Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cells ; 11(24)2022 12 08.
Article in English | MEDLINE | ID: mdl-36552728

ABSTRACT

We previously developed an in vitro model of the human blood-brain barrier (BBB) based on the use of endothelial cells derived from CD34+-hematopoietic stem cells and cultured with brain pericytes. The purpose of the present study was to provide information on the protein expression levels of the transporters, receptors, tight junction/adherence junction molecules, and transporter-associated molecules of human brain-like endothelial cells (hBLECs). The absolute protein expression levels were determined by liquid chromatography-mass spectrometry-based quantitative targeted absolute proteomics and compared with those from human brain microvessels (hBMVs). The protein levels of CD144, CD147, MRP4, Annexin A6 and caveolin-1 showed more than 3-fold abundance in hBLECs, those of MCT1, Connexin 43, TfR1, and claudin-5 showed less than 3-fold differences, and the protein levels of other drug efflux transporters and nutrient transporters were less represented in hBLECs than in hBMVs. It is noteworthy that BCRP was more expressed than MDR1 in hBLECs, as this was the case for hBMVs. These results suggest that transports mediated by MCT1, TfR1, and claudin-5-related tight junction function reflect the in vivo BBB situation. The present study provided a better characterization of hBLECs and clarified the equivalence of the transport characteristics between in vitro BBB models and in vivo BBB models using LC-MS/MS-based protein quantification.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Humans , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Chromatography, Liquid , Proteomics/methods , Claudin-5/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Tandem Mass Spectrometry/methods , Neoplasm Proteins/metabolism , Membrane Transport Proteins/metabolism , Hematopoietic Stem Cells/metabolism
2.
Pharmaceutics ; 13(6)2021 Jun 16.
Article in English | MEDLINE | ID: mdl-34208550

ABSTRACT

Central nervous system (CNS) diseases are one of the top causes of death worldwide. As there is a difficulty of drug penetration into the brain due to the blood-brain barrier (BBB), many CNS drugs treatments fail in clinical trials. Hence, there is a need to develop effective CNS drugs following strategies for delivery to the brain by better selecting them as early as possible during the drug discovery process. The use of in vitro BBB models has proved useful to evaluate the impact of drugs/compounds toxicity, BBB permeation rates and molecular transport mechanisms within the brain cells in academic research and early-stage drug discovery. However, these studies that require biological material (animal brain or human cells) are time-consuming and involve costly amounts of materials and plastic wastes due to the format of the models. Hence, to adapt to the high yields needed in early-stage drug discoveries for compound screenings, a patented well-established human in vitro BBB model was miniaturized and automated into a 96-well format. This replicate met all the BBB model reliability criteria to get predictive results, allowing a significant reduction in biological materials, waste and a higher screening capacity for being extensively used during early-stage drug discovery studies.

3.
J Alzheimers Dis ; 64(4): 1195-1211, 2018.
Article in English | MEDLINE | ID: mdl-30010117

ABSTRACT

The role of ABCA7 in brain homeostasis and Alzheimer's disease (AD) is currently under intense scrutiny, since it has been reported that polymorphisms in the Abca7 gene and a loss of function of the protein are closely linked to excessive accumulation of amyloid peptides and disturbed cholesterol homeostasis. The blood-brain barrier (BBB), which isolates the brain from the blood compartment, is involved in both of these processes. We therefore hypothesized that ABCA7 downregulation might affect cholesterol and amyloid exchanges at the BBB. Using siRNA and primary cultures of mouse endothelial cells purified from brain microvessels and seeded on Transwell ® inserts, we investigated the role of ABCA7 in cholesterol and amyloid exchanges across the BBB. Our results showed that a decrease in ABCA7 expression at the BBB provokes in vitro a reduction in ABCA1 expression and a decrease in APOE secretion. In vitro, these decreases reduce cholesterol exchange across the BBB, particularly for high-density lipoproteins and ApoA-I particles. When ABCA7 was absent, we observed a reduction in Aß peptide basolateral-to-apical transport in the presence of ApoA-I, with non-significant changes in the expression levels of Rage, Lrp1, Abcb1, Abcc1, and Abcg2. Our study in murine BBB model highlighted a putative new role for ABCA7 in AD via the protein's involvement in cholesterol metabolism and amyloid clearance at the BBB.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Cholesterol/metabolism , Homeostasis/physiology , ATP-Binding Cassette Transporters/genetics , Amyloid beta-Peptides/pharmacology , Animals , Apolipoproteins E/genetics , Blood-Brain Barrier/drug effects , Brain/cytology , Cells, Cultured , Claudins/genetics , Claudins/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Endothelial Cells/drug effects , Homeostasis/drug effects , In Vitro Techniques , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection , Zonula Occludens-1 Protein/metabolism
4.
Pharm Res ; 34(12): 2652-2662, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28952054

ABSTRACT

PURPOSE: Alzheimer's disease (AD) may disturb functions of the blood-brain barrier and change the disposition of drugs to the brain. This study assessed the disease-induced changes in drug transporters in the brain capillaries of transgenic AD mice. METHODS: Eighteen drug transporters and four tight junction-associated proteins were analyzed by RT-qPCR in cortex, hippocampus and cerebellum tissue samples of 12-16-month-old APdE9, Tg2576 and APP/PS1 transgenic mice and their healthy age-matched controls. In addition, microvessel fractions enriched from 1-3-month-old APdE9 mice were analyzed using RT-qPCR and Western blotting. Brain transport of methotrexate in APdE9 mice was assessed by in vivo microdialysis. RESULTS: The expression profiles of studied genes were similar in brain tissues of AD and control mice. Instead, in the microvessel fraction in APdE9 mice, >2-fold alterations were detected in the expressions of 11 genes but none at the protein level. In control mice strains, >5-fold changes between different brain regions were identified for Slc15a2, Slc22a3 and occludin. Methotrexate distribution into hippocampus of APdE9 mice was faster than in controls. CONCLUSIONS: The expression profile of mice carrying presenilin and amyloid precursor protein mutations is comparable to controls, but clear regional differences exist in the expression of drug transporters in brain.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Brain/metabolism , Enzyme Inhibitors/pharmacokinetics , Membrane Transport Proteins/metabolism , Methotrexate/pharmacokinetics , Tight Junction Proteins/metabolism , Alzheimer Disease/genetics , Animals , Biological Transport , Brain/blood supply , Brain/drug effects , Disease Models, Animal , Drug Delivery Systems , Drug Discovery , Enzyme Inhibitors/metabolism , Humans , Male , Membrane Transport Proteins/analysis , Membrane Transport Proteins/genetics , Methotrexate/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pharmaceutical Preparations/metabolism , Tight Junction Proteins/analysis , Tight Junction Proteins/genetics , Transcriptome
5.
Front Physiol ; 7: 185, 2016.
Article in English | MEDLINE | ID: mdl-27252658

ABSTRACT

Atherosclerosis is an inflammatory disease that leads to an aberrant accumulation of cholesterol in vessel walls forming atherosclerotic plaques. During this process, the mechanism regulating complex cellular cholesterol pools defined as the reverse cholesterol transport (RCT) is altered as well as expression and functionality of transporters involved in this process, namely ABCA1, ABCG1, and SR-BI. Macrophages, arterial endothelial and smooth muscle cells (SMCs) have been involved in the atherosclerotic plaque formation. As macrophages are widely described as the major cell type forming the foam cells by accumulating intracellular cholesterol, RCT alterations have been poorly studied at the arterial endothelial cell and SMC levels. Amongst the therapeutics tested to actively counteract cellular cholesterol accumulation, the methylated ß-cyclodextrin, KLEPTOSE® CRYSMEß, has recently shown promising effects on decreasing the atherosclerotic plaque size in atherosclerotic mouse models. Therefore we investigated in vitro the RCT process occurring in SMCs and in arterial endothelial cells (ABAE) as well as the ability of some modified ß-CDs with different methylation degree to modify RCT in these cells. To this aim, cells were incubated in the presence of different methylated ß-CDs, including KLEPTOSE® CRYSMEß. Both cell types were shown to express basal levels of ABCA1 and SR-BI whereas ABCG1 was solely found in ABAE. Upon CD treatments, the percentage of membrane-extracted cholesterol correlated to the methylation degree of the CDs independently of the lipid composition of the cell membranes. Decreasing the cellular cholesterol content with CDs led to reduce the expression levels of ABCA1 and ABCG1. In addition, the cholesterol efflux to ApoA-I and HDL particles was significantly decreased suggesting that cells forming the blood vessel wall are able to counteract the CD-induced loss of cholesterol. Taken together, our observations suggest that methylated ß-CDs can significantly reduce the cellular cholesterol content of cells forming atherosclerotic lesions and can subsequently modulate the expression of ABC transporters involved in RCT. The use of methylated ß-CDs would represent a valuable and efficient tool to interfere with atherosclerosis pathogenesis in patients, nonetheless their mode of action still needs further investigations to be fully understood and finely controlled at the cellular level.

6.
J Alzheimers Dis ; 48(3): 849-62, 2015.
Article in English | MEDLINE | ID: mdl-26402114

ABSTRACT

One of the prime features of Alzheimer's disease (AD) is the excessive accumulation of amyloid-ß (Aß) peptides in the brain. Several recent studies suggest that this phenomenon results from the dysregulation of cholesterol homeostasis in the brain and impaired bidirectional Aß exchange between blood and brain. These mechanisms appear to be closely related and are controlled by the blood-brain barrier (BBB) at the brain microvessel level. In animal models of AD, the anticancer drug bexarotene (a retinoid X receptor agonist) has been found to restore cognitive functions and decrease the brain amyloid burden by regulating cholesterol homeostasis. However, the drug's therapeutic effect is subject to debate and the exact mechanism of action has not been characterized. Therefore, the objective of this present study was to determine bexarotene's effects on the BBB. Using an in vitro model of the human BBB, we investigated the drug's effects on cholesterol exchange between abluminal and luminal compartments and the apical-to-basolateral transport of Aß peptides across the BBB. Our results demonstrated that bexarotene induces the expression of ABCA1 but not ApoE. This upregulation correlates with an increase in ApoE2-, ApoE4-, ApoA-I-, and HDL-mediated cholesterol efflux. Regarding the transport of Aß peptides, bexarotene increases the expression of ABCB1, which in turn decreases Aß apical-to-basolateral transport. Our results showed that bexarotene not only promotes the cholesterol exchange between the brain and the blood but also decreases the influx of Aß peptides across BBB, suggesting that bexarotene is a promising drug candidate for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cholesterol/metabolism , Neuroprotective Agents/pharmacology , Tetrahydronaphthalenes/pharmacology , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Anticarcinogenic Agents/pharmacology , Apolipoproteins E/metabolism , Bexarotene , Biological Transport/drug effects , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cattle , Coculture Techniques , Fetal Blood , Humans , Pericytes , Stem Cells , Tight Junctions/drug effects , Tight Junctions/metabolism
7.
Brain Res ; 1594: 15-26, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25451130

ABSTRACT

Several studies have demonstrated that the blood-brain barrier (BBB) (dynamic cellular complex composed by brain capillary endothelial cells (BCECs) and surrounded by astrocytic end feet and pericytes) regulates the exchanges of amyloid ß (Aß) peptide between the blood and the brain. Deregulation of these exchanges seems to be a key trigger for the brain accumulation of Aß peptide observed in Alzheimer's disease (AD). Whereas the involvement of receptor for advanced glycation end-products in Aß peptide transcytosis has been demonstrated in our laboratory, low-density lipoprotein receptor's role at the cellular level needs to be clarified. For this, we used an in vitro BBB model that consists of a co-culture of bovine BCECs and rat glial cells. This model has already been used to characterize low-density lipoprotein receptor-related peptide (LRP)'s involvement in the transcytosis of molecules such as tPA and angiopep-2. Our results suggest that Aß peptide efflux across the BCEC monolayer involves a transcellular transport. However, the experiments with RAP discard an involvement of LRP family members at BCECs level. In contrast, our results show a strong transcriptional expression of LRP1 in pericytes and suggest its implication in Aß endocytosis. Moreover, the observations of pericytes contraction and local downregulation of LRP1 in response to Aß treatment opens up perspectives for studying this cell type with respect to Aß peptide metabolism and AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Pericytes/metabolism , Receptors, LDL/metabolism , Alzheimer Disease/metabolism , Animals , Blotting, Western , Brain/metabolism , Cattle , Coculture Techniques , Endocytosis/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , In Vitro Techniques , Rats , Reverse Transcriptase Polymerase Chain Reaction
8.
Microvasc Res ; 91: 44-57, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24333620

ABSTRACT

Thrombolysis treatment of acute ischemic stroke is limited by the pro-edematous and hemorrhagic effects exerted by reperfusion, which disrupts the blood-brain barrier (BBB) capillary endothelium in the infarct core. Most studies of the ischemic BBB overlook the complexity of the penumbral area, where the affected brain cells are still viable following deprivation. Our present objective was to examine in vitro the kinetic impact of reoxygenation on the integrity of ischemic BBB cells after oxygen-glucose deprivation. Through the use of a co-culture of brain capillary endothelial cells and glial cells, we first showed that the transendothelial permeability increase induced by deprivation can occur with both preserved cell viability and interendothelial tight junction network. The subtle and heterogeneous alteration of the tight junctions was observable only through electron microscopy. A complete permeability recovery was then found after reoxygenation, when Vimentin and Actin networks were reordered. However, still sparse ultrastructural alterations of tight junctions suggested an acquired vulnerability. Endothelial cells were then exposed to recombinant tissue-type plasminogen activator (rtPA) to define a temporal profile for the toxic effect of this thrombolytic on transendothelial permeability. Interestingly, the reoxygenated BBB broke down with aggravated tight junction disruption when exposed to rtPA only at 4h after reoxygenation. Moreover, this breakdown was enhanced by 50% when ischemic glial cells were present during the first hours of reoxygenation. Our results suggest that post-stroke reoxygenation enables retrieval of the barrier function of brain capillary endothelium when in a non-necrotic environment, but may sensitize it to rtPA at the 4-hour time point, when both endothelial breakdown mechanisms and glial secretions could be identified and targeted in a therapeutical perspective.


Subject(s)
Blood Glucose/metabolism , Blood-Brain Barrier/drug effects , Brain/blood supply , Endothelial Cells/metabolism , Oxygen/chemistry , Adenosine Triphosphate/metabolism , Animals , Cattle , Cell Nucleus/metabolism , Cell Survival , Cytoskeleton/metabolism , Endothelium, Vascular/metabolism , Glucose/metabolism , Necrosis , Neuroglia/cytology , Neuroglia/metabolism , Oxidative Stress , Permeability , Rats , Rats, Sprague-Dawley , Stroke/physiopathology , Stroke/therapy , Time Factors , Tissue Plasminogen Activator/metabolism
9.
Brain Res ; 1517: 1-15, 2013 Jun 23.
Article in English | MEDLINE | ID: mdl-23603412

ABSTRACT

It is known that activation of the liver X receptors (LXRs) by natural or synthetic agonists decreases the amyloid burden and enhances cognitive function in transgenic murine models of Alzheimer's disease (AD). Recent evidence suggests that LXR activation may affect the transport of amyloid ß (Aß) peptides across the blood-brain barrier (the BBB, which isolates the brain from the peripheral circulation). By using a well-characterized in vitro BBB model, we demonstrated that LXR agonists (24S-hydroxycholesterol, 27-hydroxycholesterol and T0901317) modulated the expression of target genes involved in cholesterol homeostasis (such as ATP-binding cassette sub-family A member 1 (ABCA1)) and promoted cellular cholesterol efflux to apolipoprotein A-I and high density lipoproteins. Interestingly, we also observed a decrease in Aß peptide influx across brain capillary endothelial cells, although ABCA1 did not appear to be directly involved in this process. By focusing on others receptors and transporters that are thought to have major roles in Aß peptide entry into the brain, we then demonstrated that LXR stimulation provoked an increase in expression of the ABCB1 transporter (also named P-glycoprotein (P-gp)). Further investigations confirmed ABCB1's involvement in the restriction of Aß peptide influx. Taken as a whole, our results not only reinforce the BBB's key role in cerebral cholesterol homeostasis but also demonstrate the importance of the LXR/ABCB1 axis in Aß peptide influx-highlighting an attractive new therapeutic approach whereby the brain could be protected from peripheral Aß peptide entry.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Hydroxycholesterols/pharmacology , ATP Binding Cassette Transporter 1/genetics , Animals , Animals, Newborn , Anticholesteremic Agents/pharmacology , Apolipoprotein A-I/metabolism , Blood-Brain Barrier/drug effects , Brain/cytology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cattle , Cholesterol/metabolism , Cholesterol, HDL/metabolism , Coculture Techniques , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Hydrocarbons, Fluorinated/pharmacology , Neuroglia/cytology , Neuroglia/drug effects , Protein Transport/drug effects , Protein Transport/physiology , Rats , Sulfonamides/pharmacology , Tritium/metabolism , Up-Regulation/drug effects
10.
J Alzheimers Dis ; 30(3): 489-503, 2012.
Article in English | MEDLINE | ID: mdl-22433669

ABSTRACT

In brain, excess cholesterol is metabolized into 24S-hydroxycholesterol (24S-OH-chol) and eliminated into the circulation across the blood-brain barrier. 24S-OH-chol is a natural agonist of the nuclear liver X receptors (LXRs) involved in peripheral cholesterol homeostasis. The effects of this oxysterol on the pericytes embedded in the basal lamina of this barrier (close to the brain compartment) have not been previously studied. We used primary cultures of brain pericytes to demonstrate that the latter express LXR nuclear receptors and their target gene ATP-binding cassette, sub-family A, member 1 (ABCA1), known to be one of the major transporters involved in peripheral lipid homeostasis. Treatment with 24S-OH-chol caused an increase in ABCA1 expression that correlated with a reverse cholesterol transfer to apolipoprotein E, apolipoprotein A-I, and high density lipoprotein particles. Inhibition of ABCA1 decreased this efflux. As pericytes are able to internalize the amyloid-ß peptides which accumulate in brain of Alzheimer's disease patients, we then investigated the effects of 24S-OH-chol on this process. We found that the cellular accumulation process was not modified by 24S-OH-chol treatment. Overall, our results highlight the importance of the LXR/ABCA1 system in brain pericytes and suggest a new role for these cells in brain cholesterol homeostasis.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cholesterol/metabolism , Pericytes/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , Animals , Biological Transport , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/cytology , Brain/drug effects , Cattle , Cells, Cultured , Hydroxycholesterols/pharmacology , Liver X Receptors , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Pericytes/cytology , Pericytes/drug effects
11.
Curr Neurovasc Res ; 8(4): 258-69, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22023614

ABSTRACT

The blood-brain barrier (BBB) is a dynamic cellular complex that is responsible for the maintenance of brain homeostasis. To understand the BBB's key cellular and molecular mechanisms, in vitro models combining endothelial cells and astrocytes can be used to reproduce most of the barrier's in vivo features (low paracellular permeability and the expression of specific transporters). However, these models lack pericytes - a poorly characterized cell type which appears to be of crucial importance to understand BBB's function in healthy and diseased states. The present study sought to identify and characterize this cell population - which lacks a specific marker - by comparing its phenotype with that of vascular smooth muscle cells. Even if pericytes and smooth muscle cells shared many markers in vitro, our results showed that they could be distinguished by their different P-glycoprotein expression and γ-glutamyltranspeptidase activity. Two different three-cell-type culture models were described, including pericytes to mimic the neurovascular unit. In the first model, endothelial cells were cultured alone on a filter, away from glial cells and pericytes, allowing endothelial cell phenotype characterization. In the second model, glial cells were at the bottom of the well while pericytes and endothelial cells were cultured together in the filter: close interactions were observed in peg-and-socket contacts. In both models low paracellular permeability and P-glycoprotein functionality were demonstrated. These models are likely to be useful tools for understanding the pericytes' role in BBB physiology and could be of value in investigating the pericytes' influence on BBB in diseased states.


Subject(s)
Blood-Brain Barrier/physiology , Endothelial Cells/physiology , Neuroglia/physiology , Pericytes/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Actins/metabolism , Animals , Animals, Newborn , Antigens/metabolism , Biological Transport/physiology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/ultrastructure , Brain/cytology , Cattle , Cells, Cultured , Claudin-5 , Claudins/metabolism , Coculture Techniques/methods , Gene Expression/physiology , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Models, Biological , Muscle, Smooth, Vascular/cytology , Occludin , Phosphoproteins/metabolism , Proteoglycans/metabolism , Rats , Receptor, Platelet-Derived Growth Factor beta/metabolism , Rhodamine 123/metabolism , Verapamil/metabolism , Vimentin/metabolism , Zonula Occludens-1 Protein , gamma-Glutamyltransferase/metabolism
12.
J Alzheimers Dis ; 22(3): 849-59, 2010.
Article in English | MEDLINE | ID: mdl-20858979

ABSTRACT

Several studies have highlighted the close relationship between Alzheimer's disease (AD) and alterations in the bidirectional transport of amyloid-ß (Aß) peptides across the blood-brain barrier (BBB). The brain capillary endothelial cells (BCECs) that compose the BBB express the receptors and transporters that enable this transport process. There is significant in vivo evidence to suggest that P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) restrict Aß peptides entry into the brain, whereas the receptor for advanced glycation end-products (RAGE) seems to mediate apical-to-basolateral passage across the BBB. However, deciphering the molecular mechanisms underlying these in vivo processes requires further in vitro characterization. Using an in vitro BBB model and specific competition experiments against RAGE, we have observed a significant decrease in apical-to-basolateral (but not basolateral-to-apical) transport of Aß1-40 and Aß1-42 peptides through BCECs. This transport is a caveolae-dependent process and fits with the apical location of RAGE observed in confocal microscopy experiments. Inhibition of P-gp and BCRP using different inhibitors increases transport of Aß peptides suggesting that these efflux pumps are involved in Aß peptide transport at the BCECs level. Taken as a whole, these results demonstrate the involvement of the caveolae-dependent transcytosis of Aß peptides through the BBB in a RAGE-mediated transport process, reinforcing the hypothesis whereby this receptor is a potential drug target in AD.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Receptors, Immunologic/physiology , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Blood-Brain Barrier/cytology , Cattle , Coculture Techniques , Protein Transport/physiology , Receptor for Advanced Glycation End Products
SELECTION OF CITATIONS
SEARCH DETAIL
...