Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Toxicol Appl Pharmacol ; 465: 116456, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36918128

ABSTRACT

The blood-brain barrier (BBB) protects the brain from toxins but hinders the penetration of neurotherapeutic drugs. Therefore, the blood-to-brain permeability of chemotherapeutics must be carefully evaluated. Here, we aimed to establish a workflow to generate primary cultures of human brain microvascular endothelial cells (BMVECs) to study drug brain permeability and bioavailability. Furthermore, we characterized and validated this BBB model in terms of quantitative expression of junction and drug-transport proteins, and drug permeability. We isolated brain microvessels (MVs) and cultured BMVECs from glioma patient biopsies. Then, we employed targeted LC-MS proteomics for absolute protein quantification and immunostaining to characterize protein localization and radiolabeled drugs to predict drug behavior at the Human BBB. The abundance levels of ABC transporters, junction proteins, and cell markers in the cultured BMVECs were similar to the MVs and correctly localized to the cell membrane. Permeability values (entrance and exit) and efflux ratios tested in vitro using the primary BMVECs were within the expected in vivo values. They correctly reflected the transport mechanism for 20 drugs (carbamazepine, diazepam, imipramine, ketoprofen, paracetamol, propranolol, sulfasalazine, terbutaline, warfarin, cimetidine, ciprofloxacin, digoxin, indinavir, methotrexate, ofloxacin, azidothymidine (AZT), indomethacin, verapamil, quinidine, and prazosin). We established a human primary in vitro model suitable for studying blood-to-brain drug permeability with a characterized quantitative abundance of transport and junction proteins, and drug permeability profiles, mimicking the human BBB. Our results indicate that this approach could be employed to generate patient-specific BMVEC cultures to evaluate BBB drug permeability and develop personalized therapeutic strategies.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Humans , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Proteomics , ATP-Binding Cassette Transporters/metabolism , Permeability
2.
Mol Pharm ; 16(3): 1312-1326, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30721081

ABSTRACT

The effect of cannabidiol (CBD), a high-affinity agonist of the transient receptor potential vanilloid-2 (TRPV2) channel, has been poorly investigated in human brain microvessel endothelial cells (BMEC) forming the blood-brain barrier (BBB). TRPV2 expression and its role on Ca2+ cellular dynamics, trans-endothelial electrical resistance (TEER), cell viability and growth, migration, and tubulogenesis were evaluated in human primary cultures of BMEC (hPBMEC) or in the human cerebral microvessel endothelial hCMEC/D3 cell line. Abundant TRPV2 expression was measured in hCMEC/D3 and hPBMEC by qRT-PCR, Western blotting, nontargeted proteomics, and cellular immunofluorescence studies. Intracellular Ca2+ levels were increased by heat and CBD and blocked by the nonspecific TRP antagonist ruthenium red (RR) and the selective TRPV2 inhibitor tranilast (TNL) or by silencing cells with TRPV2 siRNA. CBD dose-dependently induced the hCMEC/D3 cell number (EC50 0.3 ± 0.1 µM), and this effect was fully abolished by TNL or TRPV2 siRNA. A wound healing assay showed that CBD induced cell migration, which was also inhibited by TNL or TRPV2 siRNA. Tubulogenesis of hCMEC/D3 cells in 3D matrigel cultures was significantly increased by 41 and 73% after a 7 or 24 h CBD treatment, respectively, and abolished by TNL. CBD also increased the TEER of hPBMEC monolayers cultured in transwell, and this was blocked by TNL. Our results show that CBD, at extracellular concentrations close to those observed in plasma of patients treated by CBD, induces proliferation, migration, tubulogenesis, and TEER increase in human brain endothelial cells, suggesting CBD might be a potent target for modulating the human BBB.


Subject(s)
Brain Neoplasms/blood supply , Cannabidiol/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Microvessels/pathology , TRPV Cation Channels/metabolism , Blood-Brain Barrier/metabolism , Calcium/metabolism , Cannabis/chemistry , Cell Line , Cell Survival/drug effects , Electric Impedance , Hot Temperature , Humans , Plant Extracts/pharmacology , Ruthenium Red/pharmacology , TRPV Cation Channels/antagonists & inhibitors , ortho-Aminobenzoates/pharmacology
3.
Mol Pharm ; 15(7): 2528-2538, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29874916

ABSTRACT

Variability in drug response to lithium (Li+) is poorly understood and significant, as only 40% of patients with bipolar disorder highly respond to Li+. Li+ can be transported by sodium (Na+) transporters in kidney tubules or red blood cells, but its transport has not been investigated at the blood-brain barrier (BBB). Inhibition and/or transcriptomic strategies for Na+ transporters such as NHE (SLC9), NBC (SLC4), and NKCC (SLC12) were used to assess their role on Li+ transport in human brain endothelial cells. Na+-free buffer was also used to examine Na+/Li+ countertransport (NLCT) activity. The BBB permeability of Li+ evaluated in the rat was 2% that of diazepam, a high passive diffusion lipophilic compound. Gene expression of several Na+ transporters was determined in hCMEC/D3 cells, human hematopoietic stem-cell-derived BBB models (HBLEC), and human primary brain microvascular endothelial cells (hPBMECs) and showed the following rank order with close expression profile: NHE1 > NKCC1 > NHE5 > NBCn1, while NHE2-4, NBCn2, and NBCe1-2 were barely detected. Li+ influx in hCMEC/D3 cells was increased in Na+-free buffer by 3.3-fold, while depletion of chloride or bicarbonate had no effect. DMA (NHE inhibitor), DIDS (anionic carriers inhibitor), and bumetanide (NKCC inhibitor) decreased Li+ uptake significantly in hCMEC/D3 by 52, 51, and 47%, respectively, while S0859 (NBC inhibitor) increased Li+ influx 2.3-fold. Zoniporide (NHE1 inhibitor) and siRNA against NHE1 had no effect on Li+ influx in hCMEC/D3 cells. Our study shows that NHE1 and/or NHE5, NBCn1, and NKCC1 may play a significant role in the transport of Li+ through the plasma membrane of brain endothelial cells.


Subject(s)
Antimanic Agents/pharmacology , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Lithium Compounds/pharmacology , Solute Carrier Proteins/metabolism , Animals , Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Capillary Permeability/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Endothelial Cells/drug effects , HEK293 Cells , Humans , Lithium Compounds/therapeutic use , Male , Microvessels/cytology , Microvessels/drug effects , Microvessels/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sodium/metabolism , Solute Carrier Proteins/antagonists & inhibitors
4.
Nat Microbiol ; 9(9): 2434-2447, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39103571

ABSTRACT

Loss of endothelial integrity and vascular leakage are central features of sepsis pathogenesis; however, no effective therapeutic mechanisms for preserving endothelial integrity are available. Here we show that, compared to dermal microvessels, brain microvessels resist infection by Neisseria meningitidis, a bacterial pathogen that causes sepsis and meningitis. By comparing the transcriptional responses to infection in dermal and brain endothelial cells, we identified angiopoietin-like 4 as a key factor produced by the brain endothelium that preserves blood-brain barrier integrity during bacterial sepsis. Conversely, angiopoietin-like 4 is produced at lower levels in the peripheral endothelium. Treatment with recombinant angiopoietin-like 4 reduced vascular leakage, organ failure and death in mouse models of lethal sepsis and N. meningitidis infection. Protection was conferred by a previously uncharacterized domain of angiopoietin-like 4, through binding to the heparan proteoglycan, syndecan-4. These findings reveal a potential strategy to prevent endothelial dysfunction and improve outcomes in patients with sepsis.


Subject(s)
Disease Models, Animal , Endothelial Cells , Sepsis , Animals , Sepsis/microbiology , Mice , Humans , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Neisseria meningitidis/genetics , Neisseria meningitidis/metabolism , Angiopoietin-Like Protein 4/metabolism , Angiopoietin-Like Protein 4/genetics , Blood-Brain Barrier/metabolism , Meningococcal Infections/microbiology , Brain/metabolism , Brain/microbiology , Brain/pathology , Mice, Inbred C57BL , Endothelium, Vascular/metabolism , Endothelium, Vascular/microbiology
5.
Front Cell Dev Biol ; 8: 578514, 2020.
Article in English | MEDLINE | ID: mdl-33262985

ABSTRACT

Transient receptor potential vanilloid 1-4 (TRPV1-4) expression and functionality were investigated in brain microvessel endothelial cells (BMEC) forming the blood-brain barrier (BBB) from rat and human origins. In rat, Trpv1-4 were detected by qRT-PCR in the brain cortex, brain microvessels, and in primary cultures of brain microvessel endothelial cells [rat brain microvessel endothelial cells (rPBMEC)]. A similar Trpv1-4 expression profile in isolated brain microvessels and rPBMEC was found with the following order: Trpv4 > Trpv2 > Trpv3 > Trpv1. In human, TRPV1-4 were detected in the BBB cell line human cerebral microvessel endothelial cells D3 cells (hCMEC/D3) and in primary cultures of BMEC isolated from human adult and children brain resections [human brain microvascular endothelial cells (hPBMEC)], showing a similar TRPV1-4 expression profile in both hCMEC/D3 cells and hPBMECs as follow: TRPV2 > > TRPV4 > TRPV1 > TRPV3. Western blotting and immunofluorescence experiments confirmed that TRPV2 and TRPV4 are the most expressed TRPV isoforms in hCMEC/D3 cells with a clear staining at the plasma membrane. A fluorescent dye Fluo-4 AM ester was applied to record intracellular Ca2+ levels. TRPV4 functional activity was demonstrated in mediating Ca2+ influx under stimulation with the specific agonist GSK1016790A (ranging from 3 to 1000 nM, EC50 of 16.2 ± 4.5 nM), which was inhibited by the specific TRPV4 antagonist, RN1734 (30 µM). In contrast, TRPV1 was slightly activated in hCMEC/D3 cells as shown by the weak Ca2+ influx induced by capsaicin at a high concentration (3 µM), a highly potent and specific TRPV1 agonist. Heat-induced Ca2+ influx was not altered by co-treatment with a selective potent TRPV1 antagonist capsazepine (20 µM), in agreement with the low expression of TRPV1 as assessed by qRT-PCR. Our present study reveals an interspecies difference between Rat and Human. Functional contributions of TRPV1-4 subtype expression were not identical in rat and human tissues reflective of BBB integrity. TRPV2 was predominant in the human whereas TRPV4 had a larger role in the rat. This interspecies difference from a gene expression point of view should be taken into consideration when modulators of TRPV2 or TRPV4 are investigated in rat models of brain disorders.

6.
Pharmaceutics ; 12(4)2020 Mar 28.
Article in English | MEDLINE | ID: mdl-32231079

ABSTRACT

Organic cation transporters (OCTs) participate in the handling of compounds in kidneys and at the synaptic cleft. Their role at the blood-brain barrier (BBB) in brain drug delivery is still unclear. The presence of OCT1,2,3 (SLC22A1-3) in mouse, rat and human isolated brain microvessels was investigated by either qRT-PCR, quantitative proteomics and/or functional studies. BBB transport of the prototypical substrate [3H]-1-methyl-4-phenylpyridinium ([3H]-MPP+) was measured by in situ brain perfusion in six mouse strains and in Sprague Dawley rats, in primary human brain microvascular endothelial cells seeded on inserts, in the presence or absence of OCTs and a MATE1 (SLC49A1) inhibitor. The results show negligible OCT1 (SLC22A1) and OCT2 (SLC22A2) expression in either mice, rat or human brain microvessels, while OCT3 expression was identified in rat microvessels by qRT-PCR. The in vitro human cellular uptake of [3H]-MPP+ was not modified by OCTs/MATE-inhibitor. Brain transport of [3H]-MPP+ remains unchanged between 2- and 6-month old mice, and no alteration was observed in mice and rats with inhibitors. In conclusion, the evidenced lack of expression and/or functional OCTs and MATE at the BBB allows the maintenance of the brain homeostasis and function as it prevents an easy access of their neurotoxicant substrates to the brain parenchyma.

7.
Brain Res ; 1150: 1-13, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17434463

ABSTRACT

Physiological studies of the blood-brain barrier (BBB) are often performed in rats. We describe the functional characterization of a reproducible in vitro model of the rat BBB and its validation for investigating mechanisms involved in BBB regulation. Puromycin-purified primary cultures of brain endothelial cells, co-cultured with astrocytes in the presence of hydrocortisone (HC) and cAMP, presented low sucrose permeability (< or =0.1 x 10(-3) cm/min) and high transendothelial electrical resistance (> or =270 Omega cm(2)). Expression of specific BBB markers and their transcripts was detected by immunostaining and RT-PCR, respectively: tight junction proteins (claudin-3 and -5, ZO-1 and occludin) and transporters (P-gp, Bcrp and Oatp-2). RT-PCR experiments demonstrated a role of treatment by astrocytes, HC and cAMP in regulation of the transcript level of tight junction proteins (claudin-5 and ZO-1) as well as transporters (Mdr1a, Mrp3, Mrp4, Bcrp, Glut-1), while transcript level of Mdr1b was significantly decreased. The functionality of efflux pumps (P-gp, Mrps and Bcrp) was demonstrated in the presence of specific inhibitors (PSC833, MK571 or Ko143, respectively) by (i) assessing the uptake of the common substrates rhodamine 123 and daunorubicin and (ii) evaluating apical to basolateral and basolateral to apical polarized transport of daunorubicin. In addition, a good correlation (R=0.94) was obtained between the permeability coefficients of a series of compounds of various lipophilicity and their corresponding in vivo rodent blood-brain transfer coefficients. Taken together, our results provide compelling evidence that puromycin-purified rat brain endothelial cells constitute a reliable model of the rat BBB for physiological and pharmacological characterization of BBB transporters.


Subject(s)
Astrocytes/physiology , Blood-Brain Barrier/physiology , Capillary Permeability/physiology , Endothelial Cells/physiology , Gene Expression Regulation/physiology , Analysis of Variance , Animals , Animals, Newborn , Blood-Brain Barrier/drug effects , Brain/cytology , Capillary Permeability/drug effects , Cells, Cultured , Coculture Techniques/methods , Cyclic AMP/pharmacology , Electric Impedance , Gene Expression Regulation/drug effects , Hydrocortisone/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Animal , Protein Transport/drug effects , Protein Transport/physiology , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods
8.
PLoS One ; 9(1): e84179, 2014.
Article in English | MEDLINE | ID: mdl-24392113

ABSTRACT

OBJECTIVE: The vascular system is adapted to specific functions in different tissues and organs. Vascular endothelial cells are important elements of this adaptation, leading to the concept of 'specialized endothelial cells'. The phenotype of these cells is highly dependent on their specific microenvironment and when isolated and cultured, they lose their specific features after few passages, making models using such cells poorly predictive and irreproducible. We propose a new source of specialized endothelial cells based on cord blood circulating endothelial progenitors (EPCs). As prototype examples, we evaluated the capacity of EPCs to acquire properties characteristic of cerebral microvascular endothelial cells (blood-brain barrier (BBB)) or of arterial endothelial cells, in specific inducing culture conditions. APPROACH AND RESULTS: First, we demonstrated that EPC-derived endothelial cells (EPDCs) co-cultured with astrocytes acquired several BBB phenotypic characteristics, such as restricted paracellular diffusion of hydrophilic solutes and the expression of tight junction proteins. Second, we observed that culture of the same EPDCs in a high concentration of VEGF resulted, through activation of Notch signaling, in an increase of expression of most arterial endothelial markers. CONCLUSIONS: We have thus demonstrated that in vitro culture of early passage human cord blood EPDCs under specific conditions can induce phenotypic changes towards BBB or arterial phenotypes, indicating that these EPDCs maintain enough plasticity to acquire characteristics of a variety of specialized phenotypes. We propose that this property of EPDCs might be exploited for producing specialized endothelial cells in culture to be used for drug testing and predictive in vitro assays.


Subject(s)
Arteries/cytology , Arteries/metabolism , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Phenotype , Stem Cells/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Capillary Permeability , Cell Culture Techniques , Coculture Techniques , Endothelial Cells/cytology , Fetal Blood/cytology , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Rats , Stem Cells/cytology , Transcriptome , Veins/metabolism
9.
Neurochem Int ; 56(5): 703-10, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20153394

ABSTRACT

Several in vivo studies suggest that docosahexaenoic acid (22:6 n-3), the main n-3 long-chain polyunsaturated fatty acids (LC-PUFA) of brain membranes, could be an important regulator of brain energy metabolism by affecting glucose utilization and the density of the two isoforms of the glucose transporter-1 (GLUT1) (endothelial and astrocytic). This study was conducted to test the hypothesis that 22:6 n-3 in membranes may modulate glucose metabolism in brain endothelial cells. It compared the impact of 22:6 n-3 and the other two main LC-PUFA, arachidonic acid (20:4 n-6) and eicosapentaenoic acid (20:5 n-3), on fatty acid composition of membrane phospholipids, glucose uptake and expression of 55-kDa GLUT1 isoform in two models of rat brain endothelial cells (RBEC), in primary culture and in the immortalized rat brain endothelial cell line RBE4. Without PUFA supplementation, both types of cerebral endothelial cells were depleted in 22:6 n-3, RBE4 being also particularly low in 20:4 n-6. After exposure to supplemental 20:4 n-6, 20:5 n-3 or 22:6 n-3 (15microM, i.e. a physiological dose), RBEC and RBE4 avidly incorporated these PUFA into their membrane phospholipids thereby resembling physiological conditions, i.e. the PUFA content of rat cerebral microvessels. However, RBE4 were unable to incorporate physiological level of 20:4 n-6. Basal glucose transport in RBEC (rate of [(3)H]-3-o-methylglucose uptake) was increased after 20:5 n-3 or 22:6 n-3 supplementation by 50% and 35%, respectively, whereas it was unchanged with 20:4 n-6. This increase of glucose transport was associated with an increased GLUT1 protein, while GLUT1 mRNA was not affected. The different PUFA did not impact on glucose uptake in RBE4. Due to alterations in n-6 PUFA metabolism and weak expression of GLUT1, RBE4 seems to be less adequate than RBEC to study PUFA metabolism and glucose transport in brain endothelial cells. Physiological doses of n-3 LC-PUFA have a direct and positive effect on glucose transport and GLUT1 density in RBEC that could partly explain decreased brain glucose utilization in n-3 PUFA-deprived rats.


Subject(s)
Brain Chemistry/drug effects , Endothelial Cells/metabolism , Fatty Acids, Omega-3/pharmacology , Glucose/metabolism , 3-O-Methylglucose/metabolism , Animals , Blotting, Western , Capillaries/cytology , Capillaries/drug effects , Capillaries/metabolism , Cells, Cultured , DNA Primers , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Endothelial Cells/drug effects , Fatty Acids/analysis , Fatty Acids/metabolism , Glucose Transporter Type 1/biosynthesis , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Male , Rats , Rats, Wistar
10.
J Cell Sci ; 119(Pt 22): 4634-43, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17062642

ABSTRACT

The conversion of prion protein (PrP(C)) to its protease-resistant isoform is involved in the pathogenesis of prion diseases. Although PrP(C) is highly expressed in neurons and other cell types, its physiological function still remains elusive. Here, we describe how we evaluated its expression, subcellular localization and putative function in brain endothelial cells, which constitute the blood-brain barrier. We detected its expression in microvascular endothelium in mouse brain sections and at intercellular junctions of freshly isolated brain microvessels and cultured brain endothelial cells of mouse, rat and human origin. PrP(C) co-localized with the adhesion molecule platelet endothelial cell adhesion molecule-1 (PECAM-1); moreover, both PrP(C) and PECAM-1 were present in raft membrane microdomains. Using mixed cultures of wild-type and PrP(C)-deficient mouse brain endothelial cells, we observed that PrP(C) accumulation at cell-cell contacts was probably dependent on homophilic interactions between adjacent cells. Moreover, we report that anti-PrP(C) antibodies unexpectedly inhibited transmigration of U937 human monocytic cells as well as freshly isolated monocytes through human brain endothelial cells. Significant inhibition was observed with various anti-PrP(C) antibodies or blocking anti-PECAM-1 antibodies as control. Our results strongly support the conclusion that PrP(C) is expressed by brain endothelium as a junctional protein that is involved in the trans-endothelial migration of monocytes.


Subject(s)
Brain/blood supply , Endothelial Cells/physiology , Intercellular Junctions/metabolism , Monocytes/physiology , Prions/metabolism , Animals , Cell Movement , Cells, Cultured , Humans , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/cytology , Microcirculation/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Prions/genetics , Protein Transport , Rats
SELECTION OF CITATIONS
SEARCH DETAIL