1
|
Peterson DR, Hawkins RA, Viña JR. Editorial: Organization and Functional Properties of the Blood-Brain Barrier. Front Physiol 2021; 12:796030. [PMID: 34925076 PMCID: PMC8674868 DOI: 10.3389/fphys.2021.796030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Darryl R. Peterson
- Discipline of Physiology and Biophysics, Chicago Medical School/Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Richard A. Hawkins
- Discipline of Physiology and Biophysics, Chicago Medical School/Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Juan R. Viña
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute INCLIVA, University of Valencia, Valencia, Spain
| |
Collapse
|
2
|
Proteome of the Luminal Surface of the Blood-Brain Barrier. Proteomes 2021; 9:proteomes9040045. [PMID: 34842825 PMCID: PMC8629012 DOI: 10.3390/proteomes9040045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022] Open
Abstract
Interrogation of the molecular makeup of the blood–brain barrier (BBB) using proteomic techniques has contributed to the cataloguing and functional understanding of the proteins uniquely organized at this specialized interface. The majority of proteomic studies have focused on cellular components of the BBB, including cultured brain endothelial cells (BEC). Detailed proteome mapping of polarized BEC membranes and their intracellular endosomal compartments has led to an improved understanding of the processes leading to internalization and transport of various classes of molecules across the BBB. Quantitative proteomic methods have further enabled absolute and comparative quantification of key BBB transporters and receptors in isolated BEC and microvessels from various species. However, translational studies further require in vivo/in situ analyses of the proteins exposed on the luminal surface of BEC in vessels under various disease and treatment conditions. In vivo proteomics approaches, both profiling and quantitative, usually rely on ‘capturing’ luminally-exposed proteins after perfusion with chemical labeling reagents, followed by analysis with various mass spectrometry-based approaches. This manuscript reviews recent advances in proteomic analyses of luminal membranes of BEC in vitro and in vivo and their applications in translational studies focused on developing novel delivery methods across the BBB.
Collapse
|
3
|
Pardridge WM. Brain Delivery of Nanomedicines: Trojan Horse Liposomes for Plasmid DNA Gene Therapy of the Brain. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:602236. [PMID: 35047884 PMCID: PMC8757841 DOI: 10.3389/fmedt.2020.602236] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/06/2020] [Indexed: 12/14/2022] Open
Abstract
Non-viral gene therapy of the brain is enabled by the development of plasmid DNA brain delivery technology, which requires the engineering and manufacturing of nanomedicines that cross the blood-brain barrier (BBB). The development of such nanomedicines is a multi-faceted problem that requires progress at multiple levels. First, the type of nanocontainer, e.g., nanoparticle or liposome, which encapsulates the plasmid DNA, must be developed. Second, the type of molecular Trojan horse, e.g., peptide or receptor-specific monoclonal antibody (MAb), must be selected for incorporation on the surface of the nanomedicine, as this Trojan horse engages specific receptors expressed on the BBB, and the brain cell membrane, to trigger transport of the nanomedicine from blood into brain cells beyond the BBB. Third, the plasmid DNA must be engineered without bacterial elements, such as antibiotic resistance genes, to enable administration to humans; the plasmid DNA must also be engineered with tissue-specific gene promoters upstream of the therapeutic gene, to insure gene expression in the target organ with minimal off-target expression. Fourth, upstream manufacturing of the nanomedicine must be developed and scalable so as to meet market demand for the target disease, e.g., annual long-term treatment of 1,000 patients with an orphan disease, short term treatment of 10,000 patients with malignant glioma, or 100,000 patients with new onset Parkinson's disease. Fifth, downstream manufacturing problems, such as nanomedicine lyophilization, must be solved to ensure the nanomedicine has a commercially viable shelf-life for treatment of CNS disease in humans.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
4
|
Ghaffari H, Grant SC, Petzold LR, Harrington MG. Regulation of CSF and Brain Tissue Sodium Levels by the Blood-CSF and Blood-Brain Barriers During Migraine. Front Comput Neurosci 2020; 14:4. [PMID: 32116618 PMCID: PMC7010722 DOI: 10.3389/fncom.2020.00004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/10/2020] [Indexed: 11/13/2022] Open
Abstract
Cerebrospinal fluid (CSF) and brain tissue sodium levels increase during migraine. However, little is known regarding the underlying mechanisms of sodium homeostasis disturbance in the brain during the onset and propagation of migraine. Exploring the cause of sodium dysregulation in the brain is important, since correction of the altered sodium homeostasis could potentially treat migraine. Under the hypothesis that disturbances in sodium transport mechanisms at the blood-CSF barrier (BCSFB) and/or the blood-brain barrier (BBB) are the underlying cause of the elevated CSF and brain tissue sodium levels during migraines, we developed a mechanistic, differential equation model of a rat's brain to compare the significance of the BCSFB and the BBB in controlling CSF and brain tissue sodium levels. The model includes the ventricular system, subarachnoid space, brain tissue and blood. Sodium transport from blood to CSF across the BCSFB, and from blood to brain tissue across the BBB were modeled by influx permeability coefficients PBCSFB and PBBB, respectively, while sodium movement from CSF into blood across the BCSFB, and from brain tissue to blood across the BBB were modeled by efflux permeability coefficients PBCSFB′ and PBBB′, respectively. We then performed a global sensitivity analysis to investigate the sensitivity of the ventricular CSF, subarachnoid CSF and brain tissue sodium concentrations to pathophysiological variations in PBCSFB, PBBB, PBCSFB′ and PBBB′. Our results show that the ventricular CSF sodium concentration is highly influenced by perturbations of PBCSFB, and to a much lesser extent by perturbations of PBCSFB′. Brain tissue and subarachnoid CSF sodium concentrations are more sensitive to pathophysiological variations of PBBB and PBBB′ than variations of PBCSFB and PBCSFB′ within 30 min of the onset of the perturbations. However, PBCSFB is the most sensitive model parameter, followed by PBBB and PBBB′, in controlling brain tissue and subarachnoid CSF sodium levels within 3 h of the perturbation onset.
Collapse
Affiliation(s)
- Hamed Ghaffari
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Samuel C Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL, United States
| | - Linda R Petzold
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, United States
| | - Michael G Harrington
- Neuroscience, Huntington Medical Research Institutes, Pasadena, CA, United States
| |
Collapse
|
5
|
R. Peterson D, J. Sukowski E. Prevention of Oxidative Injury Associated with Thrombolysis for Ischemic Stroke. Antioxidants (Basel) 2019. [DOI: 10.5772/intechopen.84774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
6
|
Lykke K, Assentoft M, Hørlyck S, Helms HC, Stoica A, Toft-Bertelsen TL, Tritsaris K, Vilhardt F, Brodin B, MacAulay N. Evaluating the involvement of cerebral microvascular endothelial Na +/K +-ATPase and Na +-K +-2Cl - co-transporter in electrolyte fluxes in an in vitro blood-brain barrier model of dehydration. J Cereb Blood Flow Metab 2019; 39:497-512. [PMID: 28994331 PMCID: PMC6421245 DOI: 10.1177/0271678x17736715] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The blood-brain barrier (BBB) is involved in brain water and salt homeostasis. Blood osmolarity increases during dehydration and water is osmotically extracted from the brain. The loss of water is less than expected from pure osmotic forces, due to brain electrolyte accumulation. Although the underlying molecular mechanisms are unresolved, the current model suggests the luminally expressed Na+-K+-2Cl- co-transporter 1 (NKCC1) as a key component, while the role of the Na+/K+-ATPase remains uninvestigated. To test the involvement of these proteins in brain electrolyte flux under mimicked dehydration, we employed a tight in vitro co-culture BBB model with primary cultures of brain endothelial cells and astrocytes. The Na+/K+-ATPase and the NKCC1 were both functionally dominant in the abluminal membrane. Exposure of the in vitro BBB model to conditions mimicking systemic dehydration, i.e. hyperosmotic conditions, vasopressin, or increased [K+]o illustrated that NKCC1 activity was unaffected by exposure to vasopressin and to hyperosmotic conditions. Hyperosmotic conditions and increased K+ concentrations enhanced the Na+/K+-ATPase activity, here determined to consist of the α1 β1 and α1 β3 isozymes. Abluminally expressed endothelial Na+/K+-ATPase, and not NKCC1, may therefore counteract osmotic brain water loss during systemic dehydration by promoting brain Na+ accumulation.
Collapse
Affiliation(s)
- Kasper Lykke
- 1 Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Assentoft
- 1 Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofie Hørlyck
- 2 Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Cc Helms
- 2 Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anca Stoica
- 1 Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine L Toft-Bertelsen
- 1 Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katerina Tritsaris
- 3 Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Frederik Vilhardt
- 3 Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birger Brodin
- 2 Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nanna MacAulay
- 1 Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Hartz AMS, Schulz JA, Sokola BS, Edelmann SE, Shen AN, Rempe RG, Zhong Y, Seblani NE, Bauer B. Isolation of Cerebral Capillaries from Fresh Human Brain Tissue. J Vis Exp 2018. [PMID: 30272660 DOI: 10.3791/57346] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Understanding blood-brain barrier function under physiological and pathophysiological conditions is critical for the development of new therapeutic strategies that hold the promise to enhance brain drug delivery, improve brain protection, and treat brain disorders. However, studying the human blood-brain barrier function is challenging. Thus, there is a critical need for appropriate models. In this regard, brain capillaries isolated from human brain tissue represent a unique tool to study barrier function as close to the human in vivo situation as possible. Here, we describe an optimized protocol to isolate capillaries from human brain tissue at a high yield and with consistent quality and purity. Capillaries are isolated from fresh human brain tissue using mechanical homogenization, density-gradient centrifugation, and filtration. After the isolation, the human brain capillaries can be used for various applications including leakage assays, live cell imaging, and immune-based assays to study protein expression and function, enzyme activity, or intracellular signaling. Isolated human brain capillaries are a unique model to elucidate the regulation of the human blood-brain barrier function. This model can provide insights into central nervous system (CNS) pathogenesis, which will help the development of therapeutic strategies for treating CNS disorders.
Collapse
Affiliation(s)
- Anika M S Hartz
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University of Kentucky
| | - Julia A Schulz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky
| | - Brent S Sokola
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky
| | - Stephanie E Edelmann
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University of Kentucky
| | - Andrew N Shen
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University of Kentucky
| | - Ralf G Rempe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky
| | - Yu Zhong
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University of Kentucky
| | | | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky;
| |
Collapse
|
8
|
Hladky SB, Barrand MA. Fluid and ion transfer across the blood-brain and blood-cerebrospinal fluid barriers; a comparative account of mechanisms and roles. Fluids Barriers CNS 2016; 13:19. [PMID: 27799072 PMCID: PMC5508927 DOI: 10.1186/s12987-016-0040-3] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/01/2016] [Indexed: 12/24/2022] Open
Abstract
The two major interfaces separating brain and blood have different primary roles. The choroid plexuses secrete cerebrospinal fluid into the ventricles, accounting for most net fluid entry to the brain. Aquaporin, AQP1, allows water transfer across the apical surface of the choroid epithelium; another protein, perhaps GLUT1, is important on the basolateral surface. Fluid secretion is driven by apical Na+-pumps. K+ secretion occurs via net paracellular influx through relatively leaky tight junctions partially offset by transcellular efflux. The blood-brain barrier lining brain microvasculature, allows passage of O2, CO2, and glucose as required for brain cell metabolism. Because of high resistance tight junctions between microvascular endothelial cells transport of most polar solutes is greatly restricted. Because solute permeability is low, hydrostatic pressure differences cannot account for net fluid movement; however, water permeability is sufficient for fluid secretion with water following net solute transport. The endothelial cells have ion transporters that, if appropriately arranged, could support fluid secretion. Evidence favours a rate smaller than, but not much smaller than, that of the choroid plexuses. At the blood-brain barrier Na+ tracer influx into the brain substantially exceeds any possible net flux. The tracer flux may occur primarily by a paracellular route. The blood-brain barrier is the most important interface for maintaining interstitial fluid (ISF) K+ concentration within tight limits. This is most likely because Na+-pumps vary the rate at which K+ is transported out of ISF in response to small changes in K+ concentration. There is also evidence for functional regulation of K+ transporters with chronic changes in plasma concentration. The blood-brain barrier is also important in regulating HCO3- and pH in ISF: the principles of this regulation are reviewed. Whether the rate of blood-brain barrier HCO3- transport is slow or fast is discussed critically: a slow transport rate comparable to those of other ions is favoured. In metabolic acidosis and alkalosis variations in HCO3- concentration and pH are much smaller in ISF than in plasma whereas in respiratory acidosis variations in pHISF and pHplasma are similar. The key similarities and differences of the two interfaces are summarized.
Collapse
Affiliation(s)
- Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD UK
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD UK
| |
Collapse
|
9
|
Devraj K, Poznanovic S, Spahn C, Schwall G, Harter PN, Mittelbronn M, Antoniello K, Paganetti P, Muhs A, Heilemann M, Hawkins RA, Schrattenholz A, Liebner S. BACE-1 is expressed in the blood-brain barrier endothelium and is upregulated in a murine model of Alzheimer's disease. J Cereb Blood Flow Metab 2016; 36:1281-94. [PMID: 26661166 PMCID: PMC4929696 DOI: 10.1177/0271678x15606463] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 07/21/2015] [Indexed: 01/08/2023]
Abstract
Endothelial cells of the blood-brain barrier form a structural and functional barrier maintaining brain homeostasis via paracellular tight junctions and specific transporters such as P-glycoprotein. The blood-brain barrier is responsible for negligible bioavailability of many neuroprotective drugs. In Alzheimer's disease, current treatment approaches include inhibitors of BACE-1 (β-site of amyloid precursor protein cleaving enzyme), a proteinase generating neurotoxic β-amyloid. It is known that BACE-1 is highly expressed in endosomes and membranes of neurons and glia. We now provide evidence that BACE-1 is expressed in blood-brain barrier endothelial cells of human, mouse, and bovine origin. We further show its predominant membrane localization by 3D-dSTORM super-resolution microscopy, and by biochemical fractionation that further shows an abluminal distribution of BACE-1 in brain microvessels. We confirm its functionality in processing APP in primary mouse brain endothelial cells. In an Alzheimer's disease mouse model we show that BACE-1 is upregulated at the blood-brain barrier compared to healthy controls. We therefore suggest a critical role for BACE-1 at the blood-brain barrier in β-amyloid generation and in vascular aspects of Alzheimer's disease, particularly in the development of cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Kavi Devraj
- Edinger Institute of Neurology, Goethe University Medical School, Frankfurt, Germany
| | | | - Christoph Spahn
- Institute of Physical and Theoretical Chemistry, Goethe-University, Frankfurt, Germany
| | | | - Patrick N Harter
- Edinger Institute of Neurology, Goethe University Medical School, Frankfurt, Germany
| | - Michel Mittelbronn
- Edinger Institute of Neurology, Goethe University Medical School, Frankfurt, Germany
| | | | | | | | - Mike Heilemann
- Institute of Physical and Theoretical Chemistry, Goethe-University, Frankfurt, Germany
| | - Richard A Hawkins
- Dept of Physiology/Biophysics, University of Health Sci./Chicago Medical School, Illinois, USA
| | | | - Stefan Liebner
- Edinger Institute of Neurology, Goethe University Medical School, Frankfurt, Germany
| |
Collapse
|
10
|
Worzfeld T, Schwaninger M. Apicobasal polarity of brain endothelial cells. J Cereb Blood Flow Metab 2016; 36:340-62. [PMID: 26661193 PMCID: PMC4759676 DOI: 10.1177/0271678x15608644] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/07/2015] [Indexed: 01/24/2023]
Abstract
Normal brain homeostasis depends on the integrity of the blood-brain barrier that controls the access of nutrients, humoral factors, and immune cells to the CNS. The blood-brain barrier is composed mainly of brain endothelial cells. Forming the interface between two compartments, they are highly polarized. Apical/luminal and basolateral/abluminal membranes differ in their lipid and (glyco-)protein composition, allowing brain endothelial cells to secrete or transport soluble factors in a polarized manner and to maintain blood flow. Here, we summarize the basic concepts of apicobasal cell polarity in brain endothelial cells. To address potential molecular mechanisms underlying apicobasal polarity in brain endothelial cells, we draw on investigations in epithelial cells and discuss how polarity may go awry in neurological diseases.
Collapse
Affiliation(s)
- Thomas Worzfeld
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), University of Marburg, Marburg, Germany Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany German Research Centre for Cardiovascular Research, DZHK, Lübeck, Germany
| |
Collapse
|
11
|
Synthesis of diacylated γ-glutamyl-cysteamine prodrugs, and in vitro evaluation of their cytotoxicity and intracellular delivery of cysteamine. Eur J Med Chem 2016; 109:206-15. [DOI: 10.1016/j.ejmech.2015.12.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/28/2015] [Accepted: 12/14/2015] [Indexed: 11/20/2022]
|
12
|
Deracinois B, Lenfant AM, Dehouck MP, Flahaut C. Tissue Non-specific Alkaline Phosphatase (TNAP) in Vessels of the Brain. Subcell Biochem 2016. [PMID: 26219710 DOI: 10.1007/978-94-017-7197-9_7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The microvessels of the brain represent around 3-4 % of the brain compartment but constitute the most important length (400 miles) and surface of exchange (20 m(2)) between the blood and the parenchyma of brain. Under influence of surrounding tissues, the brain microvessel endothelium expresses a specific phenotype that regulates and restricts the entry of compounds and cells from blood to brain, and defined the so-called blood-brain barrier (BBB). Evidences that alkaline phosphatase (AP) is a characteristic feature of the BBB phenotype that allows differentiating capillary endothelial cells from brain to those of the periphery have rapidly emerge. Thenceforth, AP has been rapidly used as a biomarker of the blood-brain barrier phenotype. In fact, brain capillary endothelial cells (BCECs) express exclusively tissue non-specific alkaline phosphatase (TNAP). There are several lines of evidence in favour of an important role for TNAP in brain function. TNAP is thought to be responsible for the control of transport of some compounds across the plasma membrane of the BCECs. Here, we report that levamisole-mediated inhibition of TNAP provokes an increase of the permeability to Lucifer Yellow of the endothelial monolayer. Moreover, we illustrate the disruption of the cytoskeleton organization. Interestingly, all observed effects were reversible 24 h after levamisole removal and correlated with the return of a full activity of the TNAP. This reversible effect remains to be studied in details to evaluate the potentiality of a levamisole treatment to enhance the entry of drugs in the brain parenchyma.
Collapse
|
13
|
Mokgokong R, Wang S, Taylor CJ, Barrand MA, Hladky SB. Ion transporters in brain endothelial cells that contribute to formation of brain interstitial fluid. Pflugers Arch 2014; 466:887-901. [PMID: 24022703 PMCID: PMC4006130 DOI: 10.1007/s00424-013-1342-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 08/24/2013] [Accepted: 08/24/2013] [Indexed: 02/07/2023]
Abstract
Ions and water transported across the endothelium lining the blood–brain barrier contribute to the fluid secreted into the brain and are important in maintaining appropriate volume and ionic composition of brain interstitial fluid. Changes in this secretion process may occur after stroke. The present study identifies at transcript and protein level ion transporters involved in the movement of key ions and examines how levels of certain of these alter following oxidative stress. Immunohistochemistry provides evidence for Cl−/HCO3− exchanger, AE2, and Na+, HCO3− cotransporters, NBCe1 and NBCn1, on brain microvessels. mRNA analysis by RT-PCR reveals expression of these transporters in cultured rat brain microvascular endothelial cells (both primary and immortalized GPNT cells) and also Na+/H+ exchangers, NHE1 (primary and immortalized) and NHE2 (primary cells only). Knock-down using siRNA in immortalized GPNT cells identifies AE2 as responsible for much of the Cl−/HCO3− exchange following extracellular chloride removal and NHE1 as the transporter that accounts for most of the Na+/H+ exchange following intracellular acidification. Transcript levels of both AE2 and NHE1 are increased following hypoxia/reoxygenation. Further work is now required to determine the localization of the bicarbonate transporters to luminal or abluminal membranes of the endothelial cells as well as to identify and localize additional transport mechanisms that must exist for K+ and Cl−.
Collapse
Affiliation(s)
- Ruth Mokgokong
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Shanshan Wang
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Caroline J. Taylor
- O’Brien Institute and Department of Surgery, University of Melbourne, St. Vincent’s Hospital, 42 Fitzroy Street, Fitzroy, Melbourne, VIC 3065 Australia
- Faculty of Health Sciences, Australian Catholic University, Melbourne, VIC 3065 Australia
| | - Margery A. Barrand
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| | - Stephen B. Hladky
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD UK
| |
Collapse
|
14
|
Transendothelial Transport and Its Role in Therapeutics. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:309404. [PMID: 27355037 PMCID: PMC4897564 DOI: 10.1155/2014/309404] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/13/2014] [Accepted: 06/18/2014] [Indexed: 12/17/2022]
Abstract
Present review paper highlights role of BBB in endothelial transport of various substances into the brain. More specifically, permeability functions of BBB in transendothelial transport of various substances such as metabolic fuels, ethanol, amino acids, proteins, peptides, lipids, vitamins, neurotransmitters, monocarbxylic acids, gases, water, and minerals in the peripheral circulation and into the brain have been widely explained. In addition, roles of various receptors, ATP powered pumps, channels, and transporters in transport of vital molecules in maintenance of homeostasis and normal body functions have been described in detail. Major role of integral membrane proteins, carriers, or transporters in drug transport is highlighted. Both diffusion and carrier mediated transport mechanisms which facilitate molecular trafficking through transcellular route to maintain influx and outflux of important nutrients and metabolic substances are elucidated. Present review paper aims to emphasize role of important transport systems with their recent advancements in CNS protection mainly for providing a rapid clinical aid to patients. This review also suggests requirement of new well-designed therapeutic strategies mainly potential techniques, appropriate drug formulations, and new transport systems for quick, easy, and safe delivery of drugs across blood brain barrier to save the life of tumor and virus infected patients.
Collapse
|
15
|
|
16
|
Kimura T, Takanami T, Sakashita T, Wada S, Kobayashi Y, Higashitani A. Innate Immune Genes Including a Mucin-Like Gene,mul-1, Induced by Ionizing Radiation inCaenorhabditis elegans. Radiat Res 2012; 178:313-20. [DOI: 10.1667/rr2989.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
17
|
The role of amino acid transporters in GSH synthesis in the blood-brain barrier and central nervous system. Neurochem Int 2012; 61:405-14. [PMID: 22634224 DOI: 10.1016/j.neuint.2012.05.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 04/17/2012] [Accepted: 05/15/2012] [Indexed: 12/22/2022]
Abstract
Glutathione (GSH) plays a critical role in protecting cells from oxidative stress and xenobiotics, as well as maintaining the thiol redox state, most notably in the central nervous system (CNS). GSH concentration and synthesis are highly regulated within the CNS and are limited by availability of the sulfhydryl amino acid (AA) l-cys, which is mainly transported from the blood, through the blood-brain barrier (BBB), and into neurons. Several antiporter transport systems (e.g., x(c)(-), x(-)(AG), and L) with clearly different luminal and abluminal distribution, Na(+), and pH dependency have been described in brain endothelial cells (BEC) of the BBB, as well as in neurons, astrocytes, microglia and oligodendrocytes from different brain structures. The purpose of this review is to summarize information regarding the different AA transport systems for l-cys and its oxidized form l-cys(2) in the CNS, such as expression and activity in blood-brain barrier endothelial cells, astrocytes and neurons and environmental factors that modulate transport kinetics.
Collapse
|
18
|
Rasgado-Flores H, Mokashi A, Hawkins RA. Na(+)-dependent transport of taurine is found only on the abluminal membrane of the blood-brain barrier. Exp Neurol 2011; 233:457-62. [PMID: 22123083 DOI: 10.1016/j.expneurol.2011.11.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 10/26/2011] [Accepted: 11/10/2011] [Indexed: 11/16/2022]
Abstract
Luminal and abluminal plasma membranes were isolated from bovine brain microvessels and used to identify and characterize Na(+)-dependent and facilitative taurine transport. The calculated transmembrane potential was -59 mV at time 0; external Na(+) (or choline under putative zero-trans conditions) was 126 mM (T=25 °C). The apparent affinity constants of the taurine transporters were determined over a range of taurine concentrations from 0.24 μM to 11.4 μM. Abluminal membranes had both Na(+)-dependent taurine transport as well as facilitative transport while luminal membranes only had facilitative transport. The apparent K(m) for facilitative and Na(+)-dependent taurine transport were 0.06±0.02 μM and 0.7±0.1 μM, respectively. The Na(+)-dependent transport of taurine was voltage dependent over the range of voltages studied (-25 to -101 mV). The transport was over 5 times greater at -101 mV compared to when V(m) was -25 mV. The sensitivity to external osmolality of Na(+)-dependent transport was studied over a range of osmolalities (229 to 398 mOsm/kg H(2)O) using mannitol as the osmotic agent to adjust the osmolality. For these experiments the concentration of Na(+) was maintained constant at 50mM, and the calculated transmembrane potential was -59 mV. The Na(+)-dependent transport system was sensitive to osmolality with the greatest rate observed at 229 mOsm/kg H(2)O.
Collapse
Affiliation(s)
- Hector Rasgado-Flores
- Department of Physiology & Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064-3095 USA.
| | | | | |
Collapse
|
19
|
Devraj K, Klinger ME, Myers RL, Mokashi A, Hawkins RA, Simpson IA. GLUT-1 glucose transporters in the blood-brain barrier: differential phosphorylation. J Neurosci Res 2011; 89:1913-25. [PMID: 21910135 DOI: 10.1002/jnr.22738] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/13/2011] [Accepted: 06/16/2011] [Indexed: 11/11/2022]
Abstract
Glucose is the primary metabolic fuel for the mammalian brain, and a continuous supply is required to maintain normal CNS function. The transport of glucose across the blood-brain barrier (BBB) into the brain is mediated by the facilitative glucose transporter GLUT-1. Prior studies (Simpson et al. [2001] J Biol Chem 276:12725-12729) had revealed that the conformations of the GLUT-1 transporter were different in luminal (blood facing) and abluminal (brain facing) membranes of bovine cerebral endothelial cells, based on differential antibody recognition. This study has extended these observations and, by using a combination of 2D-PAGE/Western blotting and immunogold electron microscopy, determined that these different conformations are exhibited in vivo and arise from differential phosphorylation of GLUT-1 and not from alternative splicing or altered O- or N-linked glycosylation.
Collapse
Affiliation(s)
- Kavi Devraj
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, Pennsylvania 17033, USA
| | | | | | | | | | | |
Collapse
|
20
|
Abstract
Glutamate concentrations in plasma are 50-100 micromol/L; in whole brain, they are 10,000-12,000 micromol/L but only 0.5-2 micromol/L in extracellular fluids (ECFs). The low ECF concentrations, which are essential for optimal brain function, are maintained by neurons, astrocytes, and the blood-brain barrier (BBB). Cerebral capillary endothelial cells form the BBB that surrounds the entire central nervous system. Tight junctions connect endothelial cells and separate the BBB into luminal and abluminal domains. Molecules entering or leaving the brain thus must pass 2 membranes, and each membrane has distinct properties. Facilitative carriers exist only in luminal membranes, and Na(+)-dependent glutamate cotransporters (excitatory amino acid transporters; EAATs) exist exclusively in abluminal membranes. The EAATs are secondary transporters that couple the Na(+) gradient between the ECF and the endothelial cell to move glutamate against the existing electrochemical gradient. Thus, the EAATs in the abluminal membrane shift glutamate from the ECF to the endothelial cell where glutamate is free to diffuse into blood on facilitative carriers. This organization does not allow net glutamate entry to the brain; rather, it promotes the removal of glutamate and the maintenance of low glutamate concentrations in the ECF. This explains studies that show that the BBB is impermeable to glutamate, even at high concentrations, except in a few small areas that have fenestrated capillaries (circumventricular organs). Recently, the question of whether the BBB becomes permeable in diabetes has arisen. This issue was tested in rats with diet-induced obesity and insulin resistance or with streptozotocin-induced diabetes. Neither condition produced any detectable effect on BBB glutamate transport.
Collapse
Affiliation(s)
- Richard A Hawkins
- Department of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064-3095, USA.
| |
Collapse
|
21
|
Devraj K, Geguchadze R, Klinger ME, Freeman WM, Mokashi A, Hawkins RA, Simpson IA. Improved membrane protein solubilization and clean-up for optimum two-dimensional electrophoresis utilizing GLUT-1 as a classic integral membrane protein. J Neurosci Methods 2009; 184:119-23. [PMID: 19631691 DOI: 10.1016/j.jneumeth.2009.07.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 07/09/2009] [Accepted: 07/15/2009] [Indexed: 11/18/2022]
Abstract
Two-dimensional (2-D) electrophoresis remains a primary resolving tool for proteomic analyses. The final number of proteins resolved by 2-D electrophoresis depends on their respective solubility, size, charge, and isoelectric point. While water-soluble cytosolic proteins have often been well represented in 2-D maps, the same is not true with membrane proteins. Highly hydrophobic in nature, membrane proteins are poorly resolved in 2-D gels due to problems associated primarily with sample preparation. This is of especial concern in neuroscience studies where many proteins of interest are membrane bound. In the current work, we present a substantially improved sample preparation protocol for membrane proteins utilizing the GLUT-1 glucose transporter from brain microvessels as an example of a typical membrane protein. GLUT-1 (SLC2A1; solute carrier family 2 (facilitated glucose transporter), member 1) is a 55kD glycoprotein that contains 12 membrane-spanning alpha helices that impart the protein its characteristic hydrophobicity. GLUT-1 based on its amino acid sequence has a theoretical isoelectric point (pI) of 8.94. Using a combination of the non-ionic detergents, n-dodecyl-beta-maltoside (DDM) and amido sulphobetaine-14 (ASB-14) for sample solubilization, and a modification of the Bio-Rad 2-D clean-up protocol involving trichloroacetic acid (TCA)/acetone, we obtained near complete solubilization of GLUT-1 and greater than 90% recovery of this membrane protein in 1-D and 2-D Western blots. The total number of proteins resolved also increased dramatically in Deep Purple total protein stains using our improved protocol.
Collapse
Affiliation(s)
- K Devraj
- Department of Neural & Behavioral Sciences, College of Medicine, Pennsylvania State University, United States
| | | | | | | | | | | | | |
Collapse
|
22
|
Huang BS, White RA, Jeng AY, Leenen FHH. Role of central nervous system aldosterone synthase and mineralocorticoid receptors in salt-induced hypertension in Dahl salt-sensitive rats. Am J Physiol Regul Integr Comp Physiol 2009; 296:R994-R1000. [DOI: 10.1152/ajpregu.90903.2008] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In Dahl salt-sensitive (S) rats, high salt intake increases cerebrospinal fluid (CSF) Na+ concentration ([Na+]) and blood pressure (BP). Intracerebroventricular (ICV) infusion of a mineralocorticoid receptor (MR) blocker prevents the hypertension. To assess the role of aldosterone locally produced in the brain, we evaluated the effects of chronic central blockade with the aldosterone synthase inhibitor FAD286 and the MR blocker spironolactone on changes in aldosterone and corticosterone content in the hypothalamus and the increase in CSF [Na+] and hypertension induced by high salt intake in Dahl S rats. After 4 wk of high salt intake, plasma aldosterone and corticosterone were not changed, but hypothalamic aldosterone increased by ∼35% and corticosterone tended to increase in Dahl S rats, whereas both steroids decreased by ∼65% in Dahl salt-resistant rats. In Dahl S rats fed the high-salt diet, ICV infusion of FAD286 or spironolactone did not affect the increase in CSF [Na+]. ICV infusion of FAD286 prevented the increase in hypothalamic aldosterone and 30 mmHg of the 50-mmHg BP increase induced by high salt intake. ICV infusion of spironolactone fully prevented the salt-induced hypertension. These results suggest that, in Dahl S rats, high salt intake increases aldosterone synthesis in the hypothalamus and aldosterone acts as the main MR agonist activating central pathways contributing to salt-induced hypertension.
Collapse
|
23
|
Millar ID, Wang S, Brown PD, Barrand MA, Hladky SB. Kv1 and Kir2 potassium channels are expressed in rat brain endothelial cells. Pflugers Arch 2007; 456:379-91. [DOI: 10.1007/s00424-007-0377-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2007] [Accepted: 10/23/2007] [Indexed: 12/01/2022]
|
24
|
O'Kane RL, Viña JR, Simpson I, Zaragozá R, Mokashi A, Hawkins RA. Cationic amino acid transport across the blood-brain barrier is mediated exclusively by system y+. Am J Physiol Endocrinol Metab 2006; 291:E412-9. [PMID: 16569760 DOI: 10.1152/ajpendo.00007.2006] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cationic amino acid (CAA) transport is brought about by two families of proteins that are found in various tissues: Cat (CAA transporter), referred to as system y+, and Bat [broad-scope amino acid (AA) transporter], which comprises systems b0,+, B0,+, and y+L. CAA traverse the blood-brain barrier (BBB), but experiments done in vivo have only been able to examine the BBB from the luminal (blood-facing) side. In the present study, plasma membranes isolated from bovine brain microvessels were used to identify and characterize the CAA transporter(s) on both sides of the BBB. From these studies, it was concluded that system y+ was the only transporter present, with a prevalence of activity on the abluminal membrane. System y+ was voltage dependent and had a Km of 470 +/- 106 microM (SE) for lysine, a Ki of 34 microM for arginine, and a Ki of 290 microM for ornithine. In the presence of Na+, system y+ was inhibited by several essential neutral AAs. The Ki values were 3-10 times the plasma concentrations, suggesting that system y+ was not as important a point of access for these AAs as system L1. Several small nonessential AAs (serine, glutamine, alanine,and glycine) inhibited system y+ with Ki values similar to their plasma concentrations, suggesting that system y+ may account for the permeability of the BBB to these AAs. System y+ may be important in the provision of arginine for NO synthesis. Real-time PCR and Western blotting techniques established the presence of the three known nitric oxide synthases in cerebral endothelial cells: NOS-1 (neuronal), NOS-2 (inducible), and NOS-3 (endothelial). These results confirm that system y+ is the only CAA transporter in the BBB and suggest that NO can be produced in brain endothelial cells.
Collapse
Affiliation(s)
- Robyn L O'Kane
- Natural and Applied Science Department, LaGuardia Community College/City University of New York, New York, USA
| | | | | | | | | | | |
Collapse
|
25
|
Hawkins RA, Simpson IA, Mokashi A, Viña JR. Pyroglutamate stimulates Na+ -dependent glutamate transport across the blood-brain barrier. FEBS Lett 2006; 580:4382-6. [PMID: 16844120 DOI: 10.1016/j.febslet.2006.06.097] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 06/29/2006] [Accepted: 06/30/2006] [Indexed: 10/24/2022]
Abstract
Regulation of Na(+)-dependent glutamate transport was studied in isolated luminal and abluminal plasma membranes derived from the bovine blood-brain barrier. Abluminal membranes have Na(+)-dependent glutamate transporters while luminal membranes have facilitative transporters. This organization allows glutamate to be actively removed from brain. gamma-Glutamyl transpeptidase, the first enzyme of the gamma-glutamyl cycle (GGC), is on the luminal membrane. Pyroglutamate (oxoproline), an intracellular product of GGC, stimulated Na(+)-dependent transport of glutamate by 46%, whereas facilitative glutamate uptake in luminal membranes was inhibited. This relationship between GGC and glutamate transporters may be part of a regulatory mechanism that accelerates glutamate removal from brain.
Collapse
Affiliation(s)
- Richard A Hawkins
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064-3095, USA.
| | | | | | | |
Collapse
|
26
|
Hawkins RA, O'Kane RL, Simpson IA, Viña JR. Structure of the blood-brain barrier and its role in the transport of amino acids. J Nutr 2006; 136:218S-26S. [PMID: 16365086 DOI: 10.1093/jn/136.1.218s] [Citation(s) in RCA: 300] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Brain capillary endothelial cells form the blood-brain barrier (BBB). They are connected by extensive tight junctions, and are polarized into luminal (blood-facing) and abluminal (brain-facing) plasma membrane domains. The polar distribution of transport proteins mediates amino acid (AA) homeostasis in the brain. The existence of two facilitative transporters for neutral amino acids (NAAs) on both membranes provides the brain access to essential AAs. Four Na(+)-dependent transporters of NAA exist in the abluminal membranes of the BBB. Together these systems have the capability to actively transfer every naturally occurring NAA from the extracellular fluid (ECF) to endothelial cells and from there into circulation. The presence of Na(+)-dependent carriers on the abluminal membrane provides a mechanism by which NAA concentrations in the ECF of brain are maintained at approximately 10% those of the plasma. Also present on the abluminal membrane are at least three Na(+)-dependent systems transporting acidic AAs (EAAT) and a Na(+)-dependent system transporting glutamine (N). Facilitative carriers for glutamine and glutamate are found only in the luminal membrane of the BBB. This organization promotes the net removal of acidic- and nitrogen-rich AAs from the brain and accounts for the low level of glutamate penetration into the central nervous system. The presence of a gamma-glutamyl cycle at the luminal membrane and Na(+)-dependent AA transporters at the abluminal membrane may serve to modulate movement of AAs from blood to the brain. The gamma-glutamyl cycle is expected to generate pyroglutamate (synonymous with oxyproline) within the endothelial cells. Pyroglutamate stimulates secondary active AA transporters at the abluminal membrane, thereby reducing the net influx of AAs to the brain. It is now clear that BBB participates in the active regulation of the AA content of the brain.
Collapse
Affiliation(s)
- Richard A Hawkins
- Department of Biochemistry and Molecular Biology, Facultad de Medicina y Farmacia,Universidad de Valencia,Valencia, Spain.
| | | | | | | |
Collapse
|
27
|
Amin MS, Wang HW, Reza E, Whitman SC, Tuana BS, Leenen FHH. Distribution of epithelial sodium channels and mineralocorticoid receptors in cardiovascular regulatory centers in rat brain. Am J Physiol Regul Integr Comp Physiol 2005; 289:R1787-97. [PMID: 16141309 DOI: 10.1152/ajpregu.00063.2005] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Epithelial sodium channels (ENaC) are important for regulating sodium transport across epithelia. Functional studies indicate that neural mechanisms acting through mineralocorticoid receptors (MR) and sodium channels (presumably ENaC) are crucial to the development of sympathoexcitation and hypertension in experimental models of salt-sensitive hypertension. However, expression and localization of the ENaC in cardiovascular regulatory centers of the brain have not yet been studied. RT-PCR and immunohistochemistry were performed to study ENaC and MR expression at the mRNA and protein levels, respectively. Both mRNA and protein for alpha-, beta-, and gamma-ENaC subunits and MR were found to be expressed in the rat brain. All three ENaC subunits and MR were present in the supraoptic nucleus, magnocellular paraventricular nucleus, hippocampus, choroid plexus, ependyma, and brain blood vessels, suggesting the presence of multimeric channels and possible regulation by mineralocorticoids. In most cortical areas, thalamus, amygdala, and suprachiasmatic nucleus, notable expression of gamma-ENaC was undetectable, whereas alpha- and beta-ENaC were abundantly expressed pointing to the possibility of a heterogeneous population of channels. The findings suggest that stoichiometrically different populations of ENaC may be present in both epithelial and neural components in the brain, which may contribute to regulation of cerebrospinal fluid and interstitial Na+ concentration as well as neuronal excitation.
Collapse
Affiliation(s)
- Md Shahrier Amin
- Hypertension Unit, Univ. of Ottawa Heart Institute, Ottawa, ON, Canada K1Y 4W7
| | | | | | | | | | | |
Collapse
|
28
|
Hawkins RA, Mokashi A, Simpson IA. An active transport system in the blood–brain barrier may reduce levodopa availability. Exp Neurol 2005; 195:267-71. [PMID: 15925365 DOI: 10.1016/j.expneurol.2005.04.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Revised: 04/11/2005] [Accepted: 04/13/2005] [Indexed: 11/28/2022]
Abstract
Levodopa, the primary drug used to treat patients with Parkinson's disease, is transported into the brain by the facilitative amino acid transporter (L1). We present here an unanticipated discovery: levodopa may be pumped out of the brain by a Na(+)-dependent transport system that couples the naturally occurring Na(+) gradient existing between the brain's extracellular fluid and the cytoplasm of capillary endothelial cells. The activity of this system reduces the net availability of levodopa.
Collapse
Affiliation(s)
- Richard A Hawkins
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.
| | | | | |
Collapse
|
29
|
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: 5.0] [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
|
30
|
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
|
31
|
McKay SJ, Johnsen R, Khattra J, Asano J, Baillie DL, Chan S, Dube N, Fang L, Goszczynski B, Ha E, Halfnight E, Hollebakken R, Huang P, Hung K, Jensen V, Jones SJM, Kai H, Li D, Mah A, Marra M, McGhee J, Newbury R, Pouzyrev A, Riddle DL, Sonnhammer E, Tian H, Tu D, Tyson JR, Vatcher G, Warner A, Wong K, Zhao Z, Moerman DG. Gene expression profiling of cells, tissues, and developmental stages of the nematode C. elegans. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2004; 68:159-69. [PMID: 15338614 DOI: 10.1101/sqb.2003.68.159] [Citation(s) in RCA: 239] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- S J McKay
- Genome Sciences Centre, BC Cancer Agency, Vancouver, B.C., Canada, V6T 1Z4
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
O'Kane RL, Viña JR, Simpson I, Hawkins RA. Na+ -dependent neutral amino acid transporters A, ASC, and N of the blood-brain barrier: mechanisms for neutral amino acid removal. Am J Physiol Endocrinol Metab 2004; 287:E622-9. [PMID: 15165996 DOI: 10.1152/ajpendo.00187.2004] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Four Na+ -dependent transporters of neutral amino acids (NAA) are known to exist in the abluminal membranes (brain side) of the blood-brain barrier (BBB). This article describes the kinetic characteristics of systems A, ASC, and N that, together with the recently described Na+ -dependent system for large NAA (Na+ -LNAA), provide a basis for understanding the functional organization of the BBB. The data demonstrate that system A is voltage dependent (3 positive charges accompany each molecule of substrate). Systems ASC and N are not voltage dependent. Each NAA is a putative substrate for at least one system, and several NAA are transported by as many as three. System A transports Pro, Ala, His, Asn, Ser, and Gln; system ASC transports Ser, Gly, Met, Val, Leu, Ile, Cys, and Thr; system N transports Gln, His, Ser, and Asn; Na+ -LNAA transports Leu, Ile, Val, Trp, Tyr, Phe, Met, Ala, His, Thr, and Gly. Together, these four systems have the capability to actively transfer every naturally occurring NAA from the extracellular fluid (ECF) to endothelial cells and thence to the circulation. The existence of facilitative transport for NAA (L1) on both membranes provides the brain access to essential NAA. The presence of Na+ -dependent carriers on the abluminal membrane provides a mechanism by which NAA concentrations in the ECF of brain are maintained at approximately 10% of those of the plasma.
Collapse
Affiliation(s)
- Robyn L O'Kane
- Department of Physiology and Biophysics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, IL 60064-3095, USA
| | | | | | | |
Collapse
|
33
|
Huang BS, Van Vliet BN, Leenen FHH. Increases in CSF [Na+] precede the increases in blood pressure in Dahl S rats and SHR on a high-salt diet. Am J Physiol Heart Circ Physiol 2004; 287:H1160-6. [PMID: 15130889 DOI: 10.1152/ajpheart.00126.2004] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In Dahl salt-sensitive (S) and salt-resistant (R) rats, and spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats, at 5-6 wk of age, a cannula was placed in the cisterna magna, and cerebrospinal fluid (CSF) was withdrawn continuously at 75 microl/12 h. CSF was collected as day- and nighttime samples from rats on a regular salt intake (0.6% Na+; R-Na) and then on a high salt intake (8% Na+; H-Na). In separate groups of rats, the abdominal aorta was cannulated and blood pressure (BP) and heart rate (HR) measured at 10 AM and 10 PM, with rats first on R-Na and then on H-Na. On H-Na, CSF [Na+] started to increase in the daytime of day 2 in Dahl S rats and of day 3 in SHR. BP and HR did not rise until day 3 in Dahl S rats and day 4 in SHR. In Dahl R and WKY rats, high salt did not change CSF [Na+], BP, or HR. In a third set of Dahl S rats, sampling of both CSF and BP was performed in each individual rat. Again, significant increases in CSF [Na+] were observed 1-2 days earlier than the increases in BP and HR. In a fourth set of Dahl S rats, BP and HR were recorded continuously by means of radiotelemetry for 5 days on R-Na and 8 days on H-Na. On H-Na, BP (but not HR) increased first in the nighttime of day 2. In another set of Dahl S rats, intracerebroventricular infusion of antibody Fab fragments binding ouabain-like compounds (OLC) with high affinity prevented the increase in BP and HR by H-Na but further increased CSF [Na+]. Finally, in Wistar rats on H-Na, intracerebroventricular infusion of ouabain increased BP and HR but decreased CSF [Na+]. Thus, in both Dahl S and SHR on H-Na, increases in CSF [Na+] preceded the increases in BP and HR, consistent with a primary role of increased CSF [Na+] in the salt-induced hypertension. An increase in brain OLC in response to the initial increase in CSF [Na+] appears to attenuate further increases in CSF [Na+] but at the "expense" of sympathoexcitation and hypertension.
Collapse
Affiliation(s)
- Bing S Huang
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada K1Y 4W7
| | | | | |
Collapse
|
34
|
Jiang C, Koyabu N, Yonemitsu Y, Shimazoe T, Watanabe S, Naito M, Tsuruo T, Ohtani H, Sawada Y. In vivo delivery of glial cell-derived neurotrophic factor across the blood-brain barrier by gene transfer into brain capillary endothelial cells. Hum Gene Ther 2003; 14:1181-91. [PMID: 12908969 DOI: 10.1089/104303403322168019] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The gene encoding mouse glial cell-derived neurotrophic factor (mGDNF gene) was transfected into brain capillary endothelial cells (BCECs) with the aim of delivering the gene product extensively into the brain parenchyma by making use of the secretory function of BCECs. First, we transfected mGDNF gene into cultured BCECs (MBEC4; mouse brain capillary endothelial cells) in vitro. The amount of mGDNF protein secreted from the transfected cells into the medium was 1500 to 3200 pg/mg of cell protein per day, being about sevenfold higher than that accumulated intracellularly. Furthermore, the basolateral-directed secretion of mGDNF protein from the transfected MBEC4 cells was fivefold higher than the apical-directed secretion. Next, the hemagglutination virus of Japan (HVJ)-liposomes encapsulating mGDNF gene were administered to rats in vivo via the internal carotid artery. The transfected rats showed a marked increase in the brain level of GDNF as assessed by means of enzyme-linked immunosorbent assay (ELISA) and Western blotting on day 3 after the administration, and the level remained significantly elevated for at least 12 days. Furthermore, immunohistochemical staining revealed an increase in GDNF immunoreactivity throughout the transfected forebrain. These results indicate that the gene was successfully transferred in vivo from HVJ-liposomes into BCECs, where it was expressed, and the gene product was secreted into the brain. Then, using this delivery method, we evaluated the protective effect for dopamine neuron against a retrograde 6-hydroxydopamine (6-OHDA) lesion, as assessed by behavioral and neurochemical indices.
Collapse
Affiliation(s)
- Chen Jiang
- Department of Medico-Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Huang BS, Leenen FHH. Sympathoinhibitory and depressor effects of amlodipine in spontaneously hypertensive rats. J Cardiovasc Pharmacol 2003; 42:153-60. [PMID: 12883316 DOI: 10.1097/00005344-200308000-00001] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The authors examined whether central actions contribute to the hypotensive effects of peripherally administered amlodipine, a lipophilic dihydropyridine with slow onset and long duration of action. After 5 to 6 weeks of high (8%, H-Na) or regular (0.6%, R-Na) salt intake, changes in renal sympathetic nerve activity (RSNA), mean arterial pressure (MAP), and heart rate (HR) were recorded at rest and in response to intravenous (iv) and intracerebroventricular (icv) injection, and prolonged iv infusion of amlodipine, in conscious spontaneously hypertensive rats (SHR). Iv injection of amlodipine at 50 to 100 microg/kg decreased MAP but increased RSNA and HR in a dose-related manner. In contrast, icv injection of amlodipine at 10 to 50 microg/kg caused parallel decreases in MAP, RSNA, and HR. Iv infusion of amlodipine at 50 microg/kg per hour for 3 hours followed by 100 microg/kg per hour for 2 hours also decreased in parallel RSNA, MAP, and HR. Maximal decreases in RSNA, MAP, and HR in response to icv injection and iv infusion were significantly larger in SHR on H-Na versus R-Na. All responses lasted at least 1 hour following iv and icv injection, or after the termination of iv infusion of amlodipine. These data suggest that in SHR during prolonged iv infusion, amlodipine appears to cross the blood-brain barrier, block brain l-type Ca2+ channels, and decrease sympathetic outflow and thereby BP. Central actions may prevail during iv infusion of amlodipine at low rates, and the decrease in BP is associated with sympathoinhibition. High salt intake markedly enhances its sympathoinhibitory action, likely through central mechanisms.
Collapse
Affiliation(s)
- Bing S Huang
- Hypertension Unit, University of Ottawa Heart Institute, H360, 40 Ruskin Street, Ottawa, Ontario K1Y 4W7, Canada
| | | |
Collapse
|
36
|
Tewes BJ, Galla HJ. Lipid polarity in brain capillary endothelial cells. ENDOTHELIUM : JOURNAL OF ENDOTHELIAL CELL RESEARCH 2003; 8:207-20. [PMID: 11824473 DOI: 10.1080/10623320109051566] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Brain capillary endothelial cells (BCEC) represent an epithelial like cell type with continuous tight junctions and polar distributed proteins. In this paper we investigated whether cultured BCEC show a polar distribution of membrane lipids as this was demonstrated for many epithelial cell types. Therefore we applied a high yield membrane fractionation method to isolate pure fractions of the apical and the basolateral plasma membrane (PM) domains. Using a set of methods for lipid analysis we were able to determine the total lipid composition of the whole cells and the PM fractions. Both membrane domains showed a unique lipid composition with clear differences to each other and to the whole cell composition. Three lipid species were polar distributed between the two PM domains. Phosphatidylcholine was enriched in the apical membrane whereas sphingomyelin and glucosylceramide were enriched in the basolateral membrane. The possible function of this lipid polarity for the blood-brain barrier mechanism is the generation of a suitable lipid environment for polar distributed membrane proteins and the generation of two PM domains with different biophysical properties and permeabilities.
Collapse
Affiliation(s)
- B J Tewes
- Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Germany
| | | |
Collapse
|
37
|
Copin JC, Gasche Y. [Morphology and physiology of the blood-brain barrier]. ANNALES FRANCAISES D'ANESTHESIE ET DE REANIMATION 2003; 22:202-14. [PMID: 12747988 DOI: 10.1016/s0750-7658(03)00040-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The blood-brain barrier (BBB) is a complex biological system that consists of endothelial cells, pericytes and astrocytes, which are involved in the induction and maintenance of its physiological and ultrastructural characteristics. The BBB plays a primordial role in isolating the cerebral parenchyma as well as in controlling brain homeostasis by its selective permeability to nutriments and other molecules flowing through the cerebral microcapillaries. A better knowledge of this system is crucial in order to improve the efficiency of brain penetration by drugs, and in order to prevent BBB opening, leading to brain edema, in physiopathological situations such as brain ischemia, trauma or inflammatory processes.
Collapse
Affiliation(s)
- J C Copin
- Divisions des soins intensifs de chirurgie et de médecine, division d'investigations anesthésiologiques, hôpitaux universitaires de Genève, 1211 Genève, Suisse.
| | | |
Collapse
|
38
|
Librizzi L, Janigro D, De Biasi S, de Curtis M. Blood-brain barrier preservation in the in vitro isolated guinea pig brain preparation. J Neurosci Res 2001; 66:289-97. [PMID: 11592126 DOI: 10.1002/jnr.1223] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The morphofunctional preservation of the blood-brain barrier (BBB) was evaluated in the isolated guinea pig brain maintained in vitro by arterial perfusion. Electron microscopy evaluation after 5 hr in vitro demonstrated that cerebral capillaries and BBB specializations in this preparation retain features compatible with structural integrity. BBB-impermeable and -permeable atropine derivatives arterially perfused to antagonize carbachol-induced fast oscillatory activity confirmed the functional preservation of the BBB in vitro. To study BBB function further, changes in extracellular K+ concentration during arterial perfusion of a high-K+ solution were measured with K+-sensitive electrodes positioned in the cortex and, as control, at the brain venous outlet, where the solution perfused through the brain arterial system was collected. After 5 hr in vitro, the [K+](o) values measured during high-K+ perfusion in the piriform and entorhinal cortices were 5.02 +/- 0.17 mM (mean +/- SE) and 5.2 +/- 0.21 mM, respectively (n = 6). Coperfusion of the high-K+ solution with the Na+/K+ pump blocker ouabain (10 microM; n = 4) induced consistently spreading depression preceded by a rise in [K+](o). Finally, sporadic, isolated spots of extravasation of the fluorescent marker fluorescein isothiocyanate (FITC)-dextran preferentially circumscribed to deep cortical layers was observed in brains perfused with FITC-dextran after 5 hr in vitro. The study demonstrates that the in vitro isolated guinea pig brain is viable for studying cerebrovascular interactions and BBB permeability of compounds active in the central nervous system.
Collapse
Affiliation(s)
- L Librizzi
- Department of Neurophysiology, Istituto Nazionale Neurologico, via Celoria 11, 20133 Milan, Italy
| | | | | | | |
Collapse
|
39
|
Cardelli P, Fiori A, Corleto VD, Savi MR, Granata F, Ceci F, Ferraguti G, Potenza RL, Delle Fave G, Jensen RT, Strom R. Inhibitory effect of somatostatin on neutral amino acid transport in isolated brain microvessels. J Neurochem 2001; 78:349-57. [PMID: 11461970 DOI: 10.1046/j.1471-4159.2001.00412.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the presence of somatostatin-14 or some of its receptorial agonists, the uptake of large neutral amino acids by isolated brain microvessels was found to be inhibited up to 50%, no other transport system being affected. Although the luminal and abluminal sides of brain endothelial cells are both capable of taking up large neutral amino acids, only uptake from the abluminal side appears to be inhibited by somatostatin. The involvement of a type-2 somatostatin receptor was suggested by assays with a series of receptor-specific somatostatin agonists, and was confirmed by the release of inhibition caused by a specific type-2 receptor antagonist. A type-2-specific mRNA was indeed shown to be present in both bovine brain microvessels ex vivo and primary cultures of endothelial cells from rat brain microvessels.
Collapse
Affiliation(s)
- P Cardelli
- Department of Cellular Biotechnology and Haematology, University 'La Sapienza', Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Pathological states (i.e. stroke, cardiac arrest) can lead to reduced blood flow to the brain potentially altering blood-brain barrier (BBB) permeability and regulatory transport functions. BBB disruption leads to increased cerebrovascular permeability, an important factor in the development of ischemic brain injury and edema formation. In this study, reduced flow was investigated to determine the effects on cerebral blood flow (CBF), pressure, basal BBB permeability, and transport of insulin and K+ across the BBB. Anesthetized adult female Sprague-Dawley rats were measured at normal flow (3.1 ml min(-1)), half flow (1.5 ml min(-1)), and quarter flow (0.75 ml min(-1)), using bilateral in situ brain perfusion for 20 min followed by capillary depletion analysis. Reduction in perfusion flow rates demonstrated a modest reduction in CBF (1.27-1.56 ml min(-1) g(-1)), a decrease in pressure, and no significant effect on basal BBB permeability indicating that autoregulation remained functional. In contrast, there was a concomittant decrease in BBB transport of both insulin and K+ with reduced flow. At half and quarter flow, insulin transport was significantly reduced (R(Br)%=17.2 and R(Br)%=16.2, respectively) from control (R(Br)%=30.4). Additionally, a significant reduction in [86Rb+] was observed at quarter flow (R(Br)%=2.5) as compared to control (R(Br)%=4.8) suggesting an alteration in ion homeostasis as a result of low flow. This investigation suggests that although autoregulation maintains CBF, BBB transport mechanisms were significantly compromised in states of reduced flow. These flow alterations may have a significant impact on brain homeostasis in pathological states.
Collapse
Affiliation(s)
- S Hom
- Department of Pharmacology, University of Arizona, College of Medicine, Tucson, AZ 85724, USA
| | | | | | | |
Collapse
|
41
|
Bayol-Denizot C, Daval JL, Netter P, Minn A. Xenobiotic-mediated production of superoxide by primary cultures of rat cerebral endothelial cells, astrocytes, and neurones. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1497:115-26. [PMID: 10838165 DOI: 10.1016/s0167-4889(00)00047-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Previous works of our group demonstrated that xenobiotic metabolism by brain microsomes or cultured cerebral cells may promote the formation of reactive oxygen species. In order to characterise the risk of oxidative stress to both the central nervous system and the blood-brain barrier, we measured in the present work the release of superoxide in the culture medium of rat cerebrovascular endothelial cells during the metabolism of menadione, anthraquinone, diquat or nitrofurazone. Assays were run in the same experimental conditions on primary cultures of rat neurones and astrocytes. Quinone metabolism efficiently produced superoxide, but the production of radicals during the metabolism of diquat or nitrofurazone was very low, as a probable result of their reduced transport inside the cells. In all cell types assayed, superoxide production was time- and concentration-dependent, and cultured astrocytes always produced the highest amounts of radicals. Superoxide formation by microsomes prepared from the cultured cells was decreased by immunoinhibition of NADPH-cytochrome P450 reductase or by its irreversible inhibition by diphenyliodonium chloride, suggesting the involvement of this flavoprotein in radical production. Cerebrovascular endothelial cells cultured on collagen-coated filters produced equivalent amounts of superoxide both at their luminal side and through the artificial basement membrane, suggesting that in vivo, endothelial superoxide production may endanger adjacent astrocytes and neurones.
Collapse
Affiliation(s)
- C Bayol-Denizot
- UMR CNRS-Universit¿e Henri Poincar¿e-Nancy 1 No 7561, Laboratoire de Pharmacologie, Facult¿e de M¿edecine, Vandoeuvre-l¿es-Nancy, France
| | | | | | | |
Collapse
|
42
|
Manoonkitiwongsa PS, Schultz RL, Wareesangtip W, Whitter EF, Nava PB, McMillan PJ. Luminal localization of blood-brain barrier sodium, potassium adenosine triphosphatase is dependent on fixation. J Histochem Cytochem 2000; 48:859-65. [PMID: 10820159 DOI: 10.1177/002215540004800614] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cytochemical data in the literature reporting localization of sodium, potassium adenosine triphosphatase (Na(+), K(+)-ATPase) in the blood-brain barrier (BBB) have been contradictory. Whereas some studies showed the enzyme to be located exclusively on the abluminal endothelial plasma membrane, others demonstrated it on both the luminal and abluminal membranes. The influence of fixation on localization of the enzyme was not considered a critical factor, but our preliminary studies showed data to the contrary. We therefore quantitatively investigated the effect of commonly used fixatives on the localization pattern of the enzyme in adult rat cerebral microvessels. Fixation with 1%, 2%, and 4% formaldehyde allowed deposition of reaction product on both the luminal and abluminal plasma membranes. The luminal reaction was reduced with increasing concentration of formaldehyde. Glutaraldehyde at 0.1%, 0.25%, 0.5%, in combination with 2% formaldehyde, drastically inhibited the luminal reaction. The abluminal reaction was not significantly altered in all groups. These results show that luminal localization of BBB Na(+), K(+)-ATPase is strongly dependent on fixation. The lack of luminal localization, as reported in the literature, may have been the result of fixation. The currently accepted abluminal polarity of the enzyme should be viewed with caution.
Collapse
Affiliation(s)
- P S Manoonkitiwongsa
- Department of Pathology and Human Anatomy, Division of Human Anatomy, Loma Linda University, Loma Linda, California 92350, USA
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
The blood-brain barrier (BBB) which is located in the continuous endothelial lining of cerebral blood vessels rigidly controls exchange of water soluble compounds under physiological conditions. Under pathological conditions such as trauma or ischemia, BBB permeability may increase thus allowing plasma constituents to escape into brain tissue. This "opening" of the BBB may, at least in part, be mediated by massive release of autacoids resulting in vasogenic brain edema. Five criteria have to be fulfilled by an individual autacoid to be considered a mediator candidate of cerebral edema: i) a permeability-enhancing action under physiological conditions, ii) a vasodilatory action, iii) the ability to induce vasogenic brain edema, iv) an increase of concentration in the tissue or interstitial fluid under pathological conditions, and v) a decrease of brain edema by specific interference with the release or action of a given autacoid. Among the mediator candidates considered, bradykinin is the only one to meet all criteria. Histamine, arachidonic acid and free radicals including nitric oxide may also be considered mediators of brain edema, but for each of these compounds evidence is less clear than for bradykinin. Although the concept of mediators inducing brain edema is well established by experimental studies, only a bradykinin receptor antagonist has so far gained entrance into clinical evaluation.
Collapse
Affiliation(s)
- L Schilling
- Dept. Neurosurg., Fac. Clin. Med., Mannheim, Univ. Heidelberg, Germany
| | | |
Collapse
|
44
|
Mroczkowska JE, Roux FS, Naleçz MJ, Naleçz KA. Blood-brain barrier controls carnitine level in the brain: a study on a model system with RBE4 cells. Biochem Biophys Res Commun 2000; 267:433-7. [PMID: 10623637 DOI: 10.1006/bbrc.1999.1923] [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: 11/22/2022]
Abstract
Transport of carnitine was studied with immortalized rat brain endothelial cells (RBE4), an in vitro model of the blood-brain barrier. The experiments on uptake and efflux through the luminal membrane excluded any involvement of choline and amino acids transporters, as well as that of glycoprotein P. Acetyl-, octanoylcarnitine, and betaine were without any effect; the only compound decreasing both processes was butyrobetaine. An exposure of the abluminal membrane resulted in a 40% inhibition of carnitine uptake by the substrates of neutral amino acid transporter L, while its efflux through the basolateral membrane, occurring in a form of free carnitine, was sensitive to SH group reagent, mersalyl, and was diminished by butyrobetaine. These features of carnitine transport did not fully correspond to the known characteristics of the proteins transporting carnitine in other tissues (OCTN2 and CT1); however, they did not exclude an involvement of a transporter belonging to the same superfamily. Moreover, such a protein in brain endothelium would fulfill a regulatory role in the transport of carnitine through the blood-brain barrier.
Collapse
Affiliation(s)
- J E Mroczkowska
- Nencki Institute of Experimental Biology, 3 Pasteur Street, Warsaw, 02-093, Poland
| | | | | | | |
Collapse
|
45
|
Flores LG, Kawai K, Nakagawa M, Shikano N, Jinnouchi S, Tamura S, Watanabe K, Kubodera A. A new radiopharmaceutical for the cerebral dopaminergic presynaptic function: 6-radioiodinated L-meta-tyrosine. J Cereb Blood Flow Metab 2000; 20:207-12. [PMID: 10616810 DOI: 10.1097/00004647-200001000-00026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Geometric isomers of radioiodinated L-meta-tyrosine, 6-[I-125]iodo-and 4-[1-125]iodo-L-meta-tyrosine (6-I-L-mTyr, 4-I-L-mTyr) were separated by high-performance liquid chromatography . Both 6-I- and 4-I-L-mTyr had high energy-dependent brain accumulation. 6-I- and 4-I-L-mTyr were also metabolically stable and were rapidly excreted through the urine. 6-I-L-mTyr had a predilection for the cerebral aromatic L-amino acid decarboxylase (DOPA decarboxylase), the final enzyme of dopamine biosynthesis. 6-Radioiodinated L-mTyr is a new radiopharmaceutical that can be both useful in assessing cerebral amino acid transport mechanism and quantifying metabolically active DOPA decarboxylase.
Collapse
Affiliation(s)
- L G Flores
- Department of Radiology, Miyazaki Medical College, Miyazaki-gun, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Zhang Y, Liu G. A novel method to determine the localization of high and low-affinity GABA transporters to the luminal and antiluminal membranes of brain capillary endothelial cells. BRAIN RESEARCH. BRAIN RESEARCH PROTOCOLS 1999; 4:288-94. [PMID: 10592337 DOI: 10.1016/s1385-299x(99)00031-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Some evidence has shown that the luminal plasma membrane and the antiluminal plasma membrane of the blood-brain barrier (BBB) are functionally distinct. This polarity permits brain capillary endothelial cells (BCECs) to actively regulate the internal milieu of the brain. Especially, polarity also exists concerning the transport of some endogenous substances such as amino acids and glucose. It is important to determine the luminal and antiluminal membrane localization of some transporters of endogenous substances, which will contribute to a better understanding of the transport mechanisms of the endogenous substances at the BBB. The present paper describes a novel procedure for combining isolated brain capillaries with primary cultured BCECs to determine the subcellular localization of some transporters, which is relatively simpler than the methods described previously. This method was used for the first time to determine successfully the luminal and antiluminal distribution of high and low-affinity GABA transporters. The low-affinity GABA transporter is probably localized to the luminal membrane of BCECs, and the high-affinity GABA transporter may be localized to the antiluminal membrane.
Collapse
Affiliation(s)
- Y Zhang
- Department of Pharmacology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | | |
Collapse
|
47
|
Jäger M, Wolf S. Localization of 5-oxo-L-prolinase mRNA in the murine choroid plexus by in situ hybridization. Neurosci Lett 1999; 274:171-4. [PMID: 10548417 DOI: 10.1016/s0304-3940(99)00718-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The gamma-glutamyl cycle is thought to play a significant role in the transport of amino acids between the blood and the cerebrospinal fluid. In the present study we investigated the expression of 5-oxo-L-prolinase, an enzyme of this pathway, in mouse brain and kidney by in situ hybridization using digoxygenin-labeled RNA probes. Expression of 5-OPase mRNA was found in epithelial cells of the proximal tubules in kidney as well as in the epithelium and vascular endothelial cells of the choroid plexus but not in periventricular capillaries.
Collapse
Affiliation(s)
- M Jäger
- Max-Planck-Institut für Immunbiologie, Freiburg, Germany
| | | |
Collapse
|
48
|
Keep RF, Ulanski LJ, Xiang J, Ennis SR, Lorris Betz A. Blood-brain barrier mechanisms involved in brain calcium and potassium homeostasis. Brain Res 1999; 815:200-5. [PMID: 9878735 DOI: 10.1016/s0006-8993(98)01155-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
This study examined the potential roles of the plasma membrane Ca2+-ATPase (PMCA) at the blood-CSF and blood-brain barriers in brain Ca2+ homeostasis and blood-brain barrier Na+/K+-ATPase subunits in brain K+ homeostasis. During dietary-induced hypo- and hypercalcemia (0.59+/-0.06 and 1.58+/-0.12 mM [Ca2+]) there was no significant change in choroid plexus PMCA (Western Blots) compared to normocalcemic rats (plasma [Ca2+]: 1.06+/-0.11 mM). In contrast, PMCA in cerebral microvessels isolated from hypocalcemic rats was 150% greater than that in controls (p<0.001). Comparison of the alpha3 subunit of Na+/K+-ATPase from cerebral microvessels isolated from hypo-, normo- and hyperkalemic rats (2.3+/-0.1, 3.9+/-0.1 and 7. 2+/-0.6 mM [K+]) showed a 75% reduction in the amount of this isoform during hyperkalemia. None of the other Na+/K+-ATPase isoforms varied with plasma [K+]. These results suggest that both PMCA and the alpha3 subunit of Na+/K+-ATPase at the blood-brain barrier play a role in maintaining a constant brain microenvironment during fluctuations in plasma composition.
Collapse
Affiliation(s)
- R F Keep
- Department of Surgery (Neurosurgery), University of Michigan, R5605 Kresge I, Ann Arbor, MI 48109-0532,
| | | | | | | | | |
Collapse
|
49
|
Pontiggia L, Winterhalter K, Gloor SM. Inhibition of Na,K-ATPase activity by cGMP is isoform-specific in brain endothelial cells. FEBS Lett 1998; 436:466-70. [PMID: 9801170 DOI: 10.1016/s0014-5793(98)01183-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
cGMP has been shown to either activate or inhibit Na,K-ATPase activity. Using mouse brain endothelial cells which express both ouabain-resistant alpha1 and ouabain-sensitive alpha2 and alpha3 isoforms, we show that cGMP reduces total Na,KATPase activity to about 58%. The inhibition is prevented by the protein kinase G (PKG)-specific inhibitor KT5823, indicating that cGMP-mediated activation of PKG leads to inhibition of the pump. A similar extent of inhibition is obtained with nitric oxide. cGMP-induced inhibition acts mainly on alpha1 isoforms but hardly affects alpha2/alpha3 isoforms. These data suggest that inhibition of Na,K-ATPase activity by cGMP occurs in an isoform-selective manner in brain endothelial cells.
Collapse
Affiliation(s)
- L Pontiggia
- Biochemistry I, Swiss Federal Institute of Technology, ETH Zentrum, Zürich
| | | | | |
Collapse
|
50
|
Manoonkitiwongsa PS, Whitter EF, Schultz RL. An in situ cytochemical evaluation of blood-brain barrier sodium, potassium-activated adenosine triphosphatase polarity. Brain Res 1998; 798:261-70. [PMID: 9666144 DOI: 10.1016/s0006-8993(98)00426-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is presently believed that sodium, potassium-activated adenosine triphosphatase (Na+, K+-ATPase) is localized on the abluminal plasma membrane of brain endothelial cells. But there have been contrary reports from some cytochemical studies. We examined the localization of the enzyme in rat cerebral microvessel endothelium using the in situ model originally employed to establish the abluminal polarity concept. Alterations in fixation and incubation media from the original reports were conducted to determine the effect on localization pattern. With the Ernst indirect incubation method as originally used, three types of localization patterns were obtained: abluminal only, luminal only, and on both surfaces of endothelial cells. With the direct incubation method of Mayahara, reaction product was seen on both surfaces. Reduction in fixation time followed by the use of the indirect incubation method resulted in a complete loss of the reaction product. The same reduction in fixation time followed by the use of the direct method did not alter the localization pattern of the enzyme. Our results demonstrated that Na+, K+-ATPase is localized on both surfaces of brain endothelial cells. The localization pattern of Na+, K+-ATPase is significantly dependent upon fixation and the incubation medium used in the in situ model. Data discrepancies for the enzyme as reported in the literature appear to be caused by differences in cytochemical protocols, rather than the biological reasons advocated by other investigators. We conclude that past cytochemical reports of blood-brain barrier (BBB) Na+, K+-ATPase abluminal localization were incomplete. The currently held abluminal polarity theory of the enzyme needs to be reexamined. Past basic and clinical cytochemical studies of BBB Na+, K+-ATPase should be viewed and interpreted with caution.
Collapse
Affiliation(s)
- P S Manoonkitiwongsa
- Department of Pathology and Human Anatomy, Division of Human Anatomy, Loma Linda University, Loma Linda, CA 92350, USA
| | | | | |
Collapse
|