Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 13091, 2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35906251

ABSTRACT

The measurement of tiny variations in local gravity enables the observation of subterranean features. Gravimeters have historically been extremely expensive instruments, but usable gravity measurements have recently been conducted using MEMS (microelectromechanical systems) sensors. Such sensors are cheap to produce, since they rely on the same fabrication techniques used to produce mobile phone accelerometers. A significant challenge in the development of MEMS gravimeters is maintaining stability over long time periods, which is essential for long term monitoring applications. A standard way to demonstrate gravimeter stability and sensitivity is to measure the periodic elastic distortion of the Earth due to tidal forces-the Earth tides. Here, a 19 day measurement of the Earth tides, with a correlation coefficient to the theoretical signal of 0.975, has been presented. This result demonstrates that this MEMS gravimeter is capable of conducting long-term time-lapse gravimetry, a functionality essential for applications such as volcanology.

2.
Fluids Barriers CNS ; 18(1): 36, 2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34344390

ABSTRACT

BACKGROUND: In vitro models based on brain capillary endothelial cells (BCECs) are among the most versatile tools in blood-brain barrier research for testing drug penetration into the brain and how this is affected by efflux transporters such as P-glycoprotein (Pgp). However, compared to freshly isolated brain capillaries or primary BCECs, the expression of Pgp in immortalized BCEC lines is markedly lower, which prompted us previously to transduce the widely used human BCEC line hCMEC/D3 with a doxycycline-inducible MDR1-EGFP fusion plasmid. The EGFP-labeled Pgp in these cells allows studying the localization and trafficking of the transporter and how these processes are affected by drug exposure. Here we used this strategy for the rat BCEC line RBE4 and performed a face-to-face comparison of RBE4 and hCMEC/D3 wild-type (WT) and MDR1-EGFP transduced cells. METHODS: MDR1-EGFP-transduced variants were derived from WT cells by lentiviral transduction, using an MDR1-linker-EGFP vector. Localization, trafficking, and function of Pgp were compared in WT and MDR1-EGFP transduced cell lines. Primary cultures of rat BCECs and freshly isolated rat brain capillaries were used for comparison. RESULTS: All cells exhibited typical BCEC morphology. However, significant differences were observed in the localization of Pgp in that RBE4-MDR1-EGFP cells expressed Pgp primarily at the plasma membrane, whereas in hCMEC/D3 cells, the Pgp-EGFP fusion protein was visible both at the plasma membrane and in endolysosomal vesicles. Exposure to doxorubicin increased the number of Pgp-EGFP-positive endolysosomes, indicating a lysosomotropic effect. Furthermore, lysosomal trapping of doxorubicin was observed, likely contributing to the protection of the cell nucleus from damage. In cocultures of WT and MDR1-EGFP transduced cells, intercellular Pgp-EGFP trafficking was observed in RBE4 cells as previously reported for hCMEC/D3 cells. Compared to WT cells, the MDR1-EGFP transduced cells exhibited a significantly higher expression and function of Pgp. However, the junctional tightness of WT and MDR1-EGFP transduced RBE4 and hCMEC/D3 cells was markedly lower than that of primary BCECs, excluding the use of the cell lines for studying vectorial drug transport. CONCLUSIONS: The present data indicate that MDR1-EGFP transduced RBE4 cells are an interesting tool to study the biogenesis of lysosomes and Pgp-mediated lysosomal drug trapping in response to chemotherapeutic agents and other compounds at the level of the blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Green Fluorescent Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/analysis , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Animals , Blood-Brain Barrier/chemistry , Cell Line , Cell Line, Transformed , Endothelial Cells/chemistry , Green Fluorescent Proteins/analysis , Humans , Microscopy, Fluorescence/methods , Protein Transport/physiology , Rats , Rats, Wistar , Species Specificity
3.
J Clin Pharmacol ; 61 Suppl 1: S125-S132, 2021 06.
Article in English | MEDLINE | ID: mdl-34185914

ABSTRACT

Pharmacometrics could play a key role in shifting pediatric pharmacotherapy from dosing for an average patient to individualizing dosing. Clinicians can have these quantitative tools at their disposal without requiring significant training through the development of clinical decision support systems with easy-to-use interfaces that have a back-end analysis engine or pharmacometric model that uses extensive electronic health record data to predict an individualized dose for each patient. There has been increased development of these clinical decision support systems recently, and for these tools to make the proper breakthrough into clinical practice, it is of utmost importance to perform rigorous testing to ensure adequate predictive performance. In this article, we walk through the components of a decision support tool and the testing required to determine its robustness using an example of a decision support tool we developed for vancomycin dosing in pediatrics.


Subject(s)
Decision Support Techniques , Delivery of Health Care/methods , Pediatrics/methods , Adolescent , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacokinetics , Child , Child, Preschool , Electronic Health Records , Female , Humans , Infant , Infant, Newborn , Male , Models, Biological , Pharmacokinetics , Software , Vancomycin/administration & dosage , Vancomycin/blood , Vancomycin/pharmacokinetics
4.
Fluids Barriers CNS ; 17(1): 53, 2020 Aug 26.
Article in English | MEDLINE | ID: mdl-32843059

ABSTRACT

BACKGROUND: Predictive in vitro models of the human blood-brain barrier (BBB) are essential in early drug discovery and development. Among available immortalized human brain capillary endothelial cell lines (BCECs), the hCMEC/D3 cell line has become the most widely used in vitro BBB model. However, monolayers of hCMEC/D3 cells form only moderately restrictive barriers, most likely because the major tight junction protein, claudin-5, is markedly downregulated. Thus, hCMEC/D3 monolayers cannot be used for vectorial drug transport experiments, which is a major disadvantage of this model. METHODS: Here we transduced hCMEC/D3 cells with a claudin-5 plasmid and compared the characteristics of these cells with those of hCMEC/D3 wildtype cells and primary cultured porcine BCECs. RESULTS: The claudin-5 transduced hCMEC/D3 exhibited expression levels (and junctional localization) of claudin-5 similar to those of primary cultured porcine BCECs. The transduced cells exhibited increased TEER values (211 Ω cm2) and reduced paracellular mannitol permeability (8.06%/h), indicating improved BBB properties; however, the barrier properties of porcine BCECs (TEER 1650 Ω cm2; mannitol permeability 3.95%/h) were not reached. Hence, vectorial transport of a selective P-glycoprotein substrate (N-desmethyl-loperamide) was not observed in claudin-5 transduced hCMEC/D3 (or wildtype) cells, whereas such drug transport occurred in porcine BCECs. CONCLUSIONS: The claudin-5 transduced hCMEC/D3 cells provide a tool to studying the contribution of claudin-5 to barrier tightness and how this can be further enhanced by additional transfections or other manipulations of this widely used in vitro model of the BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Claudin-5/metabolism , Drug Delivery Systems , Endothelial Cells/metabolism , Animals , Biological Transport , Cell Line , Claudin-5/genetics , Humans , Models, Neurological , Permeability , Sus scrofa , Transfection
5.
Eur J Microbiol Immunol (Bp) ; 8(3): 78-86, 2018 Sep 28.
Article in English | MEDLINE | ID: mdl-30345087

ABSTRACT

ATP-binding cassette (ABC) transporters are of major importance for the restricted access of toxins and drugs to the human body. At the body's barrier tissues like the blood-brain barrier, these transporters are highly represented. Especially, ABCB1 (P-glycoprotein) has been a priority target of pharmaceutical research, for instance, to aid chemotherapy of cancers, therapy resistant epilepsy, and lately even neurodegenerative diseases. To improve translational research, the humanization of mouse genes has become a popular tool although, like recently seen for Abcb1, not all approaches were successful. Here, we report the characterization of another unsuccessful commercially available ABCB1 humanized mouse strain. In vivo assessment of transporter activity using positron emission tomography imaging revealed a severe reduction of ABCB1 function in the brain of these mice. Analyses of brain mRNA and protein expression showed that the murine Abcb1a gene is still expressed in homozygous humanized animals while expression of the human gene is minimal. Promoter region analyses underpinned that the introduced human gene might dysregulate normal expression and provided insights into the regulation of both transcription and translation of Abcb1a. We conclude that insertion of the human coding DNA sequence (CDS) into exon 3 instead of exon 2 most probably represents a more promising strategy for Abcb1a humanization.

6.
Proc Natl Acad Sci U S A ; 115(41): E9590-E9599, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30254169

ABSTRACT

The blood-brain barrier protects the brain against a variety of potentially toxic compounds. Barrier function results from tight junctions between brain capillary endothelial cells and high expression of active efflux transporters, including P-glycoprotein (Pgp), at the apical membrane of these cells. In addition to actively transporting drugs out of the cell, Pgp mediates lysosomal sequestration of chemotherapeutic drugs in cancer cells, thus contributing to drug resistance. Here, we describe that lysosomal sequestration of Pgp substrates, including doxorubicin, also occurs in human and porcine brain endothelial cells that form the blood-brain barrier. This is followed by shedding of drug-sequestering vesicular structures, which stay attached to the apical side of the plasma membrane and form aggregates ("barrier bodies") that ultimately undergo phagocytosis by neutrophils, thus constituting an as-yet-undescribed mechanism of drug disposal. These findings introduce a mechanism that might contribute to brain protection against potentially toxic xenobiotics, including therapeutically important chemotherapeutic drugs.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Lysosomes/metabolism , Neutrophils/metabolism , Phagocytosis/drug effects , Xenobiotics/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Blood-Brain Barrier/pathology , Cell Line , Endothelial Cells/pathology , Humans , Lysosomes/pathology , Neutrophils/pathology , Swine , Xenobiotics/pharmacology
7.
Neuropharmacology ; 140: 107-120, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30081001

ABSTRACT

The mTOR signaling pathway has emerged as a possible therapeutic target for epilepsy. Clinical trials have shown that mTOR inhibitors such as everolimus reduce seizures in tuberous sclerosis complex patients with intractable epilepsy. Furthermore, accumulating preclinical data suggest that mTOR inhibitors may have anti-seizure or anti-epileptogenic actions in other types of epilepsy. However, the chronic use of rapalogs such as everolimus is limited by poor tolerability, particularly by immunosuppression, poor brain penetration and induction of feedback loops which might contribute to their limited therapeutic efficacy. Here we describe two novel, brain-permeable and well tolerated small molecule 1,3,5-triazine derivatives, the catalytic mTORC1/C2 inhibitor PQR620 and the dual pan-PI3K/mTOR inhibitor PQR530. These derivatives were compared with the mTORC1 inhibitors rapamycin and everolimus as well as the anti-seizure drugs phenobarbital and levetiracetam. The anti-seizure potential of these compounds was determined by evaluating the electroconvulsive seizure threshold in normal and epileptic mice. Rapamycin and everolimus only poorly penetrated into the brain (brain:plasma ratio 0.0057 for rapamycin and 0.016 for everolimus). In contrast, the novel compounds rapidly entered the brain, reaching brain:plasma ratios of ∼1.6. Furthermore, they significantly decreased phosphorylation of S6 ribosomal protein in the hippocampus of normal and epileptic mice, demonstrating effective mTOR inhibition. PQR620 and PQR530 significantly increased seizure threshold at tolerable doses. The effect of PQR620 was more marked in epileptic vs. nonepileptic mice, matching the efficacy of levetiracetam. Overall, the novel compounds described here have the potential to overcome the disadvantages of rapalogs for treatment of epilepsy and mTORopathies directly connected to mutations in the mTOR signaling cascade.


Subject(s)
Anticonvulsants , Azabicyclo Compounds , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Enzyme Inhibitors/pharmacology , Epilepsy/complications , Epilepsy/drug therapy , Morpholines , Pyridines , Seizures/complications , Seizures/prevention & control , Triazines , Animals , Anticonvulsants/blood , Anticonvulsants/pharmacokinetics , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Azabicyclo Compounds/blood , Azabicyclo Compounds/pharmacokinetics , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/therapeutic use , Catalysis/drug effects , Electroshock , Everolimus/blood , Everolimus/pharmacokinetics , Everolimus/pharmacology , Female , Hippocampus/drug effects , Hippocampus/metabolism , Levetiracetam/pharmacology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Mice , Morpholines/blood , Morpholines/pharmacokinetics , Morpholines/pharmacology , Morpholines/therapeutic use , Phenobarbital/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Pyridines/blood , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyridines/therapeutic use , Ribosomal Proteins/metabolism , Sirolimus/blood , Sirolimus/pharmacokinetics , Sirolimus/pharmacology , Triazines/blood , Triazines/pharmacokinetics , Triazines/pharmacology , Triazines/therapeutic use
8.
Article in English | MEDLINE | ID: mdl-29564145

ABSTRACT

BACKGROUND: Sequencing of fungal species has demonstrated the existence of thousands of putative secondary metabolite gene clusters, the majority of them harboring a unique set of genes thought to participate in production of distinct small molecules. Despite the ready identification of key enzymes and potential cluster genes by bioinformatics techniques in sequenced genomes, the expression and identification of fungal secondary metabolites in the native host is often hampered as the genes might not be expressed under laboratory conditions and the species might not be amenable to genetic manipulation. To overcome these restrictions, we developed an inducible expression system in the genetic model Aspergillus nidulans. RESULTS: We genetically engineered a strain of A. nidulans devoid of producing eight of the most abundant endogenous secondary metabolites to express the sterigmatocystin Zn(II)2Cys6 transcription factor-encoding gene aflR and its cofactor aflS under control of the nitrate inducible niiA/niaD promoter. Furthermore, we identified a subset of promoters from the sterigmatocystin gene cluster that are under nitrate-inducible AflR/S control in our production strain in order to yield coordinated expression without the risks from reusing a single inducible promoter. As proof of concept, we used this system to produce ß-carotene from the carotenoid gene cluster of Fusarium fujikuroi. CONCLUSION: Utilizing one-step yeast recombinational cloning, we developed an inducible expression system in the genetic model A. nidulans and show that it can be successfully used to produce commercially valuable metabolites.

9.
Am J Pathol ; 187(5): 1106-1120, 2017 May.
Article in English | MEDLINE | ID: mdl-28411859

ABSTRACT

Intestinal homeostasis disturbance through intestinal barrier disruption presumably plays a key role in inflammatory bowel disease (IBD) development. Genetic and candidate gene analyses in an Il10-deficient IBD mouse model system identified Cd14 as a potentially protective candidate gene. The role of Cd14 in colitis development was determined using dextran sulfate sodium (DSS)-induced acute and an Il10-deficiency-induced chronic model of intestinal inflammation. Intestinal permeability was investigated by fluorescein isothiocyanate-dextran uptake assay, quantitative RT-PCR analysis of tight junction proteins, myosin light chain kinase, and proinflammatory cytokine expression. Immunohistological staining of occludin, Ki-67, NF-κB-p65, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay was performed, and intestinal inflammation severity was evaluated histologically. Untreated B6-Cd14-/- mice and wild-type controls did not differ in intestinal barrier function. However, DSS-treated Cd14-deficient and B6-Il10-/-Cd14-/- mice exhibited more severe intestinal barrier disruption, with increased histological scores and proinflammatory cytokine expression, compared to controls. Therefore, Cd14 deficiency did not influence epithelial integrity under steady-state conditions but caused intestinal barrier dysfunction under inflammation. As expected, CD14 overexpression increased barrier integrity. No difference in intestinal epithelial NF-κB translocation was observed between the investigated groups. Intestinal myosin light chain kinase expression decreased in Cd14-deficient mice under steady-state conditions and in the chronic model, whereas no difference was detected in the DSS models. Thus, CD14 plays a protective role in IBD development by enhancing intestinal barrier function.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/physiology , Lipopolysaccharide Receptors/physiology , Acute Disease , Animals , Colitis/physiopathology , Colitis/prevention & control , Colon/metabolism , Disease Models, Animal , Interleukin-10/deficiency , Lipopolysaccharide Receptors/metabolism , Male , Mice, Inbred C57BL , Myosin-Light-Chain Kinase/metabolism , NF-kappa B/metabolism , Permeability , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/physiology , Up-Regulation
10.
Opt Lett ; 42(4): 751-754, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28198863

ABSTRACT

One of the limiting noise sources in the current generation of gravitational wave detectors, such as the advanced laser interferometer gravitational wave observatory (aLIGO), is the thermal noise in the interferometer's test mass coatings. One proposed method to reduce the coupling of this noise source to the gravitational wave readout is using a laser beam in the higher-order spatial LG33 mode within the interferometer. Here we show that the current four-mirror cavities of aLIGO are not compatible with Laguerre-Gauss modes due to astigmatism. A non-degeneracy of modes of the same order could be observed in experiment and simulation. We demonstrate that a non-planar cavity could be used instead as it compensates for the astigmatism and transmits the LG33 mode undisturbed.

11.
Sci Rep ; 6: 29253, 2016 07 04.
Article in English | MEDLINE | ID: mdl-27375084

ABSTRACT

The blood-brain barrier (BBB) controls the entry of compounds into the brain, thereby regulating brain homeostasis. Efflux transporters such as P-glycoprotein (Pgp) significantly contribute to BBB function. Multiple signaling pathways modulate the expression and activity of Pgp in response to xenobiotics and disease. A non-genetic way of intercellular transfer of Pgp occurs in cancer cells, but whether this also occurs in non-cancer cells such as endothelial cells that form the BBB is not known. A human brain endothelial cell line (hCMEC/D3) was used to study whether cell-to-cell Pgp transfer occurs during co-culturing with Pgp-EGFP expressing hCMEC/D3 cells. The Pgp-EGFP fusion protein was transferred from donor to recipient cells by cell-to-cell contact and Pgp-EGFP enriched vesicles, which were exocytosed by donor cells and endocytosed by adherent recipient cells. Flow cytometry experiments with the Pgp substrate eFLUXX-ID Gold demonstrated that the transferred Pgp is functional in the recipient cells. Exposure of the donor cells with inhibitors of histone deacetylases (HDACs) resulted in an enhanced intercellular Pgp transfer. Non-genetic transfer of a resistance phenotype and its regulation by HDACs is a novel mechanism of altering BBB functionality. This mechanism may have important implications for understanding drug-induced alterations in Pgp expression and activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/metabolism , Brain/pathology , Endothelial Cells/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Transcytosis/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Blood-Brain Barrier/drug effects , Cell Line , Cell Separation , Coculture Techniques , Flow Cytometry , Green Fluorescent Proteins/genetics , Humans , Signal Transduction , Transgenes/genetics
12.
PLoS One ; 9(2): e88154, 2014.
Article in English | MEDLINE | ID: mdl-24505408

ABSTRACT

P-glycoprotein (Pgp; ABCB1/MDR1) is a major efflux transporter at the blood-brain barrier (BBB), restricting the penetration of various compounds. In other tissues, trafficking of Pgp from subcellular stores to the cell surface has been demonstrated and may constitute a rapid way of the cell to respond to toxic compounds by functional membrane insertion of the transporter. It is not known whether drug-induced Pgp trafficking also occurs in brain capillary endothelial cells that form the BBB. In this study, trafficking of Pgp was investigated in human brain capillary endothelial cells (hCMEC/D3) that were stably transfected with a doxycycline-inducible MDR1-EGFP fusion plasmid. In the presence of doxycycline, these cells exhibited a 15-fold increase in Pgp-EGFP fusion protein expression, which was associated with an increased efflux of the Pgp substrate rhodamine 123 (Rho123). The chemotherapeutic agent mitomycin C (MMC) was used to study drug-induced trafficking of Pgp. Confocal fluorescence microscopy of single hCMEC/D3-MDR1-EGFP cells revealed that Pgp redistribution from intracellular pools to the cell surface occurred within 2 h of MMC exposure. Pgp-EGFP exhibited a punctuate pattern at the cell surface compatible with concentrated regions of the fusion protein in membrane microdomains, i.e., lipid rafts, which was confirmed by Western blot analysis of biotinylated cell surface proteins in Lubrol-resistant membranes. MMC exposure also increased the functionality of Pgp as assessed in three functional assays with Pgp substrates (Rho123, eFluxx-ID Gold, calcein-AM). However, this increase occurred with some delay after the increased Pgp expression and coincided with the release of Pgp from the Lubrol-resistant membrane complexes. Disrupting rafts by depleting the membrane of cholesterol increased the functionality of Pgp. Our data present the first direct evidence of drug-induced Pgp trafficking at the human BBB and indicate that Pgp has to be released from lipid rafts to gain its full functionality.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/drug effects , Brain/metabolism , Endothelial Cells/drug effects , Mitomycin/pharmacology , Protein Transport/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Cholesterol/metabolism , Endothelial Cells/metabolism , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Rhodamine 123
13.
Pharm Res ; 31(6): 1588-604, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24477677

ABSTRACT

PURPOSE: The expression of P-glycoprotein (Pgp) is increased in brain capillary endothelial cells (BCECs) of patients with pharmacoresistant epilepsy. This may restrict the penetration of antiepileptic drugs (AEDs) into the brain. However, the mechanisms underlying increased Pgp expression in epilepsy patients are not known. One possibility is that AEDs induce the expression and functionality of Pgp in BCECs. Several older AEDs that induce human cytochrome P450 enzymes also induce Pgp in hepatocytes and enterocytes, but whether this extends to Pgp at the human BBB and to newer AEDs is not known. METHODS: This prompted us to study the effects of various old and new AEDs on Pgp functionality in the human BCEC line, hCMEC/D3, using the rhodamine 123 (Rho123) efflux assay. For comparison, experiments were performed in two rat BCEC lines, RBE4 and GPNT, and primary cultures of rat and pig BCECs. Furthermore, known Pgp inducers, such as dexamethasone and several cytostatic drugs, were included in our experiments. RESULTS: Under control conditions, GPNT cells exhibited the highest and RBE4 the lowest Pgp expression and Rho123 efflux, while intermediate values were determined in hCMEC/D3. Known Pgp inducers increased Rho123 efflux in all cell lines, but marked inter-cell line differences in effect size were observed. Of the various AEDs examined, only carbamazepine (100 µM) moderately increased Pgp functionality in hCMEC/D3, while valproate (300 µM) inhibited Pgp. CONCLUSIONS: These data do not indicate that treatment with AEDs causes a clinically relevant induction in Pgp functionality in BCECs that form the BBB.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anticonvulsants/pharmacology , Antineoplastic Agents/pharmacology , Endothelial Cells/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Brain/metabolism , Capillaries/cytology , Capillaries/drug effects , Cell Line , Dexamethasone/pharmacology , Humans , Primary Cell Culture , Rats , Swine
14.
Int J Pharm ; 423(2): 440-51, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22197758

ABSTRACT

Curcuminoid-loaded solid lipid nanoparticles (SLN) were produced by melt-homogenization. The used lipid matrices were medium chain triglycerides, trimyristin and tristearin. The resulting nanoparticles had an anisometric shape and a platelet-like structure. Curcuminoid-loaded trimyristin particles did not solidify when stored at room temperature. The supercooled state of trimyristin was studied by DSC and (1)H NMR experiments. A partial recrystallization of the lipid matrix was detected but no change of the mobility of the lipid was noted. Nanoparticles based on tristearin had an α- and ß-modification which was subsequently converted into the stable ß-phase. Curcuminoids did neither influence the melting behavior nor the crystalline or geometric structure of the particles. The interactions between the curcuminoids and the lipid matrix were investigated by Raman and fluorescence spectroscopy. The shape of the curcuminoid bands in the Raman spectra suggested that the drug was in an amorphous state. The fluorescence spectra showed an effect of the lipid matrix on fluorescence properties of the curcuminoids. It was further demonstrated that the drug was not secluded by the solid lipid matrix, but it was influenced by the surrounding aqueous environment. Fluorescence anisotropy measurements revealed a decreased mobility of the curcuminoids within the nanodispersions. From the results of Raman and fluorescence measurements it was concluded that the drug was mainly located on the surface of the crystalline particles.


Subject(s)
Curcumin/chemistry , Drug Carriers , Lipids/chemistry , Nanoparticles , Calorimetry, Differential Scanning , Chemistry, Pharmaceutical , Crystallization , Crystallography, X-Ray , Drug Compounding , Magnetic Resonance Spectroscopy , Microscopy, Electron, Transmission , Nanotechnology , Particle Size , Spectrometry, Fluorescence , Spectrum Analysis, Raman , Surface Properties , Technology, Pharmaceutical/methods , Triglycerides/chemistry
15.
Mol Pharmacol ; 80(2): 314-20, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21540293

ABSTRACT

microRNAs (miRNAs), which contribute to the post-transcriptional processing through 3'-untranslated region-interference, have been shown to be involved in the regulation of ATP-binding cassette (ABC) membrane transporters. The aim of this study was to investigate whether ABCC2, an important efflux transporter for various endogenous and exogenous compounds at several compartment barriers, is subject to miRNA-mediated post-transcriptional gene regulation. We screened the expression of 377 human miRNAs in HepG2 cells after 48 h of treatment with 5 µM rifampicin [a pregnane X receptor (PXR) ligand] or vehicle using reverse transcription-polymerase chain reaction-based low-density arrays. Specific miRNA, ABCC2 mRNA, and protein expression were monitored in HepG2 cells undergoing rifampicin treatment for 72 h. Loss- and gain-of-function experiments and reporter gene assays were performed for further confirmation. Highly deregulated miRNAs compared with in silico data revealed miRNA (miR) 379 as candidate miRNA targeting ABCC2 mRNA. Under rifampicin treatment, ABCC2 mRNA increased significantly, with a maximal fold change of 1.56 ± 0.43 after 24 h. In addition, miR-379 increased (maximally 4.10 ± 1.33-fold after 48 h), whereas ABCC2 protein decreased with a maximal fold change of 0.47 ± 0.08 after 72 h. In contrast, transfection of miR-379 inhibitor led to an elevation of ABCC2 protein expression after rifampicin incubation for 48 h. We identify a miRNA negatively regulating ABCC2 on the post-transcriptional level and provide evidence that this miRNA impedes overexpression of ABCC2 protein after a PXR-mediated external transcriptional stimulus in HepG2 cells.


Subject(s)
Down-Regulation/drug effects , Down-Regulation/genetics , MicroRNAs/physiology , Multidrug Resistance-Associated Proteins/biosynthesis , Rifampin/pharmacology , Binding Sites/genetics , Hep G2 Cells , Humans , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , RNA Interference/physiology
16.
BMC Biotechnol ; 10: 31, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20388199

ABSTRACT

BACKGROUND: Toll-like receptor (TLR) 2 is a component of the innate immune system and senses specific pathogen associated molecular patterns (PAMPs) of both microbial and viral origin. Cell activation via TLR2 and other pattern recognition receptors (PRRs) contributes to sepsis pathology and chronic inflammation both relying on overamplification of an immune response. Intracellular antibodies expressed and retained inside the endoplasmatic reticulum (ER-intrabodies) are applied to block translocation of secreted and cell surface molecules from the ER to the cell surface resulting in functional inhibition of the target protein. Here we describe generation and application of a functional anti-TLR2 ER intrabody (alphaT2ib) which was generated from an antagonistic monoclonal antibody (mAb) towards human and murine TLR2 (T2.5) to inhibit the function of TLR2. alphaT2ib is a scFv fragment comprising the variable domain of the heavy chain and the variable domain of the light chain of mAb T2.5 linked together by a synthetic (Gly4Ser)3 amino acid sequence. RESULTS: Coexpression of alphaT2ib and mouse TLR2 in HEK293 cells led to efficient retention and accumulation of TLR2 inside the ER compartment. Co-immunoprecipitation of human TLR2 with alphaT2ib indicated interaction of alphaT2ib with its cognate antigen within cells. alphaT2ib inhibited NF-kappaB driven reporter gene activation via TLR2 but not through TLR3, TLR4, or TLR9 if coexpressed in HEK293 cells. Co-transfection of human TLR2 with increasing amounts of the expression plasmid encoding alphaT2ib into HEK293 cells demonstrated high efficiency of the TLR2-alphaT2ib interaction. The alphaT2ib open reading frame was integrated into an adenoviral cosmid vector for production of recombinant adenovirus (AdV)-alphaT2ib. Transduction with AdValphaT2ib specifically inhibited TLR2 surface expression of murine RAW264.7 and primary macrophages derived from bone marrow (BMM). Furthermore, TLR2 activation dependent TNFalpha mRNA accumulation, as well as TNFalpha translation and release by macrophages were largely abrogated upon transduction of alphaT2ib. alphaT2ib was expressed in BMM and splenocytes over 6 days upon systemic infection with AdValphaT2ib. Systemic transduction applying AdValphaT2ib rendered immune cells largely non-responsive to tripalmitoyl-peptide challenge. Our results show persistent paralysis of TLR2 activity and thus inhibition of immune activation. CONCLUSION: The generated anti-TLR2 scFv intrabody inhibits specifically and very efficiently TLR2 ligand-driven cell activation in vitro and ex vivo. This indicates a therapeutic potential of alphaT2ib in microbial or viral infections.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Macrophages/metabolism , Single-Chain Antibodies/biosynthesis , Toll-Like Receptor 2/metabolism , Adenoviridae , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Base Sequence , Cell Line , Endoplasmic Reticulum/metabolism , Genetic Vectors , Humans , Interleukin-6/analysis , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Signal Transduction , Single-Chain Antibodies/immunology , Toll-Like Receptor 2/immunology , Transfection , Tumor Necrosis Factor-alpha/analysis
17.
J Mol Med (Berl) ; 88(7): 729-40, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20354842

ABSTRACT

Tumor necrosis factor related apoptosis-inducing ligand (TRAIL) and agonistic anti-DR4/TRAIL-R1 and anti-DR5/TRAIL-R2 antibodies are currently under clinical investigation for treatment of different malignancies. TRAIL activates DR4 and DR5 and thereby triggers apoptotic and non-apoptotic signaling pathways, but possible different roles of DR4 or DR5 in these responses has poorly been addressed so far. In the present work, we analyzed cell viability, DISC formation as well as IL-8 and NF-kappaB activation side by side in responses to TRAIL and agonistic antibodies against DR4 (mapatumumab) and against DR5 (lexatumumab) in pancreatic ductal adenocarcinoma cells. We found that all three reagents are able to activate cell death and pro-inflammatory signaling. Death-inducing signaling complex (DISC) analysis revealed that mapatumumab and lexatumumab induce formation of homocomplexes of either DR4 or DR5, whereas TRAIL additionally stimulated the formation of heterocomplexes of both receptors. Notably, blocking of receptors using DR4- and DR5-specific Fab fragments indicated that TRAIL exerted its function predominantly via DR4. Interestingly, inhibition of PKC by Goe6983 enabled DR5 to trigger apoptotic signaling in response to TRAIL and also strongly enhanced lexatumumab-mediated cell death. Our results suggest the existence of mechanisms that silence DR5 for TRAIL- but not for agonistic-antibody treatment.


Subject(s)
Pancreatic Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Enzyme Inhibitors/metabolism , Humans , Immunoglobulin Fab Fragments/metabolism , Interleukin-8/metabolism , Jurkat Cells , NF-kappa B/metabolism , Protein Kinase C/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor/genetics
18.
Phys Rev E Stat Nonlin Soft Matter Phys ; 79(2 Pt 2): 026102, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19391801

ABSTRACT

Two natural and widely used representations for the community structure of networks are clusterings, which partition the vertex set into disjoint subsets, and layouts, which assign the vertices to positions in a metric space. This paper unifies prominent characterizations of layout quality and clustering quality, by showing that energy models of pairwise attraction and repulsion subsume Newman and Girvan's modularity measure. Layouts with optimal energy are relaxations of, and are thus consistent with, clusterings with optimal modularity, which is of practical relevance because the two representations are complementary and often used together.

19.
Cancer Res ; 68(5): 1443-50, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316608

ABSTRACT

Chronic inflammation has been implicated in the pathogenesis of many severe autoimmune disorders, as well as in diabetes, pulmonary diseases, and cancer. Inflammation accompanies most solid cancers including pancreatic ductal adenocarcinoma (PDAC), one of the most fatal cancers with surgery being the only curative therapeutic approach currently available. In the present work, we investigated the role of the major proinflammatory cytokine tumor necrosis factor alpha (TNFalpha) in the malignancy of PDAC cells in vitro and in vivo. In vitro, TNFalpha strongly increased invasiveness of Colo357, BxPc3, and PancTuI cells and showed only moderate antiproliferative effect. TNFalpha treatment of mice bearing orthotopically growing PDAC tumors led to dramatically enhanced tumor growth and metastasis. Notably, we found that PDAC cells themselves secrete TNFalpha. Although inhibition of TNFalpha with infliximab or etanercept only marginally affected proliferation and invasiveness of PDAC cells in vitro, both reagents exerted strong antitumoral effects in vivo. In severe combined immunodeficient mice with orthotopically growing Colo357, BxPc3, or PancTuI tumors, human-specific anti-TNF antibody infliximab reduced tumor growth and metastasis by about 30% and 50%, respectively. Importantly, in a PDAC resection model performed with PancTuI cells, we found an even stronger therapeutic effect for both anti-TNF compounds. Infliximab and etanercept reduced the number of liver metastases by 69% and 42%, respectively, as well as volumes of recurrent tumors by 73% and 51%. Thus, tumor cell-derived TNFalpha plays a profound role in malignancy of PDAC, and inhibition of TNFalpha represents a promising therapeutic option particularly in adjuvant therapy after subtotal pancreatectomy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Humans , Interleukin-8/metabolism , Mice , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL
...