1
|
Mughis H, Lye P, Imperio GE, Bloise E, Matthews SG. Hypoxia modulates P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) drug transporters in brain endothelial cells of the developing human blood-brain barrier. Heliyon 2024; 10:e30207. [PMID: 38737275 PMCID: PMC11088273 DOI: 10.1016/j.heliyon.2024.e30207] [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: 07/20/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/14/2024] Open
Abstract
P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP) multidrug resistance (MDR) transporters are localized at the luminal surface of the blood-brain barrier (BBB). They confer fetal brain protection against harmful compounds that may be circulating in the peripheral blood. The fetus develops in low oxygen levels; however, some obstetric pathologies such as pre-eclampsia, placenta accreta/previa may result in even greater fetal hypoxic states. We investigated how hypoxia impacts MDR transporters in human fetal brain endothelial cells (hfBECs) derived from early and mid-stages of pregnancy. Hypoxia decreased BCRP protein and activity in hfBECs derived in early pregnancy. In contrast, in hfBECs derived in mid-pregnancy there was an increase in P-gp and BCRP activity following hypoxia. Results suggest a hypoxia-induced reduction in fetal brain protection in early pregnancy, but a potential increase in transporter-mediated protection at the BBB during mid-gestation. This would modify accumulation of various key physiological and pharmacological substrates of P-gp and BCRP in the developing fetal brain and potentially contribute to the pathogenesis of neurodevelopmental disorders commonly associated with in utero hypoxia.
Collapse
Affiliation(s)
- Hafsah Mughis
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Phetcharawan Lye
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Guinever E. Imperio
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
| | - Enrrico Bloise
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Departmento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Stephen G. Matthews
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Sinai Health System, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Obstetrics & Gynaecology, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Milane LS, Dolare S, Ren G, Amiji M. Combination Organelle Mitochondrial Endoplasmic Reticulum Therapy (COMET) for Multidrug Resistant Breast Cancer. J Control Release 2023; 363:435-451. [PMID: 37717658 DOI: 10.1016/j.jconrel.2023.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/21/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
It is time for the story of mitochondria and intracellular communication in multidrug resistant cancer to be rewritten. Herein we characterize the extent and cellular advantages of mitochondrial network fusion in multidrug resistant (MDR) breast cancer and have designed a novel nanomedicine that disrupts mitochondrial network fusion and systematically manipulates organelle fusion and function. Combination Organelle Mitochondrial Endoplasmic reticulum Therapy (COMET) is an innovative translational nanomedicine for treating MDR triple negative breast cancer (TNBC) that has superior safety and equivalent efficacy to the current standard of care (paclitaxel). Our study has demonstrated that the increased mitochondrial networks in MDR TNBC contribute to apoptotic resistance and network fusion is mediated by mitofusin2 (MFN2) on the outer mitochondrial membrane. COMET consists of three components; Mitochondrial Network Disrupting (MiND) nanoparticles (NPs) that are loaded with an anti-MFN2 peptide, tunicamycin, and Bam7. The therapeutic rationale of COMET is to reduce the apoptotic threshold in MDR cells with MiND NPs, followed by inducing the endoplasmic reticulum mediated unfolded protein response (UPR) by stressing MDR cells with tunicamycin, and finally, directly inducing mitochondrial apoptosis with Bam7 which is a specific bcl-2 Bax activator. MiND NPs are PEGylated liposomes with the 21 amino acid (2577.98 MW) anti-MFN2 peptide compartmentalized in the aqueous core. Hypoxia (0.5% oxygen) was used to create MDR derivatives of MDA-MB-231 cells and BT-549 cells. Mitochondrial networks were quantified using 3D analysis of 60× live cell images acquired with a Keyence BZ-X710 microscope and MiND NPs effectively fragmented mitochondrial networks in drug sensitive and MDR TNBC cells. The IC50 values, combination index, and dose reduction index derived from dose response studies demonstrate that MiND NPs decrease the apoptotic threshold of both drug sensitive and MDR TNBC cells and COMET is a synergistic drug combination. Complex V (ATP synthase) extracted from bovine cardiac mitochondria was used to assess the effect of MiND NPs on OXPHOS; both MiND NPs and anti-MFN2 peptide solution significantly decrease the activity of mitochondrial complex V and decrease the capacity of OXPHOS. A BacMam viral vector based fluorescent biosensor was used to quantify the unfolded protein response (UPR) at the level of the endoplasmic reticulum and tunicamycin specifically induces the UPR in drug sensitive and MDR TNBC cells. A caspase 3 colorimetric assay demonstrated that the synergistic triple drug combination of COMET increases the ability of Bam7 to specifically induce apoptosis. Dose limiting toxicity and off target effects are a significant challenge for current chemotherapy regimens including paclitaxel. COMET has significantly lower cytotoxicity than paclitaxel in human embryonic kidney epithelial cells and has the potential to fulfill the clinical need for safer cancer therapeutics. COMET is a promising early stage translational nanomedicine for treating MDR TNBC. Manipulating intracellular communication and organelle fusion is a novel approach to treating MDR cancer. The data from this study has rewritten the story of mitochondria, organelle fusion, and intracellular communication and by targeting this intersection, COMET is an exciting new chapter in cancer therapeutics that could transform the clinical outcome of MDR TNBC.
Collapse
Affiliation(s)
- Lara Scheherazade Milane
- Northeastern University, Department of Pharmaceutical Sciences, 360 Huntington Ave, Boston, MA 02116, United States of America.
| | - Saket Dolare
- Northeastern University, Department of Pharmaceutical Sciences, 360 Huntington Ave, Boston, MA 02116, United States of America
| | - Guangwen Ren
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, United States of America
| | - Mansoor Amiji
- Northeastern University, Department of Pharmaceutical Sciences, 360 Huntington Ave, Boston, MA 02116, United States of America
| |
Collapse
|
3
|
Jeleń A, Świechowski R, Żebrowska-Nawrocka M, Sałagacka-Kubiak A, Szmajda-Krygier D, Gałecki P, Balcerczak E. Importance of selected ABCB1 SNPs for the level of severity of depressive symptoms and effectiveness of recurrent depressive disorder therapy. Gene X 2022; 851:147021. [DOI: 10.1016/j.gene.2022.147021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/07/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
|
4
|
Ronaldson PT, Davis TP. Transport Mechanisms at the Blood-Brain Barrier and in Cellular Compartments of the Neurovascular Unit: Focus on CNS Delivery of Small Molecule Drugs. Pharmaceutics 2022; 14:1501. [PMID: 35890396 PMCID: PMC9324459 DOI: 10.3390/pharmaceutics14071501] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is a primary origin of morbidity and mortality in the United States and around the world. Indeed, several research projects have attempted to discover new drugs or repurpose existing therapeutics to advance stroke pharmacotherapy. Many of these preclinical stroke studies have reported positive results for neuroprotective agents; however, only one compound (3K3A-activated protein C (3K3A-APC)) has advanced to Phase III clinical trial evaluation. One reason for these many failures is the lack of consideration of transport mechanisms at the blood-brain barrier (BBB) and neurovascular unit (NVU). These endogenous transport processes function as a "gateway" that is a primary determinant of efficacious brain concentrations for centrally acting drugs. Despite the knowledge that some neuroprotective agents (i.e., statins and memantine) are substrates for these endogenous BBB transporters, preclinical stroke studies have largely ignored the role of transporters in CNS drug disposition. Here, we review the current knowledge on specific BBB transporters that either limit drug uptake into the brain (i.e., ATP-binding cassette (ABC) transporters) or can be targeted for optimized drug delivery (i.e., solute carrier (SLC) transporters). Additionally, we highlight the current knowledge on transporter expression in astrocytes, microglia, pericytes, and neurons with an emphasis on transport mechanisms in these cell types that can influence drug distribution within the brain.
Collapse
Affiliation(s)
- Patrick T. Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724-5050, USA;
| | | |
Collapse
|
5
|
Duan Y, Bai X, Yang J, Zhou Y, Gu W, Liu G, Wang Q, Zhu J, La L, Li X. Exposure to High-Altitude Environment is Associated with Drug Transporters Change: miR-873-5p-Mediated Alteration of Function and Expression Levels of Drug Transporters under Hypoxia. Drug Metab Dispos 2021; 50:174-186. [PMID: 34844996 DOI: 10.1124/dmd.121.000681] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022] Open
Abstract
Hypoxia is the main characteristic of a high-altitude environment, affect ing drug metabolism. However, so far, the mechanism of miRNA involved in the regulation of drug metabolism and transporters under high-altitude hypoxia is still unclear. This study aims to investigate the function s and expression levels of multidrug resistance protein 1 ( MDR1 ), m ultidrug resistance-associated protein 2 ( MRP2 ), breast cancer resistance protein ( BCRP ) , peptide transport 1 (PEPT1), and organic anion-transporting polypeptides 2B1 (OATP2B1) in rats and Caco-2 cells after exposure to high - altitude hypoxia. The protein and mRNA expression of MDR1 , MRP2, BCRP, PEPT1, and OATP2B1 were determined by Western blot and qPCR. The function s of MDR1 , MRP2, BCRP, PEPT1, and OATP2B1 were evaluated by determining the effective intestinal permeability and a bsorption rate constants of their specific substrates in rats under high-altitude hypoxia , and uptake and transport studies were performed on Caco-2 cells . To screen the miRNA associated with hypoxia, Caco-2 cells were examined by high throughput sequencing . We observed that the miR-873-5p was significantly decreased under hypoxia and might target MDR1 and pregnane X receptor ( PXR). To clarify whether miR-873-5p regulates MDR1 and pregnane X receptor (PXR) under hypoxia, Caco-2 cells were transfected with mimics or inhibitors of miR-873-5p and negative control (NC). The function and expression of drug transporters were found to be significantly increased in rats and Caco-2 cells under hypoxia. We found that miR-873-5p regulated MDR1 and PXR expression. Herein, it is shown that miRNA may affect the expression of drug transporter and nuclear receptor under hypoxia. Significance Statement This study explores if alterations to the microRNAs, induced by high-altitude hypoxia, can be translated to altered drug transporters. Among miRNAs, which show a significant change in a hypoxic environment, miR-873-5p can act on the MDR1 gene; however, there are multiple miRNAs that can act on the PXR. We speculate that the miRNA-PXR-Drug transporter axis is important in the physiological disposition of drugs. The results of this study are anticipated to be helpful for rational pharmaceutical use in high - altitude environments .
Collapse
Affiliation(s)
- Yabin Duan
- Department of Clinical Pharmacy,, Qinghai University Affiliated Hospital, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Czornyj L, Auzmendi J, Lazarowski A. Transporter hypothesis in pharmacoresistant epilepsies Is it at the central or peripheral level? Epilepsia Open 2021; 7 Suppl 1:S34-S46. [PMID: 34542938 PMCID: PMC9340303 DOI: 10.1002/epi4.12537] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 12/22/2022] Open
Abstract
The multidrug‐resistance (MDR) phenotype is typically observed in patients with refractory epilepsy (RE) whose seizures are not controlled despite receiving several combinations of more than two antiseizure medications (ASMs) directed against different ion channels or neurotransmitter receptors. Since the use of bromide in 1860, more than 20 ASMs have been developed; however, historically ~30% of cases of RE with MDR phenotype remains unchanged. Irrespective of metabolic biotransformation, the biodistribution of ASMs and their metabolites depends on the functional expression of some ATP‐binding cassette transporters (ABC‐t) in different organs, such as the blood‐brain barrier (BBB), bowel, liver, and kidney, among others. ABC‐t, such as P‐glycoprotein (P‐gp), multidrug resistance–associated protein (MRP‐1), and breast cancer–resistance protein (BCRP), are mainly expressed in excretory organs and play a critical role in the pharmacokinetics (PK) of all drugs. The transporter hypothesis can explain pharmacoresistance to a broad spectrum of ASMs, even when administered simultaneously. Since ABC‐t expression can be induced by hypoxia, inflammation, or seizures, a high frequency of uncontrolled seizures increases the risk of RE. These stimuli can induce ABC‐t expression in excretory organs and in previously non‐expressing (electrically responsive) cells, such as neurons or cardiomyocytes. In this regard, an alternative mechanism to the classical pumping function of P‐gp indicates that P‐gp activity can also produce a significant reduction in resting membrane potential (ΔΨ0 = −60 to −10 mV). P‐gp expression in neurons and cardiomyocytes can produce membrane depolarization and participate in epileptogenesis, heart failure, and sudden unexpected death in epilepsy. On this basis, ABC‐t play a peripheral role in controlling the PK of ASMs and their access to the brain and act at a central level, favoring neuronal depolarization by mechanisms independent of ion channels or neurotransmitters that current ASMs cannot control.
Collapse
Affiliation(s)
- Liliana Czornyj
- Neurology Service, "Juan P. Garrahan" National Children's Hospital, Buenos Aires, Argentina
| | - Jerónimo Auzmendi
- Institute for Research in Physiopathology and Clinical Biochemistry (INFIBIOC), Clinical Biochemistry Department, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,National Council for Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Alberto Lazarowski
- Institute for Research in Physiopathology and Clinical Biochemistry (INFIBIOC), Clinical Biochemistry Department, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
7
|
Melana JP, Mignolli F, Stoyanoff T, Aguirre MV, Balboa MA, Balsinde J, Rodríguez JP. The Hypoxic Microenvironment Induces Stearoyl-CoA Desaturase-1 Overexpression and Lipidomic Profile Changes in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13122962. [PMID: 34199164 PMCID: PMC8231571 DOI: 10.3390/cancers13122962] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Clear cell renal cell carcinoma (ccRCC) is characterized by a high rate of cell proliferation and an extensive accumulation of lipids. Uncontrolled cell growth usually generates areas of intratumoral hypoxia that define the tumor phenotype. In this work, we show that, under these microenvironmental conditions, stearoyl-CoA desaturase-1 is overexpressed. This enzyme induces changes in the cellular lipidomic profile, increasing the oleic acid levels, a metabolite that is essential for cell proliferation. This work supports the idea of considering stearoyl-CoA desaturase-1 as an exploitable therapeutic target in ccRCC. Abstract Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of renal cell carcinoma (RCC). It is characterized by a high cell proliferation and the ability to store lipids. Previous studies have demonstrated the overexpression of enzymes associated with lipid metabolism, including stearoyl-CoA desaturase-1 (SCD-1), which increases the concentration of unsaturated fatty acids in tumor cells. In this work, we studied the expression of SCD-1 in primary ccRCC tumors, as well as in cell lines, to determine its influence on the tumor lipid composition and its role in cell proliferation. The lipidomic analyses of patient tumors showed that oleic acid (18:1n-9) is one of the major fatty acids, and it is particularly abundant in the neutral lipid fraction of the tumor core. Using a ccRCC cell line model and in vitro-generated chemical hypoxia, we show that SCD-1 is highly upregulated (up to 200-fold), and this causes an increase in the cellular level of 18:1n-9, which, in turn, accumulates in the neutral lipid fraction. The pharmacological inhibition of SCD-1 blocks 18:1n-9 synthesis and compromises the proliferation. The addition of exogenous 18:1n-9 to the cells reverses the effects of SCD-1 inhibition on cell proliferation. These data reinforce the role of SCD-1 as a possible therapeutic target.
Collapse
Affiliation(s)
- Juan Pablo Melana
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del Nordeste Argentino (IQUIBA-NEA), Universidad Nacional del Nordeste, Consejo Nacional de Investigaciones Científicas y Técnicas (UNNE-CONICET), Corrientes 3400, Argentina; (J.P.M.); (T.S.); (M.V.A.)
| | - Francesco Mignolli
- Instituto de Botánica del Nordeste, Facultad de Ciencias Agrarias (UNNE-CONICET), Universidad Nacional del Nordeste, Corrientes 3400, Argentina;
| | - Tania Stoyanoff
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del Nordeste Argentino (IQUIBA-NEA), Universidad Nacional del Nordeste, Consejo Nacional de Investigaciones Científicas y Técnicas (UNNE-CONICET), Corrientes 3400, Argentina; (J.P.M.); (T.S.); (M.V.A.)
| | - María V. Aguirre
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del Nordeste Argentino (IQUIBA-NEA), Universidad Nacional del Nordeste, Consejo Nacional de Investigaciones Científicas y Técnicas (UNNE-CONICET), Corrientes 3400, Argentina; (J.P.M.); (T.S.); (M.V.A.)
| | - María A. Balboa
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Jesús Balsinde
- Instituto de Biología y Genética Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 47003 Valladolid, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Correspondence: (J.B.); (J.P.R.); Tel.: +34-983-423-062 (J.B.); Tel.: +54-937-9469-4464 (J.P.R.)
| | - Juan Pablo Rodríguez
- Laboratorio de Investigaciones Bioquímicas de la Facultad de Medicina (LIBIM), Instituto de Química Básica y Aplicada del Nordeste Argentino (IQUIBA-NEA), Universidad Nacional del Nordeste, Consejo Nacional de Investigaciones Científicas y Técnicas (UNNE-CONICET), Corrientes 3400, Argentina; (J.P.M.); (T.S.); (M.V.A.)
- Correspondence: (J.B.); (J.P.R.); Tel.: +34-983-423-062 (J.B.); Tel.: +54-937-9469-4464 (J.P.R.)
| |
Collapse
|
8
|
Lithium enhances post-stroke blood-brain barrier integrity, activates the MAPK/ERK1/2 pathway and alters immune cell migration in mice. Neuropharmacology 2020; 181:108357. [PMID: 33065166 DOI: 10.1016/j.neuropharm.2020.108357] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/21/2022]
Abstract
Lithium induces neuroprotection against cerebral ischemia, although the underlying mechanisms remain elusive. We have previously suggested a role for lithium in calcium regulation and (extra)cerebral vessel relaxation under non-ischemic conditions. Herein, we aimed to investigate whether or not lithium contributes to post-stroke stabilization of the blood-brain barrier (BBB) in mice. Using an oxygen-glucose-deprivation (OGD) model, we first analyzed the impact of lithium treatment on endothelial cells (EC) in vitro. Indeed, such treatment of EC exposed to OGD resulted in increased cell survival as well as in enhanced expression of tight junction proteins and P-glycoprotein. Additional in vivo studies demonstrated an increased stabilization of the BBB upon lithium treatment in stroke mice, as shown by a reduced Evans blue extravasation and an elevation of tight junction protein expression. Furthermore, stabilization of the BBB as a consequence of lithium treatment was associated with an inhibition of matrix metalloproteinase-9 activity, independent of calveolin-1 regulation. In line with this, flow cytometry analysis revealed that lithium treatment led to a decreased neutrophil invasion and an increased T cell extravasation from the blood compartment towards the brain parenchyma. We finally identified the pro-survival MAPK/ERK1/2 pathway as the key regulator of the impact of lithium on the BBB. In conclusion, we demonstrate for the first time that lithium is able to enhance post-stroke BBB integrity. Importantly, our work delivers novel insights into the exact mechanism of lithium-induced acute neuroprotection, providing critical information for future clinical trials involving lithium treatment in stroke patients.
Collapse
|
9
|
Wang F, Ji S, Wang M, Liu L, Li Q, Jiang F, Cen J, Ji B. HMGB1 promoted P-glycoprotein at the blood-brain barrier in MCAO rats via TLR4/NF-κB signaling pathway. Eur J Pharmacol 2020; 880:173189. [PMID: 32417325 DOI: 10.1016/j.ejphar.2020.173189] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/07/2020] [Accepted: 05/10/2020] [Indexed: 01/28/2023]
Abstract
P-glycoprotein (P-gp) is located on the luminal surface of brain vascular endothelium and its status may determine the delivery of the agents into the brain tissues. Previous study showed that upregulation of P-gp at the blood-brain barrier (BBB) after ischemic stroke were mediated by nuclear factor-B (NF-kB) and tumour necrosis factor-α (TNF-α). Based on middle cerebral artery occlusion (MCAO) rats and oxygen-glucose deprivation (OGD) in co-culture of rat brain microvessel endothelial cells (rBMECs) and astrocytes system, the present data indicated that potentiated P-gp expression and activity in brain microvessels or rBMECs were associated with the increase in high-mobility group box 1 (HMGB1), Toll-like receptor 4 (TLR4) and activation of NF-kB and that HMGB1 can release from nucleus to the cytoplasm in activated astrocytes, then into the medium. Moreover, changes in TLR4, TIR domain-containing adaptor protein (TIRAP), NF-kB and P-gp in rBMECs were attenuated by addition of 1 mM ethyl pyruvate (EP), 10 μM TAK-242 and 10 μM pyrrolidine dithiocarbamate (PDTC), respectively. These results demonstrated that HMGB1 promoted P-gp at the BBB after cerebral ischemia via TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Fei Wang
- Department of Cerebral Surgery, The Second People's Hospital of Zhengzhou, Zhengzhou, 450000, People's Republic of China; Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China
| | - Shenglan Ji
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China
| | - Muxi Wang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China; Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, 61801, USA
| | - Lu Liu
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China
| | - Qiaoling Li
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China
| | - Fuxia Jiang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China.
| | - Biansheng Ji
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, 475004, People's Republic of China.
| |
Collapse
|
10
|
Gil-Martins E, Barbosa DJ, Silva V, Remião F, Silva R. Dysfunction of ABC transporters at the blood-brain barrier: Role in neurological disorders. Pharmacol Ther 2020; 213:107554. [PMID: 32320731 DOI: 10.1016/j.pharmthera.2020.107554] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/07/2020] [Indexed: 12/14/2022]
Abstract
ABC (ATP-binding cassette) transporters represent one of the largest and most diverse superfamily of proteins in living species, playing an important role in many biological processes such as cell homeostasis, cell signaling, drug metabolism and nutrient uptake. Moreover, using the energy generated from ATP hydrolysis, they mediate the efflux of endogenous and exogenous substrates from inside the cells, thereby reducing their intracellular accumulation. At present, 48 ABC transporters have been identified in humans, which were classified into 7 different subfamilies (A to G) according to their phylogenetic analysis. Nevertheless, the most studied members with importance in drug therapeutic efficacy and toxicity include P-glycoprotein (P-gp), a member of the ABCB subfamily, the multidrug-associated proteins (MPRs), members of the ABCC subfamily, and breast cancer resistance protein (BCRP), a member of the ABCG subfamily. They exhibit ubiquitous expression throughout the human body, with a special relevance in barrier tissues like the blood-brain barrier (BBB). At this level, they play a physiological function in tissue protection by reducing or limiting the brain accumulation of neurotoxins. Furthermore, dysfunction of ABC transporters, at expression and/or activity level, has been associated with many neurological diseases, including epilepsy, multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis. Additionally, these transporters are strikingly associated with the pharmacoresistance to central nervous system (CNS) acting drugs, because they contribute to the decrease in drug bioavailability. This article reviews the signaling pathways that regulate the expression and activity of P-gp, BCRP and MRPs subfamilies of transporters, with particular attention at the BBB level, and their mis-regulation in neurological disorders.
Collapse
Affiliation(s)
- Eva Gil-Martins
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Daniel José Barbosa
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal.
| | - Vera Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Fernando Remião
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| | - Renata Silva
- UCIBIO-REQUIMTE, Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
11
|
Auzmendi J, Palestro P, Blachman A, Gavernet L, Merelli A, Talevi A, Calabrese GC, Ramos AJ, Lazarowski A. Cannabidiol (CBD) Inhibited Rhodamine-123 Efflux in Cultured Vascular Endothelial Cells and Astrocytes Under Hypoxic Conditions. Front Behav Neurosci 2020; 14:32. [PMID: 32256321 PMCID: PMC7090129 DOI: 10.3389/fnbeh.2020.00032] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/17/2020] [Indexed: 12/14/2022] Open
Abstract
Despite the constant development of new antiepileptic drugs (AEDs), more than 30% of patients develop refractory epilepsy (RE) characterized by a multidrug-resistant (MDR) phenotype. The “transporters hypothesis” indicates that the mechanism of this MDR phenotype is the overexpression of ABC transporters such as P-glycoprotein (P-gp) in the neurovascular unit cells, limiting access of the AEDs to the brain. Recent clinical trials and basic studies have shown encouraging results for the use of cannabinoids in RE, although its mechanisms of action are still not fully understood. Here, we have employed astrocytes and vascular endothelial cell cultures subjected to hypoxia, to test the effect of cannabidiol (CBD) on the P-gp-dependent Rhodamine-123 (Rho-123) efflux. Results show that during hypoxia, intracellular Rho-123 accumulation after CBD treatment is similar to that induced by the P-gp inhibitor Tariquidar (Tq). Noteworthy, this inhibition is like that registered in non-hypoxia conditions. Additionally, docking studies predicted that CBD could behave as a P-gp substrate by the interaction with several residues in the α-helix of the P-gp transmembrane domain. Overall, these findings suggest a direct effect of CBD on the Rho-123 P-gp-dependent efflux activity, which might explain why the CBD add-on treatment regimen in RE patients results in a significant reduction in seizure frequency.
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Instituto de Fisiopatología y Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Pablo Palestro
- Laboratorio de Investigaciones Bioactivas y Desarrollo, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad de La Plata, La Plata, Argentina
| | - Agustín Blachman
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luciana Gavernet
- Laboratorio de Investigaciones Bioactivas y Desarrollo, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad de La Plata, La Plata, Argentina
| | - Amalia Merelli
- Instituto de Fisiopatología y Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alan Talevi
- Laboratorio de Investigaciones Bioactivas y Desarrollo, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad de La Plata, La Plata, Argentina
| | - Graciela Cristina Calabrese
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Lazarowski
- Instituto de Fisiopatología y Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
12
|
Merelli A, Ramos AJ, Lazarowski A, Auzmendi J. Convulsive Stress Mimics Brain Hypoxia and Promotes the P-Glycoprotein (P-gp) and Erythropoietin Receptor Overexpression. Recombinant Human Erythropoietin Effect on P-gp Activity. Front Neurosci 2019; 13:750. [PMID: 31379495 PMCID: PMC6652211 DOI: 10.3389/fnins.2019.00750] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/05/2019] [Indexed: 12/19/2022] Open
Abstract
Erythropoietin (EPO) is not only a hormone that promotes erythropoiesis but also has a neuroprotective effect on neurons attributed to its known anti-apoptotic action. Previously, our group has demonstrated that recombinant-human EPO (rHu-EPO) can protect neurons and recovery motor activity in a chemical focal brain hypoxia model (Merelli et al., 2011). We and others also have reported that repetitive seizures can mimic a hypoxic- like condition by HIF-1α nuclear translocation and high neuronal expression P-gp. Here, we report that a single 20-min status epilepticus (SE) induces P-gp and EPO-R expression in cortical pyramidal neurons and only P-gp expression in astrocytes. In vitro, excitotoxic stress (300 μM glutamate, 5 min), can also induce the expression of EPO-R and P-gp simultaneously with both HIF-1α and NFkB nuclear translocation in primary cortical neurons. Primary astrocytes exposed to chemical hypoxia with CoCl2 (0.3 mM, 6 h) increased P-gp expression as well as an increased efflux of Rhodamine 123 (Rho123) that is a P-gp substrate. Tariquidar, a specific 3er generation P-gp-blocker was used as an efflux inhibitor control. Astrocytes treated with rHu-EPO showed a significant recovery of the Rho123 retention in a similar way as seen by Tariquidar, demonstrating for first time that rHu-EPO can inhibit the P-gp-dependent efflux activity. Taking together, these data suggest that stimulation of EPO depending signaling system could not only play a central role in brain cell protection, but this system could be a new tool for reverse the pharmacoresistant phenotype in refractory epilepsy as well as in other pharmacoresistant hypoxic brain diseases expressing P-gp.
Collapse
Affiliation(s)
- Amalia Merelli
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Javier Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" IBCN-UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alberto Lazarowski
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jeronimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" IBCN-UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
13
|
Cen J, Zhao N, Huang WW, Liu L, Xie YY, Gan Y, Wang CJ, Ji BS. Polyamine analogue QMA attenuated ischemic injury in MCAO rats via ERK and Akt activated Nrf2/HO-1 signaling pathway. Eur J Pharmacol 2019; 844:165-174. [DOI: 10.1016/j.ejphar.2018.12.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/03/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
|
14
|
Ostrowski RP, Zhang JH. The insights into molecular pathways of hypoxia-inducible factor in the brain. J Neurosci Res 2018; 98:57-76. [PMID: 30548473 DOI: 10.1002/jnr.24366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022]
Abstract
The objectives of this present work were to review recent developments on the role of hypoxia-inducible factor (HIF) in the survival of cells under normoxic versus hypoxic and inflammatory brain conditions. The dual nature of HIF effects appears well established, based on the accumulated evidence of HIF playing both the role of adaptive factor and mediator of cell demise. Cellular HIF responses depend on pathophysiological conditions, developmental phase, comorbidities, and administered medications. In addition, HIF-1α and HIF-2α actions may vary in the same tissues. The multiple roles of HIF in stem cells are emerging. HIF not only regulates expression of target genes and thereby influences resultant protein levels but also contributes to epigenetic changes that may reciprocally provide feedback regulations loops. These HIF-dependent alterations in neurological diseases and its responses to treatments in vivo need to be examined alongside with a functional status of subjects involved in such studies. The knowledge of HIF pathways might be helpful in devising HIF-mimetics and modulating drugs, acting on the molecular level to improve clinical outcomes, as exemplified here by clinical and experimental data of selected brain diseases, occasionally corroborated by the data from disorders of other organs. Because of complex role of HIF in brain injuries, prospective therapeutic interventions need to differentially target HIF responses depending on their roles in the molecular mechanisms of neurologic diseases.
Collapse
Affiliation(s)
- Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - John H Zhang
- Departments of Anesthesiology and Physiology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
15
|
Auzmendi J, Buchholz B, Salguero J, Cañellas C, Kelly J, Men P, Zubillaga M, Rossi A, Merelli A, Gelpi RJ, Ramos AJ, Lazarowski A. Pilocarpine-Induced Status Epilepticus Is Associated with P-Glycoprotein Induction in Cardiomyocytes, Electrocardiographic Changes, and Sudden Death. Pharmaceuticals (Basel) 2018; 11:ph11010021. [PMID: 29462915 PMCID: PMC5874717 DOI: 10.3390/ph11010021] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 02/07/2018] [Accepted: 02/13/2018] [Indexed: 02/06/2023] Open
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the major cause of death in those patients suffering from refractory epilepsy (RE), with a 24-fold higher risk relative to the normal population. SUDEP risk increases with seizure frequency and/or seizure-duration as in RE and Status Epilepticus (SE). P-glycoprotein (P-gp), the product of the multidrug resistant ABCB1-MDR-1 gene, is a detoxifying pump that extrudes drugs out of the cells and can confer pharmacoresistance to the expressing cells. Neurons and cardiomyocytes normally do not express P-gp, however, it is overexpressed in the brain of patients or in experimental models of RE and SE. P-gp was also detected after brain or cardiac hypoxia. We have previously demonstrated that repetitive pentylenetetrazole (PTZ)-induced seizures increase P-gp expression in the brain, which is associated with membrane depolarization in the hippocampus, and in the heart, which is associated with fatal SE. SE can produce hypoxic-ischemic altered cardiac rhythm (HIACR) and severe arrhythmias, and both are related with SUDEP. Here, we investigate whether SE induces the expression of hypoxia-inducible transcription factor (HIF)-1α and P-gp in cardiomyocytes, which is associated with altered heart rhythm, and if these changes are related with the spontaneous death rate. SE was induced in Wistar rats once a week for 3 weeks, by lithium-pilocarpine-paradigm. Electrocardiograms, HIF-1α, and P-gp expression in cardiomyocytes, were evaluated in basal conditions and 72 h after SE. All spontaneous deaths occurred 48 h after each SE was registered. We observed that repeated SE induced HIF-1α and P-gp expression in cardiomyocytes, electrocardiographic (ECG) changes, and a high rate of spontaneous death. Our results suggest that the highly accumulated burden of convulsive stress results in a hypoxic heart insult, where P-gp expression may play a depolarizing role in cardiomyocyte membranes and in the development of the ECG changes, such as QT interval prolongation, that could be related with SUDEP. We postulate that this mechanism could explain, in part, the higher SUDEP risk in patients with RE or SE.
Collapse
Affiliation(s)
- Jerónimo Auzmendi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Bruno Buchholz
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Jimena Salguero
- Departamento de Fisicomatemática, Laboratorio de Radioisótopos, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Carlos Cañellas
- Laboratorio Tecnonuclear SA, Arias 4176, Buenos Aires C1430CRP, Argentina.
| | - Jazmín Kelly
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Paula Men
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Marcela Zubillaga
- Departamento de Fisicomatemática, Laboratorio de Radioisótopos, Cátedra de Física, Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Alicia Rossi
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Amalia Merelli
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| | - Ricardo J Gelpi
- Departamento de Patología, Instituto de Fisiopatología Cardiovascular (INFICA), Universidad de Buenos Aires, Facultad de Medicina, Buenos Aires C1121ABG, Argentina.
| | - Alberto J Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Profesor E. De Robertis" IBCN UBA-CONICET, Buenos Aires CP1121, Argentina.
| | - Alberto Lazarowski
- Departamento de Bioquímica Clínica, Instituto de Investigaciones en Fisiopatología y Bioquímica Clínica (INFIBIOC), Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Junín 956, Buenos Aires C1113AAD, Argentina.
| |
Collapse
|
16
|
Merelli A, Rodríguez JCG, Folch J, Regueiro MR, Camins A, Lazarowski A. Understanding the Role of Hypoxia Inducible Factor During Neurodegeneration for New Therapeutics Opportunities. Curr Neuropharmacol 2018; 16:1484-1498. [PMID: 29318974 PMCID: PMC6295932 DOI: 10.2174/1570159x16666180110130253] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 11/24/2017] [Accepted: 01/08/2018] [Indexed: 12/14/2022] Open
Abstract
Neurodegeneration (NDG) is linked with the progressive loss of neural function with intellectual and/or motor impairment. Several diseases affecting older individuals, including Alzheimer's disease, Amyotrophic Lateral Sclerosis, Huntington's disease, Parkinson's disease, stroke, Multiple Sclerosis and many others, are the most relevant disorders associated with NDG. Since other pathologies such as refractory epilepsy, brain infections, or hereditary diseases such as "neurodegeneration with brain iron accumulation", also lead to chronic brain inflammation with loss of neural cells, NDG can be said to affect all ages. Owing to an energy and/or oxygen supply imbalance, different signaling mechanisms including MAPK/PI3K-Akt signaling pathways, glutamatergic synapse formation, and/or translocation of phosphatidylserine, might activate some central executing mechanism common to all these pathologies and also related to oxidative stress. Hypoxia inducible factor 1-α (HIF-1α) plays a twofold role through gene activation, in the sense that this factor has to "choose" whether to protect or to kill the affected cells. Most of the afore-mentioned processes follow a protracted course and are accompanied by progressive iron accumulation in the brain. We hypothesize that the neuroprotective effects of iron chelators are acting against the generation of free radicals derived from iron, and also induce sufficient -but not excessive- activation of HIF-1α, so that only the hypoxia-rescue genes will be activated. In this regard, the expression of the erythropoietin receptor in hypoxic/inflammatory neurons could be the cellular "sign" to act upon by the nasal administration of pharmacological doses of Neuro-EPO, inducing not only neuroprotection, but eventually, neurorepair as well.
Collapse
Affiliation(s)
| | | | | | | | | | - Alberto Lazarowski
- Address correspondence to this author at the Clinical Biochemistry Department, School of Pharmacy and Biochemistry, University of Buenos Aires-Argentina, Junín 954, Buenos Aires-Argentina; Tel: +54-11-5950-8674;, E-mail:
| |
Collapse
|
17
|
Transplantation of Human Umbilical Cord Blood Mononuclear Cells Attenuated Ischemic Injury in MCAO Rats via Inhibition of NF-κB and NLRP3 Inflammasome. Neuroscience 2017; 369:314-324. [PMID: 29175152 DOI: 10.1016/j.neuroscience.2017.11.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 12/28/2022]
Abstract
Accumulated evidence displayed that transplantation of stem cells may be a promising approach for the treatment of neurological disorders. However, the underlying mechanisms remain to be well elucidated. Moreover, some investigators cannot reproduce similar results as the previous. The present results showed that transplantation of fresh human umbilical cord blood mononuclear cells (cbMNCs) attenuated ischemic damage in middle cerebral artery occlusion (MCAO) rats, accompanied with improvement of neurologic deficits, learning and memory function. The increase in neovascularization and related molecules such as vascular endothelial growth factor (VEGF), Angiopoietin-1 (Ang-1) and endothelium-specific receptor tyrosine kinase 2 (Tie-2) in the injured brain was observed in cbMNCs-treated rats. Moreover, nuclear factor-κB (NF-κB) activation and nucleotide binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome were also inhibited by the cells graft, resulting in reduction in cleaved caspase-1 and mature interleukin-1β (IL-1β) content. These results suggested that the protective actions of the cells on the cerebral ischemia may be related to inhibition of NF-κB pathway and NLRP3 inflammasome.
Collapse
|
18
|
DeMars KM, Yang C, Hawkins KE, McCrea AO, Siwarski DM, Candelario-Jalil E. Spatiotemporal Changes in P-glycoprotein Levels in Brain and Peripheral Tissues Following Ischemic Stroke in Rats. J Exp Neurosci 2017; 11:1179069517701741. [PMID: 28469478 PMCID: PMC5398227 DOI: 10.1177/1179069517701741] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/02/2017] [Indexed: 01/19/2023] Open
Abstract
P-glycoprotein (P-gp) is known to transport a diverse array of xenobiotics, including therapeutic drugs. A member of the ATP-binding cassette (ABC) transporter family, P-gp is a protein encoded by the gene Mdr1 in humans and Abcb1 in rodents (represented by 2 isoforms Abcb1a and Abcb1b). Lining the luminal and abluminal membrane of brain capillary endothelial cells, P-gp is a promiscuous efflux pump extruding a variety of exogenous toxins and drugs. In this study, we measured dynamic changes in Abcb1a and Abcb1b transcripts and P-gp protein in the brain, liver, and kidney after experimental stroke. P-glycoprotein has been shown to increase in brain endothelial cells following hypoxia in vitro or after exposure to proinflammatory cytokines. Using a rat model of ischemic stroke, we hypothesized that P-gp expression will be increased in the brain, liver, and kidney in response to neuroinflammation following ischemic stroke. Adult Sprague Dawley rats underwent middle cerebral artery occlusion (MCAO) for 90 minutes and were killed at 4, 14, 24, and 48 hours postreperfusion onset to determine the time course of P-gp expression. To mimic ischemia occurring at the blood-brain barrier, rat brain endothelial (RBE4) cells were subjected to hypoxia and low glucose (HLG) for 16 hours. Immunoblotting analyses showed P-gp increases in brain and liver following 90-minute MCAO, as well as in cultured RBE4 cells after 16-hour HLG treatment, but fluctuated in the kidney depending on the time point. The relative roles of each isoform in the protein expression were analyzed with quantitative reverse transcriptase polymerase chain reaction. Ischemic stroke leads to significant increases in P-gp levels not only in the brain but also in the liver. The increase in P-gp could dramatically reduce the bioavailability and efficacy of neuroprotective drugs. Therefore, P-gp represents a big hurdle to drug delivery to the ischemic brain.
Collapse
Affiliation(s)
- Kelly M DeMars
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Austin O McCrea
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David M Siwarski
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
19
|
Qosa H, Mohamed LA, Alqahtani S, Abuasal BS, Hill RA, Kaddoumi A. Transporters as Drug Targets in Neurological Diseases. Clin Pharmacol Ther 2016; 100:441-453. [PMID: 27447939 DOI: 10.1002/cpt.435] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 12/13/2022]
Abstract
Membrane transport proteins have central physiological function in maintaining cerebral homeostasis. These transporters are expressed in almost all cerebral cells in which they regulate the movement of a wide range of solutes, including endogenous substrates, xenobiotic, and therapeutic drugs. Altered activity/expression of central nervous system (CNS) transporters has been implicated in the onset and progression of multiple neurological diseases. Neurological diseases are heterogeneous diseases that involve complex pathological alterations with only a few treatment options; therefore, there is a great need for the development of novel therapeutic treatments. To that end, transporters have emerged recently to be promising therapeutic targets to halt or slow the course of neurological diseases. The objective of this review is to discuss implications of transporters in neurological diseases and summarize available evidence for targeting transporters as decent therapeutic approach in the treatment of neurological diseases.
Collapse
Affiliation(s)
- H Qosa
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - L A Mohamed
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - S Alqahtani
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - B S Abuasal
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - R A Hill
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA
| | - A Kaddoumi
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, USA.
| |
Collapse
|
20
|
Lam J, Baello S, Iqbal M, Kelly LE, Shannon PT, Chitayat D, Matthews SG, Koren G. The ontogeny of P-glycoprotein in the developing human blood-brain barrier: implication for opioid toxicity in neonates. Pediatr Res 2015; 78:417-21. [PMID: 26086643 DOI: 10.1038/pr.2015.119] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/23/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Neonates have been shown to have a heightened sensitivity to the central depressive effects of opioids compared to older infants and adults. The limited development of P-glycoprotein (P-gp) may limit the ability of the neonate to efflux morphine from the brain back to the systemic circulation. The objective of the study was to determine the ontogeny of P-gp in the human brain. METHODS Postmortem cortex samples from gestational age (GA) 20-26 wk, GA 36-40 wk, postnatal age (PNA) 0-3 mo, PNA 3-6 mo, and adults were immunostained for P-gp. RESULTS The intensity of P-gp staining in adults was significantly higher compared to at GA 20-26 wk (P < 0.05), GA 36-40 wk (P < 0.05), and PNA 0-3 mo (P < 0.05). P-gp intensity at GA 20-26 wk (P < 0.05), GA 36-40 wk (P < 0.05), and PNA 0-3 mo (P < 0.05) was significantly lower compared to at PNA 3-6 mo. CONCLUSION P-gp expression in the brain is limited at birth, increases with postnatal maturation, and reaches adult levels at ~3-6 mo of age. Given the immaturity of blood-brain barrier (BBB) P-gp after birth, morphine may concentrate in the brain. This provides mechanistic support to life threatening opioid toxicity seen with maternal codeine use during breastfeeding.
Collapse
Affiliation(s)
- Jessica Lam
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.,Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephanie Baello
- Department of Physiology, Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Majid Iqbal
- Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lauren E Kelly
- Department of Physiology and Pharmacology, University of Western Ontario, Toronto, Ontario, Canada
| | - Patrick T Shannon
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - David Chitayat
- Department of Obstetrics and Gynaecology, Prenatal Diagnosis and Medical Genetics Program, Mount Sinai Hospital, Toronto, Ontario, Canada.,Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephen G Matthews
- Department of Physiology, Obstetrics and Gynaecology, University of Toronto, Toronto, Ontario, Canada
| | - Gideon Koren
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.,Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Physiology and Pharmacology, University of Western Ontario, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Li W, Li J, Wang R, Xie H, Jia Z. MDR1 will play a key role in pharmacokinetic changes under hypoxia at high altitude and its potential regulatory networks. Drug Metab Rev 2015; 47:191-8. [PMID: 25639892 DOI: 10.3109/03602532.2015.1007012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Some newest studies indicated that drug transports may play the key role in pharmacokinetics changes under hypoxia at high altitude; MDR1 is now known to affect the disposition of many administered drugs and make a major contribution to absorption, distribution, metabolism, excretion. Different expression of MDR1 is frequently found in different normal tissues and tumor cells; it is important to better understand how MDR1 is regulated under hypoxia, which seems to be a complex and highly controlled process. Several signaling pathways and transcription factors have been described as being involved in the regulation of MDR1 expression, such as MAPK/ERK, nuclear factor-kappaB, hypoxia-inducible factor-1a, pregnane × receptor, constitutive androstane receptor and microRNA. Recently, researches have been increasingly appreciating long non-coding RNAs (lncRNAs) as an integral component of gene regulatory networks. lncRNAs play crucial roles in various biological processes ranging from epigenetic gene regulation, transcriptional control, post-transcriptional regulation, pre-mRNA processing and nuclear organization. A last recent research showed that H19 gene non-coding RNA is believed to induce P-glycoprotein expression under hypoxia.
Collapse
Affiliation(s)
- Wenbin Li
- Key Laboratory of the Plateau Environmental Damage Control, Lanzhou General Hospital of Lanzhou Military Command , PLA, Lanzhou , China
| | | | | | | | | |
Collapse
|
22
|
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: 4.9] [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
|
23
|
Kervezee L, Hartman R, van den Berg DJ, Shimizu S, Emoto-Yamamoto Y, Meijer JH, de Lange ECM. Diurnal variation in P-glycoprotein-mediated transport and cerebrospinal fluid turnover in the brain. AAPS JOURNAL 2014; 16:1029-37. [PMID: 24917180 PMCID: PMC4147055 DOI: 10.1208/s12248-014-9625-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 05/19/2014] [Indexed: 12/27/2022]
Abstract
Nearly all bodily processes exhibit circadian rhythmicity. As a consequence, the pharmacokinetic and pharmacodynamic properties of a drug may also vary with time of day. The objective of this study was to investigate diurnal variation in processes that regulate drug concentrations in the brain, focusing on P-glycoprotein (P-gp). This efflux transporter limits the distribution of many drugs in the brain. To this end, the exposure to the P-gp substrate quinidine was determined in the plasma and brain tissue after intravenous administration in rats at six different time points over the 24-h period. Our results indicate that time of administration significantly affects the exposure to quinidine in the brain. Upon inhibition of P-gp, exposure to quinidine in brain tissue is constant over the 24-h period. To gain more insight into processes regulating brain concentrations, we used intracerebral microdialysis to determine the concentration of quinidine in brain extracellular fluid (ECF) and cerebrospinal fluid (CSF) after intravenous administration at two different time points. The data were analyzed by physiologically based pharmacokinetic modeling using NONMEM. The model shows that the variation is due to higher activity of P-gp-mediated transport from the deep brain compartment to the plasma compartment during the active period. Furthermore, the analysis reveals that CSF flux is higher in the resting period compared to the active period. In conclusion, we show that the exposure to a P-gp substrate in the brain depends on time of administration, thereby providing a new strategy for drug targeting to the brain.
Collapse
Affiliation(s)
- Laura Kervezee
- Division of Pharmacology, LACDR, Leiden University, PO Box 9502, 2300 RA, Leiden, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
24
|
Wei J, Fang W, Sha L, Han D, Zhang R, Hao X, Li Y. XQ-1H Suppresses Neutrophils Infiltration and Oxidative Stress Induced by Cerebral Ischemia Injury Both In Vivo and In Vitro. Neurochem Res 2013; 38:2542-2549. [PMID: 24122081 DOI: 10.1007/s11064-013-1176-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 09/03/2013] [Accepted: 10/05/2013] [Indexed: 01/09/2023]
Abstract
Cerebral ischemia/reperfusion injury plays an important role in the development of tissue injury after acute stroke, including neutrophils adhesion and infiltration, inflammation and oxidative stress. 10-O-(N,N-dimethylaminoethyl)-ginkgolide B methanesulfonate (XQ-1H) is a novel ginkdolide B derivative. In this study, we investigated the anti-inflammatory and anti-oxidative activities of XQ-1H in vivo and vitro. In our study, rats were treating with XQ-1H (31.2, 15.6 and 7.8 mg/kg) after middle cerebral artery occlusion surgery. Primary cultured cortical rat neurons were treated with Na2S2O4 for 1.5 h to mimic hypoxia and reoxygenation injury in vitro. Cortical neurons were preincubated with XQ-1H (100, 10, 1 μM) 24 h before hypoxic injury. Brain edema was evaluated by brain water content. Neutrophil infiltration was determined by fluorescence imaging method and myeloperoxidase assay. Intercellular adhesion molecule 1 (ICAM-1) and matrix metallopeptidase 9 (MMP-9) expressions were examined by immunohistochemistry analysis. Neuronal injury was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide, lactate dehydrogenase releasing and lactic acid content. The anti-oxidative effects of XQ-1H were evaluated by superoxide dismutase (SOD) activity and malondialdehyde content in ischemic brain and neuron cultures subjected to hypoxia/reoxygenation procedure. Results showed that XQ-1H reduced neutrophils infiltration to ischemic brain, which might result from down regulation of inflammatory mediators, such as ICAM-1 and MMP-9. In addition, an antioxidative effect of XQ-1H was observed in cortical neuron and brain homogenates by enhancing SOD activity and inhibiting lipid peroxidation. These results indicated that XQ-1H possessed a protective effect against cerebral ischemia, especially on neutrophil infiltration and oxidative stress.
Collapse
Affiliation(s)
- Jie Wei
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
25
|
Li MS, Cen J, He L, Liu L, Ji BS. CJY, an isoflavone, interacts with ATPase of P-glycoprotein in the rat brain microvessel endothelial cells (RBMECs). J Chemother 2013; 25:347-54. [PMID: 24090809 DOI: 10.1179/1973947813y.0000000094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Our previous study reported CJY, an isoflavone, can reverse P-glycoprotein (P-gp) efflux activity in rat brain microvessel endothelial cells (RBMECs). In the present report, by assessment of ATPase activity of RBMECs, we gained further insight into the nature of the CJY interactions with P-gp. The results revealed that the basal P-gp ATPase activity was increased by CJY. Kinetic studies on ATPase activity showed the effects of Tetrandrine (Tet) on CJY-stimulated, CsA on CJY-stimulated, and CsA on Tet-stimulated P-gp ATPase activity were all non-competitive inhibition, indicating that these substrates can simultaneously but independently bind to diverse sites on P-gp. Furthermore, the combined effects of CJY with Tet, and CJY with CsA were also evaluated isobolographically. The results showed synergistic interactions in both combinations, implying that combined treatment of CJY with other modulators may exert synergistic interactions for the drug's penetration into the brain and the treatment of neurological disorders.
Collapse
|
26
|
Cen J, Liu L, Li MS, He L, Wang LJ, Liu YQ, Liu M, Ji BS. Alteration in P-glycoprotein at the blood–brain barrier in the early period of MCAO in rats. J Pharm Pharmacol 2013; 65:665-72. [DOI: 10.1111/jphp.12033] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 01/06/2013] [Indexed: 01/16/2023]
Abstract
Abstract
Objectives
The aim of this work was to investigate the alteration in P-glycoprotein (P-gp) at the blood–brain barrier (BBB) after middle cerebral artery occlusion (MCAO) in rats.
Methods
Permanent MCAO was verified via 2,3,5-triphenyltetrazolium staining and hematoxylin-eosin staining. The expression of P-gp, matrix metalloproteinase-2 (MMP-2), MMP-9, claudin-5, tumour necrosis factor-α (TNF-α) and nitric oxide synthase (NOS) at the BBB was evaluated using western blot or immunostaining analysis. The content of fluorescein sodium (NaF), rhodamine-123 and nimodipine in ischaemic brain tissues was determined using high-performance liquid chromatography.
Key findings
Elevated expression of P-gp at the BBB and decreased concentration of P-gp substrates in the ischaemic brain tissues were observed within 4 h after MCAO. However, at 6 h after MCAO, the concentration of P-gp substrates in the ischaemic hemisphere began to rise even though the expression of P-gp was still increased. Moreover, the expression of claudin-5 was decreased; contrarily, the expression of MMP-2, MMP-9, TNF-α as well as NOS gradually increased within 6 h after MCAO.
Conclusions
P-gp plays a crucial role in limiting the entrance of agents into the brain after MCAO and the specific regulation of P-gp expression/activity may provide an important approach for the improvement of pharmacotherapy in ischaemic stroke.
Collapse
Affiliation(s)
- Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, China
| | - Lu Liu
- School of Pharmacy, Henan University, Kaifeng, China
| | - Ming-Shan Li
- School of Medicine, Henan University, Kaifeng, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Li-Juan Wang
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, China
| | - Yan-qing Liu
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, China
| | - Meng Liu
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, China
| | - Bian-Sheng Ji
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng, China
| |
Collapse
|
27
|
Fang W, Lv P, Geng X, Shang E, Yang Q, Sha L, Li Y. Penetration of verapamil across blood brain barrier following cerebral ischemia depending on both paracellular pathway and P-glycoprotein transportation. Neurochem Int 2013; 62:23-30. [DOI: 10.1016/j.neuint.2012.10.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 09/19/2012] [Accepted: 10/28/2012] [Indexed: 11/25/2022]
|
28
|
Kim YO, Kim SY, Yun DH, Lee SW. Association between ABCB1 Polymorphisms and Ischemic Stroke in Korean Population. Exp Neurobiol 2012; 21:164-71. [PMID: 23319877 PMCID: PMC3538181 DOI: 10.5607/en.2012.21.4.164] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 11/16/2012] [Indexed: 01/12/2023] Open
Abstract
Neuronal expression of ATP-binding cassette, sub-family B (MDR/TAP), member 1 (ABCB1) has been demonstrated after brain ischemia. To investigate whether ABCB1 polymorphisms are associated with the development, risk factors (hypertension, dyslipidemia, and diabetes mellitus), severity (National Institutes of Health Stroke Scale, NIHSS), and sequelae (Modified Barthel Index, MBI) of ischemic stroke (IS), four single nucleotide polymorphisms (SNPs) of the ABCB1 gene [rs4148727, promoter, -154T>C; rs3213619, 5'-untranslation region (5'UTR), -129T>C); rs1128503, synonymous, Gly412 (C>T); rs3842, 3'UTR, A>G] were analyzed in 121 IS patients and 291 control subjects. SNPStats and SPSS 18.0 were used to obtain odds ratios (OR), 95% confidence intervals (CI), and p values. Multiple logistic regression models (codominant1, codominant2, dominant, recessive, and log-additive models) were applied to analyze the genetic data. The rs3842 SNP was weakly associated with the development of IS (p=0.020 in codominant1 model and p=0.028 in dominant model). In the analysis of clinical phenotypes, ABCB1 polymorphisms were nominally associated with hypertension (rs3213619 and rs3842, p<0.05), dyslipidemia (rs1128503, p<0.05), diabetes (rs3842, p<0.05), and NIHSS (rs4148727, p<0.05). Interestingly, rs3842 showed statistically strong association between IS with hypertension and IS without hypertension (Fisher's exact p=0.003, OR=0.11, 95% CI=0.03-0.51 in recessive model). These results suggest that the ABCB1 gene may be associated with the development and clinical phenotypes of IS in Korean population.
Collapse
Affiliation(s)
- Young-Ock Kim
- Department of Herbal Crop Research, National Institute of Horticultural & Herbal Science, Rural Development Administration, Eumseong 369-873, Korea
| | | | | | | |
Collapse
|
29
|
Cen J, Liu L, He L, Liu M, Wang CJ, Ji BS. N(1)-(quinolin-2-ylmethyl)butane-1,4-diamine, a polyamine analogue, attenuated injury in in vitro and in vivo models of cerebral ischemia. Int J Dev Neurosci 2012; 30:584-95. [PMID: 22982502 DOI: 10.1016/j.ijdevneu.2012.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 08/31/2012] [Accepted: 08/31/2012] [Indexed: 02/07/2023] Open
Abstract
It has been widely recognized that glutamate (Glu)-induced cytotoxicity, intracellular calcium overload and excessive free radical production are the key players in the development and progression of ischemic brain injury. Since MK-801, an antagonist of N-methyl-d-aspartate (NMDA) receptor, showed many adverse reactions that hampered its clinical applications, development of safe and effective agent for the treatment of cerebral ischemia is eagerly required. This study was to investigate the effects of N(1)-(quinolin-2-ylmethyl)butane-1,4-diamine (QMA), a polyamine analogue, on the in vitro and in vivo models of cerebral ischemic damage. The results revealed that pretreatment with QMA could attenuate Glu, putrescine (Put) and oxygen-glucose deprivation (OGD)-induced cell death, lipid peroxidation as well as the elevation of reactive oxygen species (ROS) and intracellular [Ca(2+)](i) in pheochromocytoma (PC12) cells and in rat primary cortical neurons. The results also demonstrated that QMA could inhibit NMDA-mediated intracellular [Ca(2+)](i) accumulation in rat primary cortical neurons and reduce brain infarct volume in middle cerebral artery occlusion (MCAO) rats. The present report suggested that polyamines played a crucial role in the pathological processes of cerebral ischemic damage and that QMA or other novel polyamine analogues could be promising therapeutic candidates for stroke by virtue of their anti-hypoxia and antioxidation property.
Collapse
Affiliation(s)
- Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, Henan University, Kaifeng 475004, China
| | | | | | | | | | | |
Collapse
|
30
|
O'Brien FE, Dinan TG, Griffin BT, Cryan JF. Interactions between antidepressants and P-glycoprotein at the blood-brain barrier: clinical significance of in vitro and in vivo findings. Br J Pharmacol 2012; 165:289-312. [PMID: 21718296 DOI: 10.1111/j.1476-5381.2011.01557.x] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The drug efflux pump P-glycoprotein (P-gp) plays an important role in the function of the blood-brain barrier by selectively extruding certain endogenous and exogenous molecules, thus limiting the ability of its substrates to reach the brain. Emerging evidence suggests that P-gp may restrict the uptake of several antidepressants into the brain, thus contributing to the poor success rate of current antidepressant therapies. Despite some inconsistency in the literature, clinical investigations of potential associations between functional single nucleotide polymorphisms in ABCB1, the gene which encodes P-gp, and antidepressant response have highlighted a potential link between P-gp function and treatment-resistant depression (TRD). Therefore, co-administration of P-gp inhibitors with antidepressants to patients who are refractory to antidepressant therapy may represent a novel therapeutic approach in the management of TRD. Furthermore, certain antidepressants inhibit P-gp in vitro, and it has been hypothesized that inhibition of P-gp by such antidepressant drugs may play a role in their therapeutic action. The present review summarizes the available in vitro, in vivo and clinical data pertaining to interactions between antidepressant drugs and P-gp, and discusses the potential relevance of these interactions in the treatment of depression.
Collapse
Affiliation(s)
- Fionn E O'Brien
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
31
|
Experimental evidence of the potential use of erythropoietin by intranasal administration as a neuroprotective agent in cerebral hypoxia. ACTA ACUST UNITED AC 2011; 26:65-9. [DOI: 10.1515/dmdi.2011.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
32
|
Merelli A, Caltana L, Girimonti P, Ramos AJ, Lazarowski A, Brusco A. Recovery of Motor Spontaneous Activity After Intranasal Delivery of Human Recombinant Erythropoietin in a Focal Brain Hypoxia Model Induced by CoCl2 in Rats. Neurotox Res 2010; 20:182-92. [DOI: 10.1007/s12640-010-9233-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 11/10/2010] [Accepted: 11/11/2010] [Indexed: 02/04/2023]
|
33
|
Fang W, Geng X, Deng Y, Li Y, Shang E, Cen J, Lv P. Platelet activating factor induces blood brain barrier permeability alteration in vitro. J Neuroimmunol 2010; 230:42-7. [PMID: 20870297 DOI: 10.1016/j.jneuroim.2010.08.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/29/2010] [Accepted: 08/24/2010] [Indexed: 01/26/2023]
Abstract
The purposes of this article were to investigate whether blood brain barrier (BBB) permeability is altered after platelet activating factor (PAF) induced injury in vitro and elucidate the preliminary possible mechanisms of it. MTT method was used to observe cell damage after PAF incubation with rat brain microvessel endothelial cells (RBMECs). Intracellular concentrations of Nimodipine in normal and PAF injured RBMECs were estimated by LC-MS/MS analytical method to estimate BBB permeability. Accumulation of P-glycoprotein (P-gp) substrate rhodamine 123 in normal or PAF injured RBMECs was measured with Poly Immune Analysis System-1420 to evaluate the function of P-gp on RBMECs. Intercellular adhesion molecule-1 (ICAM-1) mRNA and protein expression levels in RBMECs were assayed by RT-PCR and flow cytometry respectively. Results showed that after RBMECs were incubated with 1 μM PAF for 24h, cell survival rate was decreased, and intracellular concentrations of Nimodipine were increased evidently. Rhodamine 123 accumulation between normal and PAF injured cells has no significant difference, but ICAM-1 mRNA and protein expression were increased remarkably in PAF injured cells, which could be inhibited by PAF antagonists. In conclusion, the present study demonstrated that BBB permeability was increased after PAF incubation, and which may be due to ICAM-1 up-regulating but not P-glycoprotein function alteration.
Collapse
Affiliation(s)
- Weirong Fang
- Department of Physiology, China Pharmaceutical University, Nanjing 210009, PR China
| | | | | | | | | | | | | |
Collapse
|
34
|
Aviles-Reyes RX, Angelo MF, Villarreal A, Rios H, Lazarowski A, Ramos AJ. Intermittent hypoxia during sleep induces reactive gliosis and limited neuronal death in rats: implications for sleep apnea. J Neurochem 2010; 112:854-69. [DOI: 10.1111/j.1471-4159.2009.06535.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
35
|
Fifth Santorini Conference Biologie Prospective 2010. Clin Chem Lab Med 2010. [DOI: 10.1515/cclm.2010.551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
36
|
Neuroprotective effects of probenecid in a transgenic animal model of Huntington's disease. J Neural Transm (Vienna) 2009; 116:1079-86. [PMID: 19551467 DOI: 10.1007/s00702-009-0253-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Accepted: 06/02/2009] [Indexed: 01/04/2023]
Abstract
Huntington's disease (HD) is an autosomal dominantly inherited disorder, caused by an expanded polyglutamine region of a protein called huntingtin. The excitotoxicity, oxidative damage and altered membrane transport may have an important role in the pathogenesis of HD. Probenecid is a non-selective inhibitor of multidrug resistance-associated proteins, but it also inhibits organic anion transporters. In this study, we examined the effects of probenecid on the survival, behaviour and immunohistochemical changes in the N171-82Q transgenic mouse model of HD. After probenecid administration, the duration of survival improved by 35%. The motor activity was significantly ameliorated as compared with the control transgenic group. Probenecid treatment significantly reduced the neuronal loss and the number of neuronal intranuclear aggregates. These results suggest that probenecid may exert a neuroprotective effect by increasing the membrane transport of protective compounds, and/or inhibiting the toxic compounds.
Collapse
|
37
|
Caltana L, Merelli A, Lazarowski A, Brusco A. Neuronal and Glial Alterations Due to Focal Cortical Hypoxia Induced by Direct Cobalt Chloride (CoCl2) Brain Injection. Neurotox Res 2009; 15:348-58. [DOI: 10.1007/s12640-009-9038-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Revised: 01/09/2009] [Accepted: 02/24/2009] [Indexed: 01/19/2023]
|
38
|
Murozono M, Matsumoto S, Okada S, Nagaoka D, Isshiki A, Watanabe Y. Reduction of brain infarction induced by a transient brain ischemia in mdr1a knockout mice. Neurochem Res 2009; 34:1555-61. [PMID: 19277863 DOI: 10.1007/s11064-009-9943-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 02/26/2009] [Indexed: 01/16/2023]
Abstract
In order to evaluate the functional role of P-glycoprotein (P-gp) in cerebral ischemia, both multidrug resistance 1a knockout (KO) mice and wild-type mice were subjected to transient focal ischemia under a constant body and brain temperature about 37 degrees C. The results showed that the volume of brain infarction induced by middle cerebral artery occlusion in KO mice was significantly smaller than that seen in wild-type mice, although there were no significant differences in cerebral blood flow, physiological data and on anatomical analysis of cerebrovasculature between both groups. We suggest that multidrug resistance 1a P-gp plays a role for adjusting the expressions of endogenous neuronal cell modulating substances, such as cytokines, neuronal peptides, and others, in the brain, which is consistent with a previous paper (Bobrov et al. Neurosci Lett 24: 6-11, 2008).
Collapse
|
39
|
Roberts DJ, Goralski KB. A critical overview of the influence of inflammation and infection on P-glycoprotein expression and activity in the brain. Expert Opin Drug Metab Toxicol 2008; 4:1245-64. [DOI: 10.1517/17425255.4.10.1245] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
40
|
Daood MJ, Tsai C, Ahdab-Barmada M, Watchko JF. ABC transporter (P-gp/ABCB1, MRP1/ABCC1, BCRP/ABCG2) expression in the developing human CNS. Neuropediatrics 2008; 39:211-8. [PMID: 19165709 PMCID: PMC2821654 DOI: 10.1055/s-0028-1103272] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
P-glycoprotein (P-gp/ABCB1), multidrug resistance associated protein 1 (MRP1/ABCC1), and breast cancer resistance protein (BCRP/ABCG2) are plasma membrane efflux pumps that limit the intracellular uptake and retention of numerous lipophilic, amphipathic xeno- and endobiotics. Little is known about the neonatal and developmental expression of P-gp/ABCB1, MRP1/ABCC1, and BCRP/ABCG2 in the human central nervous system (CNS), therefore post-mortem CNS tissue from infants born at 22 (0/7)-42 (0/7) weeks of gestation and adults was immunostained to determine their ontogeny and cellular localization. P-gp/ABCB1 immunostaining was observed in microvessel endothelial cells as early as 22 (0/7) weeks, increasing in prevalence and intensity with maturation, and later in gestation in large pyramidal neurons. MRP1/ABCC1 immunostaining was prominent early in the choroid plexus and ventricular ependyma, and noted later in large pyramidal neurons. BCRP/ABCG2 expression was limited to microvessel endothelial cells. P-gp/ABCB1, MRP1/ABCC1 and BCRP/ABCG2 in adult brain matched term newborn CNS but with more intense immunostaining. We conclude that P-gp/ABCB1, MRP1/ABCC1, and BCRP/ABCG2 are expressed in a developmental, cell specific, fashion in the human CNS. The complementary pattern of P-gp/ABCB1 and BCRP/ABCG2 at the blood-brain with MRP1/ABCC1 at the blood-CSF barriers may limit CNS uptake and retention of drugs and toxins in neonates.
Collapse
Affiliation(s)
- Monica J. Daood
- Division of Newborn Medicine Department of Pediatrics University of Pittsburgh School of Medicine Magee-Womens Research Institute Pittsburgh, Pennsylvania USA
| | - Cathy Tsai
- Division of Newborn Medicine Department of Pediatrics University of Pittsburgh School of Medicine Magee-Womens Research Institute Pittsburgh, Pennsylvania USA
| | | | - Jon F. Watchko
- Division of Newborn Medicine Department of Pediatrics University of Pittsburgh School of Medicine Magee-Womens Research Institute Pittsburgh, Pennsylvania USA
| |
Collapse
|
41
|
Robey RW, Lazarowski A, Bates SE. P-glycoprotein--a clinical target in drug-refractory epilepsy? Mol Pharmacol 2008; 73:1343-6. [PMID: 18314494 DOI: 10.1124/mol.108.046680] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
ATP-binding cassette transporters such as P-glycoprotein (Pgp), multidrug resistance-associated protein, and breast cancer resistance protein are known to transport a wide range of substrates and are highly expressed in the capillary endothelial cells that form part of the blood-brain barrier. It is noteworthy that P-glycoprotein has been shown to be up-regulated in animal models of refractory epilepsy, and adding a Pgp inhibitor to treatment regimens has been shown to reverse the drug-resistant phenotype. Limited data have suggested a role for Pgp in epilepsy in humans as well. However, few epilepsy drugs have been shown to be transported by Pgp, leading to controversy over whether Pgp actually plays a role in drug-resistant epilepsy. In this issue of Molecular Pharmacology, Bauer et al. (p. 1444) demonstrate that glutamate can cause localized up-regulation of Pgp via cyclooxygenase-2 (COX-2) and that this phenomenon can be prevented with COX-2 inhibitors. Localized rather than global up-regulation of Pgp may explain some of the difficulty investigators have had in proving a role for Pgp in epilepsy. The results add new support for future clinical trials targeting Pgp expression in drug-refractory epilepsy.
Collapse
Affiliation(s)
- Robert W Robey
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
42
|
Xiao-Dong L, Zhi-Hong Y, Hui-Wen Y. Repetitive/temporal hypoxia increased P-glycoprotein expression in cultured rat brain microvascular endothelial cells in vitro. Neurosci Lett 2008; 432:184-7. [PMID: 18241990 DOI: 10.1016/j.neulet.2007.12.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2007] [Revised: 12/07/2007] [Accepted: 12/10/2007] [Indexed: 10/22/2022]
Abstract
The aim of the study was to investigate whether repetitive/temporal hypoxia up-regulated P-glycoprotein (P-gp) in cultured rat brain microvascular endothelial cells (rBMECs). Cultured rBMECs were used as in vitro blood brain barrier (BBB) model. Cells reached confluence were subjected to temporal hypoxic exposure. Under free-glucose cultured medium, the cells were covered by sterile paraffin oil for 15 min, inducing temporal hypoxic exposure. The hypoxic-exposure was carried out once every day up to 8 days, leading to the repetitive/temporal hypoxia in rBMECs. The cell viability was tested using CCK-8 kit, function and levels of P-gp in the cells were measured using rhodamine 123 uptake and western blot, respectively. It was found that 8-temporal hypoxic exposure induced 1.6-fold increase of P-gp level in cells, accompanied by decrease of cellular accumulation of rhodamine 123. Cellular accumulation of phenobarbital was also decreased. These findings indicated that repetitive/temporal hypoxia may be one of the factors resulting in P-gp overexpression in refractory epilepsy.
Collapse
Affiliation(s)
- Liu Xiao-Dong
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China.
| | | | | |
Collapse
|
43
|
Pariante CM. The role of multi-drug resistance p-glycoprotein in glucocorticoid function: studies in animals and relevance in humans. Eur J Pharmacol 2008; 583:263-71. [PMID: 18275949 DOI: 10.1016/j.ejphar.2007.11.067] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2007] [Revised: 10/01/2007] [Accepted: 11/07/2007] [Indexed: 01/16/2023]
Abstract
Entry of glucocorticoid hormones into cells is tightly regulated by membrane transporters. One of these transporters, the multi-drug resistance p-glycoprotein, has been extensively described to confer treatment resistance to tumour cells as well as to regulate the intracellular levels of glucocorticoid hormones. Moreover, multi-drug resistance p-glycoprotein is also present on the endothelial cells of the blood-brain-barrier, and in neurones, where it limits the access of glucocorticoids to the brain. Finally, this transporter also has the ability to limit the entry of some antidepressants to the brain, with potential consequences for the clinical therapeutic effects of these drugs. This review will focus on the studies that have used multi-drug resistance p-glycoprotein knockout animals in such context, and will discuss the potential clinical relevance of these transporters for psychiatric disorders. In particular, we will discuss the reciprocal interactions between this transporter and antidepressants, both as its inhibitors and as its substrates. We believe that the interaction between antidepressants and multi-drug resistance p-glycoprotein is one of the most potentially exciting developments in psychopharmacological research.
Collapse
Affiliation(s)
- Carmine M Pariante
- Section and Laboratory of Stress, Psychiatry and Immunology (SPI-Lab), Institute of Psychiatry, Kings College London, United Kingdom.
| |
Collapse
|
44
|
Ishmael JE, Löhr CV, Fischer K, Kioussi C. Localization of myosin II regulatory light chain in the cerebral vasculature. Acta Histochem 2007; 110:172-7. [PMID: 17961638 DOI: 10.1016/j.acthis.2007.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 08/13/2007] [Accepted: 08/21/2007] [Indexed: 10/22/2022]
Abstract
The cytoskeleton of cerebral microvascular endothelial cells is a critical determinant of blood-brain barrier (BBB) function. Barrier integrity appears to be particularly sensitive to the phosphorylation state of specific residues within myosin regulatory light chain (RLC), one of two accessory light chains of the myosin II motor complex. Phosphorylation of myosin RLC by myosin light chain kinase (MLCK) has been implicated in BBB dysfunction associated with alcohol abuse and hypoxia, whereas dephosphorylation may enhance BBB integrity following exposure to lipid-lowering statin drugs. Using immunohistochemistry we provide evidence of widespread myosin II RLC distribution throughout the cerebral vasculature of the mouse. Light microscopy revealed immunolocalization of myosin II RLC protein in the endothelium of brain capillaries, the endothelial cell layer of arterioles and in association with venules. Immunolabeling of myosin RLC in non-muscle endothelial cells could be distinguished from myosin RLC immunoreactivity associated with the smooth muscle layer of the tunica media in larger muscular arterioles. These findings support an emerging role for myosin II RLC as a component of the actomyosin cytoskeleton of cerebral endothelial cells with the potential to contribute to the selective vulnerability of the brain in vivo.
Collapse
|