12 research outputs found

    Efficient synthesis of pseudopeptidic molecular cages

    No full text
    Pseudopeptidic cages have been efficiently prepared by combining a dynamic covalent procedure with the suitable preorganization of the building blocks by a conformational bias or an anion templation (see figure). © 2012 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.Financial support from the Spanish MICINN (CTQ2009-14366-C02 project) is gratefully acknowledged.Peer Reviewe

    Compuestos para la inhibición de la apoptosis

    Get PDF
    La presente invención se refiere a compuestos que actúan como inhibidores de la apoptosis, así como a procedimientos para su preparación, a composiciones farmacéuticas que los contienen y a su uso en medicina.Peer reviewedLaboratorios SALVAT SA, Consejo Superior de Investigaciones Científicas (España)T3 Traducción de patente europe

    Anti-Tat and anti-HIV activities of trimers of n-alkylglycines

    No full text
    9 pages, 7 figures.Transcription of human immunodeficiency virus (HIV-1) is activated by viral Tat protein which regulates HIV–LTR transcription and elongation. In the present report, the evaluation of the anti-Tat activity of a combinatorial library composed of 5120 N-trialkylglycines is reported. The antiviral activity was studied through luciferase-based assays targeting the HIV-1 promoter activation induced by the HIV-1 Tat protein. We identified five peptoids with specific anti-HIV-1 Tat activity; none of these peptoids affected the binding of HIV-1 Tat protein to the viral TAR RNA. Using a recombinant-virus assay in which luciferase activity correlates with the rate of HIV-1 transcription we have detected that one of the five selected peptoids, NC37-37-15C, is a potent inhibitor of HIV-1–LTR transcription in both primary T lymphocytes and transformed cell lines. The inhibitory effect of NC37-37-15C, which is additive with azidothymidine (AZT), correlates with its ability to inhibit CTD phosphorylation and shows a suitable profile for development of novel anti-HIV-1 drugs. Likewise, the structural simplicity of N-alkylglycine oligomers makes these peptidomimetics amenable to structural manipulation, thus facilitating the optimisation of lead molecules for drug-like properties.This work was supported by Ministerio de Educación y Ciencia Grant SAF2004-00926 and Fondo de Investigación Sanitaria PI040526 to EM, and Ministerio de Ciencia y Tecnología SAF2001-2286 and Fundació Marató TV3 (2004) to AM.Peer reviewe

    Cationic Peptides and Peptidomimetics Bind Glycosaminoglycans as Potential Sema3A Pathway Inhibitors

    No full text
    Semaphorin3A (Sema3A) is a vertebrate-secreted protein that was initially characterized as a repulsive-guidance cue. Semaphorins have crucial roles in several diseases; therefore, the development of Sema3A inhibitors is of therapeutic interest. Sema3A interacts with glycosaminoglycans (GAGs), presumably through its C-terminal basic region. We used different biophysical techniques (i.e., NMR, surface plasmon resonance, isothermal titration calorimetry, fluorescence, and UV-visible spectroscopy) to characterize the binding of two Sema3A C-terminus-derived basic peptides (FS2 and NFS3) to heparin and chondroitin sulfate A. We found that these peptides bind to both GAGs with affinities in the low-micromolar range. On the other hand, a peptoid named SICHI (semaphorin-induced chemorepulsion inhibitor), which is positively charged at physiological pH, was first identified by our group as being able to block Sema3A chemorepulsion and growth-cone collapse in axons at the extracellular level. To elucidate the direct target for the reported SICHI inhibitory effect in the Sema3A signaling pathway, we looked first to the protein-protein interaction between secreted Sema3A and the Nrp1 receptor. However, our results show that SICHI does not bind directly to the Sema3A sema domain or to Nrp1 extracellular domains. We evaluated a new, to our knowledge, hypothesis, according to which SICHI binds to GAGs, thereby perturbing the Sema3A-GAG interaction. By using the above-mentioned techniques, we observed that SICHI binds to GAGs and competes with Sema3A C-terminus-derived basic peptides for binding to GAGs. These data support the ability of SICHI to block the biologically relevant interaction between Sema3A and GAGs, thus revealing SICHI as a new, to our knowledge, class of inhibitors that target the GAG-protein interaction. © 2016 Biophysical Society.This work was supported by the European Union Seventh Framework Programme (FP7/2007-2013) under Project VISION, grant No. 304884.Peer reviewe

    Semaphorin 3A—Glycosaminoglycans Interaction as Therapeutic Target for Axonal Regeneration

    No full text
    Semaphorin 3A (Sema3A) is a cell-secreted protein that participates in the axonal guidance pathways. Sema3A acts as a canonical repulsive axon guidance molecule, inhibiting CNS regenerative axonal growth and propagation. Therefore, interfering with Sema3A signaling is proposed as a therapeutic target for achieving functional recovery after CNS injuries. It has been shown that Sema3A adheres to the proteoglycan component of the extracellular matrix (ECM) and selectively binds to heparin and chondroitin sulfate-E (CS-E) glycosaminoglycans (GAGs). We hypothesize that the biologically relevant interaction between Sema3A and GAGs takes place at Sema3A C-terminal polybasic region (SCT). The aims of this study were to characterize the interaction of the whole Sema3A C-terminal polybasic region (Sema3A 725–771) with GAGs and to investigate the disruption of this interaction by small molecules. Recombinant Sema3A basic domain was produced and we used a combination of biophysical techniques (NMR, SPR, and heparin affinity chromatography) to gain insight into the interaction of the Sema3A C-terminal domain with GAGs. The results demonstrate that SCT is an intrinsically disordered region, which confirms that SCT binds to GAGs and helps to identify the specific residues involved in the interaction. NMR studies, supported by molecular dynamics simulations, show that a new peptoid molecule (CSIC02) may disrupt the interaction between SCT and heparin. Our structural study paves the way toward the design of new molecules targeting these protein–GAG interactions with potential therapeutic applications.This work was funded by the European Union Seventh Framework Programme (FP7/2007–2013) under Project VISION, grant No. 304884, the Spanish Ministry of Science and Innovation/Spanish Research Agency (MCI/AEI/FEDER, RTI2018–096182-B-I00) and AGAUR (2017 SGR 208).Peer reviewe

    Chemical Modulation of Peptoids: Synthesis and Conformational Studies on Partially Constrained Derivatives

    No full text
    The high conformational flexibility of peptoids can generate problems in biomolecular selectivity as a result of undesired off-target interactions. This drawback can be counterbalanced by restricting the original flexibility to a certain extent, thus leading to new peptidomimetics. By starting from the structure of an active peptoid as an apoptosis inhibitor, we designed two families of peptidomimetics that bear either 7-substituted perhydro- 1,4-diazepine-2,5-dione 2 or 3-substituted 1,4-piperazine-2,5-dione 3 moieties. We report an efficient, solid-phasebased synthesis for both peptidomimetic families 2 and 3 from a common intermediate. An NMR spectroscopic study of 2a,b and 3a,b showed two species in solution in different solvents that interconvert slowly on the NMR timescale. The cis/trans isomerization around the exocyclic tertiary amide bond is responsible for this conformational behavior. The cis isomers are more favored in nonpolar environments, and this preference is higher for the six-membered-ring derivative 3a,b. We propose that the hydrogen-bonding pattern could play an important role in the cis/trans equilibrium process. These hydrogen bonds were characterized in solution, in the solid state (i.e., by using X-ray studies), and by molecularmodeling of simplified systems. A comparative study of a model peptoid 10 containing the isolated tertiary amide bond under study outlined the importance of the heterocyclic moiety for the prevalence of the cis configuration in 2a and 3a. The kinetics of the cis/trans interconversion in 2a, 3a, and 10 was also studied by variable-temperature NMR spectroscopic analysis. The full line-shape analysis of the NMR spectra of 10 revealed negligible entropic contribution to the energetic barrier in this conformational process. A theoretical analysis of 10 supported the results observed by NMR spectroscopic analysis. Overall, these results are relevant for the study of the peptidomimetic/biological- target interactions.Support from MICINN (Grants CTQ2005-00995, SAF2008-00048, BIO2007 60066) is acknowledged. The authors thank F. J. Morales and M. Camargo for technical assistance. A JAE-CSIC fellowship to A.M. is also acknowledged.Peer reviewe

    Protein-protein interaction antagonists as novel inhibitors of non-canonical polyubiquitylation

    Get PDF
    [Background]: Several pathways that control cell survival under stress, namely RNF8-dependent DNA damage recognition and repair, PCNA-dependent DNA damage tolerance and activation of NF-kB by extrinsic signals, are regulated by the tagging of key proteins with lysine 63-based polyubiquitylated chains, catalyzed by the conserved ubiquitin conjugating heterodimeric enzyme Ubc13-Uev. [Methodology/Principal Findings]: By applying a selection based on in vivo protein-protein interaction assays of compounds from a combinatorial chemical library followed by virtual screening, we have developed small molecules that efficiently antagonize the Ubc13-Uev1 protein-protein interaction, inhibiting the enzymatic activity of the heterodimer. In mammalian cells, they inhibit lysine 63-type polyubiquitylation of PCNA, inhibit activation of NF-kB by TNF-a and sensitize tumor cells to chemotherapeutic agents. One of these compounds significantly inhibited invasiveness, clonogenicity and tumor growth of prostate cancer cells. [Conclusions/Significance]: This is the first development of pharmacological inhibitors of non-canonical polyubiquitylation that show that these compounds produce selective biological effects with potential therapeutic applications.This work was funded by grants from the Consejo Superior de Investigaciones Cientificas (PIF200580 to T.M.T., A.R.O. and A. Messeguer), the Ministerio de Educacion y Ciencia (SAF2005-05109-CO2-01 to T.M.T.; CTQ2005-00995 and GEN2003-20642-C09-09 to A. Messeguer; BIO2005-0576, and GEN2003-206420-C09-08 to A.R.O.), the Comunidad de Madrid (GR/SAL/0306/2004 and 200520M157 to A.R.O.), and an institutional grant from the Fundacion Ramon Areces. J.S. and M.G.R. are receipients of fellowships from the Ministerio de Educacion y Ciencia, and A. Moure of a I3P fellowship from the Consejo Superior de Investigaciones Cientificas.Peer reviewe

    Compuestos con actividad inhibidora de las interaciones UBC13-UEV, composiciones farmacéuticas y aplicaciones terapéuticas

    No full text
    Fecha de solicitud: 04-02-2009.- Titular: Consejo Superior de Investigaciones Científicas (CSIC)The invention relates to a compound R-(CR 1 R 2 )q-CO-N(R 3 )-C(R 4R 5 )-CO-NH 2 (I), wherein: R is a heterocyclyl radical; R 1 and R 2 independently represent H or alkyl; R 3 is H, alkyl, cycloalkyl, cycloalkylalkyl, alkenyl, aryl, arylalkyl, heterocyclyl or heterocyclylalkyl; R 4 and R 5 independently represent H or alkyl; q is a number between 0 and 1; and the salts, solvates, pro-drugs or stereoisomers thereof, which can inhibit UBC13-UEV interactions and be used in the production of pharmaceutical compositions intended for antitumour therapy or the treatment and/or prophylaxis of diseases associated with metabolic routes involving the UBC13 enzyme, metabolic routes involving transcriptional factor NF-kappaB or routes involving PCNA or RAD6.La invención relaciona a un r del compuesto (CR 1 R 2) q-CO-n (R 3) - C (R 4 R 5) - CO-NH 2 (i), en donde: R es un heterociclil radical R 1 y R 2 representan independientemente H o el alquílico R 3 es H, alquílico, cicloalquil, cicloalquilalquilo, alquenil, arílico, arilalquilo, heterociclil o heterocyclylalkyl R 4 y R 5 representan independientemente H o el alquílico q es un número entre 0 y 1 y las sales, los solvates, los pro-drugs o los stereoisomers de eso, que el bote inhibe interacciones de UBC13-UEV y sea utilizado en la producción de composiciones farmacéuticas destinadas para la terapia antitumour o el tratamiento y/o la profilaxis de las enfermedades asociadas con las vías metabólicas que implican la enzima UBC13, las vías metabólicas que implican el factor transcripcional N-F-kappaB o las vías que implican PCNA o RAD6.Peer reviewe

    Polymer conjugate compounds for inhibition of apoptosis

    No full text
    Fecha de presentación internacional 21.05.2007.-- Titulares: Laboratorios Salvat, S.A., Consejo Superior de Investigaciones Científicas.The present invention relates to polymer cdonjugates compounds, as well as processes for their preparation, to pharmatheutical compositions containing them and their use in medicine.Peer reviewe

    Triazine-Based Vanilloid 1 Receptor Open Channel Blockers: Design, Synthesis, Evaluation, and SAR Analysis

    No full text
    The thermosensory transient receptor potential vanilloid 1 channel (TRPV1) is a polymodal receptor activated by physical and chemical stimuli. TRPV1 activity is drastically potentiated by proinflammatory agents released upon tissue damage. Given the pivotal role of TRPV1 in human pain, there is pressing need for improved TRPV1 antagonists, the development of which will require identification of new pharmacophore scaffolds. Uncompetitive antagonists acting as open-channel blockers might serve as activity-dependent blockers that preferentially modulate the activity of overactive channels, thus displaying fewer side effects than their competitive counterparts. Herein we report the design, synthesis, biological evaluation, and SAR analysis of a family of triazine-based compounds acting as TRPV1 uncompetitive antagonists. We identified the triazine 8aA as a potent, pure antagonist that inhibits TRPV1 channel activity with nanomolar efficacy and strong voltage dependency. It represents a new class of activity-dependent TRPV1 antagonists and may serve as the basis for lead optimization in the development of new analgesics.This work was supported by grants from Spanish Ministry of Science and Innovation (Grant SAF2008-00048 to A.M., Grant BFU2009-08346 to A.F.-M., CONSOLIDER-INGENIO 2010 (Grant CSD2008-00005) to A.F.-M., J.M.G.-R., and A.M.), from Fundació La Marató de TV3 (to A.F.-M. and A.M.), and from PROMETEO/2010/046 from the GVA to A.F.-M.Peer reviewe
    corecore