Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters








Publication year range
1.
Mol Med ; 30(1): 132, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187765

ABSTRACT

Cell-based therapeutic strategies have been proposed as an alternative for brain and blood vessels repair after stroke, but their clinical application is hampered by potential adverse effects. We therefore tested the hypothesis that secretome of these cells might be used instead to still focus on cell-based therapeutic strategies. We therefore characterized the composition and the effect of the secretome of brain microvascular endothelial cells (BMECs) on primary in vitro human models of angiogenesis and vascular barrier. Two different secretome batches produced in high scale (scHSP) were analysed by mass spectrometry. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used as well as in vitro models of EC monolayer (CMECs) and blood-brain barrier (BBB). Cells were also exposed to oxygen-glucose deprivation (OGD) conditions and treated with scHSP during reoxygenation. Protein yield and composition of scHSP batches showed good reproducibility. scHSP increased CD34+-EC proliferation, tubulogenesis, and migration. Proteomic analysis of scHSP revealed the presence of growth factors and proteins modulating cell metabolism and inflammatory pathways. scHSP improved the integrity of CMECs, and upregulated the expression of junctional proteins. Such effects were mediated through the activation of the interferon pathway and downregulation of Wnt signalling. Furthermore, OGD altered the permeability of both CMECs and BBB, while scHSP prevented the OGD-induced vascular leakage in both models. These effects were mediated through upregulation of junctional proteins and regulation of MAPK/VEGFR2. Finally, our results highlight the possibility of using secretome from BMECs as a therapeutic alternative to promote brain angiogenesis and to protect from ischemia-induced vascular leakage.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Proteomics , Humans , Endothelial Cells/metabolism , Blood-Brain Barrier/metabolism , Proteomics/methods , Secretome/metabolism , Capillary Permeability , Brain/metabolism , Brain/blood supply , Brain/pathology , Cell Hypoxia , Proteome/metabolism , Cells, Cultured , Microvessels/metabolism , Microvessels/cytology
3.
Exp Hematol Oncol ; 12(1): 104, 2023 Dec 10.
Article in English | MEDLINE | ID: mdl-38072918

ABSTRACT

BACKGROUND: Triple-Negative Breast Cancer is particularly aggressive, and its metastasis to the brain has a significant psychological impact on patients' quality of life, in addition to reducing survival. The development of brain metastases is particularly harmful in triple-negative breast cancer (TNBC). To date, the mechanisms that induce brain metastasis in TNBC are poorly understood. METHODS: Using a human blood-brain barrier (BBB) in vitro model, an in vitro 3D organotypic extracellular matrix, an ex vivo mouse brain slices co-culture and in an in vivo xenograft experiment, key step of brain metastasis were recapitulated to study TNBC behaviors. RESULTS: In this study, we demonstrated for the first time the involvement of the precursor of Nerve Growth Factor (proNGF) in the development of brain metastasis. More importantly, our results showed that proNGF acts through TrkA independent of its phosphorylation to induce brain metastasis in TNBC. In addition, we found that proNGF induces BBB transmigration through the TrkA/EphA2 signaling complex. More importantly, our results showed that combinatorial inhibition of TrkA and EphA2 decreased TBNC brain metastasis in a preclinical model. CONCLUSIONS: These disruptive findings provide new insights into the mechanisms underlying brain metastasis with proNGF as a driver of brain metastasis of TNBC and identify TrkA/EphA2 complex as a potential therapeutic target.

4.
Methods Mol Biol ; 2492: 277-288, 2022.
Article in English | MEDLINE | ID: mdl-35733051

ABSTRACT

Studying the mechanisms of breast cancer cells in brain metastases is challenging, considering the high specificity of the blood-brain barrier (BBB) with whom breast cancer cells have to interact and cross in order to reach the brain parenchyma. While numerous in vitro BBB models are available, the setting of the model and phenotype of the endothelial cells (ECs) of the BBB model are essential to obtain relevant results.In this chapter, we describe a method to establish a human in vitro BBB model to study adhesion of breast cancer cells and the adaptation of the method for trans-endothelial migration assay keeping the appropriate BBB phenotype of the ECs.


Subject(s)
Brain Neoplasms , Breast Neoplasms , Biological Transport , Blood-Brain Barrier , Brain/pathology , Brain Neoplasms/genetics , Breast Neoplasms/pathology , Endothelial Cells , Female , Humans
5.
Int J Pharm ; 621: 121780, 2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35504427

ABSTRACT

Treatment for CNS related diseases are limited by the difficulty of the drugs to cross the blood-brain barrier (BBB). The functionalization of polymeric nanoparticles (NPs) coated with the surfactants polysorbate 80 (PS80) and poloxamer 188 (P188), have shown promising results as drugs carriers are able to cross the BBB on animal models. In this study, poly(lactide-co-glycolide) (PLGA) NPs coated with PS80 and P188, labelled with a fluorescent dye were tested on human pre-clinical in vitro model to evaluate and compare their uptake profiles, mechanisms of transport and crossing over human brain-like endothelial cells (BLECs) mimicking the human BBB. In addition, these NPs were produced using a method facilitating their reproducible production at high scale, the MicroJet reactor® technology. Results showed that both formulations were biocompatible and able to be internalized within the BLECs in different uptake profiles depending on their coating: P188 NP showed higher internalization capacity than PS80 NP. Both NPs uptakes were ATP-dependent, following more than one endocytosis pathway with colocalization in the early endosomes, ending with a NPs release in the brain compartment. Thus, both surfactant-coated PLGA NPs are interesting formulations for delivery to the brain through the BBB, presenting different uptake profiles.


Subject(s)
Nanoparticles , Pulmonary Surfactants , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Drug Carriers/metabolism , Endothelial Cells/metabolism , Excipients/metabolism , Humans , Poloxamer/metabolism , Polysorbates , Pulmonary Surfactants/metabolism , Surface-Active Agents/metabolism
6.
J Exp Clin Cancer Res ; 41(1): 110, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35346305

ABSTRACT

BACKGROUND: CD44 is a multifunctional membrane glycoprotein. Through its heparan sulfate chain, CD44 presents growth factors to their receptors. We have shown that CD44 and Tropomyosin kinase A (TrkA) form a complex following nerve growth factor (NGF) induction. Our study aimed to understand how CD44 and TrkA interact and the consequences of inhibiting this interaction regarding the pro-tumoral effect of NGF in breast cancer. METHODS: After determining which CD44 isoforms (variants) are involved in forming the TrkA/CD44 complex using proximity ligation assays, we investigated the molecular determinants of this interaction. By molecular modeling, we isolated the amino acids involved and confirmed their involvement using mutations. A CD44v3 mimetic peptide was then synthesized to block the TrkA/CD44v3 interaction. The effects of this peptide on the growth, migration and invasion of xenografted triple-negative breast cancer cells were assessed. Finally, we investigated the correlations between the expression of the TrkA/CD44v3 complex in tumors and histo-pronostic parameters. RESULTS: We demonstrated that isoform v3 (CD44v3), but not v6, binds to TrkA in response to NGF stimulation. The final 10 amino acids of exon v3 and the TrkA H112 residue are necessary for the association of CD44v3 with TrkA. Functionally, the CD44v3 mimetic peptide impairs not only NGF-induced RhoA activation, clonogenicity, and migration/invasion of breast cancer cells in vitro but also tumor growth and metastasis in a xenograft mouse model. We also detected TrkA/CD44v3 only in cancerous cells, not in normal adjacent tissues. CONCLUSION: Collectively, our results suggest that blocking the CD44v3/TrkA interaction can be a new therapeutic option for triple-negative breast cancers.


Subject(s)
Breast Neoplasms , Hyaluronan Receptors , Nerve Growth Factor , Receptor, trkA , Animals , Breast Neoplasms/genetics , Female , Humans , Hyaluronan Receptors/metabolism , Mice , Nerve Growth Factor/pharmacology , Protein Isoforms , Receptor, trkA/metabolism
7.
J Vis Exp ; (177)2021 11 30.
Article in English | MEDLINE | ID: mdl-34927613

ABSTRACT

The delivery of drugs to the brain remains a challenge due to the blood-brain barrier's (BBB) highly specific and restrictive properties, which controls and restrict access to the brain parenchyma. However, with the development of nanotechnologies, large panels of new nanomaterials were developed to improve drug delivery, highlighting the need for reliable in vitro microsystems to predict brain penetration in the frame of preclinical assays. Here is a straightforward method to set up a microphysiological system to model the BBB using solely human cells. In its configuration, the model consists of a triple culture including brain-like endothelial cells (BLECs), pericytes, and astrocytes, the three main BBB cellular actors necessary to induce and regulate the BBB properties in a more physiological manner without the requirement of tightening compounds. The model developed in a 12-well plate format, ready after 6 days of triple culture, is characterized in physical properties, gene, and protein expressions and used for polymeric nanogel transport measurement. The model can be used for an extensive range of experiments in healthy and pathological conditions and represents a valuable tool for preclinical assessments of molecule and particle transport, as well as inter-and intracellular trafficking.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Astrocytes , Biological Transport/physiology , Blood-Brain Barrier/metabolism , Coculture Techniques , Endothelial Cells/metabolism , Humans , Pericytes/metabolism
8.
Stem Cell Res Ther ; 12(1): 552, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702368

ABSTRACT

BACKGROUND: Cell-based therapeutic strategies have been proposed as an alternative for brain repair after stroke, but their clinical application has been hampered by potential adverse effects in the long term. The present study was designed to test the effect of the secretome of endothelial progenitor cells (EPCs) from stroke patients (scCM) on in vitro human models of angiogenesis and vascular barrier. METHODS: Two different scCM batches were analysed by mass spectrometry and a proteome profiler. Human primary CD34+-derived endothelial cells (CD34+-ECs) were used for designing angiogenesis studies (proliferation, migration, and tubulogenesis) or in vitro models of EC monolayer (confluent monolayer ECs-CMECs) and blood-brain barrier (BBB; brain-like ECs-BLECs). Cells were treated with scCM (5 µg/mL) or protein-free endothelial basal medium (scEBM-control). CMECs or BLECs were exposed (6 h) to oxygen-glucose deprivation (OGD) conditions (1% oxygen and glucose-free medium) or normoxia (control-5% oxygen, 1 g/L of glucose) and treated with scCM or scEBM during reoxygenation (24 h). RESULTS: The analysis of different scCM batches showed a good reproducibility in terms of protein yield and composition. scCM increased CD34+-EC proliferation, tubulogenesis, and migration compared to the control (scEBM). The proteomic analysis of scCM revealed the presence of growth factors and molecules modulating cell metabolism and inflammatory pathways. Further, scCM decreased the permeability of CMECs and upregulated the expression of the junctional proteins such as occludin, VE-cadherin, and ZO-1. Such effects were possibly mediated through the activation of the interferon pathway and a moderate downregulation of Wnt signalling. Furthermore, OGD increased the permeability of both CMECs and BLECs, while scCM prevented the OGD-induced vascular leakage in both models. These effects were possibly mediated through the upregulation of junctional proteins and the regulation of MAPK/VEGFR2 activity. CONCLUSION: Our results suggest that scCM promotes angiogenesis and the maturation of newly formed vessels while restoring the BBB function in ischemic conditions. In conclusion, our results highlight the possibility of using EPC-secretome as a therapeutic alternative to promote brain angiogenesis and protect from ischemia-induced vascular leakage.


Subject(s)
Endothelial Progenitor Cells , Stroke , Blood-Brain Barrier , Humans , Hypoxia , Proteomics , Reproducibility of Results
9.
Fluids Barriers CNS ; 17(1): 37, 2020 Jun 02.
Article in English | MEDLINE | ID: mdl-32487241

ABSTRACT

BACKGROUND: Pediatric diffuse intrinsic pontine glioma (DIPG) represents one of the most devastating and lethal brain tumors in children with a median survival of 12 months. The high mortality rate can be explained by the ineligibility of patients to surgical resection due to the diffuse growth pattern and midline localization of the tumor. While the therapeutic strategies are unfortunately palliative, the blood-brain barrier (BBB) is suspected to be responsible for the treatment inefficiency. Located at the brain capillary endothelial cells (ECs), the BBB has specific properties to tightly control and restrict the access of molecules to the brain parenchyma including chemotherapeutic compounds. However, these BBB specific properties can be modified in a pathological environment, thus modulating brain exposure to therapeutic drugs. Hence, this study aimed at developing a syngeneic human blood-brain tumor barrier model to understand how the presence of DIPG impacts the structure and function of brain capillary ECs. METHODS: A human syngeneic in vitro BBB model consisting of a triple culture of human (ECs) (differentiated from CD34+-stem cells), pericytes and astrocytes was developed. Once validated in terms of BBB phenotype, this model was adapted to develop a blood-brain tumor barrier (BBTB) model specific to pediatric DIPG by replacing the astrocytes by DIPG-007, -013 and -014 cells. The physical and metabolic properties of the BBTB ECs were analyzed and compared to the BBB ECs. The permeability of both models to chemotherapeutic compounds was evaluated. RESULTS: In line with clinical observation, the integrity of the BBTB ECs remained intact until 7 days of incubation. Both transcriptional expression and activity of efflux transporters were not strongly modified by the presence of DIPG. The permeability of ECs to the chemotherapeutic drugs temozolomide and panobinostat was not affected by the DIPG environment. CONCLUSIONS: This original human BBTB model allows a better understanding of the influence of DIPG on the BBTB ECs phenotype. Our data reveal that the chemoresistance described for DIPG does not come from the development of a "super BBB". These results, validated by the absence of modification of drug transport through the BBTB ECs, point out the importance of understanding the implication of the different protagonists in the pathology to have a chance to significantly improve treatment efficiency.


Subject(s)
Antineoplastic Agents/pharmacology , Blood-Brain Barrier , Brain Neoplasms , Diffuse Intrinsic Pontine Glioma , Drug Resistance, Neoplasm , Models, Neurological , Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Cells, Cultured , Diffuse Intrinsic Pontine Glioma/drug therapy , Endothelial Cells , Humans , Panobinostat/pharmacology , Temozolomide/pharmacology
10.
FEBS Open Bio ; 8(4): 494-501, 2018 04.
Article in English | MEDLINE | ID: mdl-29632803

ABSTRACT

This study describes feedback on the effects of changes introduced in our teaching practices for an introductory biochemistry course in the Life Sciences curriculum. Students on this course have diverse educational qualifications and are taught in large learning groups, creating challenges for the management of individual learning. We used the constructive alignment principle, refining the learning contract and re-drafting the teaching program to introduce active learning and an organization of activities that promotes the participation of all the students and helps their understanding. We also created teaching resources available through the university virtual work environment. Our research aimed to measure the effects of those changes on the students' success. Monitoring of the student performance showed a continuous increase in the percentage of students who passed the course, from 2.13% to 33.5% in 4 years. Analysis of student perceptions highlighted that the teaching methodology was greatly appreciated by the students, whose attendance also improved. The recent introduction of clickers-questions constituted a complementary leverage. The active involvement of the students and better results for summative assessments are altogether a strong motivation for teaching staff to continue to make improvements.

11.
Int J Mol Sci ; 17(8)2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27529215

ABSTRACT

The ST6GALNAC5 gene that encodes an α2,6-sialyltransferase involved in the biosynthesis of α-series gangliosides, was previously identified as one of the genes that mediate breast cancer metastasis to the brain. We have shown that the expression of ST6GALNAC5 in MDA-MB-231 breast cancer cells resulted in the expression of GD1α ganglioside at the cell surface. By using a human blood-brain barrier in vitro model recently developed, consisting in CD34⁺ derived endothelial cells co-cultivated with pericytes, we show that ST6GALNAC5 expression decreased the interactions between the breast cancer cells and the human blood-brain barrier.


Subject(s)
Blood-Brain Barrier/metabolism , Breast Neoplasms/metabolism , Sialyltransferases/metabolism , Animals , Antigens, CD34/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Female , G(M1) Ganglioside/analogs & derivatives , G(M1) Ganglioside/metabolism , Humans , Mice , Pericytes/metabolism , Sialyltransferases/genetics
12.
J Alzheimers Dis ; 53(2): 677-91, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27232214

ABSTRACT

Amyloid-ß (Aß) accumulation in Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA) is likely caused by the impairment of its brain clearance that partly occurs through the blood-brain barrier (BBB). In this context, an in vitro BBB model is a valuable tool for studying the molecular mechanisms that regulate this process. This study assessed brain Aß elimination across the BBB and its modulation by the natural chaperones Apolipoprotein A1 (ApoA1) and Apolipoprotein J/Clusterin (ApoJ). The model was based on primary cerebral endothelial cells that were cultured on Matrigel-coated Transwells and treated with fluorescently labeled-Aß1-40 to track its efflux across the BBB, which corresponds to trafficking from the basolateral (brain) to apical (blood) compartments. We observed that the transport of basolateral Aß1-40 was enhanced when it was complexed to rApoJ, whereas the complex formed with rApoA1 did not influence Aß1-40 efflux. However, the presence of rApoA1 in the apical compartment was able to mobilize Aß1-40 from the basolateral side. We also observed that both rApoA1 and rApoJ moderately crossed the monolayer (from blood to brain) through a mechanism involving the LDL receptor-related protein family. In contrast to the increased rApoJ efflux when complexed to Aß1-40, rApoA1 trafficking was restricted when it was bound to the Aß peptide. In summary, the present study highlights the role of ApoJ and ApoA1 in the in vitro modulation of Aß elimination across the BBB.


Subject(s)
Amyloid beta-Peptides/metabolism , Apolipoprotein A-I/metabolism , Blood-Brain Barrier/metabolism , Clusterin/metabolism , Endothelial Cells/metabolism , Peptide Fragments/metabolism , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Line, Transformed , Cerebral Cortex/cytology , Clusterin/genetics , Clusterin/pharmacology , Endothelial Cells/drug effects , HEK293 Cells , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger/metabolism , Transfection , Zonula Occludens-1 Protein/metabolism
13.
PLoS One ; 11(3): e0151155, 2016.
Article in English | MEDLINE | ID: mdl-26958843

ABSTRACT

Around 7-17% of metastatic breast cancer patients will develop brain metastases, associated with a poor prognosis. To reach the brain parenchyma, cancer cells need to cross the highly restrictive endothelium of the Blood-Brain Barrier (BBB). As treatments for brain metastases are mostly inefficient, preventing cancer cells to reach the brain could provide a relevant and important strategy. For that purpose an in vitro approach is required to identify cellular and molecular interaction mechanisms between breast cancer cells and BBB endothelium, notably at the early steps of the interaction. However, while numerous studies are performed with in vitro models, the heterogeneity and the quality of BBB models used is a limitation to the extrapolation of the obtained results to in vivo context, showing that the choice of a model that fulfills the biological BBB characteristics is essential. Therefore, we compared pre-established and currently used in vitro models from different origins (bovine, mice, human) in order to define the most appropriate tool to study interactions between breast cancer cells and the BBB. On each model, the BBB properties and the adhesion capacities of breast cancer cell lines were evaluated. As endothelial cells represent the physical restriction site of the BBB, all the models consisted of endothelial cells from animal or human origins. Among these models, only the in vitro BBB model derived from human stem cells both displayed BBB properties and allowed measurement of meaningful different interaction capacities of the cancer cell lines. Importantly, the measured adhesion and transmigration were found to be in accordance with the cancer cell lines molecular subtypes. In addition, at a molecular level, the inhibition of ganglioside biosynthesis highlights the potential role of glycosylation in breast cancer cells adhesion capacities.


Subject(s)
Blood-Brain Barrier/cytology , Breast Neoplasms/pathology , Animals , Antigens, CD34/metabolism , Cattle , Cell Differentiation/physiology , Cell Line, Tumor , Cells, Cultured , Female , Fetal Blood/cytology , Humans , Mice , Neuroglia/cytology , Neuroglia/metabolism , Pericytes/cytology , Pericytes/metabolism , Rats
14.
Lab Invest ; 96(5): 588-98, 2016 05.
Article in English | MEDLINE | ID: mdl-26901835

ABSTRACT

Although brain metastases are the most common brain tumors in adults, there are few treatment options in this setting. To colonize the brain, circulating tumor cells must cross the blood-brain barrier (BBB), which is situated within specialized, restrictive microvascular endothelium. Understanding how cancer cells manage to transmigrate through the BBB might enable this process to be prevented. In vitro models are dedicated tools for characterizing the cellular and molecular mechanisms that underlie transendothelial migration process, as long as they accurately mimic the brain endothelium's in vivo characteristics. The objective of the present study was to adapt an existing in vitro model of the human BBB for use in studying cancer cell transmigration. The model is based on the coculture of endothelial cells (ECs, derived from cord blood hematopoietic stem cells) and brain pericytes. To allow the migration of cancer cells into the lower compartment, our model had to be transposed onto inserts with a larger pore size. However, we encountered a problem when culturing ECs on large (3-µm)-pore inserts: the cells crossed the membrane and formed a non-physiological second layer on the lower face of the insert. Using 3-µm-pore inserts (in a 12-well plate format), we report here on a method that enables the maintenance of a single monolayer of ECs on the insert's upper face only. Under these chosen conditions, the ECs exhibited typical BBB properties found in the original model (including restricted paracellular permeability and the expression of continuous tight junctions). This modified in vitro model of the human BBB enabled us to investigate the migratory potential of the MDA-MB-231 cell line (derived from highly metastatic human breast cancer cells). Last, the results obtained were compared with the rate of transmigration through endothelia with no BBB features.


Subject(s)
Blood-Brain Barrier/physiology , Neoplastic Cells, Circulating/pathology , Transendothelial and Transepithelial Migration/physiology , Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cell Line, Tumor , Coculture Techniques/instrumentation , Coculture Techniques/methods , Endothelium, Vascular/physiology , Female , Human Umbilical Vein Endothelial Cells , Humans , Models, Biological , Pericytes/physiology , Porosity
15.
Molecules ; 20(4): 6913-24, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25913930

ABSTRACT

α-Series gangliosides define a particular sub-class of glycosphingolipids containing sialic acid α2,6-linked to GalNAc residue that was isolated as a minor compound from the brain. The sialyltransferase ST6GalNAc V was cloned from mouse brain and showed α2,6-sialyltransferase activity almost exclusively for GM1b, to form GD1α and is considered as the main enzyme involved in the biosynthesis of α-series gangliosides. Recently, ST6GALNAC5 was identified as one of the genes over-expressed in breast cancer cell populations selected for their ability to produce brain metastasis. However, the capacity of human breast cancer cells to produce α-series gangliosides has never been clearly demonstrated. Here, we show by stable transfection and MS-MS analysis of total glycosphingolipids that ST6GALNAC5 expressing MDA-MB-231 breast cancer cells accumulate GD1α ganglioside (IV3Neu5Ac1, III6Neu5Ac1Gg4-Cer).


Subject(s)
Breast Neoplasms/metabolism , G(M1) Ganglioside/analogs & derivatives , Sialyltransferases/genetics , Breast Neoplasms/genetics , Cell Line, Tumor , Female , G(M1) Ganglioside/metabolism , Humans , Mass Spectrometry/methods , Sialyltransferases/metabolism
16.
Arch Biochem Biophys ; 554: 28-35, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24823859

ABSTRACT

Carnitine (3-hydroxy-4-trimethylammoniobutyrate) is necessary for transfer of fatty acids through the inner mitochondrial membrane. Carnitine, not synthesized in the brain, is delivered there through the strongly polarized blood-brain barrier (BBB). Expression and presence of two carnitine transporters - organic cation/carnitine transporter (OCTN2) and amino acid transporter B(0,+) (ATB(0,+)) have been demonstrated previously in an in vitro model of the BBB. Due to potential protein kinase C (PKC) phosphorylation sites within ATB(0,+) sequence, the present study verified effects of this kinase on transporter function and localization in the BBB. ATB(0,+) can be regulated by estrogen receptor α and up-regulated in vitro, therefore its presence in vivo was verified with the transmission electron microscopy. The analyses of brain slices demonstrated ATB(0,+) luminal localization in brain capillaries, confirmed by biotinylation experiments in an in vitro model of the BBB. Brain capillary endothelial cells were shown to control carnitine gradient. ATB(0,+) was phosphorylated by PKC, what correlated with inhibition of carnitine transport. PKC activation did not change the amount of ATB(0,+) present in the apical membrane of brain endothelial cells, but resulted in transporter exclusion from raft microdomains. ATB(0,+) inactivation by a lateral movement in plasma membrane after transporter phosphorylation has been postulated.


Subject(s)
Blood-Brain Barrier/metabolism , Carnitine/metabolism , Neurotransmitter Transport Proteins/metabolism , Protein Kinase C/metabolism , Animals , Biological Transport, Active , Brain/blood supply , Brain/metabolism , Brain/ultrastructure , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Male , Membrane Microdomains/metabolism , Microscopy, Electron, Transmission , Models, Neurological , Organic Cation Transport Proteins/metabolism , Phosphorylation , Rats , Rats, Wistar , Solute Carrier Family 22 Member 5 , Tetradecanoylphorbol Acetate/pharmacology
17.
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
18.
J Cereb Blood Flow Metab ; 34(1): 95-107, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24084699

ABSTRACT

The disappointing clinical outcomes of neuroprotectants challenge the relevance of preclinical stroke models and data in defining early cerebrovascular events as potential therapeutic targets. The kinetics of blood-brain barrier (BBB) leakage after reperfusion and the link with parenchymal lesion remain debated. By using in vivo and in vitro approaches, we conducted a kinetic analysis of BBB dysfunction during early reperfusion. After 60 minutes of middle cerebral artery occlusion followed by reperfusion times up to 24 hours in mice, a non-invasive magnetic resonance imaging method, through an original sequence of diffusion-weighted imaging, determined brain water mobility in microvascular compartments (D*) apart from parenchymal compartments (apparent diffusion coefficient). An increase in D* found at 4 hours post reperfusion concurred with the onset of both Evans blue/Dextran extravasations and in vitro BBB opening under oxygen-glucose deprivation and reoxygenation (R). The BBB leakage coincided with an emerging cell death in brain tissue as well as in activated glial cells in vitro. The co-culture of BBB endothelial and glial cells evidenced a recovery of endothelium tightness when glial cells were absent or non-injured during R. Preserving the ischemic brain parenchymal cells within 4 hours of reperfusion may improve therapeutic strategies for cerebrovascular protection against stroke.


Subject(s)
Blood-Brain Barrier/physiopathology , Capillary Permeability/physiology , Stroke/physiopathology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cell Death , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Glucose/metabolism , Kinetics , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Neuroglia/ultrastructure , Oxygen/metabolism , Stroke/metabolism , Stroke/pathology
19.
Acta Neuropathol ; 125(3): 395-412, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23269317

ABSTRACT

The migration of polymorphonuclear granulocytes (PMN) into the brain parenchyma and release of their abundant proteases are considered the main causes of neuronal cell death and reperfusion injury following ischemia. Yet, therapies targeting PMN egress have been largely ineffective. To address this discrepancy we investigated the temporo-spatial localization of PMNs early after transient ischemia in a murine transient middle cerebral artery occlusion (tMCAO) model and human stroke specimens. Using specific markers that distinguish PMN (Ly6G) from monocytes/macrophages (Ly6C) and that define the cellular and basement membrane boundaries of the neurovascular unit (NVU), histology and confocal microscopy revealed that virtually no PMNs entered the infarcted CNS parenchyma. Regardless of tMCAO duration, PMNs were mainly restricted to luminal surfaces or perivascular spaces of cerebral vessels. Vascular PMN accumulation showed no spatial correlation with increased vessel permeability, enhanced expression of endothelial cell adhesion molecules, platelet aggregation or release of neutrophil extracellular traps. Live cell imaging studies confirmed that oxygen and glucose deprivation followed by reoxygenation fail to induce PMN migration across a brain endothelial monolayer under flow conditions in vitro. The absence of PMN infiltration in infarcted brain tissues was corroborated in 25 human stroke specimens collected at early time points after infarction. Our observations identify the NVU rather than the brain parenchyma as the site of PMN action after CNS ischemia and suggest reappraisal of targets for therapies to reduce reperfusion injury after stroke.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain/pathology , Granulocytes/pathology , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Animals , Antigens, CD/metabolism , Antigens, Ly/metabolism , Blood Vessels/pathology , Blood Vessels/physiopathology , Blood-Brain Barrier/pathology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Disease Models, Animal , Endothelium/pathology , Functional Laterality , Gene Expression Regulation/physiology , Glucose/deficiency , Humans , Hypoxia , Male , Mice , Mice, Inbred C57BL , Models, Biological , Oxygen/administration & dosage
20.
Biol Aujourdhui ; 206(3): 161-76, 2012.
Article in French | MEDLINE | ID: mdl-23171839

ABSTRACT

Since it was discovered and its brain-protective role characterized, the blood-brain barrier (BBB), through the permeability-restricting action of the brain capillary endothelial cells, has been representing a hurdle for 95% of new medical compounds targeting the central nervous system. Recently, a BBB dysfunction is being found in an increasing number of pathologies such as brain ischaemic stroke, whose only therapy consists in a pharmacological thrombolysis limited to a small percentage of the admitted patients, because of the toxical effects of thrombolytics. And since the clinical failure of promising neuroprotectants, numerous studies of brain ischaemia were carried out, with physiopathological or pharmacological approaches refocused on the BBB, whose structural complexity is now expanded to perivascular cells, all forming a functional unit named the neurovascular unit (NVU). Nevertheless, in spite of the numerous molecular mechanisms identified, the process of BBB dysfunction in the ischaemia/reperfusion cascade remains insufficiently established to explain the pleiotropic action exerted by new pharmacological compounds, possibly protecting the entire NVU and representing potential treatments.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain Ischemia/drug therapy , Endothelial Cells/physiology , Fibrinolytic Agents/adverse effects , Humans , Neuroprotective Agents , Reperfusion Injury/physiopathology , Stroke/drug therapy , Thrombolytic Therapy
SELECTION OF CITATIONS
SEARCH DETAIL