Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Publication year range
1.
Pharm Res ; 39(7): 1321-1341, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35411506

ABSTRACT

PURPOSE: More than 15 years have passed since the first description of the unbound brain-to-plasma partition coefficient (Kp,uu,brain) by Prof. Margareta Hammarlund-Udenaes, which was enabled by advancements in experimental methodologies including cerebral microdialysis. Since then, growing knowledge and data continue to support the notion that the unbound (free) concentration of a drug at the site of action, such as the brain, is the driving force for pharmacological responses. Towards this end, Kp,uu,brain is the key parameter to obtain unbound brain concentrations from unbound plasma concentrations. METHODS: To understand the importance and impact of the Kp,uu,brain concept in contemporary drug discovery and development, a survey has been conducted amongst major pharmaceutical companies based in Europe and the USA. Here, we present the results from this survey which consisted of 47 questions addressing: 1) Background information of the companies, 2) Implementation, 3) Application areas, 4) Methodology, 5) Impact and 6) Future perspectives. RESULTS AND CONCLUSIONS: From the responses, it is clear that the majority of the companies (93%) has established a common understanding across disciplines of the concept and utility of Kp,uu,brain as compared to other parameters related to brain exposure. Adoption of the Kp,uu,brain concept has been mainly driven by individual scientists advocating its application in the various companies rather than by a top-down approach. Remarkably, 79% of all responders describe the portfolio impact of Kp,uu,brain implementation in their companies as 'game-changing'. Although most companies (74%) consider the current toolbox for Kp,uu,brain assessment and its validation satisfactory for drug discovery and early development, areas of improvement and future research to better understand human brain pharmacokinetics/pharmacodynamics translation have been identified.


Subject(s)
Blood-Brain Barrier , Central Nervous System Agents , Drug Discovery , Brain , Drug Discovery/methods , Humans
2.
J Pharmacol Exp Ther ; 365(2): 336-345, 2018 05.
Article in English | MEDLINE | ID: mdl-29511033

ABSTRACT

Little is known about the impact of the blood-nerve barrier (BNB) on drug distribution into peripheral nerves. In this study, we examined the peripheral nerve penetration in rats of 11 small-molecule drugs possessing diverse physicochemical and transport properties and ProTx-II, a tarantula venom peptide with molecular mass of 3826 Daltons. Each drug was administered as constant rate intravenous infusion for 6 hours (small molecules) or 24 hours (ProTx-II). Blood and tissues including brain, spinal cord, sciatic nerve, and dorsal root ganglion (DRG) were collected for drug concentration measurements. Unbound fractions of a set of compounds were determined by equilibrium dialysis method in rat blood, brains, spinal cords, sciatic nerves, and DRG. We also investigated the influence of N-[4-[2-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-yl)ethyl]phenyl]-5-methoxy-9-oxo-10H-acridine-4-carboxamide (GF120918), a P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) inhibitor, on the peripheral nerve and central nervous system (CNS) tissue penetration of imatinib. We found that: 1) the unbound fraction in brain tissue homogenate highly correlates with that in the spinal cord, sciatic nerve, and DRG for a set of compounds and thus provides a good surrogate for spinal cord and peripheral nerve tissues, 2) small-molecule drugs investigated can penetrate the DRG and sciatic nerve, 3) P-gp and BCRP have a limited impact on the distribution of small-molecule drugs into peripheral nerves, and 4) DRG is permeable to ProTx-II, but its distribution into sciatic nerve and CNS tissues is restricted. These results demonstrate that small-molecule drugs investigated can penetrate peripheral nerve tissues, and P-gp/BCRP may not be a limiting factor at the BNB. Biologics as large as ProTx-II can access the DRG but not sciatic nerve and CNS tissues.


Subject(s)
Peripheral Nerves/metabolism , Pharmaceutical Preparations/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley , Small Molecule Libraries/metabolism
3.
Biochim Biophys Acta ; 1854(8): 979-86, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25882196

ABSTRACT

Evaluation of drug-target interaction kinetics is becoming increasingly important during the drug-discovery process to investigate selectivity of a drug and predict in vivo target occupancy. To date, it remains challenging to obtain kinetic information for interactions between G-protein-coupled receptors (GPCRs) and small-molecule ligands in a label-free manner. Often GPCRs need to be solubilized or even stabilized by mutations which can be difficult and is time consuming. In addition, it is often unclear if the native conformation of the receptors is sustained. In this study, surface plasmon resonance (SPR) and surface acoustic wave (SAW) technologies have been used to detect ligand binding to the GPCR chemokine (C-X-C motif) receptor 4 (CXCR4) expressed in lipoparticles. We first evaluated different strategies to immobilize CXCR4-expressing lipoparticles. The highest small-molecule binding signal in SPR and SAW was achieved with a matrix-free carboxymethylated sensor chip coated with wheat germ agglutinin for lipoparticle capturing. Next, the binding kinetics of the anti-CXCR4 antibody 12G5 raised against a conformational epitope (k(on)=1.83×10(6)M(-1)s(-1), k(off)=2.79×10(-4) s(-1)) and the small molecule AMD3100 (k(on)=5.46×10(5)M(-1)s(-1), k(off)=1.01×10(-2)s(-1)) were assessed by SAW. Our kinetic and affinity data are consistent with previously published radioligand-binding experiments using cells and label-free experiments with solubilized CXCR4. This is the first study demonstrating label-free kinetic characterization of small-molecule binding to a GPCR in the membrane environment. The presented method holds the potential to greatly facilitate label-free assay development for GPRCs that can be expressed at high levels in lipoparticles.


Subject(s)
Cell Membrane/chemistry , Heterocyclic Compounds/chemistry , Receptors, CXCR4/chemistry , Surface Plasmon Resonance , Benzylamines , Cell Membrane/genetics , Cell Membrane/metabolism , Cyclams , HEK293 Cells , Heterocyclic Compounds/metabolism , Humans , Kinetics , Protein Binding , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism
4.
FASEB J ; 25(6): 1983-2000, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21393573

ABSTRACT

Humanin (HN) is a 24-residue peptide displaying a protective activity in vitro against a range of cytotoxic and neurotoxic insults, as well as mediating in vivo amelioration of Alzheimer disease (AD)-related memory impairment in experimental models. Published evidence suggests that the mechanisms through which HN exerts its cyto- and neuroprotective activity may include its secretion and binding to membrane-associated receptors. Here, we describe the identification of a new modulator of HN neuroprotective activity, V-set and transmembrane domain containing 2 like (VSTM2L), previously known as C20orf102. VSTM2L interacts with HN in both yeast and mammalian cells, is secreted in cultured cells, is present in serum, and is selectively expressed in the central nervous system. VSTM2L colocalizes with HN in distinct brain areas as well as in primary cultured neurons, where it plays a role in the modulation of neuronal viability. When tested in HN neuroprotection bioassays, VSTM2L acts as a strong antagonist of HN neuroprotective activity. In summary, VSTM2L is the first example of a secreted antagonist of HN and may play a role in the modulation of HN biological functions.


Subject(s)
Gene Expression Regulation/physiology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Brain/cytology , Brain/metabolism , Cell Line , Cloning, Molecular , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Rats , Rats, Wistar , Saccharomyces cerevisiae , Spinal Cord/metabolism , Two-Hybrid System Techniques
5.
Circ Res ; 106(12): 1839-48, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20431062

ABSTRACT

RATIONALE: Integrins cooperate with growth factor receptors to promote downstream signaling for cell proliferation and migration. However, the mechanism of receptor activation is still unknown. OBJECTIVE: To analyze the mechanism of phosphorylation of the vascular endothelial growth factor receptor (VEGFR)-3 by cell adhesion. METHODS AND RESULTS: We show that VEGFR-3 phosphorylation, induced by cell attachment to the extracellular matrix, is independent from the intrinsic kinase activity of the receptor, as evidenced from phosphorylation cell adhesion experiments with a mutant kinase dead receptor or in the presence of the specific kinase inhibitor MAZ 51. Cell adhesion experiments in the presence of the c-Src inhibitor PP2 or in fibroblast triple knockout for c-Src, Yes, and Fyn (SYF) demonstrate that VEGFR-3 phosphorylation, induced by extracellular matrix, is mediated by c-Src. Kinase assays in vitro with recombinant c-Src show that VEGFR-3 is a direct c-Src target and mass spectrometry analysis identified the sites phosphorylated by c-Src as tyrosine 830, 833, 853, 1063, 1333, and 1337, demonstrating that integrin-mediated receptor phosphorylation induces a phosphorylation pattern that is distinct from that induced by growth factors. Furthermore, pull-down assays show that integrin-mediated VEGFR-3 phosphorylation activates the recruitment to the receptor of the adaptor proteins CRKI/II and SHC inducing activation of JNK. CONCLUSIONS: These data suggest that cell adhesion to extracellular matrix induces a downstream signaling using the tyrosine kinase receptor VEGFR-3 as scaffold.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extracellular Matrix/metabolism , Protein-Tyrosine Kinases/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Adhesion , Cell Line , Cells, Cultured , Collagen Type I/metabolism , Humans , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Models, Animal , Phosphorylation , Proto-Oncogene Proteins c-crk/metabolism , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , Shc Signaling Adaptor Proteins/metabolism , src-Family Kinases
6.
Methods Mol Biol ; 2454: 521-530, 2022.
Article in English | MEDLINE | ID: mdl-33689164

ABSTRACT

Development of central nervous system (CNS) therapeutics and their brain delivery is impeded by the presence of the blood-brain barrier (BBB). In vitro BBB models, in particular human in vitro BBB models, are critical tools for CNS drug research and development. However, the availability of primary human microvascular endothelial cells is very limited for in vitro modeling. Advances in human induced pluripotent stem cell (hiPSC) technologies provide reproducible human cell resources for scientific research, regenerative medicine, and in vitro modeling. In particular, the differentiation of hiPSC into brain endothelial cells provides scalable, renewable and unlimited cells for in vitro BBB modeling that enables rapid screening of CNS drugs in terms of their BBB permeability. The following protocols provide a general guideline for hiPSC culture, differentiation of hiPSC into endothelial cells (hiPSC-ECs), generation of rat primary astrocytes, and establishment of a two-chamber co-culture in vitro BBB model.


Subject(s)
Blood-Brain Barrier , Induced Pluripotent Stem Cells , Animals , Cell Differentiation/physiology , Coculture Techniques , Endothelial Cells , Humans , Rats
7.
Drug Discov Today ; 27(11): 103338, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35973661

ABSTRACT

Significant efforts have been channeled into developing antibodies for the treatment of CNS indications. Disappointment with the first generation of clinical Tau antibodies in Alzheimer's disease has highlighted the challenges in understanding whether an antibody can reach or affect the target in the compartment where it is involved in pathological processes. Here, we highlight different aspects essential for improving translatability of Tau-based immunotherapy.

8.
iScience ; 25(8): 104787, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35992086

ABSTRACT

Despite much progress in developing better drugs, many patients with acute myeloid leukemia (AML) still die within a year of diagnosis. This is partly because it is difficult to identify therapeutic targets that are effective across multiple AML subtypes. One common factor across AML subtypes is the presence of a block in differentiation. Overcoming this block should allow for the identification of therapies that are not dependent on a specific mutation for their efficacy. Here, we used a phenotypic screen to identify compounds that stimulate differentiation in genetically diverse AML cell lines. Lead compounds were shown to decrease tumor burden and to increase survival in vivo. Using multiple complementary target deconvolution approaches, these compounds were revealed to be anti-mitotic tubulin disruptors that cause differentiation by inducing a G2-M mitotic arrest. Together, these results reveal a function for tubulin disruptors in causing differentiation of AML cells.

9.
Nat Rev Drug Discov ; 20(5): 362-383, 2021 05.
Article in English | MEDLINE | ID: mdl-33649582

ABSTRACT

Achieving sufficient delivery across the blood-brain barrier is a key challenge in the development of drugs to treat central nervous system (CNS) disorders. This is particularly the case for biopharmaceuticals such as monoclonal antibodies and enzyme replacement therapies, which are largely excluded from the brain following systemic administration. In recent years, increasing research efforts by pharmaceutical and biotechnology companies, academic institutions and public-private consortia have resulted in the evaluation of various technologies developed to deliver therapeutics to the CNS, some of which have entered clinical testing. Here we review recent developments and challenges related to selected blood-brain barrier-crossing strategies - with a focus on non-invasive approaches such as receptor-mediated transcytosis and the use of neurotropic viruses, nanoparticles and exosomes - and analyse their potential in the treatment of CNS disorders.


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System Agents/pharmacokinetics , Central Nervous System Agents/therapeutic use , Central Nervous System Diseases/drug therapy , Drug Delivery Systems , Animals , Humans
10.
Nanoscale Adv ; 3(9): 2488-2500, 2021 May 04.
Article in English | MEDLINE | ID: mdl-36134165

ABSTRACT

Hollow viral vectors, such as John Cunningham virus-like particles (JC VLPs), provide a unique opportunity to deliver drug cargo into targeted cells and tissue. Current understanding of the entry of JC virus in brain cells has remained insufficient. In particular, interaction of JC VLPs with the blood-brain barrier (BBB) has not been analyzed in detail. Thus, JC VLPs were produced in this study for investigating the trafficking across the BBB. We performed a carotid artery injection procedure for mouse brain to qualitatively study JC VLPs' in vivo binding and distribution and used in vitro approaches to analyze their uptake and export kinetics in brain endothelial cells. Our results show that clathrin-dependent mechanisms contributed to the entry of VLPs into brain endothelial cells, and exocytosis or transcytosis of VLPs across the BBB was observed in vitro. VLPs were found to interact with sialic acid glycans in mouse brain endothelia. The ability of JC VLPs to cross the BBB can be useful in developing a delivery system for transport of genes and small molecule cargoes to the brain.

11.
J Neurochem ; 112(6): 1539-51, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20050968

ABSTRACT

To investigate the role of the Wnt inhibitor Dickkopf-1 (DKK-1) in the pathophysiology of neurodegenerative diseases, we analysed DKK-1 expression and localization in transgenic mouse models expressing familial Alzheimer's disease mutations and a frontotemporal dementia mutation. A significant increase of DKK-1 expression was found in the diseased brain areas of all transgenic lines, where it co-localized with hyperphosphorylated tau-bearing neurons. In TgCRND8 mice, DKK-1 immunoreactivity was detected in neurons surrounding amyloid deposits and within the choline acetyltransferase-positive neurons of the basal forebrain. Active glycogen synthase kinase-3 (GSK-3) was found to co-localize with DKK-1 and phospho-tau staining. Downstream to GSK-3, a significant reduction in beta-catenin translocation to the nucleus, indicative of impaired Wnt signaling functions, was found as well. Cumulatively, our findings indicate that DKK-1 expression is associated with events that lead to neuronal death in neurodegenerative diseases and support a role for DKK-1 as a key mediator of neurodegeneration with therapeutic potential.


Subject(s)
Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Neurodegenerative Diseases/metabolism , Age Factors , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antibodies, Monoclonal/metabolism , Brain/cytology , Brain/metabolism , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/metabolism , Peptide Fragments/metabolism , Phosphopyruvate Hydratase/metabolism , Presenilin-1/genetics , beta Catenin/metabolism , tau Proteins/metabolism
12.
Drug Discov Today ; 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32971235

ABSTRACT

Investment in phenotypic drug discovery has led to increased demand for rapid and robust target deconvolution to aid successful drug development. Although methods for target identification and mechanism of action (MoA) discovery are flourishing, they typically lead to lists of putative targets. Validating which target(s) are involved in the therapeutic mechanism of a compound poses a significant challenge, requiring direct binding, target engagement, and functional studies in relevant physiological contexts. A combination of orthogonal approaches can allow target identification beyond the proteome as well as aid prioritisation for resource-intensive target validation studies.

13.
BMC Bioinformatics ; 10: 141, 2009 May 11.
Article in English | MEDLINE | ID: mdl-19432989

ABSTRACT

BACKGROUND: Quantitative measurements of specific protein phosphorylation sites, as presented here, can be used to investigate signal transduction pathways, which is an important aspect of cell dynamics. The presented method quantitatively compares peptide abundances from experiments using 18O/16O labeling starting from elaborated MS spectra. It was originally developed to study signaling cascades activated by amyloid-beta treatment of neurons used as a cellular model system with relevance to Alzheimer's disease, but is generally applicable. RESULTS: The presented method assesses, in complete cell lysates, the degree of phosphorylation of specific peptide residues from MS spectra using 18O/16O labeling. The abundance of each observed phospho-peptide from two cell states was estimated from three overlapping isotope contours. The influence of peptide-specific labeling efficiency was removed by performing a label swapped experiment and assuming that the labeling efficiency was unchanged upon label swapping. Different degrees of phosphorylation were reported using the fold change measure which was extended with a confidence interval found to reflect the quality of the underlying spectra. Furthermore a new way of method assessment using simulated data is presented. Using simulated data generated in a manner mimicking real data it was possible to show the method's robustness both with increasing noise levels and with decreasing labeling efficiency. CONCLUSION: The fold change error assessable on simulated data was on average 0.16 (median 0.10) with an error-to-signal ratio and labeling efficiency distributions similar to the ones found in the experimentally observed spectra. Applied to experimentally observed spectra a very good match was found to the model (<10% error for 85% of spectra) with a high degree of robustness, as assessed by data removal. This new method can thus be used for quantitative signal cascade analysis of total cell extracts in a high throughput mode.


Subject(s)
Mass Spectrometry/methods , Phosphopeptides/analysis , Computational Biology/methods , Oxygen Isotopes/chemistry , Phosphopeptides/chemistry , Proteome/analysis , Proteomics/methods
14.
J Pharmacol Exp Ther ; 329(2): 459-68, 2009 May.
Article in English | MEDLINE | ID: mdl-19223665

ABSTRACT

The alpha7 nicotinic acetylcholine receptor (nAChR) is a promising target for treatment of cognitive dysfunction associated with Alzheimer's disease and schizophrenia. Here, we report the pharmacological properties of 5-morpholin-4-yl-pentanoic acid (4-pyridin-3-yl-phenyl)-amide [SEN12333 (WAY-317538)], a novel selective agonist of alpha7 nAChR. SEN12333 shows high affinity for the rat alpha7 receptor expressed in GH4C1 cells (K(i) = 260 nM) and acts as full agonist in functional Ca(2+) flux studies (EC(50) = 1.6 microM). In whole-cell patch-clamp recordings, SEN12333 activated peak currents and maximal total charges similar to acetylcholine (EC(50) = 12 microM). The compound did not show agonist activity at other nicotinic receptors tested and acted as a weak antagonist at alpha3-containing receptors. SEN12333 treatment (3 mg/kg i.p.) improved episodic memory in a novel object recognition task in rats in conditions of spontaneous forgetting as well as cognitive disruptions induced via glutamatergic [5H-dibenzo[a,d]cyclohepten-5,10-imine (dizocilpine maleate); MK-801] or cholinergic (scopolamine) mechanisms. This improvement was blocked by the alpha7-selective antagonist methyllycaconitine, indicating that it is mediated by alpha7 activation. SEN12333 also prevented a scopolamine-induced deficit in a passive avoidance task. In models targeting other cognitive domains, including attention and perceptual processing, SEN12333 normalized the apomorphine-induced deficit of prepulse inhibition. Neuroprotection of SEN12333 was demonstrated in quisqualate-lesioned animals in which treatment with SEN12333 (3 mg/kg/day i.p.) resulted in a significant protection of choline acetyltransferase-positive neurons in the lesioned hemisphere. Cumulatively, our results demonstrate that the novel alpha7 nAChR agonist SEN12333 has procognitive and neuroprotective properties, further demonstrating utility of alpha7 agonists for treatment of neurodegenerative and cognitive disorders.


Subject(s)
Cognition Disorders/drug therapy , Morpholines/therapeutic use , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Nicotinic Agonists/therapeutic use , Pyridines/therapeutic use , Receptors, Nicotinic/metabolism , Animals , Behavior, Animal/drug effects , Calcium/metabolism , Cell Line , Cognition/drug effects , Cognition Disorders/metabolism , Humans , Male , Membrane Potentials/drug effects , Molecular Structure , Morpholines/chemistry , Morpholines/pharmacokinetics , Morpholines/pharmacology , Motor Activity/drug effects , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Nicotinic Agonists/chemistry , Nicotinic Agonists/pharmacokinetics , Nicotinic Agonists/pharmacology , Patch-Clamp Techniques , Protein Binding , Pyridines/chemistry , Pyridines/pharmacokinetics , Pyridines/pharmacology , Radioligand Assay , Rats , Rats, Long-Evans , Rats, Wistar , alpha7 Nicotinic Acetylcholine Receptor
15.
Methods Mol Biol ; 1994: 31-39, 2019.
Article in English | MEDLINE | ID: mdl-31124102

ABSTRACT

To better understand and model neurological, in particular neurodegenerative diseases, human induced pluripotent stem cells (hiPSCs) offer a great source for generation of neural cells. We provide a protocol for the differentiation of hiPSc-derived astrocytes in vitro. This protocol not only is chemically defined, that is, it does not use serum, but also allows for the expansion of astrocyte progenitor cells and mature astrocytes. Large batches of hiPSc-derived astrocytes can be stored and used for defined in vitro disease models.


Subject(s)
Astrocytes/cytology , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Cell Culture Techniques , Cell Differentiation/drug effects , Cells, Cultured , Culture Media , Humans , Induced Pluripotent Stem Cells/drug effects , Models, Biological , Neural Stem Cells/drug effects , Neurodegenerative Diseases , Neurogenesis/drug effects
16.
Neurosci Bull ; 35(6): 996-1010, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31079318

ABSTRACT

An in vitro blood-brain barrier (BBB) model is critical for enabling rapid screening of the BBB permeability of the drugs targeting on the central nervous system. Though many models have been developed, their reproducibility and renewability remain a challenge. Furthermore, drug transport data from many of the models do not correlate well with the data for in vivo BBB drug transport. Induced-pluripotent stem cell (iPSC) technology provides reproducible cell resources for in vitro BBB modeling. Here, we generated a human in vitro BBB model by differentiating the human iPSC (hiPSC) line GM25256 into brain endothelial-type cells. The model displayed BBB characteristics including tight junction proteins (ZO-1, claudin-5, and occludin) and endothelial markers (von Willebrand factor and Ulex), as well as high trans-endothelial electrical resistance (TEER) (1560 Ω.cm2 ± 230 Ω.cm2) and γ-GTPase activity. Co-culture with primary rat astrocytes significantly increased the TEER of the model (2970 Ω.cm2 to 4185 Ω.cm2). RNAseq analysis confirmed the expression of key BBB-related genes in the hiPSC-derived endothelial cells in comparison with primary human brain microvascular endothelial cells, including P-glycoprotein (Pgp) and breast cancer resistant protein (BCRP). Drug transport assays for nine CNS compounds showed that the permeability of non-Pgp/BCRP and Pgp/BCRP substrates across the model was strongly correlated with rodent in situ brain perfusion data for these compounds (R2 = 0.982 and R2 = 0.9973, respectively), demonstrating the functionality of the drug transporters in the model. Thus, this model may be used to rapidly screen CNS compounds, to predict the in vivo BBB permeability of these compounds and to study the biology of the BBB.


Subject(s)
Blood-Brain Barrier/physiology , Capillary Permeability/physiology , In Vitro Techniques/methods , Induced Pluripotent Stem Cells/physiology , ATP-Binding Cassette Transporters , Animals , Astrocytes , Cell Differentiation , Cell Line , Cells, Cultured , Coculture Techniques , Endothelial Cells , Humans , Permeability , Rats , Rats, Sprague-Dawley
17.
Proteomics ; 8(5): 930-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18219698

ABSTRACT

Proteomics studies employing primary neurons are difficult due to the neurons' characteristics. We have developed a detergent-based fractionation method which reduces complexity of the protein extracts, is sufficiently fast to allow differential proteomics analysis after treatments of neurons for short time periods, can be applied to small numbers of cells directly in culture plates, and allows differential extraction of proteins in a compartment-specific manner. The sequential use of detergent-containing buffers on neurons in culture plates yields four extracts enriched in cytosolic, membrane-bound or enclosed, nuclear, and cytoskeletal proteins. Fractionation of neurons was validated by comparison of the distribution of known subcellular marker proteins in the four extracts using Western blotting. Comparison of extracts by DIGE showed a clear difference in protein composition demonstrating significant variations with a fold change (FC) of at least 1.20 for 82% of the detected spots. Using proteins identified in these spots that could be assigned a subcellular localization based on descriptions in the Uniprot database, an extraction efficiency of 85% was calculated for cytosolic proteins in extract 1, 90% for membrane-bound and membrane-enclosed proteins in extract 2, 82% for nuclear proteins in extract 3 and 38% for cytoskeletal and RAFT proteins in extract 4.


Subject(s)
Cell Fractionation/methods , Detergents , Neurons/chemistry , Proteomics , Animals , Cell Culture Techniques , Electrophoresis, Gel, Two-Dimensional , Rats
18.
Neurobiol Dis ; 31(1): 145-58, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18571100

ABSTRACT

In a comprehensive proteomics study aiming at the identification of proteins associated with amyloid-beta (Abeta)-mediated toxicity in cultured cortical neurons, we have identified Thimet oligopeptidase (THOP1). Functional modulation of THOP1 levels in primary cortical neurons demonstrated that its overexpression was neuroprotective against Abeta toxicity, while RNAi knockdown made neurons more vulnerable to amyloid peptide. In the TgCRND8 transgenic mouse model of amyloid plaque deposition, an age-dependent increase of THOP1 expression was found in brain tissue, where it co-localized with Abeta plaques. In accordance with these findings, THOP1 expression was significantly increased in human AD brain tissue as compared to non-demented controls. These results provide compelling evidence for a neuroprotective role of THOP1 against toxic effects of Abeta in the early stages of AD pathology, and suggest that the observed increase in THOP1 expression might be part of a compensatory defense mechanism of the brain against an increased Abeta load.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/toxicity , Cerebral Cortex/enzymology , Metalloendopeptidases/biosynthesis , Neurons/enzymology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Blotting, Western , Cells, Cultured , Cerebral Cortex/pathology , Female , Gene Expression , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Middle Aged , Neurons/pathology , Plaque, Amyloid/metabolism , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transfection
19.
Assay Drug Dev Technol ; 6(2): 181-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18471073

ABSTRACT

Heterologous functional expression of alpha7 nicotinic acetylcholine receptors (nAChRs) is difficult to achieve in mammalian cell lines, and the reasons have been associated with a lack of expression of the putative chaperone factor RIC-3. Here, we describe the generation and functional and pharmacological characterization of a Chinese hamster ovary (CHO)-K1 cell line co-expressing the human alpha7 nAChR and RIC-3. Stable recombinant cells expressing alpha7 nAChR on the plasma membrane were selected by binding of fluorochrome-conjugated alpha-bungarotoxin and fluorescence-activated cell sorting. The presence of functional alpha7 channels was demonstrated by whole cell patch clamp recordings. Nicotine and acetylcholine induced rapid desensitizing currents with 50% effective concentration values of 14 and 37 microM, respectively, with agonist-evoked currents detected in approximately 75% of the cell population. Surprisingly, when tested in a FLIPR (Molecular Devices, Sunnyvale, CA) Ca(2+) assay, activation of alpha7 nAChRs was measured only when nicotinic agonists were applied either in the presence of the positive allosteric modulator (PAM) PNU-120596 or after pretreatment of cells with the tyrosine kinase inhibitor genistein. No Ca(2+) influx was measured upon addition of agonists alone or together with allosteric potentiators such as 5-hydroxyindole that predominantly increase the apparent peak amplitude without robustly affecting the current desensitization rate, as exemplified by PNU-120596. These results show that functional alpha7 nAChRs can stably be expressed in the non-neuronal CHO-K1 cell line. This recombinant cell system is useful for characterization of alpha7 nAChRs and to study the mechanism of action of chemical modulators, in particular the detection of PAMs capable of slowing receptor desensitization kinetics.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Receptors, Nicotinic/physiology , Animals , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Electrophysiology , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Genistein/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Isoxazoles/pharmacology , Kinetics , Microscopy, Fluorescence , Nicotinic Agonists/pharmacology , Phenylurea Compounds/pharmacology , RNA/biosynthesis , RNA/genetics , Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/drug effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Transfection , alpha7 Nicotinic Acetylcholine Receptor
20.
Bioorg Med Chem ; 16(4): 2043-52, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18024137

ABSTRACT

A parallel synthesis of aryl azoles with neuroprotective activity is described. All compounds obtained were evaluated in an in vitro assay using a NMDA toxicity paradigm showing a neuroprotective activity between 15% and 40%. The potential biological target of the active compounds was investigated by extensive literature searches based around similar scaffolds with reported neuroprotective activity. The most interesting molecules active in the NMDA toxicity assay (3a and 2g) showed moderate but significant activity in the inhibition of the Site 2 Sodium Channel binding assay at 10 microM. To confirm our hypothesis compounds 3a, c, f and 2g were tested in the Veratridine assay which is one of the excitotoxicity assays of relevance to NaV channels. The compounds tested showed an activity between 40% and 70%. The identification of neuroprotective small molecules and the identification of NaV channels as the potential site of action were the most important goals of this work.


Subject(s)
Azoles/pharmacology , Neuroprotective Agents/chemical synthesis , Animals , Azoles/chemical synthesis , Humans , N-Methylaspartate/toxicity , Neuroprotective Agents/pharmacology , Sodium Channel Blockers/chemical synthesis , Sodium Channel Blockers/pharmacology , Sodium Channels/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL