21 research outputs found

    Abstracts from the 20th International Symposium on Signal Transduction at the Blood-Brain Barriers

    Full text link
    https://deepblue.lib.umich.edu/bitstream/2027.42/138963/1/12987_2017_Article_71.pd

    Transcytosis through the blood brain barrier : in vitro studies of the transport of low density lipoprotein and ß-amyloid peptide

    No full text
    La Barrière Hémato-Encéphalique (BHE) est une interface localisée au niveau des cellules endothéliales des capillaires cérébraux (CECs). Elle présente des caractéristiques structurales et métaboliques spécifiques restreignant considérablement les échanges entre le sang et le cerveau dans le but de maintenir l’homéostasie du système nerveux central (SNC). La présence de transporteurs et récepteurs au niveau de CECs permet l’apport de nutriments essentiels au fonctionnement cérébral. L’expression de ces propriétés est induite par l’environnement cérébral et notamment par la population astrocytaire. Dans notre laboratoire, la BHE est reconstituée in vitro en co-cultivant des CECs bovines et des cellules gliales primaires de rat reproduisant les principales caractéristiques de la BHE in vivo. Son utilisation a permis de mettre en évidence une voie originale de transcytose (récepteur-dépendante) assurant le transport des lipoprotéines de basse densité (LDL) vers le parenchyme cérébral. Nous avons caractérisé les premières étapes de ce processus et démontré que celles-ci impliquent des structures cellulaires spécialisées comme les cavéoles et les cavéosomes, permettant aux lipoprotéines d’être relarguées intactes vers le compartiment cérébral. Récemment, de nombreux travaux ont placé la BHE au centre des échanges des peptides β-amyloïde (Aβ) entre le sang et le cerveau. Un défaut des mécanismes de transport est suspecté d’être à l’origine de l’accumulation cérébrale de ces peptides responsables de la maladie d’Alzheimer (MA). En utilisant notre modèle in vitro de BHE, nous avons étudié l’implication des CECs dans les échanges de ces peptides. Nous avons démontré l’implication du récepteur « receptor for advanced glycation end-products » (RAGE) dans l’entrée vers le compartiment cérébral des peptides Aβ. Ce transport est spécifique et implique la voie des cavéoles. L’implication de pompes d’efflux telles que la P-glycoprotéine (P-gp) et la « breast cancer resistance protein » (BCRP) dans la restriction de l’influx des peptides Aβ a également été mise en évidence. D’autre part, des résultats préliminaires suggèrent que le récepteur « low density lipoprotein receptor-related 1 » (LRP1) n’est pas impliqué dans l’efflux des peptides Aβ. Ces résultats contribuent à apporter une meilleure compréhension du rôle de la BHE dans la MA et permettent d’envisager de nouvelles approches thérapeutiques.The blood-brain barrier (BBB) is a dynamic interface located at the brain capillary endothelial cells (BCECs) level. This barrier possesses some morphological and enzymatic properties whose aim is to maintain homeostasis of central nervous system (CNS) by firmly reducing the passages between blood and brain. BCECs express carrier-mediated transporters and receptors allowing the income of nutrients that are essential to brain function. These BBB properties come from brain environment and especially from the astrocytic population. In our laboratory, we have developed an in vitro BBB model consisting of a co-culture of bovine BCECs and new-born rat glial cells that closely mimics the in vivo situation. In this in vitro model, an original transcytosis pathway (receptor-dependent) that ensures low density lipoproteins (LDL) transport into the brain parenchyma has been discovered. We have characterized the first steps of this transport and we have shown that involve specialized cellular structures such as caveolae and caveosomes, allowing intact lipoproteins to be released into the brain compartment. Recently, many studies have considered the BBB like a very suitable site for the exchanges of amyloid β (Aβ) peptides between blood and brain. An alteration in the mechanisms of transport is suspected to result in cerebral accumulation of these peptides which are responsible for Alzheimer's Disease (AD). Using our in vitro model, we investigated the involvement of the BCECs in the exchanges of these peptides. Our works show an asymmetrical transport across the BBB suggesting the involvement of specific receptors and transporters. We have demonstrated the involvement of « receptor for advanced glycation end products » (RAGE) in the entry into the brain compartment. This transport is specific and involves the caveolae’s pathway. The involvement of P-glycoprotein (P-gp) and « breast cancer resistance protein » (BCRP) efflux pumps in restricting the influx of Aβ peptides was also highlighted. Moreover, preliminary results suggest that « low density lipoprotein receptor related-1 » (LRP1) is not involved in the efflux of Aβ peptides. These studies help to provide a better understanding of the role of the BBB in AD and allow to consider new therapeutic approaches

    Transcytose à travers la barrière hémato-encéphalique (étude in vitro du transport des lipoproteines de basse densité et du peptide ß-amyloïde)

    No full text
    La Barrière Hémato-Encéphalique (BHE) est une interface localisée au niveau des cellules endothéliales des capillaires cérébraux (CECs). Elle présente des caractéristiques structurales et métaboliques spécifiques restreignant considérablement les échanges entre le sang et le cerveau dans le but de maintenir l homéostasie du système nerveux central (SNC). La présence de transporteurs et récepteurs au niveau de CECs permet l apport de nutriments essentiels au fonctionnement cérébral. L expression de ces propriétés est induite par l environnement cérébral et notamment par la population astrocytaire. Dans notre laboratoire, la BHE est reconstituée in vitro en co-cultivant des CECs bovines et des cellules gliales primaires de rat reproduisant les principales caractéristiques de la BHE in vivo. Son utilisation a permis de mettre en évidence une voie originale de transcytose (récepteur-dépendante) assurant le transport des lipoprotéines de basse densité (LDL) vers le parenchyme cérébral. Nous avons caractérisé les premières étapes de ce processus et démontré que celles-ci impliquent des structures cellulaires spécialisées comme les cavéoles et les cavéosomes, permettant aux lipoprotéines d être relarguées intactes vers le compartiment cérébral. Récemment, de nombreux travaux ont placé la BHE au centre des échanges des peptides b-amyloïde (Ab) entre le sang et le cerveau. Un défaut des mécanismes de transport est suspecté d être à l origine de l accumulation cérébrale de ces peptides responsables de la maladie d Alzheimer (MA). En utilisant notre modèle in vitro de BHE, nous avons étudié l implication des CECs dans les échanges de ces peptides. Nous avons démontré l implication du récepteur receptor for advanced glycation end-products (RAGE) dans l entrée vers le compartiment cérébral des peptides Ab. Ce transport est spécifique et implique la voie des cavéoles. L implication de pompes d efflux telles que la P-glycoprotéine (P-gp) et la breast cancer resistance protein (BCRP) dans la restriction de l influx des peptides Ab a également été mise en évidence. D autre part, des résultats préliminaires suggèrent que le récepteur low density lipoprotein receptor-related 1 (LRP1) n est pas impliqué dans l efflux des peptides Ab. Ces résultats contribuent à apporter une meilleure compréhension du rôle de la BHE dans la MA et permettent d envisager de nouvelles approches thérapeutiques.The blood-brain barrier (BBB) is a dynamic interface located at the brain capillary endothelial cells (BCECs) level. This barrier possesses some morphological and enzymatic properties whose aim is to maintain homeostasis of central nervous system (CNS) by firmly reducing the passages between blood and brain. BCECs express carrier-mediated transporters and receptors allowing the income of nutrients that are essential to brain function. These BBB properties come from brain environment and especially from the astrocytic population. In our laboratory, we have developed an in vitro BBB model consisting of a co-culture of bovine BCECs and new-born rat glial cells that closely mimics the in vivo situation. In this in vitro model, an original transcytosis pathway (receptor-dependent) that ensures low density lipoproteins (LDL) transport into the brain parenchyma has been discovered. We have characterized the first steps of this transport and we have shown that involve specialized cellular structures such as caveolae and caveosomes, allowing intact lipoproteins to be released into the brain compartment. Recently, many studies have considered the BBB like a very suitable site for the exchanges of amyloid b (Ab) peptides between blood and brain. An alteration in the mechanisms of transport is suspected to result in cerebral accumulation of these peptides which are responsible for Alzheimer's Disease (AD). Using our in vitro model, we investigated the involvement of the BCECs in the exchanges of these peptides. Our works show an asymmetrical transport across the BBB suggesting the involvement of specific receptors and transporters. We have demonstrated the involvement of receptor for advanced glycation end products (RAGE) in the entry into the brain compartment. This transport is specific and involves the caveolae s pathway. The involvement of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) efflux pumps in restricting the influx of Ab peptides was also highlighted. Moreover, preliminary results suggest that low density lipoprotein receptor related-1 (LRP1) is not involved in the efflux of Ab peptides. These studies help to provide a better understanding of the role of the BBB in AD and allow to consider new therapeutic approaches.ARRAS-Bib.electronique (620419901) / SudocSudocFranceF

    La barrière hémato-encéphalique

    No full text
    Le cerveau est isolé du reste de l’organisme par la barrière hémato-encéphalique (BHE) qui contrôle étroitement les échanges entre les compartiments sanguin et cérébral. En raison de son rôle essentiel dans le maintien de l’homéostasie cérébrale, cette barrière est impliquée dans de nombreuses maladies neurodégénératives telles que la maladie d’Alzheimer (MA) dont l’origine est attribuée à l’accumulation et l’agrégation excessives de peptides Aβ. Le rôle de la BHE dans le métabolisme et le transport de ces peptides est aujourd’hui étudié avec plus d’attention et les premiers résultats obtenus permettent d’affirmer sans ambiguïté que cette barrière est un acteur majeur de cette pathologie complexe et suggèrent qu’elle pourrait être une nouvelle cible thérapeutique dans la maladie d’Alzheimer

    TNF-α and IL-1β Modulate Blood-Brain Barrier Permeability and Decrease Amyloid-β Peptide Efflux in a Human Blood-Brain Barrier Model

    No full text
    International audienceThe blood-brain barrier (BBB) is a selective barrier and a functional gatekeeper for the central nervous system (CNS), essential for maintaining brain homeostasis. The BBB is composed of specialized brain endothelial cells (BECs) lining the brain capillaries. The tight junctions formed by BECs regulate paracellular transport, whereas transcellular transport is regulated by specialized transporters, pumps and receptors. Cytokine-induced neuroinflammation, such as the tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β), appear to play a role in BBB dysfunction and contribute to the progression of Alzheimer’s disease (AD) by contributing to amyloid-β (Aβ) peptide accumulation. Here, we investigated whether TNF-α and IL-1β modulate the permeability of the BBB and alter Aβ peptide transport across BECs. We used a human BBB in vitro model based on the use of brain-like endothelial cells (BLECs) obtained from endothelial cells derived from CD34+ stem cells cocultivated with brain pericytes. We demonstrated that TNF-α and IL-1β differentially induced changes in BLECs’ permeability by inducing alterations in the organization of junctional complexes as well as in transcelluar trafficking. Further, TNF-α and IL-1β act directly on BLECs by decreasing LRP1 and BCRP protein expression as well as the specific efflux of Aβ peptide. These results provide mechanisms by which CNS inflammation might modulate BBB permeability and promote Aβ peptide accumulation. A future therapeutic intervention targeting vascular inflammation at the BBB may have the therapeutic potential to slow down the progression of A

    Oxysterols decrease apical-to-basolateral transport of Aß peptides via an ABCB1-mediated process in an in vitro Blood-brain barrier model constituted of bovine brain capillary endothelial cells

    No full text
    International audienceIt 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

    Transcriptional profiles of receptors and transporters involved in brain cholesterol homeostasis at the blood-brain barrier: Use of an in vitro model

    No full text
    International audienceBrain is the most cholesterol rich organ of the whole body and recent studies suggest a role for the blood-brain barrier (BBB) in cerebral cholesterol homeostasis. Low density lipoprotein receptor (LDLR)-related receptors and ATP-binding Cassette (ABC) transporters play an important role in peripheral sterol homeostasis. The purpose of this study was to determine the mRNAexpression profiles of ABC transporters (ABCA1, 2, 3, 7 and ABCG1) and low density lipoprotein receptor (LDLR)-related receptors (LDLR, vLDLR, LRP1, LRP2 and LRP8) in BBB endothelium using an in vitro co-culture model of bovine brain capillary endothelial cells (BCECs) and rat glial cells. All transcripts tested are expressed by BCECs and in capillary extract, except vLDLR. Glial cells influence ABCG1, A1, 2, 7 and LRP1 transcription, suggesting a role in cerebral lipid supply/elimination through the modulation of the expression of these transporters and receptors by these cells. Altogether, these results highlight the importance of glial input in the BBB transport phenotype for sterol homeostasis in the central nervous system, and confirm the importance of the BBB in this process

    Impact of Pesticide Residues on the Gut-Microbiota–Blood–Brain Barrier Axis: A Narrative Review

    No full text
    International audienceAccumulating evidence indicates that chronic exposure to a low level of pesticides found in diet affects the human gut-microbiota-blood-brain barrier (BBB) axis. This axis describes the physiological and bidirectional connection between the microbiota, the intestinal barrier (IB), and the BBB. Preclinical observations reported a gut microbial alteration induced by pesticides, also known as dysbiosis, a condition associated not only with gastrointestinal disorders but also with diseases affecting other distal organs, such as the BBB. However, the interplay between pesticides, microbiota, the IB, and the BBB is still not fully explored. In this review, we first consider the similarities/differences between these two physiological barriers and the different pathways that link the gut microbiota and the BBB to better understand the dialogue between bacteria and the brain. We then discuss the effects of chronic oral pesticide exposure on the gut-microbiota-BBB axis and raise awareness of the danger of chronic exposure, especially during the perinatal period (pregnant women and offspring)

    Ketone Bodies Promote Amyloid-β1–40 Clearance in a Human in Vitro Blood–Brain Barrier Model

    No full text
    International audienceAlzheimer's disease (AD) is characterized by the abnormal accumulation of amyloid-β (Aβ) peptides in the brain. The pathological process has not yet been clarified, although dysfunctional transport of Aβ across the blood-brain barrier (BBB) appears to be integral to disease development. At present, no effective therapeutic treatment against AD exists, and the adoption of a ketogenic diet (KD) or ketone body (KB) supplements have been investigated as potential new therapeutic approaches. Despite experimental evidence supporting the hypothesis that KBs reduce the Aβ load in the AD brain, little information is available about the effect of KBs on BBB and their effect on Aβ transport. Therefore, we used a human in vitro BBB model, brain-like endothelial cells (BLECs), to investigate the effect of KBs on the BBB and on Aβ transport. Our results show that KBs do not modify BBB integrity and do not cause toxicity to BLECs. Furthermore, the presence of KBs in the culture media was combined with higher MCT1 and GLUT1 protein levels in BLECs. In addition, KBs significantly enhanced the protein levels of LRP1, P-gp, and PICALM, described to be involved in Aβ clearance. Finally, the combined use of KBs promotes Aβ efflux across the BBB. Inhibition experiments demonstrated the involvement of LRP1 and P-gp in the efflux. This work provides evidence that KBs promote Aβ clearance from the brain to blood in addition to exciting perspectives for studying the use of KBs in therapeutic approaches
    corecore