1
|
Loss of Blood-Brain Barrier Integrity in an In Vitro Model Subjected to Intermittent Hypoxia: Is Reversion Possible with a HIF-1α Pathway Inhibitor? Int J Mol Sci 2023; 24:ijms24055062. [PMID: 36902491 PMCID: PMC10003655 DOI: 10.3390/ijms24055062] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/11/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Several sleep-related breathing disorders provoke repeated hypoxia stresses, which potentially lead to neurological diseases, such as cognitive impairment. Nevertheless, consequences of repeated intermittent hypoxia on the blood-brain barrier (BBB) are less recognized. This study compared two methods of intermittent hypoxia induction on the cerebral endothelium of the BBB: one using hydralazine and the other using a hypoxia chamber. These cycles were performed on an endothelial cell and astrocyte coculture model. Na-Fl permeability, tight junction protein, and ABC transporters (P-gp and MRP-1) content were evaluated with or without HIF-1 inhibitors YC-1. Our results demonstrated that hydralazine as well as intermittent physical hypoxia progressively altered BBB integrity, as shown by an increase in Na-Fl permeability. This alteration was accompanied by a decrease in concentration of tight junction proteins ZO-1 and claudin-5. In turn, microvascular endothelial cells up-regulated the expression of P-gp and MRP-1. An alteration was also found under hydralazine after the third cycle. On the other hand, the third intermittent hypoxia exposure showed a preservation of BBB characteristics. Furthermore, inhibition of HIF-1α with YC-1 prevented BBB dysfunction after hydralazine treatment. In the case of physical intermittent hypoxia, we observed an incomplete reversion suggesting that other biological mechanisms may be involved in BBB dysfunction. In conclusion, intermittent hypoxia led to an alteration of the BBB model with an adaptation observed after the third cycle.
Collapse
|
2
|
Bagatella S, Haghayegh Jahromi N, Monney C, Polidori M, Gall FM, Marchionatti E, Serra F, Riedl R, Engelhardt B, Oevermann A. Bovine neutrophil chemotaxis to Listeria monocytogenes in neurolisteriosis depends on microglia-released rather than bacterial factors. J Neuroinflammation 2022; 19:304. [PMID: 36527076 PMCID: PMC9758797 DOI: 10.1186/s12974-022-02653-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Listeria monocytogenes (Lm) is a bacterial pathogen of major concern for humans and ruminants due to its neuroinvasive potential and its ability to cause deadly encephalitis (neurolisteriosis). On one hand, polymorphonuclear neutrophils (PMN) are key players in the defense against Lm, but on the other hand intracerebral infiltration with PMN is associated with significant neural tissue damage. Lm-PMN interactions in neurolisteriosis are poorly investigated, and factors inducing PMN chemotaxis to infectious foci containing Lm in the central nervous system (CNS) remain unidentified. METHODS In this study, we assessed bovine PMN chemotaxis towards Lm and supernatants of infected endogenous brain cell populations in ex vivo chemotaxis assays, to identify chemotactic stimuli for PMN chemotaxis towards Lm in the brain. In addition, microglial secretion of IL-8 was assessed both ex vivo and in situ. RESULTS Our data show that neither Lm cell wall components nor intact bacteria elicit chemotaxis of bovine PMN ex vivo. Moreover, astrocytes and neural cells fail to induce bovine PMN chemotaxis upon infection. In contrast, supernatant from Lm infected microglia readily induced chemotaxis of bovine PMN. Microglial expression and secretion of IL-8 was identified during early Lm infection in vitro and in situ, although IL-8 blocking with a specific antibody could not abrogate PMN chemotaxis towards Lm infected microglial supernatant. CONCLUSIONS These data provide evidence that host-derived rather than bacterial factors trigger PMN chemotaxis to bacterial foci in the CNS, that microglia have a primary role as initiators of bovine PMN chemotaxis into the brain during neurolisteriosis and that blockade of these factors could be a therapeutic target to limit intrathecal PMN chemotaxis and PMN associated damage in neurolisteriosis.
Collapse
Affiliation(s)
- Stefano Bagatella
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland ,grid.5734.50000 0001 0726 5157Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Neda Haghayegh Jahromi
- grid.5734.50000 0001 0726 5157Theodor Kocher Institute (TKI), University of Bern, Bern, Switzerland
| | - Camille Monney
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland
| | - Margherita Polidori
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland ,grid.5734.50000 0001 0726 5157Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Flavio Max Gall
- grid.19739.350000000122291644Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | - Emma Marchionatti
- grid.5734.50000 0001 0726 5157Clinic for Ruminants, Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Rainer Riedl
- grid.19739.350000000122291644Institute of Chemistry and Biotechnology, Competence Center for Drug Discovery, Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | - Britta Engelhardt
- grid.5734.50000 0001 0726 5157Theodor Kocher Institute (TKI), University of Bern, Bern, Switzerland
| | - Anna Oevermann
- grid.5734.50000 0001 0726 5157Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, CH-3012 Bern, Switzerland
| |
Collapse
|
3
|
Versele R, Sevin E, Gosselet F, Fenart L, Candela P. TNF-α and IL-1β Modulate Blood-Brain Barrier Permeability and Decrease Amyloid-β Peptide Efflux in a Human Blood-Brain Barrier Model. Int J Mol Sci 2022; 23:ijms231810235. [PMID: 36142143 PMCID: PMC9499506 DOI: 10.3390/ijms231810235] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The blood-brain barrier (BBB) is a selective barrier and a functional gatekeeper for the central nervous system (CNS), essential for maintaining brain homeostasis. The BBB is composed of specialized brain endothelial cells (BECs) lining the brain capillaries. The tight junctions formed by BECs regulate paracellular transport, whereas transcellular transport is regulated by specialized transporters, pumps and receptors. Cytokine-induced neuroinflammation, such as the tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), appear to play a role in BBB dysfunction and contribute to the progression of Alzheimer’s disease (AD) by contributing to amyloid-β (Aβ) peptide accumulation. Here, we investigated whether TNF-α and IL-1β modulate the permeability of the BBB and alter Aβ peptide transport across BECs. We used a human BBB in vitro model based on the use of brain-like endothelial cells (BLECs) obtained from endothelial cells derived from CD34+ stem cells cocultivated with brain pericytes. We demonstrated that TNF-α and IL-1β differentially induced changes in BLECs’ permeability by inducing alterations in the organization of junctional complexes as well as in transcelluar trafficking. Further, TNF-α and IL-1β act directly on BLECs by decreasing LRP1 and BCRP protein expression as well as the specific efflux of Aβ peptide. These results provide mechanisms by which CNS inflammation might modulate BBB permeability and promote Aβ peptide accumulation. A future therapeutic intervention targeting vascular inflammation at the BBB may have the therapeutic potential to slow down the progression of AD.
Collapse
Affiliation(s)
- Romain Versele
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Emmanuel Sevin
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
| | - Laurence Fenart
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
| | - Pietra Candela
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), UR 2465, Université d’Artois, F-62300 Lens, France
- Correspondence:
| |
Collapse
|
4
|
Jamieson JJ, Lin Y, Malloy N, Soto D, Searson PC, Gerecht S. Hypoxia-induced blood-brain barrier dysfunction is prevented by pericyte-conditioned media via attenuated actomyosin contractility and claudin-5 stabilization. FASEB J 2022; 36:e22331. [PMID: 35476363 PMCID: PMC9060394 DOI: 10.1096/fj.202200010rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/12/2022] [Accepted: 04/15/2022] [Indexed: 11/11/2022]
Abstract
The blood-brain barrier (BBB) regulates molecular and cellular entry from the cerebrovasculature into the surrounding brain parenchyma. Many diseases of the brain are associated with dysfunction of the BBB, where hypoxia is a common stressor. However, the contribution of hypoxia to BBB dysfunction is challenging to study due to the complexity of the brain microenvironment. In this study, we used a BBB model with brain microvascular endothelial cells and pericytes differentiated from iPSCs to investigate the effect of hypoxia on barrier function. We found that hypoxia-induced barrier dysfunction is dependent upon increased actomyosin contractility and is associated with increased fibronectin fibrillogenesis. We propose a role for actomyosin contractility in mediating hypoxia-induced barrier dysfunction through modulation of junctional claudin-5. Our findings suggest pericytes may protect brain microvascular endothelial cells from hypoxic stresses and that pericyte-derived factors could be candidates for treatment of pathological barrier-forming tissues.
Collapse
Affiliation(s)
- John J Jamieson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - YingYu Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicholas Malloy
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel Soto
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Biomedical Engineering, Duke University, Duke, North Carolina, USA
| |
Collapse
|
5
|
Hypoxia increases expression of selected blood-brain barrier transporters GLUT-1, P-gp, SLC7A5 and TFRC, while maintaining barrier integrity, in brain capillary endothelial monolayers. Fluids Barriers CNS 2022; 19:1. [PMID: 34983574 PMCID: PMC8725498 DOI: 10.1186/s12987-021-00297-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/15/2021] [Indexed: 01/01/2023] Open
Abstract
Background Brain capillary endothelial cells (BCECs) experience hypoxic conditions during early brain development. The newly formed capillaries are tight and functional before astrocytes and pericytes join the capillaries and establish the neurovascular unit. Brain endothelial cell phenotype markers P-gp (ABCB1), LAT-1(SLC7A5), GLUT-1(SLC2A1), and TFR(TFRC) have all been described to be hypoxia sensitive. Therefore, we hypothesized that monolayers of BCECs, cultured under hypoxic conditions, would show an increase in LAT-1, GLUT-1 and TFR expression and display tight endothelial barriers. Methods and results Primary bovine BCECs were cultured under normoxic and hypoxic conditions. Chronic hypoxia induced HIF-1α stabilization and translocation to the nucleus, as judged by immunocytochemistry and confocal laser scanning imaging. Endothelial cell morphology, claudin-5 and ZO-1 localization and barrier integrity were unaffected by hypoxia, indicating that the tight junctions in the BBB model were not compromised. SLC7A5, SLC2A1, and TFRC-mRNA levels were increased in hypoxic cultures, while ABCB1 remained unchanged as shown by real-time qPCR. P-gp, TfR and GLUT-1 were found to be significantly increased at protein levels. An increase in uptake of [3H]-glucose was demonstrated, while a non-significant increase in the efflux ratio of the P-gp substrate [3H]-digoxin was observed in hypoxic cells. No changes were observed in functional LAT-1 as judged by uptake studies of [3H]-leucine. Stabilization of HIF-1α under normoxic conditions with desferrioxamine (DFO) mimicked the effects of hypoxia on endothelial cells. Furthermore, low concentrations of DFO caused an increase in transendothelial electrical resistance (TEER), suggesting that a slight activation of the HIF-1α system may actually increase brain endothelial monolayer tightness. Moreover, exposure of confluent monolayers to hypoxia resulted in markedly increase in TEER after 24 and 48 h, which corresponded to a higher transcript level of CLDN5. Conclusions Our findings collectively suggest that hypoxic conditions increase some BBB transporters' expression via HIF-1α stabilization, without compromising monolayer integrity. This may in part explain why brain capillaries show early maturation, in terms of barrier tightness and protein expression, during embryogenesis, and provides a novel methodological tool for optimal brain endothelial culture. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00297-6.
Collapse
|
6
|
Abstract
Human immunodeficiency virus type-1(HIV-1)-associated neurocognitive disorder (HAND) remains an important neurological manifestation in HIV-1-infected (HIV+) patients. Furthermore, the HIV-1 matrix protein p17 (p17) detection in the central nervous system (CNS) and its ability to form toxic assemblies in the brain has been recently confirmed. Here we show for the first time using both an in vitro blood-brain barrier (BBB) model and in vivo biodistribution studies in healthy mice that p17 can cross the BBB. There is fast brain uptake with 0.35 ± 0.19% of injected activity per gram of tissue (I.A./g) two minutes after administration, followed by brain accumulation with 0.28 ± 0.09% I.A./g after 1 h. The interaction of p17 with the chemokine receptor 2 (CXCR2) at the surface of brain endothelial cells triggers transcytosis. The present study supports the hypothesis of a direct role of free p17 in neuronal dysfunction in HAND by demonstrating its intrinsic ability to reach the CNS. IMPORTANCE The number of patients affected by HIV-1-associated neurocognitive disorder (HAND) ranges from 30 to 50% of HIV-infected (HIV+) patients. The mechanisms leading to HAND development need to be elucidated, but the role of secreted viral proteins, chemokines, and proinflammatory molecules appears to be clear. In particular, the blood-brain barrier (BBB) represents a route for entry into the central nervous system (CNS) thus playing an important role in HAND. Several findings suggest a key role for the HIV-1 matrix protein p17 (p17) as a microenvironmental factor capable of inducing neurocognitive disorders. Here we show, the ability of the p17 to cross the BBB and to reach the CNS thus playing a crucial role in neuronal dysfunction in HAND.
Collapse
|
7
|
Yang Y, Torbey MT. Angiogenesis and Blood-Brain Barrier Permeability in Vascular Remodeling after Stroke. Curr Neuropharmacol 2020; 18:1250-1265. [PMID: 32691713 PMCID: PMC7770645 DOI: 10.2174/1570159x18666200720173316] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/27/2020] [Accepted: 07/11/2020] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis, the growth of new blood vessels, is a natural defense mechanism helping to restore oxygen and nutrient supply to the affected brain tissue following an ischemic stroke. By stimulating vessel growth, angiogenesis may stabilize brain perfusion, thereby promoting neuronal survival, brain plasticity, and neurologic recovery. However, therapeutic angiogenesis after stroke faces challenges: new angiogenesis-induced vessels have a higher than normal permeability, and treatment to promote angiogenesis may exacerbate outcomes in stroke patients. The development of therapies requires elucidation of the precise cellular and molecular basis of the disease. Microenvironment homeostasis of the central nervous system is essential for its normal function and is maintained by the blood-brain barrier (BBB). Tight junction proteins (TJP) form the tight junction (TJ) between vascular endothelial cells (ECs) and play a key role in regulating the BBB permeability. We demonstrated that after stroke, new angiogenesis-induced vessels in peri-infarct areas have abnormally high BBB permeability due to a lack of major TJPs in ECs. Therefore, promoting TJ formation and BBB integrity in the new vessels coupled with speedy angiogenesis will provide a promising and safer treatment strategy for improving recovery from stroke. Pericyte is a central neurovascular unite component in vascular barriergenesis and are vital to BBB integrity. We found that pericytes also play a key role in stroke-induced angiogenesis and TJ formation in the newly formed vessels. Based on these findings, in this article, we focus on regulation aspects of the BBB functions and describe cellular and molecular special features of TJ formation with an emphasis on role of pericytes in BBB integrity during angiogenesis after stroke.
Collapse
Affiliation(s)
- Yi Yang
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| | - Michel T Torbey
- Department of Neurology, University of New Mexico Health Sciences Center; Albuquerque, New Mexico, 87131, United States
| |
Collapse
|
8
|
GM1 Oligosaccharide Crosses the Human Blood-Brain Barrier In Vitro by a Paracellular Route. Int J Mol Sci 2020; 21:ijms21082858. [PMID: 32325905 PMCID: PMC7215935 DOI: 10.3390/ijms21082858] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 01/08/2023] Open
Abstract
Ganglioside GM1 (GM1) has been reported to functionally recover degenerated nervous system in vitro and in vivo, but the possibility to translate GM1′s potential in clinical settings is counteracted by its low ability to overcome the blood–brain barrier (BBB) due to its amphiphilic nature. Interestingly, the soluble and hydrophilic GM1-oligosaccharide (OligoGM1) is able to punctually replace GM1 neurotrophic functions alone, both in vitro and in vivo. In order to take advantage of OligoGM1 properties, which overcome GM1′s pharmacological limitations, here we characterize the OligoGM1 brain transport by using a human in vitro BBB model. OligoGM1 showed a 20-fold higher crossing rate than GM1 and time–concentration-dependent transport. Additionally, OligoGM1 crossed the barrier at 4 °C and in inverse transport experiments, allowing consideration of the passive paracellular route. This was confirmed by the exclusion of a direct interaction with the active ATP-binding cassette (ABC) transporters using the “pump out” system. Finally, after barrier crossing, OligoGM1 remained intact and able to induce Neuro2a cell neuritogenesis by activating the TrkA pathway. Importantly, these in vitro data demonstrated that OligoGM1, lacking the hydrophobic ceramide, can advantageously cross the BBB in comparison with GM1, while maintaining its neuroproperties. This study has improved the knowledge about OligoGM1′s pharmacological potential, offering a tangible therapeutic strategy.
Collapse
|
9
|
Tornabene E, Helms HCC, Pedersen SF, Brodin B. Effects of oxygen-glucose deprivation (OGD) on barrier properties and mRNA transcript levels of selected marker proteins in brain endothelial cells/astrocyte co-cultures. PLoS One 2019; 14:e0221103. [PMID: 31425564 PMCID: PMC6699694 DOI: 10.1371/journal.pone.0221103] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022] Open
Abstract
Ischemic stroke has been shown to induce breakdown of the blood-brain barrier, although these changes are not fully characterized. Oxygen-glucose deprivation (OGD) has been used to investigate the effects of ischemia in cultured brain capillary endothelial cells, however this involves a change of medium which in itself may affect the cells. The aim of the present study was to investigate the effect of OGD and simple medium exchange followed by 48 h of reperfusion on barrier properties of primary bovine endothelial cells co-cultured with rat astrocytes. Barrier properties were evaluated by transendothelial electrical resistance measurements, passive permeability of flux markers, RT-qPCR and immunocytochemistry. Both OGD and simple medium exchange caused an increase in endothelial monolayer permeability. This correlated with reduced transcript levels of a number of tight junction and tight junction-associated proteins (claudin-1, claudin-5, occludin, ZO-1, tricellulin, marveld3 and PECAM-1), as well as with altered transcript level of several transporters and receptors (GLUT-1, HB-EGF, InsR, TfR, two members of the low density lipoprotein receptor family, LDLR and LRP-1, and the efflux transporter BCRP). In contrast, effects induced specifically by OGD were transient de-localization of claudin-5 from the junction zone, increased InsR localization at the plasma membrane and transient downregulation of MRP-1 and P-gp transcript levels. In conclusion, OGD caused changes in claudin-5 and InsR localization, as well as in MRP-1 and P-gp transcript levels. Our results however also indicated that medium exchange alone caused changes in functional barrier properties and expression levels of wide range of proteins.
Collapse
Affiliation(s)
- Erica Tornabene
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Stine Falsig Pedersen
- Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Birger Brodin
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
10
|
Oxygen-Glucose Deprivation/Reoxygenation-Induced Barrier Disruption at the Human Blood–Brain Barrier is Partially Mediated Through the HIF-1 Pathway. Neuromolecular Med 2019; 21:414-431. [DOI: 10.1007/s12017-019-08531-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
|
11
|
Chen S, Chen Z, Cui J, McCrary ML, Song H, Mobashery S, Chang M, Gu Z. Early Abrogation of Gelatinase Activity Extends the Time Window for tPA Thrombolysis after Embolic Focal Cerebral Ischemia in Mice. eNeuro 2018; 5:ENEURO.0391-17.2018. [PMID: 29963617 PMCID: PMC6021166 DOI: 10.1523/eneuro.0391-17.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 04/28/2018] [Accepted: 05/22/2018] [Indexed: 02/02/2023] Open
Abstract
Acute ischemic stroke (AIS) is caused by clotting in the cerebral arteries, leading to brain oxygen deprivation and cerebral infarction. Recombinant human tissue plasminogen activator (tPA) is currently the only Food and Drug Administration-approved drug for ischemic stroke. However, tPA has to be administered within 4.5 h from the disease onset and delayed treatment of tPA can increase the risk of neurovascular impairment, including neuronal cell death, blood-brain barrier (BBB) disruption, and hemorrhagic transformation. A key contributing factor for tPA-induced neurovascular impairment is activation of matrix metalloproteinase-9 (MMP-9). We used a clinically-relevant mouse embolic model of focal-cerebral ischemia by insertion of a single embolus of blood clot to block the right middle cerebral artery. We showed that administration of the potent and highly selective gelatinase inhibitor SB-3CT extends the time window for administration of tPA, attenuating infarct volume, mitigating BBB disruption, and antagonizing the increase in cerebral hemorrhage induced by tPA treatment. We demonstrated that SB-3CT attenuates tPA-induced expression of vascular MMP-9, prevents gelatinase-mediated cleavage of extracellular laminin, rescues endothelial cells, and reduces caveolae-mediated transcytosis of endothelial cells. These results suggest that abrogation of MMP-9 activity mitigates the detrimental effects of tPA treatment, thus the combination treatment holds great promise for extending the therapeutic window for tPA thrombolysis, which opens the opportunity for clinical recourse to a greater number of patients.
Collapse
Affiliation(s)
- Shanyan Chen
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, Columbia, MO 65212
- Interdisciplinary Neuroscience Program, University of Missouri at Columbia, Columbia, MO 65212
| | - Zhenzhou Chen
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, Columbia, MO 65212
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, Columbia, MO 65212
- Harry S. Truman Memorial Veterans' Hospital Research Service, Columbia, MO 65201
| | - Myah L. McCrary
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, Columbia, MO 65212
| | - Hailong Song
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, Columbia, MO 65212
- Interdisciplinary Neuroscience Program, University of Missouri at Columbia, Columbia, MO 65212
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri at Columbia, Columbia, MO 65212
- Harry S. Truman Memorial Veterans' Hospital Research Service, Columbia, MO 65201
| |
Collapse
|
12
|
Tornabene E, Brodin B. Stroke and Drug Delivery--In Vitro Models of the Ischemic Blood-Brain Barrier. J Pharm Sci 2016; 105:398-405. [PMID: 26869407 DOI: 10.1016/j.xphs.2015.11.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/20/2015] [Accepted: 11/20/2015] [Indexed: 12/11/2022]
Abstract
Stroke is a major cause of death and disability worldwide. Both cerebral hypoperfusion and focal cerebral infarcts are caused by a reduction of blood flow to the brain, leading to stroke and subsequent brain damage. At present, only few medical treatments of stroke are available, with the Food and Drug Administration-approved tissue plasminogen activator for treatment of acute ischemic stroke being the most prominent example. A large number of potential drug candidates for treatment of ischemic brain tissue have been developed and subsequently failed in clinical trials. A deeper understanding of permeation pathways across the barrier in ischemic and postischemic brain endothelium is important for development of new medical treatments. The blood-brain barrier, that is, the endothelial monolayer lining the brain capillaries, changes properties during an ischemic event. In vitro models of the blood-brain barrier are useful tools to investigate the effects of induced ischemia under controlled conditions. In the present mini review, we aim to give a brief overview of the in vitro models of ischemia. Special focus is given to the expression of uptake and efflux transport pathways in the ischemic and postischemic endothelium. Finally, we will point toward future challenges within the field of in vitro models of brain ischemia.
Collapse
Affiliation(s)
- Erica Tornabene
- Section of Pharmaceutical Design and Drug Delivery, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Birger Brodin
- Section of Pharmaceutical Design and Drug Delivery, Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
13
|
Kuntz M, Candela P, Saint-Pol J, Lamartinière Y, Boucau MC, Sevin E, Fenart L, Gosselet F. Bexarotene Promotes Cholesterol Efflux and Restricts Apical-to-Basolateral Transport of Amyloid-β Peptides in an In Vitro Model of the Human Blood-Brain Barrier. J Alzheimers Dis 2016; 48:849-62. [PMID: 26402114 DOI: 10.3233/jad-150469] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
One of the prime features of Alzheimer's disease (AD) is the excessive accumulation of amyloid-β (Aβ) peptides in the brain. Several recent studies suggest that this phenomenon results from the dysregulation of cholesterol homeostasis in the brain and impaired bidirectional Aβ exchange between blood and brain. These mechanisms appear to be closely related and are controlled by the blood-brain barrier (BBB) at the brain microvessel level. In animal models of AD, the anticancer drug bexarotene (a retinoid X receptor agonist) has been found to restore cognitive functions and decrease the brain amyloid burden by regulating cholesterol homeostasis. However, the drug's therapeutic effect is subject to debate and the exact mechanism of action has not been characterized. Therefore, the objective of this present study was to determine bexarotene's effects on the BBB. Using an in vitro model of the human BBB, we investigated the drug's effects on cholesterol exchange between abluminal and luminal compartments and the apical-to-basolateral transport of Aβ peptides across the BBB. Our results demonstrated that bexarotene induces the expression of ABCA1 but not ApoE. This upregulation correlates with an increase in ApoE2-, ApoE4-, ApoA-I-, and HDL-mediated cholesterol efflux. Regarding the transport of Aβ peptides, bexarotene increases the expression of ABCB1, which in turn decreases Aβ apical-to-basolateral transport. Our results showed that bexarotene not only promotes the cholesterol exchange between the brain and the blood but also decreases the influx of Aβ peptides across BBB, suggesting that bexarotene is a promising drug candidate for the treatment of AD.
Collapse
|
14
|
Yang X, Chu H, Tang Y, Dong Q. The role of connexin43 in hemorrhagic transformation after thrombolysis in vivo and in vitro. Neuroscience 2016; 329:54-65. [PMID: 27138645 DOI: 10.1016/j.neuroscience.2016.04.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/25/2016] [Accepted: 04/26/2016] [Indexed: 11/17/2022]
Abstract
Thrombolysis with recombinant tissue plasminogen activator (rtPA) is the most effective drug treatment for acute ischemic stroke within 4.5h after symptom onset. However, the use of rtPA may increase the risk of hemorrhagic transformation (HT), particularly when it is administered after the first 4.5h. However, no effective treatments are available to reduce the HT risk. Disruption of the blood-brain barrier (BBB) is central to the genesis of HT. Connexin43 (Cx43)-mediated gap junction intercellular communication (GJIC) has been demonstrated to regulate the integrity of the BBB in ischemia. We investigated the effect of Cx43 on BBB permeability during rtPA-induced HT. Spontaneously hypertensive rats (SHRs) underwent a 1.5-h middle cerebral artery occlusion and were treated with rtPA at 4.5h. The rats were sacrificed at 24h, and their brains were evaluated for BBB permeability and the expression of tight junction (TJ) proteins and Cx43. We examined whether the effects were Cx43 dependent using multiple Cx43 inhibitors. Phosphorylated Cx43 (p-Cx43) but not total Cx43 protein expression was increased after rtPA treatment. Delayed rtPA administration induced significant HT and BBB disruption. These effects were attenuated by inhibitors that blocked GJIC and Cx43 phosphorylation and expression but not Cx43 redistribution. Additionally, rtPA administration upregulated p-Cx43 expression in hypoxia/reoxygenation (H/R)-exposed brain endothelial cells. These effects were suppressed by the phosphatidylinositol 3'-kinase (PI3K) inhibitor LY294002 and the extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitor U0126. We suggest that rtPA-associated hemorrhage due to an alteration in the integrity of the BBB is highly associated with an increase in p-Cx43 resulting from the activation of the PI3K and ERK pathways.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China; Department of Neurology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Heling Chu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yuping Tang
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
De Bock M, Van Haver V, Vandenbroucke RE, Decrock E, Wang N, Leybaert L. Into rather unexplored terrain-transcellular transport across the blood-brain barrier. Glia 2016; 64:1097-123. [DOI: 10.1002/glia.22960] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 11/16/2015] [Accepted: 12/03/2015] [Indexed: 01/22/2023]
Affiliation(s)
- Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences; Ghent University; Ghent Belgium
| | - Valérie Van Haver
- Physiology Group, Department of Basic Medical Sciences; Ghent University; Ghent Belgium
| | - Roosmarijn E. Vandenbroucke
- Inflammation Research Center, VIB; Ghent Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent Belgium
| | - Elke Decrock
- Physiology Group, Department of Basic Medical Sciences; Ghent University; Ghent Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences; Ghent University; Ghent Belgium
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences; Ghent University; Ghent Belgium
| |
Collapse
|
16
|
Neves V, Aires-da-Silva F, Corte-Real S, Castanho MA. Antibody Approaches To Treat Brain Diseases. Trends Biotechnol 2016. [DOI: 10.1016/j.tibtech.2015.10.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Neuhaus W, Gaiser F, Mahringer A, Franz J, Riethmüller C, Förster C. The pivotal role of astrocytes in an in vitro stroke model of the blood-brain barrier. Front Cell Neurosci 2014; 8:352. [PMID: 25389390 PMCID: PMC4211409 DOI: 10.3389/fncel.2014.00352] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/07/2014] [Indexed: 12/14/2022] Open
Abstract
Stabilization of the blood-brain barrier during and after stroke can lead to less adverse outcome. For elucidation of underlying mechanisms and development of novel therapeutic strategies validated in vitro disease models of the blood-brain barrier could be very helpful. To mimic in vitro stroke conditions we have established a blood-brain barrier in vitro model based on mouse cell line cerebEND and applied oxygen/glucose deprivation (OGD). The role of astrocytes in this disease model was investigated by using cell line C6. Transwell studies pointed out that addition of astrocytes during OGD increased the barrier damage significantly in comparison to the endothelial monoculture shown by changes of transendothelial electrical resistance as well as fluorescein permeability data. Analysis on mRNA and protein levels by qPCR, western blotting and immunofluorescence microscopy of tight junction molecules claudin-3,-5,-12, occludin and ZO-1 revealed that their regulation and localisation is associated with the functional barrier breakdown. Furthermore, soluble factors of astrocytes, OGD and their combination were able to induce changes of functionality and expression of ABC-transporters Abcb1a (P-gp), Abcg2 (bcrp), and Abcc4 (mrp4). Moreover, the expression of proteases (matrixmetalloproteinases MMP-2, MMP-3, MMP-9, and t-PA) as well as of their endogenous inhibitors (TIMP-1, TIMP-3, PAI-1) was altered by astrocyte factors and OGD which resulted in significant changes of total MMP and t-PA activity. Morphological rearrangements induced by OGD and treatment with astrocyte factors were confirmed at a nanometer scale using atomic force microscopy. In conclusion, astrocytes play a major role in blood-brain barrier breakdown during OGD in vitro.
Collapse
Affiliation(s)
- Winfried Neuhaus
- Department of Pharmaceutical Chemistry, University of Vienna Vienna, Austria ; Department of Anesthesia and Critical Care, University Hospital Würzburg Würzburg, Germany
| | - Fabian Gaiser
- Department of Anesthesia and Critical Care, University Hospital Würzburg Würzburg, Germany
| | - Anne Mahringer
- Department of Pharmaceutical Technology and Biopharmacy, Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg Heidelberg, Germany
| | - Jonas Franz
- Serend-ip GmbH, Centre for Nanotechnology Münster, Germany
| | | | - Carola Förster
- Department of Anesthesia and Critical Care, University Hospital Würzburg Würzburg, Germany
| |
Collapse
|
18
|
Diesel exhaust particles impair endothelial progenitor cells, compromise endothelial integrity, reduce neoangiogenesis, and increase atherogenesis in mice. Cardiovasc Toxicol 2014; 13:290-300. [PMID: 23584878 DOI: 10.1007/s12012-013-9208-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mechanisms of the harmful cardiovascular effects of small particulate matter are incompletely understood. Endothelial progenitor cells (EPCs) predict outcome of patients with vascular disease. The aim of our study was to examine the effects of diesel exhaust particles (DEP) on EPC and on the associated vascular damage in mice. C57Bl/6 mice were exposed to DEP. 2 μg DEP/day was applicated intranasally for 3 weeks. Exposure to DEP reduced DiLDL/lectin positive EPC to 58.4 ± 5.6% (p < 0.005). Migratory capacity was reduced to 65.8 ± 3.9% (p < 0.0001). In ApoE(-/-) mice, DEP application reduced the number of EPC to 75.6 ± 6.4% (p < 0.005) and EPC migration to 58.5 ± 6.8% (p < 0.005). Neoangiogenesis was reduced to 39.5 ± 14.6% (p < 0.005). Atherogenesis was profoundly increased by DEP treatment (157.7 ± 18.1% vs. controls, p < 0.05). In cultured human EPC, DEP (0.1-100 μg/mL) reduced migratory capacity to 25 ± 2.6% (p < 0.001). The number of colony-forming units was reduced to 8.8 ± 0.9% (p < 0.001) and production of reactive oxygen species was elevated by DEP treatment (p < 0.001). Furthermore, DEP treatment increased apoptosis of EPC (to 266 ± 62% of control, p < 0.05). In a blood-brain barrier model, DEP treatment impaired endothelial cell integrity during oxygen-glucose deprivation (p < 0.001). Diesel exhaust particles impair endothelial progenitor cell number and function in vivo and in vitro. The reduction in EPC was associated with impaired neoangiogenesis and a marked increase in atherosclerotic lesion formation.
Collapse
|
19
|
Kuntz M, Mysiorek C, Pétrault O, Boucau MC, Aijjou R, Uzbekov R, Bérézowski V. Transient oxygen-glucose deprivation sensitizes brain capillary endothelial cells to rtPA at 4h of reoxygenation. Microvasc Res 2013; 91:44-57. [PMID: 24333620 DOI: 10.1016/j.mvr.2013.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2013] [Revised: 11/28/2013] [Accepted: 12/03/2013] [Indexed: 01/11/2023]
Abstract
Thrombolysis treatment of acute ischemic stroke is limited by the pro-edematous and hemorrhagic effects exerted by reperfusion, which disrupts the blood-brain barrier (BBB) capillary endothelium in the infarct core. Most studies of the ischemic BBB overlook the complexity of the penumbral area, where the affected brain cells are still viable following deprivation. Our present objective was to examine in vitro the kinetic impact of reoxygenation on the integrity of ischemic BBB cells after oxygen-glucose deprivation. Through the use of a co-culture of brain capillary endothelial cells and glial cells, we first showed that the transendothelial permeability increase induced by deprivation can occur with both preserved cell viability and interendothelial tight junction network. The subtle and heterogeneous alteration of the tight junctions was observable only through electron microscopy. A complete permeability recovery was then found after reoxygenation, when Vimentin and Actin networks were reordered. However, still sparse ultrastructural alterations of tight junctions suggested an acquired vulnerability. Endothelial cells were then exposed to recombinant tissue-type plasminogen activator (rtPA) to define a temporal profile for the toxic effect of this thrombolytic on transendothelial permeability. Interestingly, the reoxygenated BBB broke down with aggravated tight junction disruption when exposed to rtPA only at 4h after reoxygenation. Moreover, this breakdown was enhanced by 50% when ischemic glial cells were present during the first hours of reoxygenation. Our results suggest that post-stroke reoxygenation enables retrieval of the barrier function of brain capillary endothelium when in a non-necrotic environment, but may sensitize it to rtPA at the 4-hour time point, when both endothelial breakdown mechanisms and glial secretions could be identified and targeted in a therapeutical perspective.
Collapse
Affiliation(s)
- Mélanie Kuntz
- Univ Lille Nord de France, UArtois, LBHE, EA 2465, Faculté des Sciences Jean Perrin, rue Jean Souvraz, S.P.18, F-62307 Lens cedex, France; Institut de Médecine Prédictive et de Recherche Thérapeutique, Place de Verdun, F-59045 Lille cedex, France; Institut Fédératif de Recherche 114, Place de Verdun, F-59045 Lille cedex, France.
| | - Caroline Mysiorek
- Univ Lille Nord de France, UArtois, LBHE, EA 2465, Faculté des Sciences Jean Perrin, rue Jean Souvraz, S.P.18, F-62307 Lens cedex, France; Institut de Médecine Prédictive et de Recherche Thérapeutique, Place de Verdun, F-59045 Lille cedex, France; Institut Fédératif de Recherche 114, Place de Verdun, F-59045 Lille cedex, France.
| | - Olivier Pétrault
- Univ Lille Nord de France, UArtois, LBHE, EA 2465, Faculté des Sciences Jean Perrin, rue Jean Souvraz, S.P.18, F-62307 Lens cedex, France; Institut de Médecine Prédictive et de Recherche Thérapeutique, Place de Verdun, F-59045 Lille cedex, France; Institut Fédératif de Recherche 114, Place de Verdun, F-59045 Lille cedex, France.
| | - Marie-Christine Boucau
- Univ Lille Nord de France, UArtois, LBHE, EA 2465, Faculté des Sciences Jean Perrin, rue Jean Souvraz, S.P.18, F-62307 Lens cedex, France; Institut de Médecine Prédictive et de Recherche Thérapeutique, Place de Verdun, F-59045 Lille cedex, France; Institut Fédératif de Recherche 114, Place de Verdun, F-59045 Lille cedex, France.
| | - Rachid Aijjou
- Univ Lille Nord de France, UArtois, LBHE, EA 2465, Faculté des Sciences Jean Perrin, rue Jean Souvraz, S.P.18, F-62307 Lens cedex, France; Institut de Médecine Prédictive et de Recherche Thérapeutique, Place de Verdun, F-59045 Lille cedex, France; Institut Fédératif de Recherche 114, Place de Verdun, F-59045 Lille cedex, France.
| | - Rustem Uzbekov
- Département des Microscopies, Université François Rabelais, F-37100 Tours, France; Faculty of Bioengineering & Bioinformatics, Moscow State University, 119991 Moscow, Russia.
| | - Vincent Bérézowski
- Univ Lille Nord de France, UArtois, LBHE, EA 2465, Faculté des Sciences Jean Perrin, rue Jean Souvraz, S.P.18, F-62307 Lens cedex, France; Institut de Médecine Prédictive et de Recherche Thérapeutique, Place de Verdun, F-59045 Lille cedex, France; Institut Fédératif de Recherche 114, Place de Verdun, F-59045 Lille cedex, France.
| |
Collapse
|
20
|
Teng F, Beray-Berthat V, Coqueran B, Lesbats C, Kuntz M, Palmier B, Garraud M, Bedfert C, Slane N, Bérézowski V, Szeremeta F, Hachani J, Scherman D, Plotkine M, Doan BT, Marchand-Leroux C, Margaill I. Prevention of rt-PA induced blood-brain barrier component degradation by the poly(ADP-ribose)polymerase inhibitor PJ34 after ischemic stroke in mice. Exp Neurol 2013; 248:416-28. [PMID: 23876515 DOI: 10.1016/j.expneurol.2013.07.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/28/2013] [Accepted: 07/12/2013] [Indexed: 01/24/2023]
Abstract
Recombinant tissue plasminogen activator (rt-PA) is the only pharmacological treatment approved for thrombolysis in patients suffering from ischemic stroke, but its administration aggravates the risk of hemorrhagic transformations. Experimental data demonstrated that rt-PA increases the activity of poly(ADP-ribose)polymerase (PARP). The aim of the present study was to investigate whether PJ34, a potent (PARP) inhibitor, protects the blood-brain barrier components from rt-PA toxicity. In our mouse model of cerebral ischemia, administration of rt-PA (10 mg/kg, i.v.) 6h after ischemia aggravated the post-ischemic degradation of ZO-1, claudin-5 and VE-cadherin, increased the hemorrhagic transformations (assessed by brain hemoglobin content and magnetic resonance imaging). Furthermore, rt-PA also aggravated ischemia-induced functional deficits. Combining PJ34 with rt-PA preserved the expression of ZO-1, claudin-5 and VE-cadherin, reduced the hemorrhagic transformations and improved the sensorimotor performances. In vitro studies also demonstrated that PJ34 crosses the blood-brain barrier and may thus exert its protective effect by acting on endothelial and/or parenchymal cells. Thus, co-treatment with a PARP inhibitor seems to be a promising strategy to reduce rt-PA-induced vascular toxicity after stroke.
Collapse
Affiliation(s)
- Fei Teng
- Equipe de recherche "Pharmacologie de la Circulation Cérébrale" EA4475, Université Paris Descartes, Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Saint-Pol J, Candela P, Boucau MC, Fenart L, Gosselet F. Oxysterols decrease apical-to-basolateral transport of Aß peptides via an ABCB1-mediated process in an in vitro Blood-brain barrier model constituted of bovine brain capillary endothelial cells. Brain Res 2013; 1517:1-15. [PMID: 23603412 DOI: 10.1016/j.brainres.2013.04.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 04/04/2013] [Accepted: 04/09/2013] [Indexed: 01/03/2023]
Abstract
It is known that activation of the liver X receptors (LXRs) by natural or synthetic agonists decreases the amyloid burden and enhances cognitive function in transgenic murine models of Alzheimer's disease (AD). Recent evidence suggests that LXR activation may affect the transport of amyloid ß (Aß) peptides across the blood-brain barrier (the BBB, which isolates the brain from the peripheral circulation). By using a well-characterized in vitro BBB model, we demonstrated that LXR agonists (24S-hydroxycholesterol, 27-hydroxycholesterol and T0901317) modulated the expression of target genes involved in cholesterol homeostasis (such as ATP-binding cassette sub-family A member 1 (ABCA1)) and promoted cellular cholesterol efflux to apolipoprotein A-I and high density lipoproteins. Interestingly, we also observed a decrease in Aß peptide influx across brain capillary endothelial cells, although ABCA1 did not appear to be directly involved in this process. By focusing on others receptors and transporters that are thought to have major roles in Aß peptide entry into the brain, we then demonstrated that LXR stimulation provoked an increase in expression of the ABCB1 transporter (also named P-glycoprotein (P-gp)). Further investigations confirmed ABCB1's involvement in the restriction of Aß peptide influx. Taken as a whole, our results not only reinforce the BBB's key role in cerebral cholesterol homeostasis but also demonstrate the importance of the LXR/ABCB1 axis in Aß peptide influx-highlighting an attractive new therapeutic approach whereby the brain could be protected from peripheral Aß peptide entry.
Collapse
|
22
|
Bérézowski V, Mysiorek C, Kuntz M, Pétrault O, Cecchelli R. [Dysfunction of the blood-brain barrier during ischaemia: a therapeutic concern]. Biol Aujourdhui 2012; 206:161-76. [PMID: 23171839 DOI: 10.1051/jbio/2012020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Indexed: 11/14/2022]
Abstract
Since it was discovered and its brain-protective role characterized, the blood-brain barrier (BBB), through the permeability-restricting action of the brain capillary endothelial cells, has been representing a hurdle for 95% of new medical compounds targeting the central nervous system. Recently, a BBB dysfunction is being found in an increasing number of pathologies such as brain ischaemic stroke, whose only therapy consists in a pharmacological thrombolysis limited to a small percentage of the admitted patients, because of the toxical effects of thrombolytics. And since the clinical failure of promising neuroprotectants, numerous studies of brain ischaemia were carried out, with physiopathological or pharmacological approaches refocused on the BBB, whose structural complexity is now expanded to perivascular cells, all forming a functional unit named the neurovascular unit (NVU). Nevertheless, in spite of the numerous molecular mechanisms identified, the process of BBB dysfunction in the ischaemia/reperfusion cascade remains insufficiently established to explain the pleiotropic action exerted by new pharmacological compounds, possibly protecting the entire NVU and representing potential treatments.
Collapse
|
23
|
Bahjat FR, Gesuete R, Stenzel-Poore MP. Steps to translate preconditioning from basic research to the clinic. Transl Stroke Res 2012; 4:89-103. [PMID: 23504609 DOI: 10.1007/s12975-012-0223-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Efforts to treat cardiovascular and cerebrovascular diseases often focus on the mitigation of ischemia-reperfusion (I/R) injury. Many treatments or "preconditioners" are known to provide substantial protection against the I/R injury when administered prior to the event. Brief periods of ischemia itself have been validated as a means to achieve neuroprotection in many experimental disease settings, in multiple organ systems, and in multiple species suggesting a common pathway leading to tolerance. In addition, pharmacological agents that act as potent preconditioners have been described. Experimental induction of neuroprotection using these various preconditioning paradigms has provided a unique window into the brain's endogenous protective mechanisms. Moreover, preconditioning agents themselves hold significant promise as clinical-stage therapies for prevention of I/R injury. The aim of this article is to explore several key steps involved in the preclinical validation of preconditioning agents prior to the conduct of clinical studies in humans. Drug development is difficult, expensive and relies on multi-factorial analysis of data from diverse disciplines. Importantly, there is no single path for the preclinical development of a novel therapeutic and no proven strategy to ensure success in clinical translation. Rather, the conduct of a diverse array of robust preclinical studies reduces the risk of clinical failure by varying degrees depending upon the relevance of preclinical models and drug pharmacology to humans. A strong sense of urgency and high tolerance of failure are often required to achieve success in the development of novel treatment paradigms for complex human conditions.
Collapse
Affiliation(s)
- Frances R Bahjat
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon
| | | | | |
Collapse
|
24
|
Roussel BD, Mysiorek C, Rouhiainen A, Jullienne A, Parcq J, Hommet Y, Culot M, Berezowski V, Cecchelli R, Rauvala H, Vivien D, Ali C. HMGB-1 promotes fibrinolysis and reduces neurotoxicity mediated by tissue plasminogen activator. J Cell Sci 2011; 124:2070-6. [DOI: 10.1242/jcs.084392] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Owing to its ability to generate the clot-dissolving protease plasmin, tissue plasminogen activator (tPA) is the only approved drug for the acute treatment of ischemic stroke. However, tPA also promotes hemorrhagic transformation and excitotoxic events. High mobility group box-1 protein (HMGB-1) is a non-histone transcription factor and a pro-inflammatory cytokine, which has also been shown to bind to both tPA and plasminogen. We thus investigated the cellular and molecular effects through which HMGB-1 could influence the vascular and parenchymal effects of tPA during ischemia. We demonstrate that HMGB-1 not only increases clot lysis by tPA, but also reduces the passage of vascular tPA across the blood–brain barrier, as well as tPA-driven leakage of the blood–brain barrier. In addition, HMGB-1 prevents the pro-neurotoxic effect of tPA, by blocking its interaction with N-methyl-D-aspartate (NMDA) receptors and the attendant potentiation of NMDA-induced neuronal Ca2+ influx. In conclusion, we show in vitro that HMGB-1 can promote the beneficial effects of tPA while counteracting its deleterious properties. We suggest that derivatives of HMGB-1, devoid of pro-inflammatory properties, could be used as adjunctive therapies to improve the overall benefit of tPA-mediated thrombolysis following stroke.
Collapse
Affiliation(s)
| | - Caroline Mysiorek
- Univ Lille Nord de France, UArtois, Laboratoire de Physiopathologie de la Barrière Hémato-Encéphalique, EA 2465, IMPRT-IFR114, 62303 Lens, France
| | - Ari Rouhiainen
- Neuroscience Center, University of Helsinki, Helsinki 00014, Finland
| | - Amandine Jullienne
- INSERM U919 ‘Serine proteases and pathophysiology of the neurovascular unit’, UMR CNRS 6232 Cinaps, GIP Cyceron, University of Caen, 14032 Caen, France
| | - Jerome Parcq
- INSERM U919 ‘Serine proteases and pathophysiology of the neurovascular unit’, UMR CNRS 6232 Cinaps, GIP Cyceron, University of Caen, 14032 Caen, France
| | - Yannick Hommet
- INSERM U919 ‘Serine proteases and pathophysiology of the neurovascular unit’, UMR CNRS 6232 Cinaps, GIP Cyceron, University of Caen, 14032 Caen, France
| | - Maxime Culot
- Univ Lille Nord de France, UArtois, Laboratoire de Physiopathologie de la Barrière Hémato-Encéphalique, EA 2465, IMPRT-IFR114, 62303 Lens, France
| | - Vincent Berezowski
- Univ Lille Nord de France, UArtois, Laboratoire de Physiopathologie de la Barrière Hémato-Encéphalique, EA 2465, IMPRT-IFR114, 62303 Lens, France
| | - Romeo Cecchelli
- Univ Lille Nord de France, UArtois, Laboratoire de Physiopathologie de la Barrière Hémato-Encéphalique, EA 2465, IMPRT-IFR114, 62303 Lens, France
| | - Heikki Rauvala
- Neuroscience Center, University of Helsinki, Helsinki 00014, Finland
| | - Denis Vivien
- INSERM U919 ‘Serine proteases and pathophysiology of the neurovascular unit’, UMR CNRS 6232 Cinaps, GIP Cyceron, University of Caen, 14032 Caen, France
| | - Carine Ali
- INSERM U919 ‘Serine proteases and pathophysiology of the neurovascular unit’, UMR CNRS 6232 Cinaps, GIP Cyceron, University of Caen, 14032 Caen, France
| |
Collapse
|
25
|
Gesuete R, Orsini F, Zanier ER, Albani D, Deli MA, Bazzoni G, De Simoni MG. Glial Cells Drive Preconditioning-Induced Blood-Brain Barrier Protection. Stroke 2011; 42:1445-53. [DOI: 10.1161/strokeaha.110.603266] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Raffaella Gesuete
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Franca Orsini
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Elisa R. Zanier
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Diego Albani
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Maria A. Deli
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Gianfranco Bazzoni
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Maria-Grazia De Simoni
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| |
Collapse
|
26
|
Zehendner CM, Librizzi L, de Curtis M, Kuhlmann CRW, Luhmann HJ. Caspase-3 contributes to ZO-1 and Cl-5 tight-junction disruption in rapid anoxic neurovascular unit damage. PLoS One 2011; 6:e16760. [PMID: 21364989 PMCID: PMC3043060 DOI: 10.1371/journal.pone.0016760] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 12/29/2010] [Indexed: 12/11/2022] Open
Abstract
Background Tight-junction (TJ) protein degradation is a decisive step in hypoxic blood-brain barrier (BBB) breakdown in stroke. In this study we elucidated the impact of acute cerebral ischemia on TJ protein arrangement and the role of the apoptotic effector protease caspase-3 in this context. Methodology/Principal Findings We used an in vitro model of the neurovascular unit and the guinea pig whole brain preparation to analyze with immunohistochemical methods the BBB properties and neurovascular integrity. In both methodological approaches we observed rapid TJ protein disruptions after 30 min of oxygen and glucose deprivation or middle cerebral artery occlusion, which were accompanied by strong caspase-3 activation in brain endothelial cells (BEC). Surprisingly only few DNA-fragmentations were detected with TUNEL stainings in BEC. Z-DEVD-fmk, an irreversible caspase-3 inhibitor, partly blocked TJ disruptions and was protective on trans-endothelial electrical resistance. Conclusions/Significance Our data provide evidence that caspase-3 is rapidly activated during acute cerebral ischemia predominantly without triggering DNA-fragmentation in BEC. Further we detected fast TJ protein disruptions which could be partly blocked by caspase-3 inhibition with Z-DEVD-fmk. We suggest that the basis for clinically relevant BBB breakdown in form of TJ disruptions is initiated within minutes during ischemia and that caspase-3 contributes to this process.
Collapse
Affiliation(s)
- Christoph M. Zehendner
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Laura Librizzi
- Unit of Experimental Neurophysiology and Epileptology, Fondazione Istituto Neurologico Carlo Besta, Milano, Italy
| | - Marco de Curtis
- Unit of Experimental Neurophysiology and Epileptology, Fondazione Istituto Neurologico Carlo Besta, Milano, Italy
| | - Christoph R. W. Kuhlmann
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
| | - Heiko J. Luhmann
- Institute of Physiology and Pathophysiology, University Medical Center of the Johannes Gutenberg-University of Mainz, Mainz, Germany
- * E-mail:
| |
Collapse
|
27
|
|
28
|
Wolburg H, Wolburg-Buchholz K, Fallier-Becker P, Noell S, Mack AF. Structure and functions of aquaporin-4-based orthogonal arrays of particles. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 287:1-41. [PMID: 21414585 DOI: 10.1016/b978-0-12-386043-9.00001-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Orthogonal arrays or assemblies of intramembranous particles (OAPs) are structures in the membrane of diverse cells which were initially discovered by means of the freeze-fracturing technique. This technique, developed in the 1960s, was important for the acceptance of the fluid mosaic model of the biological membrane. OAPs were first described in liver cells, and then in parietal cells of the stomach, and most importantly, in the astrocytes of the brain. Since the discovery of the structure of OAPs and the identification of OAPs as the morphological equivalent of the water channel protein aquaporin-4 (AQP4) in the 1990s, a plethora of morphological work on OAPs in different cells was published. Now, we feel a need to balance new and old data on OAPs and AQP4 to elucidate the interrelationship of both structures and molecules. In this review, the identity of OAPs as AQP4-based structures in a diversity of cells will be described. At the same time, arguments are offered that under pathological or experimental circumstances, AQP4 can also be expressed in a non-OAP form. Thus, we attempt to project classical work on OAPs onto the molecular biology of AQP4. In particular, astrocytes and glioma cells will play the major part in this review, not only due to our own work but also due to the fact that most studies on structure and function of AQP4 were done in the nervous system.
Collapse
Affiliation(s)
- Hartwig Wolburg
- Institute of Pathology, University of Tübingen, Tübingen, Germany
| | | | | | | | | |
Collapse
|
29
|
Experimental models for assaying microvascular endothelial cell pathophysiology in stroke. Molecules 2010; 15:9104-34. [PMID: 21150829 PMCID: PMC6259215 DOI: 10.3390/molecules15129104] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 11/29/2010] [Accepted: 12/08/2010] [Indexed: 02/06/2023] Open
Abstract
It is important to understand the molecular mechanisms underlying neuron death following stroke in order to develop effective neuroprotective strategies. Since studies on human stroke are extremely limited due to the difficulty in collecting post-mortem tissue at different time points after the onset of stroke, brain ischaemia research focuses on information derived from in-vitro models of neuronal death through ischaemic injury [1]. This review aims to provide an update on the different in-vitro stroke models with brain microvascular endothelial cells that are currently being used. These models provide a physiologically relevant tool to screen potential neuroprotective drugs in stroke and to study the molecular mechanisms involved in brain ischaemia.
Collapse
|
30
|
Cowan KM, Easton AS. Neutrophils block permeability increases induced by oxygen glucose deprivation in a culture model of the human blood-brain barrier. Brain Res 2010; 1332:20-31. [PMID: 20346925 DOI: 10.1016/j.brainres.2010.03.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 02/23/2010] [Accepted: 03/17/2010] [Indexed: 12/22/2022]
Abstract
Experimentally, oxygen glucose deprivation (OGD) has been widely used to mimic the environmental conditions present during cerebral ischemia-reperfusion (IR) injury. OGD is known to increase permeability across cultured cerebral endothelial cells, which models the effect of IR on permeability across the blood-brain barrier (BBB); however, studies have yet to be performed in a human model. The effect of neutrophils on the increase in BBB permeability associated with IR injury has yet to be modeled in vitro. To address these questions, the human brain endothelial cell line hCMEC/D3 was exposed to OGD with reoxygenation, and permeability was measured for a range of OGD exposure times (1-24h). One hour of exposure to OGD induced a reversible increase in permeability, unassociated with cytotoxicity (assessed from lactate dehydrogenase release). However, 12-24h OGD exposures induced sustained increases in permeability associated with cytotoxicity. The 1h permeability increase was inhibited with the nitric oxide synthase inhibitors l-NAME (10(-)(7)mol/l) and 1400W (10(-)(7)mol/l). Neutrophils (5x10(6)/ml) blocked the permeability increase associated with 1h OGD, whether applied during or after OGD exposure. Permeability remained low if neutrophils were activated with leukotriene (Lt)B(4) (10(-)(7)mol/l) or exposed to a transendothelial gradient of LtB(4), while neutrophil activation with phorbyl myristate acetate (4x10(-)(8)mol/l) induced a small increase. Neutrophils had no effect on the permeability increase induced by 12h OGD exposure. This study finds that OGD induces reversible increases in permeability linked to nitric oxide synthesis in a human culture model of the BBB and shows that neutrophils mitigate permeability increases in this context.
Collapse
Affiliation(s)
- Krystyna M Cowan
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | |
Collapse
|
31
|
Liang S, Pong K, Gonzales C, Chen Y, Ling HP, Mark RJ, Boschelli F, Boschelli DH, Ye F, Barrios Sosa AC, Mansour TS, Frost P, Wood A, Pangalos MN, Zaleska MM. Neuroprotective profile of novel SRC kinase inhibitors in rodent models of cerebral ischemia. J Pharmacol Exp Ther 2009; 331:827-35. [PMID: 19741150 DOI: 10.1124/jpet.109.156562] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Src kinase signaling has been implicated in multiple mechanisms of ischemic injury, including vascular endothelial growth factor (VEGF)-mediated vascular permeability that leads to vasogenic edema, a major clinical complication in stroke and brain trauma. Here we report the effects of two novel Src kinase inhibitors, 4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-7-[3-(4-methyl-1-piperazinyl)propoxy]-3-quinolinecarbonitrile (SKI-606) and 4-[(2,4-dichloro-5-methoxyphenyl)amino]-6-methoxy-7-[4-(4-methypiperazin-1-yl)but-1-ynyl]-3-quinolinecarbonitrile (SKS-927), on ischemia-induced brain infarction and short- and long-term neurological deficits. Two well established transient [transient middle cerebral artery occlusion (tMCAO)] and permanent [permanent middle cerebral artery occlusion (pMCAO)] focal ischemia models in the rat were used with drug treatments initiated up to 6 h after onset of stroke to mimic the clinical scenario. Brain penetration of Src inhibitors, their effect on blood-brain barrier integrity and VEGF signaling in human endothelial cells were also evaluated. Our results demonstrate that both agents potently block VEGF-mediated signaling in human endothelial cells, penetrate rat brain upon systemic administration, and inhibit postischemic Src activation and vascular leakage. Treatment with SKI-606 or SKS-927 (at the doses of 3-30 mg/kg i.v.) resulted in a dose-dependent reduction in infarct volume and robust protection from neurological impairments even when the therapy was initiated up to 4- to 6-h after tMCAO. Src blockade after pMCAO resulted in accelerated improvement in recovery from motor, sensory, and reflex deficits during a long-term (3 weeks) testing period poststroke. These data demonstrate that the novel Src kinase inhibitors provide effective treatment against ischemic conditions within a clinically relevant therapeutic window and may constitute a viable therapy for acute stroke.
Collapse
Affiliation(s)
- Shi Liang
- Discovery Neuroscience, Wyeth Research, CN-8000, Princeton, NJ 08543, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Culot M, Mysiorek C, Renftel M, Roussel BD, Hommet Y, Vivien D, Cecchelli R, Fenart L, Berezowski V, Dehouck MP, Lundquist S. Cerebrovascular protection as a possible mechanism for the protective effects of NXY-059 in preclinical models: An in vitro study. Brain Res 2009; 1294:144-52. [DOI: 10.1016/j.brainres.2009.07.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 07/10/2009] [Accepted: 07/14/2009] [Indexed: 10/20/2022]
|
33
|
Pottiez G, Flahaut C, Cecchelli R, Karamanos Y. Understanding the blood-brain barrier using gene and protein expression profiling technologies. ACTA ACUST UNITED AC 2009; 62:83-98. [PMID: 19770003 DOI: 10.1016/j.brainresrev.2009.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 08/31/2009] [Accepted: 09/12/2009] [Indexed: 12/24/2022]
Abstract
The blood-brain barrier (BBB) contributes to the brain homeostasis by regulating the passage of endogenous and exogenous compounds. This function is in part due to well-known proteins such as tight junction proteins, plasma membrane transporters and metabolic barrier proteins. Over the last decade, genomics and proteomics have emerged as supplementary tools for BBB research. The development of genomic and proteomic technologies has provided several means to extend the BBB knowledge and to investigate additional routes for the bypass of this barrier. These profiling technologies have been used on BBB models to decipher the physiological characteristics and, under stress conditions, to understand the molecular mechanisms of brain diseases. In this review, we will report and discuss the genomic and proteomic studies recently carried out to enhance the understanding of BBB features.
Collapse
|
34
|
Kim JH, Yu YS, Kim JH, Kim KW, Min BH. The Role of Clusterin inIn VitroIschemia of Human Retinal Endothelial Cells. Curr Eye Res 2009; 32:693-8. [PMID: 17852194 DOI: 10.1080/02713680701487871] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Clusterin has been reported to be up-regulated in diverse pathophysiological stresses, but its role is controversial. In this study, we investigated the role of clusterin under in vitro ischemia of human retinal endothelial cells (HRECs). When HRECs were exposed to oxygen-glucose deprivation (OGD), clusterin expression increased, whereas von Willebrand factor (vWF), occludin, and zonula occludens (ZO-1) markedly decreased. Interestingly, loss of tight junction proteins and death of HRECs in OGD conditions were restored by clusterin treatment. Our results suggest that the enhanced clusterin in OGD conditions may play a protective role against ischemia-induced tight junction protein loss and HRECs death.
Collapse
Affiliation(s)
- Jeong Hun Kim
- Department of Ophthalmology, Seoul National University College of Medicine, & Seoul Artificial Eye Center, Clinical Research Institute, Seoul National University Hospital, Seoul, Korea
| | | | | | | | | |
Collapse
|
35
|
Wolburg H, Noell S, Wolburg-Buchholz K, Mack A, Fallier-Becker P. Agrin, aquaporin-4, and astrocyte polarity as an important feature of the blood-brain barrier. Neuroscientist 2009; 15:180-93. [PMID: 19307424 DOI: 10.1177/1073858408329509] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier (BBB) does not exclusively refer to brain endothelial cells, which are the site of the barrier proper. In the past few years, it has become increasingly clear that BBB endothelial cells depend considerably on the brain microenvironment to a degree exceeding the environmental influence in other organs. The concept of the BBB has been continuously developed over the decades, culminating now in the recognition that endothelial cell function in the brain is not limited to simply mediating energy and oxygen transfer between blood and neural tissue. Endothelial cells are rather "Janus-headed beings" that are active partners of both luminal molecules and cells, as well as subendothelial cells such as pericytes, astrocytes, and neurons. In this overview, the authors present and discuss both the role of astroglial cells in managing the BBB and aspects of pathological alterations in the brain as far as the BBB is involved. After a brief introduction of the BBB that describes the structure and function of the brain capillary endothelial cells, the authors report on both the water channel protein aquaporin-4 (AQP4) in astrocytes and the extracellular matrix between astrocytes/pericytes and endothelial cells. The AQP4 has an important impact on the homeostasis in the brain parenchyma; however, the mechanistic cascade from the composition of the astrocyte membrane to the maintenance of BBB properties in the endothelial cells, including their tight junction formation, is still completely unknown.
Collapse
Affiliation(s)
- Hartwig Wolburg
- Institute of Pathology, University of Tübingen, Tübingen, Germany.
| | | | | | | | | |
Collapse
|
36
|
Li YN, Qin XJ, Kuang F, Wu R, Duan XL, Ju G, Wang BR. Alterations of Fc gamma receptor I and Toll-like receptor 4 mediate the antiinflammatory actions of microglia and astrocytes after adrenaline-induced blood-brain barrier opening in rats. J Neurosci Res 2009; 86:3556-65. [PMID: 18756515 DOI: 10.1002/jnr.21810] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Blood-brain barrier (BBB) opening occurs under many physiological and pathological conditions. BBB opening will lead to the leakage of large circulating molecules into the brain parenchyma. These invasive molecules will induce immune responses. Microglia and astrocytes are the two major cell types responsible for immune responses in the brain, and Fc gamma receptor I (FcgammaRI) and Toll-like receptor 4 (TLR4) are the two important receptors mediating these processes. Data suggest that activation of the FcgammaRI pathway mediates antiinflammatory processes, whereas activation of TLR4 pathway leads to proinflammatory activities. In the present study, we tested the hypothesis that BBB opening could lead to alterations in FcgammaRI and TLR4 pathways in microglia and astrocytes, thus limiting excessive inflammation in the brain. The transient BBB opening was induced by adrenaline injection through a caudal vein in Sprague-Dawley rats. We found that the FcgammaRI pathway was significantly activated in both microglia and astrocytes, as exhibited by the up-regulation of FcgammaRI and its key downstream molecule Syk, as well as the increased production of the effector cytokines, interleukin (IL)-10 and IL-4. Interestingly, after transient BBB opening, TLR4 expression was also increased. However, the expression of MyD88, the central adapter of the TLR4 pathway, was significantly inhibited, with decreased production of the effector cytokines IL-12a and IL-1beta. These results indicate that, after transient BBB opening, FcgammaRI-mediated antiinflammatory processes were activated, whereas TLR4-mediated proinflammatory activities were inhibited in microglia and astrocytes. This may represent an important neuroprotective mechanism of microglia and astrocytes that limits excessive inflammation after BBB opening.
Collapse
Affiliation(s)
- Ying-Na Li
- Institute of Neuroscience, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | | | | | | | | | | | | |
Collapse
|
37
|
Vemula S, Roder KE, Yang T, Bhat GJ, Thekkumkara TJ, Abbruscato TJ. A functional role for sodium-dependent glucose transport across the blood-brain barrier during oxygen glucose deprivation. J Pharmacol Exp Ther 2009; 328:487-95. [PMID: 18981287 PMCID: PMC2630371 DOI: 10.1124/jpet.108.146589] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 10/30/2008] [Indexed: 11/22/2022] Open
Abstract
In the current study, we determined the functional significance of sodium-dependent/-independent glucose transporters at the neurovasculature during oxygen glucose deprivation (OGD). Confluent brain endothelial cells cocultured with astrocytes were exposed to varying degrees of in vitro stroke conditions. Glucose transporter (GLUT) 1 and sodium glucose cotransporter (SGLT) activity were investigated by luminal membrane uptake and transport studies using [(3)H]D-glucose and also by [(14)C]alpha-methyl D-glucopyranoside (AMG), a specific, nonmetabolized substrate of SGLT. In vivo middle cerebral artery occlusion experiments were tested to determine whether blood-brain barrier (BBB) SGLT activity was induced during ischemia. Increases in luminal D-glucose and AMG uptake and transport were observed with in vitro stroke conditions. Specific inhibitor experiments suggest a combined role for both SGLT and GLUT1 at the BBB during OGD. A time-dependent increase in the uptake of AMG was also seen in mice exposed to permanent focal ischemia, and this increase was sensitive to the SGLT inhibitor, phlorizin. Infarct and edema ratio during ischemia were significantly decreased by the inhibition of this transporter. These results show that both GLUT1 and SGLT play a role at the BBB in the blood-to-brain transport of glucose during ischemic conditions, and inhibition of SGLT during stroke has the potential to improve stroke outcome. Pharmacological modulation of this novel BBB transporter could prove to be a brain vascular target in stroke.
Collapse
Affiliation(s)
- Sharanya Vemula
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79016, USA
| | | | | | | | | | | |
Collapse
|
38
|
Beyond vessels: occurrence and regional clustering of vascular endothelial (VE-)cadherin-containing junctions in non-endothelial cells. Cell Tissue Res 2008; 335:49-65. [DOI: 10.1007/s00441-008-0718-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2008] [Accepted: 10/02/2008] [Indexed: 10/21/2022]
|
39
|
The development of stroke therapeutics: promising mechanisms and translational challenges. Neuropharmacology 2008; 56:329-41. [PMID: 19007799 DOI: 10.1016/j.neuropharm.2008.10.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 09/29/2008] [Accepted: 10/06/2008] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is the second most common cause of death worldwide and a major cause of disability. Intravenous thrombolysis with rt-PA remains the only available acute therapy in patients who present within 3h of stroke onset other than the recently approved mechanical MERCI device, substantiating the high unmet need in available stroke therapeutics. The development of successful therapeutic strategies remains challenging, as evidenced by the continued failures of new therapies in clinical trials. However, significant lessons have been learned and this knowledge is currently being incorporated into improved pre-clinical and clinical design. Furthermore, advancements in imaging technologies and continued progress in understanding biological pathways have established a prolonged presence of salvageable penumbral brain tissue and have begun to elucidate the natural repair response initiated by ischemic insult. We review important past and current approaches to drug development with an emphasis on implementing principles of translational research to achieve a rigorous conversion of knowledge from bench to bedside. We highlight current strategies to protect and repair brain tissue with the promise to provide longer therapeutic windows, preservation of multiple tissue compartments and improved clinical success.
Collapse
|
40
|
Vincent T, Saikali P, Cayrol R, Roth AD, Bar-Or A, Prat A, Antel JP. Functional Consequences of Neuromyelitis Optica-IgG Astrocyte Interactions on Blood-Brain Barrier Permeability and Granulocyte Recruitment. THE JOURNAL OF IMMUNOLOGY 2008; 181:5730-7. [DOI: 10.4049/jimmunol.181.8.5730] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
41
|
Wolburg H, Noell S, Mack A, Wolburg-Buchholz K, Fallier-Becker P. Brain endothelial cells and the glio-vascular complex. Cell Tissue Res 2008; 335:75-96. [DOI: 10.1007/s00441-008-0658-9] [Citation(s) in RCA: 297] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 06/03/2008] [Indexed: 02/01/2023]
|
42
|
Cronin M, Anderson PN, Cook JE, Green CR, Becker DL. Blocking connexin43 expression reduces inflammation and improves functional recovery after spinal cord injury. Mol Cell Neurosci 2008; 39:152-60. [PMID: 18617007 DOI: 10.1016/j.mcn.2008.06.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 06/01/2008] [Accepted: 06/04/2008] [Indexed: 11/17/2022] Open
Abstract
After traumatic CNS injury, a cascade of secondary events expands the initial lesion. The gap-junction protein connexin43 (Cx43), which is transiently up-regulated, has been implicated in the spread of 'bystander' damage. We have used an antisense oligodeoxynucleotide (asODN) to suppress Cx43 up-regulation in two rat models of spinal cord injury. Within 24 h of compression injury, rats treated with Cx43-asODN scored higher than sense-ODN and vehicle-treated controls on behavioural tests of locomotion. Their spinal cords showed less swelling and tissue disruption, less up-regulation of astrocytic GFAP, and less extravasation of fluorescently-labelled bovine serum albumin and neutrophils. The locomotor improvement was sustained over at least 4 weeks. Following partial spinal cord transection, Cx43-asODN treatment reduced GFAP immunoreactivity, neutrophil recruitment, and the activity of OX42(+) microglia in and around the lesion site. Cx43 has many potential roles in the pathophysiology of CNS injury and may be a valuable target for therapeutic intervention.
Collapse
Affiliation(s)
- Michael Cronin
- Research Department of Cell and Developmental Biology, University College London, London, UK
| | | | | | | | | |
Collapse
|
43
|
Expression of transferrin binding protein in the capillaries of the brain in the developing chick embryo. Neurochem Res 2008; 33:2288-93. [PMID: 18459044 DOI: 10.1007/s11064-008-9716-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Accepted: 04/10/2008] [Indexed: 10/22/2022]
Abstract
Transferrin-binding protein (TfBP) has been shown to be a novel protein, structurally related to the chicken heat shock protein 108. The physiological function of this protein, however, has not yet been established. Antiserum to TfBP selectively stains transferrin- and iron-rich oligodendrocytes and choroidal epithelium in the adult and embryonic chick brain, suggesting a role for this protein in transferrin and iron storage in these cells. In this study, we further demonstrate TfBP-immunoreactivity (IR) in the blood vessels of the embryonic chick central nervous system. A strong TfBP-IR was present in blood vessels from E6, declined from E10 and was absent by E18. Thus, the expression of the TfBP in the blood vessels precedes its expression in the oligodendrocytes. At the subcellular level, TfBP-IR was confined to the cytoplasm of capillary pericytes while the Tf-receptor IR was associated with the capillary endothelium of the brain. The up-regulated expression of TfBP, together with the Tf-receptor of the brain capillaries, suggests that pericytes may be associated with the high iron uptake required for the metabolic demands of the developing brain.
Collapse
|
44
|
Culot M, Lundquist S, Vanuxeem D, Nion S, Landry C, Delplace Y, Dehouck MP, Berezowski V, Fenart L, Cecchelli R. An in vitro blood-brain barrier model for high throughput (HTS) toxicological screening. Toxicol In Vitro 2008; 22:799-811. [PMID: 18280105 DOI: 10.1016/j.tiv.2007.12.016] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Revised: 12/14/2007] [Accepted: 12/20/2007] [Indexed: 11/18/2022]
Abstract
There is a growing interest to use in vitro BBB cell assays in early safety assessment of compounds. By modifying a well-validated co-culture model of brain capillary endothelial and glial cells, developed by Dehouck et al. [Dehouck, M.P., Meresse, S., Delorme, P., Fruchart, J.C., Cecchelli, R., 1990. An easier, reproducible, and mass-production method to study the blood-brain barrier in vitro. Journal of Neurochemistry 54 (5), 1798-1801], it has been possible to develop a new in vitro BBB system suitable for high throughput screening (HTS). In addition, this new procedure substantially reduces the use of experimental animals and considerably facilitates the process of obtaining a functional in vitro BBB model. The model is ready to use after only 4 days of culture and then shows the typical expression and localization of tight junction proteins. The function of the P-glycoprotein and the transcriptional expression of other efflux transporters such as MRP 1, 4 and 5 have been demonstrated. In addition, the model produces a good in vitro/in vivo correlation for 10 compounds (R2=0.81). Furthermore, studies were undertaken within the European ACuteTox consortium with the objective to assess BBB toxicity and make risk assessments of potentially toxic compounds according to their predicted ability to reach the CNS compartment. These investigations demonstrated that the results produced in the HTS BBB model were similar to the standard co-culture model.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/biosynthesis
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Blood-Brain Barrier/drug effects
- Blotting, Western
- Carrier Proteins/metabolism
- Cattle
- Cells, Cultured
- Chemistry, Pharmaceutical
- Data Interpretation, Statistical
- Drug Evaluation, Preclinical/methods
- Endothelial Cells/drug effects
- Endothelium, Vascular/cytology
- Endothelium, Vascular/drug effects
- Microscopy, Fluorescence
- Permeability/drug effects
- Pharmaceutical Preparations/metabolism
- RNA/biosynthesis
- RNA/isolation & purification
- Reverse Transcriptase Polymerase Chain Reaction
- Tight Junctions/drug effects
Collapse
Affiliation(s)
- Maxime Culot
- Laboratoire de Physiopathologie de la Barrière Hémato Encéphalique, Université d'Artois, EA 2465-IMPRT-IFR 114, Faculté Jean Perrin, Rue Jean Souvraz, SP 18 62307 Lens, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Miecz D, Januszewicz E, Czeredys M, Hinton BT, Berezowski V, Cecchelli R, Nałecz KA. Localization of organic cation/carnitine transporter (OCTN2) in cells forming the blood-brain barrier. J Neurochem 2007; 104:113-23. [PMID: 17995936 DOI: 10.1111/j.1471-4159.2007.05024.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Carnitine beta-hydroxy-gamma-(trimethylammonio)butyrate - a compound necessary in the peripheral tissues for a transfer of fatty acids for their oxidation within the cell, accumulates in the brain despite low beta-oxidation in this organ. In order to enter the brain, carnitine has to cross the blood-brain barrier formed by capillary endothelial cells which are in close interaction with astrocytes. Previous studies, demonstrating expression of mRNA coding two carnitine transporters - organic cation/carnitine transporter 2 (OCTN2) and B(0,+) in endothelial cells, did not give any information on carnitine transporters polarity in endothelium. Therefore more detailed experiments were performed on expression and localization of a high affinity carnitine transporter OCTN2 in an in vitro model of the blood-brain barrier by real-time PCR, western blot analysis, and immunocytochemistry. The amount of mRNA was comparable in endothelial cells and kidney, when referred to house-keeping genes, it was, however, significantly lower in astrocytes. Polarity of OCTN2 localization was further studied in an in vitro model of the blood-brain barrier with use of anti-OCTN2 antibodies. Z-axis analysis of the confocal microscope pictures of endothelial cells, with anti-P-glycoprotein antibodies as the marker of apical membrane, showed OCTN2 localization at the basolateral membrane and in the cytoplasmic region in the vicinity of nuclei. Localization of OCTN2 suggest that carnitine can be also transported from the brain, playing an important role in removal of certain acyl esters.
Collapse
Affiliation(s)
- Dorota Miecz
- Nencki Institute of Experimental Biology, Warszawa, Poland
| | | | | | | | | | | | | |
Collapse
|
46
|
Dohgu S, Nishioku T, Sumi N, Takata F, Nakagawa S, Naito M, Tsuruo T, Yamauchi A, Shuto H, Kataoka Y. Adverse effect of cyclosporin A on barrier functions of cerebral microvascular endothelial cells after hypoxia-reoxygenation damage in vitro. Cell Mol Neurobiol 2007; 27:889-99. [PMID: 17934807 PMCID: PMC11517116 DOI: 10.1007/s10571-007-9209-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Accepted: 08/28/2007] [Indexed: 10/22/2022]
Abstract
Hypoxia and post-hypoxic reoxygenation induces disruption of the blood-brain barrier (BBB). Alterations of the BBB function after hypoxia/reoxygenation (H/R) injury remain unclear. Cyclosporin A (CsA), a potent immunosuppressant, induces neurotoxic effects by entering the brain, although the transport of CsA across the BBB is restricted by P-glycoprotein (P-gp), a multidrug efflux pump, and tight junctions of the brain capillary endothelial cells. The aim of this study was to evaluate whether the BBB after H/R damage is vulnerable to CsA-induced BBB dysfunction. We attempted to establish a pathophysiological BBB model with immortalized mouse brain capillary endothelial (MBEC4) cells. The effects of CsA on permeability and P-gp activity of the MBEC4 cells were then examined. Exposure to hypoxia for 4 h and reoxygenation for 1 h (H/R (4 h/1 h)) produced a significant decrease in P-gp function of MBEC4 cells, without changing cell viability and permeability for sodium fluorescein and Evan's blue-albumin at 7 days after H/R (4 h/1 h). CsA-induced hyperpermeability and P-gp dysfunction in MBEC4 monolayers at 7 days after H/R (4 h/1 h) were exacerbated. The possibility that CsA penetrates the BBB with incomplete functions in the vicinity of cerebral infarcts to induce neurotoxicity has to be considered.
Collapse
Affiliation(s)
- Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| | - Tsuyoshi Nishioku
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| | - Noriko Sumi
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| | - Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| | - Shinsuke Nakagawa
- Department of Pharmacology 1, Graduate School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523 Japan
| | - Mikihiko Naito
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032 Japan
| | - Takashi Tsuruo
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo, 113-0032 Japan
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-10-6 Ariake, Koto-ku, Tokyo, 135-8550 Japan
| | - Atsushi Yamauchi
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| | - Hideki Shuto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| | - Yasufumi Kataoka
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180 Japan
| |
Collapse
|
47
|
Brillault J, Lam TI, Rutkowsky JM, Foroutan S, O'Donnell ME. Hypoxia effects on cell volume and ion uptake of cerebral microvascular endothelial cells. Am J Physiol Cell Physiol 2007; 294:C88-96. [PMID: 17942640 DOI: 10.1152/ajpcell.00148.2007] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increased transport of Na across an intact blood-brain barrier (BBB) contributes to cerebral edema formation in ischemic stroke. Our previous studies have shown that ischemic factors stimulate activity of a luminal BBB Na-K-Cl cotransporter, and we have hypothesized that during ischemia, the cotransporter together with the abluminal Na/K pump mediates increased transport of Na from blood into the brain. However, it is possible that elevated Na-K-Cl cotransporter activity could also cause cell swelling if it outpaces ion efflux pathways. The present study was conducted to evaluate the effects of hypoxia on intracellular volume of BBB cells. Cerebral microvascular endothelial cell (CMEC) monolayers were exposed to varying levels of hypoxia for 1 to 5 h in an O(2)-controlled glove box, and cell volume was assessed using 3-O-methyl-D-[(3)H]glucose and [(14)C]sucrose as markers of total and extracellular water space, respectively. Cells exposed to either 7.5%, 3%, or 1% O(2) showed gradual increases in volume (compared with 19% O(2) normoxic controls) that became significant after 3 or more hours. By ion chromatography methods, we also found that a 30-min exposure to 7.5% O(2) caused an increase in bumetanide-sensitive net Na uptake by the cells without increasing cell Na content. CMEC Na content was significantly increased, however, following 3 or more hours of exposure to 7.5% O(2). These findings are consistent with the hypothesis that during cerebral ischemia, the BBB Na-K-Cl cotransporter is stimulated to mediate transendothelial uptake of Na into the brain and that increased cotransporter activity also contributes to gradual swelling of the cells.
Collapse
Affiliation(s)
- Julien Brillault
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
48
|
Noell S, Fallier-Becker P, Beyer C, Kröger S, Mack AF, Wolburg H. Effects of agrin on the expression and distribution of the water channel protein aquaporin-4 and volume regulation in cultured astrocytes. Eur J Neurosci 2007; 26:2109-18. [PMID: 17927773 DOI: 10.1111/j.1460-9568.2007.05850.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Agrin is a heparan sulfate proteoglycan of the extracellular matrix and is known for organizing the postsynaptic differentiation of the neuromuscular junction. Increasing evidence also suggests roles for agrin in the developing CNS, including the formation and maintenance of the blood-brain barrier. Here we describe effects of agrin on the expression and distribution of the water channel protein aquaporin-4 (AQP4) and on the swelling capacity of cultured astrocytes of newborn mice. If astrocytes were cultured on a substrate containing poly DL-ornithine, anti-AQP4 immunoreactivity was evenly and diffusely distributed. If, however, astrocytes were cultured in the presence of agrin-conditioned medium, we observed an increase in the intensity of AQP4-specific membrane-associated staining. Freeze-fracture studies revealed a clustering of orthogonal arrays of particles, representing a structural equivalent of AQP4, when exogenous agrin was present in the astrocyte cultures. Neuronal and non-neuronal agrin isoforms (agrin A0B0 and agrin A4B8, respectively) were able to induce membrane-associated AQP4 staining. Water transport capacity as well as the density of orthogonal arrays of intramembranous particles was increased in astrocytes cultured with the neuronal agrin isoform A4B8, but not with the endothelial and meningeal isoform A0B0. RT-PCR demonstrated that agrin A4B8 increased the level of the M23 splice variant of AQP4 and decreased the level of the M1 splice variant of AQP4. Implications for the regulation and maintenance of the blood-brain barrier including oedema formation under pathological conditions are discussed.
Collapse
Affiliation(s)
- Susan Noell
- Institute of Pathology, University of Tübingen, Liebermeisterstrasse 8, D-72076 Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Cecchelli R, Berezowski V, Lundquist S, Culot M, Renftel M, Dehouck MP, Fenart L. Modelling of the blood–brain barrier in drug discovery and development. Nat Rev Drug Discov 2007; 6:650-61. [PMID: 17667956 DOI: 10.1038/nrd2368] [Citation(s) in RCA: 458] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The market for neuropharmaceuticals is potentially one of the largest sectors of the global pharmaceutical market owing to the increase in average life expectancy and the fact that many neurological disorders have been largely refractory to pharmacotherapy. The brain is a delicate organ that can efficiently protect itself from harmful compounds and precisely regulate its microenvironment. Unfortunately, the same mechanisms can also prove to be formidable hurdles in drug development. An improved understanding of the regulatory interfaces that exist between blood and brain may provide novel and more effective strategies to treat neurological disorders.
Collapse
Affiliation(s)
- Romeo Cecchelli
- Laboratoire de physiopathologie de la barrière hémato-encéphalique E.A.2465, IMPRT IFR-114, Université d'Artois, Faculté Jean Perrin, 62307 Lens, France.
| | | | | | | | | | | | | |
Collapse
|
50
|
López-Atalaya JP, Roussel BD, Ali C, Maubert E, Petersen KU, Berezowski V, Cecchelli R, Orset C, Vivien D. Recombinant Desmodus rotundus salivary plasminogen activator crosses the blood-brain barrier through a low-density lipoprotein receptor-related protein-dependent mechanism without exerting neurotoxic effects. Stroke 2007; 38:1036-43. [PMID: 17325305 DOI: 10.1161/01.str.0000258100.04923.84] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Desmoteplase, a recombinant form of the plasminogen activator DSPAalpha1 from Desmodus rotundus, may offer improved clinical benefits for acute ischemic stroke treatment over the current therapy, recombinant tissue plasminogen activator (rtPA). Accumulating evidence suggests that clinical use of rtPA could be limited by unfavorable properties, including its ability to cross the blood-brain barrier (BBB), thus potentially adding to the pro-excitotoxic effect of endogenous tPA in cerebral parenchyma. Here, to investigate whether desmoteplase may display a safer profile than the structurally-related tPA, both agents were compared for their ability to cross the BBB and promote neurotoxicity. METHODS First, the passage of vascular DSPA and rtPA was investigated in vitro in a model of BBB, subjected or not to oxygen and glucose deprivation. Second, we studied DSPA- and rtPA-mediated effects in an in vivo paradigm of excitotoxic necrosis. RESULTS The rtPA and desmoteplase cross the intact BBB by LRP-mediated transcytosis. Under conditions of oxygen and glucose deprivation, translocation rates of both compounds increased; however, unlike rtPA, desmoteplase transport remained LRP-dependent. Additionally, neither intracerebral nor intravenous desmoteplase administration enhanced NMDA-induced excitotoxic striatal damage in vivo. Interestingly, intravenous but not intrastriatal coadministration of desmoteplase and rtPA reduced the pro-excitotoxic effect of rtPA. CONCLUSIONS We show that desmoteplase crosses the BBB but does not promote neuronal death. Moreover, intravenous administration of desmoteplase antagonizes the neurotoxicity induced by vascular rtPA. This action may be caused by competition of desmoteplase with rtPA for LRP binding at the BBB, thus effectively blocking rtPA access to the brain parenchyma.
Collapse
|