Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Fabien Gosselet is active.

Publication


Featured researches published by Fabien Gosselet.


Journal of Alzheimer's Disease | 2010

Apical-to-Basolateral Transport of Amyloid-β Peptides through Blood-Brain Barrier Cells is Mediated by the Receptor for Advanced Glycation End-Products and is Restricted by P-Glycoprotein

Pietra Candela; Fabien Gosselet; Julien Saint-Pol; Emmanuel Sevin; Marie-Christine Boucau; Eric Boulanger; Roméo Cecchelli; Laurence Fenart

Several studies have highlighted the close relationship between Alzheimers disease (AD) and alterations in the bidirectional transport of amyloid-β (Aβ) 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 Aβ 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 Aβ1-40 and Aβ1-42 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 Aβ peptides suggesting that these efflux pumps are involved in Aβ peptide transport at the BCECs level. Taken as a whole, these results demonstrate the involvement of the caveolae-dependent transcytosis of Aβ peptides through the BBB in a RAGE-mediated transport process, reinforcing the hypothesis whereby this receptor is a potential drug target in AD.


Endothelium-journal of Endothelial Cell Research | 2008

Physiological Pathway for Low-Density Lipoproteins across the Blood-Brain Barrier: Transcytosis through Brain Capillary Endothelial Cells In Vitro

Pietra Candela; Fabien Gosselet; Florence Miller; Valérie Buée-Scherrer; Gérard Torpier; Roméo Cecchelli; Laurence Fenart

Although an immense knowledge has accumulated concerning regulation of cholesterol homeostasis in the body, this does not include the brain, where details are just emerging. Using an in vitro blood-brain barrier model, the authors have demonstrated that low-density lipoprotein (LDL) underwent transcytosis through the endothelial cells (ECs) by a receptor-mediated process, bypassing the lysosomal compartment. Moreover, caveolae might be involved in these blood-borne molecule transports from the blood to the brain. Although several ligands are known to be internalized through cell surface caveolae, the subsequent intracellular pathways have remained elusive. By cell fractionation experiment and Western blot, the authors have demonstrated that the LDL receptor is located in the caveolae membrane fraction. Then, LDLs internalized were detected by electron microscopy in multivesicular bodies. The authors identified in brain capillary ECs a novel endosomal compartment, mildly acidic, positive for marker Lamp-1 but devoid of any degradative capability. From the point of view of pH, cellular location, and caveolae-derived formation, the multivesicular organelles described here can be related to the caveosome structure. These results could provide clues to physiological functions of caveolae-caveosome transcellular pathway in brain capillary ECs and may help in the rational design of more effective therapeutic drugs to the brain.


Brain Research | 2009

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

Fabien Gosselet; Pietra Candela; Emmanuel Sevin; Vincent Berezowski; Roméo Cecchelli; Laurence Fenart

Brain 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 mRNA expression 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.


Photochemical and Photobiological Sciences | 2003

UVB-induced mutations in human key gatekeeper genes governing signalling pathways and consequences for skin tumourigenesis

Jean-Claude Ehrhart; Fabien Gosselet; Raphaël M. Culerrier; Alain Sarasin

The UVB component of the solar spectrum induces DNA lesions that, in the absence of error-free DNA repair, may give rise during DNA replication to mutations in caretaker and gatekeeper genes. The DNA repair genes are the best candidates for caretaker genes as exemplified by the human hereditary xeroderma pigmentosum (XP) syndrome. Cultured XP cells are hypermutable after UVB irradiation. This increased mutation frequency is also found in gatekeeper genes, which govern signalling pathways implicated in the control of cellular proliferation, differentiation and survival of human epidermal keratinocytes. We describe and discuss the role of mutated gatekeeper genes in five specific signalling pathways which have been implicated in skin carcinogenesis. The pathways we focus on in this review are: (i) P16(INK4A)-CDK4/6-RB; (ii) P14(ARF)-HDM2-P53; (iii) Sonic hedgehog (SHH)/GLI; (iv) WNT/beta-catenin; and (v) Bone Morphogenetic Protein (BMP)/SMAD. 70-80% of XP skin cancers exhibit one or several mutations in the P53, PTCH-1, SMO or CDKN2A genes, the type and frequency of mutated genes being different between squamous cell (SCCs) and basal cell carcinomas (BCCs). In XP cancers, the typically UVB-induced CC to TT tandem transitions represent approximately 60% of total mutations compared to 10-15% in skin tumours from DNA repair-proficient patients. Acquired activation of the pathways described herein can alter proliferation and differentiation of keratinocytes, allowing a damaged cell to replicate and give rise to mutated daughter cells, then eventually to the development of the carcinogenic process following clonal selection.


Current Alzheimer Research | 2013

Amyloid-β peptides, Alzheimer's disease and the blood-brain barrier.

Fabien Gosselet; Julien Saint-Pol; Pietra Candela; Laurence Fenart

Ever since amyloid-β (Aβ) peptides were first identified in cerebral plaques in patients with Alzheimers disease (AD), much research work has focused on the complex mechanisms through which these peptides are synthesized, transported and degraded. Although new information emerges on a regular basis, we consider that the importance of the blood-brain barrier (BBB) in the pathogenesis of AD has been underestimated. In fact, there are a number of obstacles that make it difficult to convince specialists in AD that the BBB indeed plays a key role in this disease: these include the complex physiology of the BBB and the technical difficulty of studying the barrier in vivo and reproducing its main properties in vitro. With these considerations in mind, the present review sets out summarize our current knowledge about the physiology of the BBB and describe recent research findings on the barriers role in Aβ peptide proteostasis and thus in the mechanism of AD.


Toxicology in Vitro | 2015

Evaluation of drug-induced neurotoxicity based on metabolomics, proteomics and electrical activity measurements in complementary CNS in vitro models

Luise Schultz; Marie-Gabrielle Zurich; Maxime Culot; Anaelle da Costa; Christophe Landry; Patricia Bellwon; Theresa Kristl; Katrin Hörmann; Silke Ruzek; Stephan Aiche; Knut Reinert; Chris Bielow; Fabien Gosselet; Roméo Cecchelli; Christian G. Huber; Olaf H.-U. Schroeder; Alexandra Gramowski-Voss; Dieter G. Weiss; Anna K. Bal-Price

The present study was performed in an attempt to develop an in vitro integrated testing strategy (ITS) to evaluate drug-induced neurotoxicity. A number of endpoints were analyzed using two complementary brain cell culture models and an in vitro blood-brain barrier (BBB) model after single and repeated exposure treatments with selected drugs that covered the major biological, pharmacological and neuro-toxicological responses. Furthermore, four drugs (diazepam, cyclosporine A, chlorpromazine and amiodarone) were tested more in depth as representatives of different classes of neurotoxicants, inducing toxicity through different pathways of toxicity. The developed in vitro BBB model allowed detection of toxic effects at the level of BBB and evaluation of drug transport through the barrier for predicting free brain concentrations of the studied drugs. The measurement of neuronal electrical activity was found to be a sensitive tool to predict the neuroactivity and neurotoxicity of drugs after acute exposure. The histotypic 3D re-aggregating brain cell cultures, containing all brain cell types, were found to be well suited for OMICs analyses after both acute and long term treatment. The obtained data suggest that an in vitro ITS based on the information obtained from BBB studies and combined with metabolomics, proteomics and neuronal electrical activity measurements performed in stable in vitro neuronal cell culture systems, has high potential to improve current in vitro drug-induced neurotoxicity evaluation.


Journal of Alzheimer's Disease | 2012

Brain pericytes ABCA1 expression mediates cholesterol efflux but not cellular amyloid-β peptide accumulation.

Julien Saint-Pol; Marie-Christine Boucau; Pietra Candela; Lucie Dehouck; Roméo Cecchelli; Marie-Pierre Dehouck; Laurence Fenart; Fabien Gosselet

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-β peptides which accumulate in brain of Alzheimers 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.


Biochemical and Biophysical Research Communications | 2014

Effects of oxysterols on the blood–brain barrier: Implications for Alzheimer’s disease

Fabien Gosselet; Julien Saint-Pol; Laurence Fenart

Altered brain cholesterol homeostasis plays a key role in neurodegenerative diseases such as Alzheimers disease (AD). For a long time, the blood-brain barrier (BBB) was basically considered as a barrier isolating the brain from circulating cholesterol, however, several lines of evidence now suggest that the BBB strictly regulates the exchanges of sterol between the brain and the peripheral circulation. Oxysterols, synthesized by neurons or by peripheral cells, cross the BBB easily and modulate the expression of several enzymes, receptors and transporters which are involved not only in cholesterol metabolism but also in other brain functions. This review article deals with the way oxysterols impact BBB cells. These perspectives open new routes for designing certain therapeutical approaches that target the BBB so that the onset and/or progression of brain diseases such as AD may be modulated.


Molecular Neurobiology | 2016

Efficient Docosahexaenoic Acid Uptake by the Brain from a Structured Phospholipid

Mayssa Hachem; Alain Géloën; Amanda Lo Van; Baptiste Foumaux; Laurence Fenart; Fabien Gosselet; Pedro Da Silva; Gildas Breton; Michel Lagarde; Madeleine Picq; Nathalie Bernoud-Hubac

Docosahexaenoic acid (DHA) is the main essential omega-3 fatty acid in brain tissues required for normal brain development and function. An alteration of brain DHA in neurodegenerative diseases such as Alzheimer’s and Parkinson’s is observed. Targeted intake of DHA to the brain could compensate for these deficiencies. Blood DHA is transported across the blood–brain barrier more efficiently when esterified at the sn-2 position of lyso-phosphatidylcholine. We used a structured phosphatidylcholine to mimic 2-docosahexaenoyl-lysoPC (lysoPC-DHA), named AceDoPC (1-acetyl,2-docosahexaenoyl-glycerophosphocholine), that may be considered as a stabilized form of the physiological lysoPC-DHA and that is neuroprotective in experimental ischemic stroke. The aim of the present study was to investigate whether AceDoPC is a relevant delivery form of DHA to the brain in comparison with other forms of the fatty acid. By combining in vitro and in vivo experiments, our findings report for the first time that AceDoPC is a privileged and specific carrier of DHA to the brain, when compared with DHA-containing PC and non-esterified DHA. We also show that AceDoPC was hydrolyzed, in part, into lysoPC-DHA. Ex vivo autoradiography of rat brain reveals that DHA from AceDoPC was localized in specific brain regions playing key roles in memory, thoughts, and cognitive functions. Finally, using molecular modeling approaches, we demonstrate that electrostatic and lipophilic potentials are distributed very similarly at the surfaces of AceDoPC and lysoPC-DHA. Our findings identify AceDoPC as an efficient way to specifically target DHA to the brain, which would allow potential preventive and therapeutic approaches for neurological diseases.


Brain Research | 2015

In vitro discrimination of the role of LRP1 at the BBB cellular level: Focus on brain capillary endothelial cells and brain pericytes

Pietra Candela; Julien Saint-Pol; Mélanie Kuntz; Marie-Christine Boucau; Yordenca Lamartinière; Fabien Gosselet; Laurence Fenart

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 β (Aβ) peptide between the blood and the brain. Deregulation of these exchanges seems to be a key trigger for the brain accumulation of Aβ peptide observed in Alzheimers disease (AD). Whereas the involvement of receptor for advanced glycation end-products in Aβ peptide transcytosis has been demonstrated in our laboratory, low-density lipoprotein receptors 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 Aβ 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 Aβ endocytosis. Moreover, the observations of pericytes contraction and local downregulation of LRP1 in response to Aβ treatment opens up perspectives for studying this cell type with respect to Aβ peptide metabolism and AD.

Collaboration


Dive into the Fabien Gosselet's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge