Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 37(2): e22770, 2023 02.
Article in English | MEDLINE | ID: mdl-36688807

ABSTRACT

The search for reliable human blood-brain barrier (BBB) models represents a challenge for the development/testing of strategies aiming to enhance brain delivery of drugs. Human-induced pluripotent stem cells (hiPSCs) have raised hopes in the development of predictive BBB models. Differentiating strategies are thus required to generate endothelial cells (ECs), a major component of the BBB. Several hiPSC-based protocols have reported the generation of in vitro models with significant differences in barrier properties. We studied in depth the properties of iPSCs byproducts from two protocols that have been established to yield these in vitro barrier models. Our analysis/study reveals that iPSCs derivatives endowed with EC features yield high permeability models while the cells that exhibit outstanding barrier properties show principally epithelial cell-like (EpC) features. We found that models containing EpC-like cells express tight junction proteins, transporters/efflux pumps and display a high functional tightness with very low permeability, which are features commonly shared between BBB and epithelial barriers. Our study demonstrates that hiPSC-based BBB models need extensive characterization beforehand and that a reliable human BBB model containing EC-like cells and displaying low permeability is still needed.


Subject(s)
Blood-Brain Barrier , Induced Pluripotent Stem Cells , Humans , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Cell Differentiation , Induced Pluripotent Stem Cells/metabolism , Permeability
2.
Sci Rep ; 9(1): 9183, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235716

ABSTRACT

The blood-brain barrier (BBB) regulates the traffic of molecules into the central nervous system (CNS) and also limits the drug delivery. Due to their flexible properties, liposomes are an attractive tool to deliver drugs across the BBB. We previously characterized gH625, a peptide derived from Herpes simplex virus 1. The present study investigates the efficiency of liposomes functionalized on their surface with gH625 to promote the brain uptake of neuroprotective peptide PACAP (pituitary adenylate cyclase-activating polypeptide). Using a rat in vitro BBB model, we showed that the liposomes preparations were non-toxic for the endothelial cells, as assessed by analysis of tight junction protein ZO1 organization and barrier integrity. Next, we found that gH625 improves the transfer of liposomes across endothelial cell monolayers, resulting in both low cellular uptake and increased transport of PACAP. Finally, in vivo results demonstrated that gH625 ameliorates the efficiency of liposomes to deliver PACAP to the mouse brain after intravenous administration. gH625-liposomes improve both PACAP reaching and crossing the BBB, as showed by the higher number of brain cells labelled with PACAP. gH625-liposomes represent a promising strategy to deliver therapeutic agents to CNS and to provide an effective imaging and diagnostic tool for the brain.


Subject(s)
Blood-Brain Barrier/drug effects , Drug Delivery Systems , Liposomes/pharmacokinetics , Peptides/pharmacokinetics , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacokinetics , Viral Envelope Proteins/pharmacokinetics , Administration, Intravenous , Animals , Biological Transport , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Mice , Pituitary Adenylate Cyclase-Activating Polypeptide/administration & dosage , Rats , Rats, Wistar
3.
Stem Cells Int ; 2017: 1478606, 2017.
Article in English | MEDLINE | ID: mdl-28698717

ABSTRACT

Stem cell-based therapies critically rely on selective cell migration toward pathological or injured areas. We previously demonstrated that human olfactory ectomesenchymal stem cells (OE-MSCs), derived from an adult olfactory lamina propria, migrate specifically toward an injured mouse hippocampus after transplantation in the cerebrospinal fluid and promote functional recoveries. However, the mechanisms controlling their recruitment and homing remain elusive. Using an in vitro model of blood-brain barrier (BBB) and secretome analysis, we observed that OE-MSCs produce numerous proteins allowing them to cross the endothelial wall. Then, pan-genomic DNA microarrays identified signaling molecules that lesioned mouse hippocampus overexpressed. Among the most upregulated cytokines, both recombinant SPP1/osteopontin and CCL2/MCP-1 stimulate OE-MSC migration whereas only CCL2 exerts a chemotactic effect. Additionally, OE-MSCs express SPP1 receptors but not the CCL2 cognate receptor, suggesting a CCR2-independent pathway through other CCR receptors. These results confirm that OE-MSCs can be attracted by chemotactic cytokines overexpressed in inflamed areas and demonstrate that CCL2 is an important factor that could promote OE-MSC engraftment, suggesting improvement for future clinical trials.

4.
FASEB J ; 31(5): 1807-1827, 2017 05.
Article in English | MEDLINE | ID: mdl-28108572

ABSTRACT

The blood-brain barrier (BBB) prevents the entry of many drugs into the brain and, thus, is a major obstacle in the treatment of CNS diseases. There is some evidence that the LDL receptor (LDLR) is expressed at the BBB and may participate in the transport of endogenous ligands from blood to brain, a process referred to as receptor-mediated transcytosis. We previously described a family of peptide vectors that were developed to target the LDLR. In the present study, in vitro BBB models that were derived from wild-type and LDLR-knockout animals (ldlr-/- ) were used to validate the specific LDLR-dependent transcytosis of LDL via a nondegradative route. We next showed that LDLR-targeting peptide vectors, whether in fusion or chemically conjugated to an Ab Fc fragment, promote binding to apical LDLR and transendothelial transfer of the Fc fragment across BBB monolayers via the same route as LDL. Finally, we demonstrated in vivo that LDLR significantly contributes to the brain uptake of vectorized Fc. We thus provide further evidence that LDLR is a relevant receptor for CNS drug delivery via receptor-mediated transcytosis and that the peptide vectors we developed have the potential to transport drugs, including proteins or Ab based, across the BBB.-Molino, Y., David, M., Varini, K., Jabès, F., Gaudin, N., Fortoul, A., Bakloul, K., Masse, M., Bernard, A., Drobecq, L., Lécorché, P., Temsamani, J., Jacquot, G., Khrestchatisky, M. Use of LDL receptor-targeting peptide vectors for in vitro and in vivo cargo transport across the blood-brain barrier.


Subject(s)
Antibodies/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Endocytosis/physiology , Receptors, LDL/metabolism , Animals , Biological Transport/physiology , Drug Delivery Systems/methods , Humans , Mice , Rats , Receptors, LDL/deficiency
5.
J Neuroinflammation ; 13(1): 290, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27832801

ABSTRACT

BACKGROUND: The heterogeneity of endothelial cell types underlies their remarkable ability to sub-specialize and provide specific requirements for a given vascular bed. Here, we compared rat microvascular endothelial cells (MECs) derived from the brain and spinal cord in both basal and inflammatory conditions. METHODS: We used whole rat genome microarrays to compare, at different time points, basal and TNF-α-induced gene expression of rat MECs from in vitro models of the blood-brain barrier (BBB) and blood-spinal cord barrier (BSCB). Validation at both messenger RNA (mRNA) and protein levels was performed on freshly extracted microvessels (MVs) from the brain and spinal cord (BMVs and SCMVs, respectively), as these were considered the closest in vivo tissues to cultured MECs. RESULTS: Most of the genes encoding adhesion/tight junction molecules and known endothelial markers were similarly expressed in brain and spinal cord MECs (BMECs and SCMECs, respectively). However, one striking finding was the higher expression of several Hox genes, which encode transcription factors involved in positional identity. The differential expression of Hoxa9 and Hoxb7 at the mRNA levels as well as protein levels was confirmed in BMVs and SCMVs. Although the TNF-α response was in general higher in BMECs than in SCMECs at 12 h, the opposite was observed at 48 h. Furthermore, we found that expression of Tnfrsf1a and Tnfrsf1b encoding the TNF receptor super-family member 1a/TNFR1 and 1b/TNFR2, respectively, were constitutively higher in BMVs compared to SCMVs. However, only Tnfrsf1b was induced in SCMECs in response to TNF-α at 24 and 48 h. CONCLUSIONS: Our results support a role for HOX members in defining the positional identities of MECs in vivo. Our data also suggest that the delayed transcriptional activation upon TNF-α treatment in SCMECs results from the requirement of the TNF-induced expression of Tnfrsf1b. In contrast, its high basal expression in BMECs might be sufficient to confer an immediate and efficient TNF-α response.


Subject(s)
Brain/cytology , Endothelial Cells/drug effects , Gene Expression/drug effects , Homeodomain Proteins/metabolism , Spinal Cord/cytology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Gene Expression Profiling , Homeodomain Proteins/genetics , Microarray Analysis , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Time Factors
6.
J Cell Physiol ; 230(10): 2415-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25728249

ABSTRACT

Pantethine, a natural low-molecular-weight thiol, shows a broad activity in a large range of essential cellular pathways. It has been long known as a hypolipidemic and hypocholesterolemic agent. We have recently shown that it exerts a neuroprotective action in mouse models of cerebral malaria and Parkinson's disease through multiple mechanisms. In the present study, we looked at its effects on membrane lipid rafts that serve as platforms for molecules engaged in cell activity, therefore providing a target against inappropriate cell response leading to a chronic inflammation. We found that pantethine-treated cells showed a significant change in raft fatty acid composition and cholesterol content, with ultimate downregulation of cell adhesion, CXCL12-driven chemotaxis, and transendothelial migration of various T cell types, including human Jurkat cell line and circulating effector T cells. The mechanisms involved include the alteration of the following: (i) CXCL12 binding to its target cells; (ii) membrane dynamics of CXCR4 and CXCR7, the two CXCL12 receptors; and (iii) cell redox status, a crucial determinant in the regulation of the chemokine system. In addition, we considered the linker for activation of T cells molecule to show that pantethine effects were associated with the displacement from the rafts of the acylated signaling molecules which had their palmitoylation level reduced.. In conclusion, the results presented here, together with previously published findings, indicate that due to its pleiotropic action, pantethine can downregulate the multifaceted process leading to pathogenic T cell activation and migration.


Subject(s)
Cell Movement/drug effects , Cholesterol/metabolism , Lipids/biosynthesis , Lymphocyte Activation/drug effects , Pantetheine/analogs & derivatives , T-Lymphocytes/drug effects , Animals , Chemokine CXCL12/metabolism , Down-Regulation , Humans , Jurkat Cells , Pantetheine/pharmacology , Rats , Signal Transduction/drug effects , T-Lymphocytes/metabolism
7.
J Vis Exp ; (88): e51278, 2014 Jun 28.
Article in English | MEDLINE | ID: mdl-24998179

ABSTRACT

The blood brain barrier (BBB) specifically regulates molecular and cellular flux between the blood and the nervous tissue. Our aim was to develop and characterize a highly reproducible rat syngeneic in vitro model of the BBB using co-cultures of primary rat brain endothelial cells (RBEC) and astrocytes to study receptors involved in transcytosis across the endothelial cell monolayer. Astrocytes were isolated by mechanical dissection following trypsin digestion and were frozen for later co-culture. RBEC were isolated from 5-week-old rat cortices. The brains were cleaned of meninges and white matter, and mechanically dissociated following enzymatic digestion. Thereafter, the tissue homogenate was centrifuged in bovine serum albumin to separate vessel fragments from nervous tissue. The vessel fragments underwent a second enzymatic digestion to free endothelial cells from their extracellular matrix. The remaining contaminating cells such as pericytes were further eliminated by plating the microvessel fragments in puromycin-containing medium. They were then passaged onto filters for co-culture with astrocytes grown on the bottom of the wells. RBEC expressed high levels of tight junction (TJ) proteins such as occludin, claudin-5 and ZO-1 with a typical localization at the cell borders. The transendothelial electrical resistance (TEER) of brain endothelial monolayers, indicating the tightness of TJs reached 300 ohm x cm(2) on average. The endothelial permeability coefficients (Pe) for lucifer yellow (LY) was highly reproducible with an average of 0.26 ± 0.11 x 10(-3) cm/min. Brain endothelial cells organized in monolayers expressed the efflux transporter P-glycoprotein (P-gp), showed a polarized transport of rhodamine 123, a ligand for P-gp, and showed specific transport of transferrin-Cy3 and DiILDL across the endothelial cell monolayer. In conclusion, we provide a protocol for setting up an in vitro BBB model that is highly reproducible due to the quality assurance methods, and that is suitable for research on BBB transporters and receptors.


Subject(s)
Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Cell Culture Techniques/methods , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Biological Transport, Active , Carbocyanines/chemistry , Carbocyanines/pharmacokinetics , Cell Membrane Permeability , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Male , Models, Animal , Rats , Rats, Wistar , Reproducibility of Results , Rhodamine 123/chemistry , Rhodamine 123/pharmacokinetics
8.
Int J Nanomedicine ; 9: 575-88, 2014.
Article in English | MEDLINE | ID: mdl-24531257

ABSTRACT

AIM: The aim of this study was to develop poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) for delivery of a protein - tissue inhibitor of matrix metalloproteinases 1 (TIMP-1) - across the blood-brain barrier (BBB) to inhibit deleterious matrix metalloproteinases (MMPs). MATERIALS AND METHODS: The NPs were formulated by multiple-emulsion solvent-evaporation, and for enhancing BBB penetration, they were coated with polysorbate 80 (Ps80). We compared Ps80-coated and uncoated NPs for their toxicity, binding, and BBB penetration on primary rat brain capillary endothelial cell cultures and the rat brain endothelial 4 cell line. These studies were followed by in vivo studies for brain delivery of these NPs. RESULTS: Results showed that neither Ps80-coated nor uncoated NPs caused significant opening of the BBB, and essentially they were nontoxic. NPs without Ps80 coating had more binding to endothelial cells compared to Ps80-coated NPs. Penetration studies showed that TIMP-1 NPs + Ps80 had 11.21%± 1.35% penetration, whereas TIMP-1 alone and TIMP-1 NPs without Ps80 coating did not cross the endothelial monolayer. In vivo studies indicated BBB penetration of intravenously injected TIMP-1 NPs + Ps80. CONCLUSION: The study demonstrated that Ps80 coating of NPs does not cause significant toxic effects to endothelial cells and that it can be used to enhance the delivery of protein across endothelial cell barriers, both in vitro and in vivo.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Delivery Systems , Lactic Acid/chemistry , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Tissue Inhibitor of Metalloproteinase-1/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Cell Line , Cells, Cultured , Chemistry, Pharmaceutical , Delayed-Action Preparations , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nanomedicine , Nanoparticles/toxicity , Nanotechnology , Polylactic Acid-Polyglycolic Acid Copolymer , Polysorbates/chemistry , Rats , Surface-Active Agents/chemistry , Tissue Inhibitor of Metalloproteinase-1/pharmacokinetics
9.
Tissue Eng ; 9(2): 201-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12740083

ABSTRACT

Tissue engineering is founded on the concept of controlling the behavior of individual cells to stimulate tissue formation. This control is achieved by mimicking signals that manage natural tissue development or repair. These interdependent signals include cytokine delivery, extracellular matrix interactions, and cell-cell communication. Here, we report on the effect of spatial guidance as a signal for nerve tissue regeneration, using a simple in vitro model. We observe the acceleration of neurite extension from rat dorsal root ganglia within micron-scale tubes. Within these hydrogel-filled conduits, neurites were observed to extend more rapidly than when cultured within the hydrogel alone. The spatial cue also induced a change in tissue architecture, with the cabling of cells within the microconduit. The acceleration of neurite extension was found to be independent of conduit diameter within the range of 200 to 635 microm. Finally, our in vitro model enabled quantification of the effect of combining spatial control and localized nerve growth factor delivery.


Subject(s)
Ganglia, Spinal/cytology , Implants, Experimental , Nerve Regeneration , Neurites/physiology , Neurons, Afferent/physiology , Tissue Engineering/methods , Animals , Collagen Type I , Glass , Hydrogel, Polyethylene Glycol Dimethacrylate , Image Processing, Computer-Assisted , Nerve Growth Factor/administration & dosage , Nerve Growth Factor/pharmacology , Nerve Regeneration/drug effects , Neurites/drug effects , Neurons, Afferent/ultrastructure , Porosity , Rats , Rats, Wistar , Time Factors , Tissue Engineering/instrumentation
SELECTION OF CITATIONS
SEARCH DETAIL
...