52 research outputs found

    ComplexTrans Total Ground Mobility Solution Based on Mutual Adaptation and Deep Cooperation of Road and Rail

    Get PDF
    Current land transport is not optimal. Road transport is congested and rail transport is under-utilised and unprofitable. Land transport is based on the burning of fossil fuels and contributes to climate change. Hence the EU’s desire to push for electric propulsion on the road and to make rail the backbone of Europe’s transport system. Developments in transport are solving some problems but creating others. The ComplexTrans project addresses private and public transport of people and freight in and between cities and removes current and upcoming transport problems in a natural way (without restrictions and subsidies), based on the mutual adaptation of electric road and rail vehicles and their deep intermodal and multimodal cooperation and using fast mixed passenger/freight trains. The solution for land transport is not competition but cooperation between road and rail

    Cancer-associated mutations in the ribosomal protein L5 gene dysregulate the HDM2/p53-mediated ribosome biogenesis checkpoint

    No full text
    Perturbations in ribosome biogenesis have been associated with cancer. Such aberrations activate p53 through the RPL5/RPL11/5S rRNA complex-mediated inhibition of HDM2. Studies using animal models have suggested that this signaling pathway might constitute an important anticancer barrier. To gain a deeper insight into this issue in humans, here we analyze somatic mutations in RPL5 and RPL11 coding regions, reported in The Cancer Genome Atlas and International Cancer Genome Consortium databases. Using a combined computational and statistical approach, complemented by a range of biochemical and functional analyses in human cancer cell models, we demonstrate the existence of several mechanisms by which RPL5 mutations may impair wild-type p53 upregulation and ribosome biogenesis. Unexpectedly, the same approach provides only modest evidence for a similar role of RPL11, suggesting that RPL5 represents a preferred target during human tumorigenesis in cancers with wild-type p53. Furthermore, we find that several functional cancer-associated RPL5 somatic mutations occur as rare germline variants in general population. Our results shed light on the so-far enigmatic role of cancer-associated mutations in genes encoding ribosomal proteins, with implications for our understanding of the tumor suppressive role of the RPL5/RPL11/5S rRNA complex in human malignancies

    Transport of olomoucine II at concentrations of 100(A, B, C), 1 µM (D, E, F) and 10 µM (G, H, I) across monolayers of MDCKII-ABCG2 (A, D, G), MDCKII-ABCB1 (B, E, H) and MDCKII-par (C, F, I) cells.

    No full text
    <p>5 µM fumitremorgin C (FTC) was used as a specific ABCG2 inhibitor in MDCKII-ABCG2 cells. 1 µM LY335979 (LY) was employed as a specific ABCB1 and endogenous canine Abcb1 inhibitor in MDCKII-ABCB1 and MDCKII-par cells, respectively. Ratios of olomoucine II transport across cell monolayers (olomoucine II transport in basolateral to apical direction divided by transport in apical to basolateral direction) with or without inhibitor were calculated two hours after olomoucine II addition and statistically compared (see insets). Due to the generation of sulfated conjugate of olomoucine II, transport ratios were determined at 2 h interval to reduce the misrepresenting effect of the metabolite. In basolateral to apical transport direction, olomoucine II was added into the basolateral compartment and its concentrations were determined in the apical compartment. In the opposite transport direction, olomoucine II was applied into the apical compartment and its concentrations were analyzed in the basolateral compartment. ▴, basolateral to apical transport without inhibitor; ▾, apical to basolateral transport without inhibitor; ▵, basolateral to apical transport with inhibitor; ▿, apical to basolateral transport with inhibitor. Data are expressed as means ± SD of three independent experiments. *p<0.05; **p<0.01; ***p<0.001.</p

    Chromatograms of samples from MDCKII-par cells six hours after olomoucine II addition.

    No full text
    <p>(A) olomoucine II was added into apical compartment while olomoucine II and its sulfated conjugate were analyzed in acceptor basolateral compartment, (B) olomoucine II was added into apical compartment while olomoucine II and its sulfated conjugate were analyzed in donor apical compartment, (C) olomoucine II was added into basolateral compartment while olomoucine II and its sulfated conjugate were analyzed in acceptor apical compartment, (D) olomoucine II was added into basolateral compartment while olomoucine II and its sulfated conjugate were analyzed in donor basolateral compartment. This analysis with end point samples was performed for all olomoucine II transport experiments.</p

    Time-dependent generation of sulfated conjugate of olomoucine II in MDCKII-ABCG2 (A, D, G), MDCKII-ABCB1 (B, E, H) and MDCKII-par (C, F, I) cells and its distribution into the apical and basolateral compartments.

    No full text
    <p>Relative quantification of sulfated olomoucine II was calculated as a ratio between peak area of sulfated olomoucine II and the peak area of internal standard (IS). 5 µM fumitremorgin C (FTC), a specific ABCG2 inhibitor, was used in MDCKII-ABCG2 cells for the assessment of possible involvement of ABCG2 in the transport of sulfated metabolite. 1 µM LY335979 (LY) was employed as a specific ABCB1 and endogenous canine Abcb1 inhibitor in MDCKII-ABCB1 and MDCKII-par cells, respectively. Data come from transport experiments with olomoucine II at concentrations of 100 nM (A, B, C), 1 µM (D, E, F) and 10 µM (G, H, I). In basolateral to apical transport direction, olomoucine II was added into the basolateral compartment and its sulfate conjugate was determined in the apical compartment. In the opposite transport direction, olomoucine II was applied into the apical compartment and its sulfated metabolite was analyzed in the basolateral compartment. ▴, transport into apical compartment without inhibitor; ▾, transport into basolateral compartment without inhibitor; ▵, transport into apical compartment with inhibitor; ▿, transport into basolateral compartment with inhibitor. Values are expressed as means ± SD of three independent experiments.</p

    Mass spectra of an unknown peak eluted in the fifth minute of HPLC analysis of olomoucine II transport.

    No full text
    <p>(A) spectrum in positive mode, (B) MS<sup>2</sup> in positive mode, (C) negative mode, (D) MS<sup>2</sup> in negative mode. Based on the nominal mass shift (+80 Da) from parent compound and the collision spectra in negative as well as positive mode the compound was identified as a sulfated conjugate of olomoucine II.</p

    Transport of purvalanol A at concentrations of 1 µM (A, B, C) and 10 µM (D, E, F) across monolayers of MDCKII-ABCG2 (A, D), MDCKII-ABCB1 (B, E) and MDCKII-par (C, F) cells.

    No full text
    <p>5 µM fumitremorgin C (FTC) was used as a specific ABCG2 inhibitor in MDCKII-ABCG2 cells. 1 µM LY335979 (LY) was employed as a specific ABCB1 inhibitor in MDCKII-ABCB1 cells. Ratios of purvalanol A transport across cell monolayers (purvalanol A transport in basolateral to apical direction divided by transport in apical to basolateral direction) with or without inhibitor were calculated and statistically compared (see insets). Transport ratios were determined 6 h after purvalanol A addition. In basolateral to apical transport direction, purvalanol A was added into the basolateral compartment and its concentrations were determined in the apical compartment. In the opposite transport direction, purvalanol A was applied into the apical compartment and its concentrations were analyzed in the basolateral compartment. ▴, basolateral to apical transport without inhibitor; ▾, apical to basolateral transport without inhibitor; ▵, basolateral to apical transport with inhibitor; ▿, apical to basolateral transport with inhibitor. Data are expressed as means ± SD of three independent experiments.</p

    Toll-like receptor dual-acting agonists are potent inducers of PBMC-produced cytokines that inhibit hepatitis B virus production in primary human hepatocytes

    Get PDF
    International audienceRecombinant interferon-α (IFN-α) treatment functionally cures chronic hepatitis B virus (HBV) infection in some individuals and suppresses virus replication in hepatocytes infected in vitro. We studied the antiviral effect of conditioned media (CM) from peripheral blood mononuclear cells (PBMCs) stimulated with agonists of Toll-like receptors (TLRs) 2, 7, 8 and 9. We found that CM from PBMCs stimulated with dual-acting TLR7/8 (R848) and TLR2/7 (CL413) agonists were more potent drivers of inhibition of HBe and HBs antigen secretion from HBV-infected primary human hepatocytes (PHH) than CM from PBMCs stimulated with single-acting TLR7 (CL264) or TLR9 (CpG-B) agonists. Inhibition of HBV in PHH did not correlate with the quantity of PBMC-produced IFN-α, but it was a complex function of multiple secreted cytokines. More importantly, we found that the CM that efficiently inhibited HBV production in freshly isolated PHH via various cytokine repertoires and mechanisms did not reduce covalently closed circular (ccc)DNA levels. We confirmed our data with a cell culture model based on HepG2-NTCP cells and the plasmacytoid dendritic cell line GEN2.2. Collectively, our data show the importance of dual-acting TLR agonists inducing broad cytokine repertoires. The development of poly-specific TLR agonists provides novel opportunities towards functional HBV cure

    Olomoucine II, but not purvalanol A, is transported by breast cancer resistance protein (ABCG2) and P-glycoprotein (ABCB1).

    Get PDF
    Contains fulltext : 125702.pdf (publisher's version ) (Open Access)Purine cyclin-dependent kinase inhibitors have been recognized as promising candidates for the treatment of various cancers; nevertheless, data regarding interaction of these substances with drug efflux transporters is still lacking. Recently, we have demonstrated inhibition of breast cancer resistance protein (ABCG2) by olomoucine II and purvalanol A and shown that these compounds are able to synergistically potentiate the antiproliferative effect of mitoxantrone, an ABCG2 substrate. In this follow up study, we investigated whether olomoucine II and purvalanol A are transported by ABCG2 and ABCB1 (P-glycoprotein). Using monolayers of MDCKII cells stably expressing human ABCB1 or ABCG2, we demonstrated that olomoucine II, but not purvalanol A, is a dual substrate of both ABCG2 and ABCB1. We, therefore, assume that pharmacokinetics of olomoucine II will be affected by both ABCB1 and ABCG2 transport proteins, which might potentially result in limited accumulation of the compound in tumor tissues or lead to drug-drug interactions. Pharmacokinetic behavior of purvalanol A, on the other hand, does not seem to be affected by either ABCG2 or ABCB1, theoretically favoring this drug in the potential treatment of efflux transporter-based multidrug resistant tumors. In addition, we observed intensive sulfatation of olomoucine II in MDCKII cell lines with subsequent active efflux of the metabolite out of the cells. Therefore, care should be taken when performing pharmacokinetic studies in MDCKII cells, especially if radiolabeled substrates are used; the generated sulfated conjugate may largely contaminate pharmacokinetic analysis and result in misleading interpretation. With regard to chemical structures of olomoucine II and purvalanol A, our data emphasize that even drugs with remarkable structure similarity may show different pharmacokinetic behavior such as interactions with ABC transporters or biotransformation enzymes
    • …
    corecore