDNAs. The derived rabbit GLUT 3 peptide revealed 84% homology to the mouse, 82% to the rat, human, dog, and sheep, and 69% to the chicken GLUT 3 peptides. Using Northern blot analysis, we investigated the tissue and brain cellular distribution of GLUT 1 and GLUT 3 expression. In addition, we examined the effect of development and hypoxic-ischemia upon brain GLUT 1 and GLUT 3 mRNA levels. While GLUT 1 mRNA was observed in most tissues, GLUT 3 was expressed predominantly in the brain, placenta, stomach, and lung with minor amounts in the heart, kidney and skeletal muscle. In the brain, both GLUT 1 and GLUT 3 were noted in neuron- and glial-enriched cultures. Both GLUT 1 and GLUT 3 mRNA levels demonstrated a similar developmental progression (p<0.05) secondary to post-transcriptional mechanisms. Further, while hypoxic-ischemia did not significantly affect brain GLUT 1 mRNA and protein, it altered GLUT 3 mRNA levels in a region-specific manner, with a three-fold increase in the cerebral cortex, a two-fold increase in the hippocampus, and a 50% increase in the caudate nucleus (p<0.05). We conclude, that the rabbit GLUT 3 peptide sequence exhibits 82-84% homology to that of other species in the coding region with a 62-89% sequence identity in the 3'-untranslated region. The tissue-specific expression of rabbit GLUT 3 mimics that of the human closely. Postnatal development and hypoxic-ischemia with reperfusion injury cause an increase in brain GLUT 3 expression, as a response to synaptogenesis and substrate deprivation, respectively.
Collapse
Affiliation(s)
- S U Devaskar
- Division of Neonatology and Developmental Biology, Department of Pediatrics, 300 Halket Street, University of Pittsburgh, Magee-Womens Research Institute, Pittsburgh, PA, 15213-3180, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Virgintino D, Robertson D, Monaghan P, Errede M, Ambrosi G, Roncali L, Bertossi M. Glucose transporter GLUT1 localization in human foetus telencephalon. Neurosci Lett 1998; 256:147-50. [PMID: 9855361 DOI: 10.1016/s0304-3940(98)00769-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The endothelial cells of the mature cerebral microvessels, provided with barrier devices (blood-brain barrier, BBB), selectively express the glucose transporter isoform 1 (GLUT1). Presence and localization of the GLUT1 were studied by immunogold silver staining (IGSS) labelling on ultrathin sections of foetal human telencephalon tissue embedded in Lowicryl HM20 according to the progressive lowering of temperature (PLT) method. In the microvascular endothelial cells of the human telencephalon GLUT1 molecules are detected at the 12th gestational week and their expression is increased at the 18th week. In both ages, the transporter is mainly localized on the ablumenal and lateral endothelial cell membranes, and at 18 weeks a greater number of GLUT1 antigenic sites are also seen at the lumenal membrane. Our findings demonstrate both the expression and subcellular localization of GLUT1 be developmentally regulated and suggest an early functioning of the BBB-GLUT1 transporter in the developing human brain.
Collapse
Affiliation(s)
- D Virgintino
- Institute of Human Anatomy, Histology and Embryology, University of Bari School of Medicine, Italy.
| | | | | | | | | | | | | |
Collapse
|
37
|
Ramsauer M, Kunz J, Krause D, Dermietzel R. Regulation of a blood-brain barrier-specific enzyme expressed by cerebral pericytes (pericytic aminopeptidase N/pAPN) under cell culture conditions. J Cereb Blood Flow Metab 1998; 18:1270-81. [PMID: 9809517 DOI: 10.1097/00004647-199811000-00014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In this study we show that the aminopeptidase N of cerebral pericytes (pAPN) associated with the blood-brain barrier (BBB) is downregulated in pericytic cell cultures. This observation is in accordance with previous data describing comparable in vitro effects for BBB-specific enzymes of endothelial or pericytic origin, such as gamma-glutamyl transpeptidase or alkaline phosphatase. By polymerase chain reaction and in situ hybridization we were able to determine that the down-regulation of pAPN occurs at the posttranscriptional level. The mRNA of pAPN was found to be constitutively expressed even when the protein is no longer detectable. Culturing the pericytes in an endothelial cell-conditioned medium allowed pAPN to be reexpressed. However, the reexpression effect depended largely on the culturing conditions of the pericytes. Although purified pericytes deprived of endothelial cells did not reveal a reexpression effect, pericytes that were kept in contact with endothelial cells were able to acquire a pAPN-positive phenotype, indicating that endothelial cells constitute an essential requirement for the in vitro reexpression of pAPN. Astrocytes, however, were insufficient in exerting any reexpression effect.
Collapse
Affiliation(s)
- M Ramsauer
- Institut für Anatomie, Ruhr-Universität Bochum, Germany
| | | | | | | |
Collapse
|
38
|
Probst-Cousin S, Villagran-Lillo R, Lahl R, Bergmann M, Schmid KW, Gullotta F. Secretory meningioma: clinical, histologic, and immunohistochemical findings in 31 cases. Cancer 1997; 79:2003-15. [PMID: 9149029 DOI: 10.1002/(sici)1097-0142(19970515)79:10<2003::aid-cncr23>3.0.co;2-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Secretory meningioma is a rare histologic variant characterized by a unique epithelial differentiation of meningothelial cells resulting in the production of hyaline inclusions. Most previous reports have presented single case observations. The authors selected 31 cases for a clinicopathologic study to characterize this type of tumor further. METHODS Clinical data were compiled and the extent of peritumoral edema was assessed from preoperative computed tomography or magnetic resonance imaging scans. Preparations of surgical specimens of all tumors were studied after both conventional histologic and immunohistochemical preparations were made. Immunostaining was performed by either the avidin-biotin complex method or the alkaline phosphatase-antialkaline phosphatase method using 22 primary antibodies. RESULTS In the tumor collection used in this study, secretory meningiomas represented 3% of meningiomas. The female-to-male ratio was 9:1. Most tumors were located at the sphenoid ridge or at the frontal convexity, and recurrences were not observed. Eighty-four percent of tumors presented with slight to marked peritumoral edema. The MIB-1 staining index showed a mean of 3.8%. Inclusions and surrounding cells consistently expressed epithelial membrane antigen, cytokeratins, carcinoembryonic antigen, and carbohydrate antigen 19-9. In decreasing frequency, they also contained alpha1-antitrypsin, immunoglobulin (Ig)A, alpha1-antichymotrypsin, IgM, and IgG. Cells positive for vimentin and S-100 did not contain inclusions. All tumors were positive for progesterone receptors. Macrophages were stained with antibodies to factor XIIIa, human leukocyte antigen-DR, and alpha1-antitrypsin. In 64% of cases, tumor vessels lacked expression of glucose transporter protein 1. CONCLUSIONS The classification of secretory meningioma as a distinct variant has been justified on clinical, histologic, and immunohistochemical grounds. The unique epithelial features call attention to the broad spectrum of differentiation properties found in meningiomas.
Collapse
Affiliation(s)
- S Probst-Cousin
- Institute of Neuropathology, University Hospital, Münster, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Schroeder RE, Rajakumar PA, Devaskar SU. Effect of streptozotocin-induced maternal diabetes on fetal rat brain glucose transporters. Pediatr Res 1997; 41:346-52. [PMID: 9078533 DOI: 10.1203/00006450-199703000-00007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Glucose, an essential substrate for brain oxidative metabolism, is transported across the blood-brain barrier and into neuronal and glial cells via Glut 1 and Glut 3 facilitative glucose transporter isoforms. To examine the effect of excessive circulating glucose on fetal brain glucose transporter expression, we investigated the effect of streptozotocin-induced maternal diabetes (SEVERE-D; n = 29) on the 20-d gestation fetal rat brain Glut 1 and Glut 3. We studied the effect of streptozotocin alone (STZ-ND; n = 12) in a nondiabetic state as well, along with vehicle injected controls (C; n = 24). In the presence of fetal hyperglycemia (12.63 +/- 0.82 nM-SEVERE-D versus 2.35 +/- 0.28-STZ-ND and 2.42 +/- 0.16-C; p < 0.001) and hypoinsulinemia (0.38 +/- 0.03 nM-SEVERE-D versus 0.50 +/- 0.07-STZ-ND and 0.55 +/- 0.06-C; p < 0.02), no detectable change in fetal brain Glut 1 and Glut 3 pretranslational expression (transcription/elongation rates and corresponding steady state mRNA levels) was noted when simultaneously compared with the STZ-ND and C groups. In contrast, a trend toward a decline in Glut 1 (approximately 25 to 30%, p = 0.05) and a substantive decrease in Glut 3 (approximately 35 to 50%, p = 0.0006) protein concentrations was present in both the STZ-ND and SEVERE-D groups when compared with the C group. These observations support a chemical effect of streptozotocin independent of maternal diabetes upon the translation or posttranslational processing of fetal brain glucose transporters. Maternal diabetes with fetal hyperglycemia, however, failed to substantively alter fetal brain glucose transporters independent of the streptozotocin effects upon neuroectodermally derived tissues. We conclude that maternal diabetes with associated overt fetal hyperglycemia does not significantly change fetal brain glucose transporter levels.
Collapse
Affiliation(s)
- R E Schroeder
- Department of Pediatrics, St. Louis University School of Medicine, Missouri 63110, USA
| | | | | |
Collapse
|
40
|
Aigner A, Wolf S, Gassen HG. Transport und Entgiftung: Grundlagen, Ansätze und Perspektiven für die Erforschung der Blut-Hirn-Schranke. Angew Chem Int Ed Engl 1997. [DOI: 10.1002/ange.19971090105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
41
|
Takata K, Hirano H, Kasahara M. Transport of glucose across the blood-tissue barriers. INTERNATIONAL REVIEW OF CYTOLOGY 1997; 172:1-53. [PMID: 9102392 DOI: 10.1016/s0074-7696(08)62357-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In specialized parts of the body, free exchange of substances between blood and tissue cells is hindered by the presence of a barrier cell layer(s). Specialized milieu of the compartments provided by these "blood-tissue barriers" seems to be important for specific functions of the tissue cells guarded by the barriers. In blood-tissue barriers, such as the blood-brain barrier, blood-cerebrospinal fluid barrier, blood-nerve barrier, blood-retinal barrier, blood-aqueous barrier, blood-perilymph barrier, and placental barrier, endothelial or epithelial cells sealed by tight junctions, or a syncytial cell layer(s), serve as a structural basis of the barrier. A selective transport system localized in the cells of the barrier provides substances needed by the cells inside the barrier. GLUT1, an isoform of facilitated-diffusion glucose transporters, is abundant in cells of the barrier. GLUT1 is concentrated at the critical plasma membranes of cells of the barriers and thereby constitutes the major machinery for the transport of glucose across these barriers where transport occurs by a transcellular mechanism. In the barrier composed of double-epithelial layers, such as the epithelium of the ciliary body in the case of the blood-aqueous barrier, gap junctions appear to play an important role in addition to GLUT1 for the transfer of glucose across the barrier.
Collapse
Affiliation(s)
- K Takata
- Laboratory of Molecular and Cellular Morphology, Gunma University, Japan
| | | | | |
Collapse
|
42
|
Miragall F, Simbürger E, Dermietzel R. Mitral and tufted cells of the mouse olfactory bulb possess gap junctions and express connexin43 mRNA. Neurosci Lett 1996; 216:199-202. [PMID: 8897492 DOI: 10.1016/0304-3940(96)13042-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Analyses of freeze-fracture replicas of mouse olfactory bulb reveal the presence of gap junctions in the plasma membranes of the cell bodies of mitral cells. Due to their localization and morphology we presume that they interconnect mitral and granule cells. Since the quality of electrical transmission between neurons is considered to be determined by the biochemical nature of the gap junction channel forming proteins (connexins) we performed immunohistochemistry and in situ hybridization using probes for connexin43 (Cx43), the most abundant connexin in brain tissue. Attribution of Cx43 immunolabel to specific neurons could not definitely be assessed by means of immunohistochemistry. In situ hybridization, however, using a specific cRNA probe for Cx43 revealed a label confined to cell bodies of mitral and tufted cells of the olfactory bulb. These data indicate that Cx43 is expressed by bulbar neurons and suggest that Cx43 is a molecular constituent of gap junction channels in neurons.
Collapse
Affiliation(s)
- F Miragall
- Institut für Anatomie, Universität Regensburg, Germany
| | | | | |
Collapse
|
43
|
Abstract
Astrocytes form functional networks that participate in active signaling in which external stimuli are generated and amplified in many of the same ways as in neurons. Gap junctions between astrocytes offer the structural avenue by which the electrical and metabolic signals are propagated from one cell to another. Little is known about the trafficking, assembly, and degradation mechanisms of the major astrocytic gap junction protein connexin43. We have studied a glial cell line transfected with the C-erbB2/neu oncogene (neu+), finding severe interruption of gap junctional communication after stable transfection. Evidence from Western blotting and phosphorylation studies showed that the processing of connexin43 to its higher phosphorylated isoforms is disturbed. Confocal laser imaging indicates that the major deficit in the neu+ cells is attributable to a lack in plaque assembly of connexin43. Because the neu+ cells also lack N-CAM proteins and because work from others has indicated a close relationship between communication competence and constitutive CAM expression, our data suggest that expression of C-erbB2/neu oncogene alters cell-cell association via CAM proteins, which thereby affects gap junction plaque assembly and appropriate phosphorylation of connexin43.
Collapse
|
44
|
Affiliation(s)
- D V Rayner
- Division of Biochemical Sciences, Rowett Research Institute, Bucksburn, Aberdeen
| |
Collapse
|
45
|
Zeller K, Vogel J, Kuschinsky W. Postnatal distribution of Glut1 glucose transporter and relative capillary density in blood-brain barrier structures and circumventricular organs during development. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1996; 91:200-8. [PMID: 8852370 DOI: 10.1016/0165-3806(95)00177-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the adult brain, Glut1 is associated with capillaries that form a tight barrier whereas Glut1 is lacking in capillaries with non-barrier properties, i.e. the circumventricular organs. In the present study the postnatal developmental changes of brain capillaries and Glut1 were compared in different tight and non-barrier structures. Rats were investigated at birth, 5th postnatal day (P5), P10, P15, P20, P30 and at the age of one year. Antibody stains of brain capillaries (fibronectin) and of Glut1 were visualized by fluorescent microscopy in identical brain cryosections. All brain capillaries of structures that have a tight barrier in adult animals showed the existence of Glut1 during postnatal development. Most non-barrier structures lacked Glut1 in their capillary endothelium after birth although Glut1 was found in the area postrema and subfornical organ at P0 and disappeared thereafter. The relative capillary density in tight barrier structures of the gray matter was more than doubled from birth to P20 with minor changes later. In contrast white matter structures missed any significant increase during development. It is concluded that Glut1, as an indicator of barrier properties, is existing in all blood-brain barrier structures at birth already. The capillary densities observed in different brain structures at birth are not related to the values found in adult animals.
Collapse
Affiliation(s)
- K Zeller
- Department of Physiology, University of Heidelberg, Germany
| | | | | |
Collapse
|
46
|
ZHANG MEI, OLSSON YNGVE. Immunohistochemical observations. APMIS 1996. [DOI: 10.1111/j.1699-0463.1996.tb00720.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
47
|
Zeller K, Duelli R, Vogel J, Schröck H, Kuschinsky W. Autoradiographic analysis of the regional distribution of Glut3 glucose transporters in the rat brain. Brain Res 1995; 698:175-9. [PMID: 8581478 DOI: 10.1016/0006-8993(95)00888-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glut3 is a glucose transporter protein which facilitates the transport of glucose across the neuronal membranes. The local distribution of Glut3 in the brain is not well known. The present study had the aim to verify the local distribution of Glut3 in the brain and to compare it with the local glucose utilization. A polyclonal antibody directed against the C-terminal peptide sequence of Glut3 was applied to cryosections of rat brains. A secondary antibody was added which had been coupled to 35S. Using autoradiography and radioactive standards, 17 cerebral structures were investigated. The results show moderate differences of Glut3 density in the structures investigated ranging from -23% to +41% of the mean density. The pineal gland was an exception with a density 66% lower than mean. Local cerebral glucose utilization (LCGU) was analyzed in identical brain structures by application of the quantitative autoradiographic 2-deoxyglucose method to conscious rats. The range of LCGU was from -59% to +55% of the mean. No correlation was found between the moderately heterogeneous Glut3 transporter density and the strongly heterogeneous local cerebral glucose utilization. The results show that the local density of Glut3 glucose transporter protein does not reflect the local level of glucose utilization in the brain.
Collapse
Affiliation(s)
- K Zeller
- Department of Physiology, Univeristy of Heidelberg, Germany
| | | | | | | | | |
Collapse
|
48
|
Bauer H, Sonnleitner U, Lametschwandtner A, Steiner M, Adam H, Bauer HC. Ontogenic expression of the erythroid-type glucose transporter (Glut 1) in the telencephalon of the mouse: correlation to the tightening of the blood-brain barrier. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1995; 86:317-25. [PMID: 7656423 DOI: 10.1016/0165-3806(95)00044-e] [Citation(s) in RCA: 54] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Since Glut 1 was shown to be highly abundant in brain microvessels, its distribution during early developmental stages seems of importance in respect to the timing of blood-brain barrier (bbb) formation in the developing CNS. Here we have followed the temporal expression of the erythroid-type glucose transporter Glut 1 in the telencephalon of the embryonic and newborn mouse, beginning at the 9th intrauterine day. Glut 1 immunofluorescence staining was done on cryosections using a rabbit polyclonal antiserum to purified human erythrocyte glucose transporter. Endothelial cells resp. capillaries were detected by staining with a rhodamin-coupled Bandeiraea simplicifolia lectin (BSL). In parallel, the developmental tightening of the embryonic bbb was assessed by perfusion of mouse embryos with Trypan blue and horse radish-peroxidase. At E9, prior to the onset of intraneural neovascularization, strong Glut 1 immunoreactivity was found in the whole neuroectoderm but only minor staining was seen in the perineural domain. Glut 1 expression remained uniformly distributed in the intraneural tissue at E10, the beginning of intraneural neovascularization in the mouse. From E11 onwards, Glut 1 immunoreactivity was invisible in neuroepithelial cells, but appeared tightly associated with intraneural capillaries. Perfusion of E12 embryos using trypan blue solution and HRP revealed that most parts of the CNS and spinal cord were impermeable to the tracer substances at that stage. Thus, we suggest that the bbb is established very early in CNS development, probably in the course of intraneural neovascularization. In addition, our data indicate that the restriction of Glut 1 expression to the intraneural capillaries reflects the onset of bbb function in the mouse embryo.
Collapse
Affiliation(s)
- H Bauer
- Institute of Zoology, University of Salzburg, Austria
| | | | | | | | | | | |
Collapse
|
49
|
Qin Y, Sato TN. Mouse multidrug resistance 1a/3 gene is the earliest known endothelial cell differentiation marker during blood-brain barrier development. Dev Dyn 1995; 202:172-80. [PMID: 7734734 DOI: 10.1002/aja.1002020209] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Molecular mechanisms of endothelial cell differentiation during blood-brain barrier (BBB) development is not well understood due to the lack of specific molecular markers. Here we describe that expression of the mouse multidrug resistance 1a/3 (mdr1a/3) gene can be detected specifically in subsets of vascular endothelial cells associated with neural tissues at as early as embryonic day 10.5 (E10.5). This onset of mdr1a/3 gene expression coincides with the previously described first appearance of morphologically distinct endothelial cells in neural tissues during BBB development. To our knowledge, the mdr1a/3 gene is the earliest endothelial cell differentiation marker gene during BBB development described thus far. In addition, we have found that neither the level nor pattern of mdr1a/3 gene expression in BBB endothelial cells is affected by aberrant cortical neuronal layers in mutant mouse reeler.
Collapse
Affiliation(s)
- Y Qin
- Roche Institute of Molecular Biology, Roche Research Center, Nutley, New Jersey 07110, USA
| | | |
Collapse
|
50
|
Rosenstein JM, More NS. Immunocytochemical expression of the blood-brain barrier glucose transporter (GLUT-1) in neural transplants and brain wounds. J Comp Neurol 1994; 350:229-40. [PMID: 7884040 DOI: 10.1002/cne.903500207] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The present study examined the immunocytochemical expression of the blood-brain barrier glucose transporter (GLUT-1) in a series of fetal neocortical transplants, autonomic tissue transplants, and stab wounds to the rat brain. GLUT-1 is one of a family of different glucose transporters and is found exclusively on barrier-type endothelial cells. In the brain it is responsible for the regulated facilitative diffusion of glucose across the blood-brain barrier. This investigation is the first to determine if this important molecule is altered during the process of angiogenesis that occurs following neural transplantation procedures or direct brain injury. Beginning in late fetal brain, e.g., E18 and continuing into maturity, GLUT-1 was strongly and exclusively expressed on normal cerebral vessels. In solid fetal central nervous system (CNS) transplants up to around 3 weeks postoperative, GLUT-1 was only weakly expressed, particularly as exemplified by colloidal gold immunostaining when compared with the host. At later times examined, up to 15 months postoperative, GLUT-1 immunoexpression was comparable with the normal adjacent brain. In autonomic tissue transplants, where the vessels do not have a blood-brain barrier, as expected, GLUT-1 was not expressed. In stab wounds, at 1 week there was extensive gliosis, and the injured vessels appeared fragmented and collapsed but still expressed GLUT-1, although to a somewhat lesser extent than normal brain. Between 3 and 6 weeks, GLUT-1 was expressed on tortuous vessels and in apparently fibrillar processes in the wound vicinity with a similar pattern to astrocyte (GFAP) reactivity. These results suggest the occurrence of a down-regulation of GLUT-1 in early transplants, perhaps related to reduced glycolytic activity or transient ischemia, or possibly due to the utilization of alternative energy sources. That GLUT-1 expression was not entirely lost in stab wounds to the mature brain suggests that the protein may be more labile in fetal or perinatal brain than in the adult and may not be affected by direct injury. Coupled with previous transplantation studies that have shown reduced neuronal glycolysis and potential barrier alterations, the reduction of GLUT-1 activity within nearly the identical time frame could indicate a relatively early critical period in cellular metabolism following transplantation of CNS tissue.
Collapse
Affiliation(s)
- J M Rosenstein
- Department of Anatomy, George Washington University Medical Center, Washington, D.C. 20037
| | | |
Collapse
|