Por favor, use este identificador para citar o enlazar a este item: http://hdl.handle.net/10261/141879
COMPARTIR / EXPORTAR:
logo share SHARE BASE
Visualizar otros formatos: MARC | Dublin Core | RDF | ORE | MODS | METS | DIDL | DATACITE

Invitar a revisión por pares abierta
Campo DC Valor Lengua/Idioma
dc.contributor.advisorDíaz, José Fernando-
dc.contributor.advisorBarasoain, Isabel-
dc.contributor.authorTrigili, Chiaraes_ES
dc.date.accessioned2016-12-23T15:23:17Z-
dc.date.available2016-12-23T15:23:17Z-
dc.date.issued2013-
dc.identifier.urihttp://hdl.handle.net/10261/141879-
dc.description343 p.-82 fig.-34 tab.es_ES
dc.description.abstractIntroduction Microtubules and actin filaments play important biological functions in connection with mitosis, cytokinesis, cell signaling, intracellular transport, and cell motility of eukaryotic cells (1, 2). Any external element that disrupts the correct functioning of tubulin and actin causes harmful effects to the body. Thus, it is reasonable that a majority of pharmacological research has focused on reactive products against proteins, many of which are natural compounds or analogues. There are several clinical drugs based on the stabilization (paclitaxel-like behavior) or destabilization (either vinca-like or colchicine-like behavior) of microtubules with regard to their heterodimeric component, α,β- tubulin (3). On the other hand, none actin-targeting drug has entered yet in clinical studies. Research content and conclusions This work takes a comprehensive approach to structure/function/ mechanistic interaction of different molecules which targets the cytoskeleton. Different compounds received were classified into three groups of different families according to their mechanism of action: microtubule stabilizing agents (dictyostatin and discodermolide family; peloruside A family), microtubule destabilizing compounds (pironetin family; podophyllotoxin family) and ligands acting at the level of actin filaments (amphidinolides family). Given the interest, we have investigated the influence of chemical modifications in the binding affinity of the compounds for tubulin and actin. We employed biochemical and biophysical techniques to determine binding constants and stoichiometries as well as NMR techniques to determine the binding epitopes and bioactive conformations of the compounds. Also we used cell biology techniques to evaluate their effects on a panel of cells both sensitive to chemotherapy and resistant to it through three different mechanisms: overexpression of P-glycoprotein, overexpression of βIII-tubulin isotypes and mutations in the β-tubulin gene.es_ES
dc.description.abstractDictyostatin and discodermolide family. Since gaining FDA approval in 1992, paclitaxel and subsequently its semisynthetic analogue taxotere (docetaxel) have been widely used in clinical studies in regards to oncology treatments, including breast, ovarian, and lung cancers (4-7). Although the cytotoxic drugs of taxanes have great utility as chemotherapeutic agents, they suffer from low aqueous solubility and tend to induce drug resistance in patients (8), further underlining the continued need for the identification of new microtubule stabilizing agents. This need has led to the search for structurally novel natural product scaffolds that share the same mode of action as the taxanes but are more effective to overcome drug resistance. The naturally occurring compounds discodermolide (9) and dictyostatin (10) bind to microtubules, cause cell cycle arrest in G2/M at nanomolar concentrations, and retain antiproliferative activity in paclitaxel-resistant cell lines (11, 12), making these compounds attractive candidates for development as antineoplastic agents. In this study, we examined a series of dictyostatin analogs and discodermolide/dictyostatina hybrids to probe biological and biochemical structure-activity relationships. A set of ten derivatives of dictyostatin and five hybrids of discodermolide and dictyostatin have been evaluated to determine the structural features required to improve the interaction of these compounds with microtubules. The activity of the agents was evaluated against drug-sensitive and drugresistant cancer cell lines. The compounds studied are cytotoxic and less susceptible than paclitaxel to multidrug resistance arising from overexpression of the P-glycoprotein efflux pump, exhibit remarkable potency against cell lines which have the isotype of βIII-tubulin and are not affected by mutations that affect the taxoid binding site of β-tubulin. In cell proliferation assay, statistically significant synergy was found between some compounds investigated and paclitaxel, and between some of these compounds and the peloruside A. These results confirm that the discodermolide/dictiostatina family's compounds, when added in combination with other microtubule stabilizing agents, act synergistically to enhance the antimitotic action of the drugs, but also highlight the complexity of drug interactions in intact cells. In vitro studies with purified tubulin indicate that these agents bind to the paclitaxel site (the binding is exothermic) and directly induce tubulin polymerization. Removal of the methyl group of dictyostatin at position 6, hydrogenation of the (Z)-alkene between positions 2 and 3, substitution of the hydroxyl at position 9 with a methoxy group and addiction of a double bound between positions 4 and 5 resulted in significant enhancements of dictyostatin and discodermolide’s binding affinity for the site and citotoxicity.es_ES
dc.description.abstractThe binding interaction of discodermolide to unassembled α/β-tubulin heterodimers and microtubules has been studied using biochemical and NMR techniques. The use of discodermolide as a water-soluble paclitaxel biomimetic and extensive NMR experiments allowed the detection of binding of microtubule stabilizing agents to unassembled tubulin α/β-heterodimers. The bioactive 3D structure of discodermolide bound to α/β-heterodimers was elucidated and compared to those bound to microtubules. Moreover, the combination of experimental TR-NOE and STD NMR data with CORCEMA-ST calculations indicate that discodermolide targets an additional binding site at the pore of the microtubules, which is different from the internal binding site at the lumen previously determined by electron crystallography. Binding to this pore site can then be considered as the first ligand-protein recognition event that takes place in advance of the drug internalization process and interaction with the lumen of the microtubules (13). Peloruside A family. Peloruside A (14) is a microtubule-stabilizing agent that targets the same site as laulimalide. It binds to microtubules with a 1:1 stoichiometry and with a binding affinity in the low-nM range (15, 16). It reduces the number of microtubular protofilaments in the same way as paclitaxel. Although the binding affinity of the compound is comparable to that of the low-affinity stabilizing agent sarcodictyin (17), peloruside is more active in inducing microtubule assembly and is more cytotoxic to tumor cells; this suggests that the peloruside site is a more effective site for stabilizing microtubules. Acetylation of the C24 hydroxyl group results in inactive compounds. Using data-directed molecular docking simulations, we confirm that peloruside A binds within a pocket on the exterior of β-tubulin at a previously unknown ligand site (18), rather than on α-tubulin as suggested in earlier studies (19).es_ES
dc.description.abstractPironetin family. The action mechanism of a series of pironetin analogues with simplified structure is described in this work. Their cytotoxic activity and their interactions with tubulin have been investigated. It has been found that, while less active than the parent molecule (20-22), the pironetin analogues still share the mechanism of action of the latter and compete for the same binding site to α- tubulin. Variations in the configurations of their stereocenters do not translate into relevant differences between biological activities (23). Podophyllotoxins family. Several pinacol derivatives of podophyllotoxins bearing different side chains and functions at C7 were synthesized through reductive cross-coupling of podophyllotoxone and several aldehydes and ketones. While possessing a hydroxylated chain at C7, the compounds retained their respective hydroxyl group with either the 7α (podo) or 7β (epipodo) configuration. Cytotoxicities against neoplastic cells followed by cell cycle arrest and cellular microtubule disruption were evaluated and mechanistically characterized through tubulin polymerization inhibition and assays of binding to the colchicine site. Compounds of the epipodopinacol (7β-OH) series behaved similarly to podophyllotoxin in all the assays and proved to be the most potent inhibitors. Significantly, 7α-isopropyl-7-deoxypodophyllotoxin (41), without any hydroxyl function, appeared as a promising lead compound for a novel type of tubulin polymerization inhibitors. Experimental results were in overall agreement with modeling and docking studies performed on representative compounds of each series (24).es_ES
dc.description.abstractAmphidinolides family. Biological studies of four small amphidinolides of the amphidinolide family have been reported. Their effect on the proliferation of A2780 (human ovarian carcinoma) and of LoVo (human colon carcinoma) cell lines, as well as on the cytoskeleton proteins tubulin, actin, and intermediate filaments of A549 lung carcinoma cells and PtK2 cells have been investigated. Their effect on actin polymerization was then studied in vitro. The findings indicate that the actin filament cytoskeleton rather than the microtubule cytoskeleton is the biological target of the compounds. We believe that this may also be the case for other small macrolides of the amphidinolide family. The influence of the ligand on the actin assembly in vitro is consistent with the actin disassembly effect observed in treated cells. As the ligands interacted with the disassembled actin (G-actin) in vitro, but they did not show any effect on the polymerized actin (F-actin), we suggest that their mechanism of action involves the inhibition of actin monomer additions to pre-existing filaments. Such an inhibition is similar to, albeit weaker than, that induced by cytochalasin B. Docking calculations of compounds indicate that they may emulate cytochalasin D and B in the inhibition of actin assembly (25). References: (1) Chen, H., Bernstein, B.W., and Bamburg, J.R. (2000). Regulating actin-filament dynamics in vivo. Trends Biochem. Sci 25(1), 19–23. (2) Desai, A., and Mitchison, T.J. (1997). Microtubule polymerization dynamics. Annu Rev Cell Dev Biol 13, 83-117. (3) Perez, E.A. (2009). Microtubule inhibitors: Differentiating tubulininhibiting agents based on mechanisms of action, clinical activity, and resistance. Mol. Cancer Ther 8, 2086–2095. (4) Jordan, M.A., and Wilson, L. (2004). Microtubules as a target for anticancer drugs. Nat Rev Cancer 4, 253-265. (5) Montero, A., Fossella, F., Hortobagyi, G., and Valero, V. (2005). Docetaxel for treatment of solid tumours: a systematic review of clinical data. Lancet Oncol 6, 229-239.es_ES
dc.description.abstract(6) Gridelli, C., Aapro, M., Ardizzoni, A., Balducci, L., De Marinis, F., Kelly, K., Le Chevalier, T., Manegold, C., Perrone, F., Rosell, R., et al. (2005). Treatment of advanced non-small-cell lung cancer in the elderly: results of an international expert panel. J Clin Oncol 23, 3125-3137. (7) Markman, M. (2008). Antineoplastic agents in the management of ovarian cancer: current status and emerging therapeutic strategies. Trends Pharmacol Sci 29, 515-519. (8) Orr, G.A., Verdier-Pinard, P., McDaid, H., Horwitz, S.B. (2003) Mechanisms of Taxol resistance related to microtubules. Oncogene 22(47), 7280-95. (9) Gunasekera, S.P., Gunasekera, M., Longley, R.E., and Schulte, G.K. (1990). Discodermolide - a new bioactive polyhydroxylated lactone from the marine sponge Discodermia dissoluta. J Org Chem 55, 4912-4915. (10) Pettit, G., Cichacz, Z., Gao, F., Boyd, M., and Schmidt, J. (1994). Isolation and structure of the cancer cell growth inhibitor dictyostatin 1. J Chem Soc Chem Commun, 1111-1112. (11) Isbrucker, R.A., Cummins, J., Pomponi, S.A., Longley, R.E., and Wright, A.E. (2003). Tubulin polymerizing activity of dictyostatin-1, a polyketide of marine sponge origin. Biochem Pharmacol 66, 75-82. (12) Madiraju, C., Edler, M.C., Hamel, E., Raccor, B.S., Balachandran, R., Zhu, G., Giuliano, K.A., Vogt, A., Shin, Y., Fournier, J.H., et al. (2005). Tubulin assembly, taxoid site binding, and cellular effects of the microtubulestabilizing agent dictyostatin. Biochemistry 44, 15053-15063. (13) Canales, A., Salarichs, J.R., Trigili, C., Nieto, L., Coderch, C., Andreu, J.M., Paterson, I., Jiménez-Barbero, J., and Díaz, J.F. (2011). Insights into the interaction of discodermolide and docetaxel with dimeric tubulin. Mapping the binding sites of microtubule-stabilizing agents using an integrated NMR and computational approach. ACS Chemical Biology 6, 789-799. (14) West, L.M., Northcote, P.T., and Battershill, C.N. (2000). Peloruside A: A potent cytotoxic macrolide isolated from the New Zealand marine sponge Mycale sp. J Org Chem 65, 445-449. (15) Mooberry, S.L., Tien, G., Hernandez, A.H., Plubrukarn, A., and Davidson, B.S. (1999). Laulimalide and isolaulimalide, new paclitaxel-like microtubule-stabilizing agents. Cancer Res.59(3),653-60. (16) Gaitanos, T.N., Buey, R.M., Díaz, J.F., Northcote, P.T., Teesdale- Spittle, P., Andreu, J.M., and Miller, J.H. (2004). Peloruside A does not bind to the taxoid site on beta-tubulin and retains its activity in multidrug-resistant cell lines. Cancer Res 64, 5063-5067.es_ES
dc.description.abstract(17) Buey, R.M., Barasoain, I., Jackson, E., Meyer, A., Giannakakou, P., Paterson, I., Mooberry, S., Andreu, J.M., and Díaz, J.F. (2005). Microtubule interactions with chemically diverse stabilizing agents: thermodynamics of binding to the paclitaxel site predicts cytotoxicity. Chem. Biol. 12, 1269-1279. (18) Pera, B., Razzak, M., Trigili, C., Pineda, O., Canales, A., Buey, R.M., Jiménez-Barbero, J., Northcote, P.T., Paterson, I., Barasoain, I., et al. (2010). Molecular Recognition of Peloruside A by Microtubules. The C24 Primary Alcohol is Essential for Biological Activity. Chembiochem 11, 1669-1678. (19) Pineda, O., Farras, J., Maccari, L., Manetti, F., Botta, M., and Vilarrasa, J. (2004). Computational comparison of microtubule-stabilising agents laulimalide and peloruside with taxol and colchicine. Bioorg Med Chem Lett 14, 4825-4829. (20) Yoshida, M., Matsui, Y., Ikarashi, Y., Usui, T., Osada, H., and Wakasugi, H. (2007). Antiproliferating activity of the mitotic inhibitor pironetin against vindesine- and paclitaxel-resistant human small cell lung cancer H69 cells. Anticancer Res 27, 729-736. (21) Kondoh, M., Usui, T., Kobayashi, S., Tsuchiya, K., Nishikawa, K., Nishikiori, T., Mayumi, T., and Osada, H. (1998). Cell cycle arrest and antitumor activity of pironetin and its derivatives. Cancer Lett 126, 29-32. (22) Kondoh, M., Usui, T., Nishikiori, T., Mayumi, T., and Osada, H. (1999). Apoptosis induction via microtubule disassembly by an antitumour compound, pironetin. Biochem J 340 (Pt 2), 411-416. (23) Marco, J.A., García-Pla, J., Carda, M., Murga, J., Falomir, E., Trigili, C., Notararigo, S., Díaz, J.F., Barasoain, I. (2011). Design and synthesis of pironetin analogues with simplified structure and study of their interactions with microtubules. Eur J Med Chem 46(5),1630-7. (24) Abad, A., Lopez-Perez, J.L., del Olmo, E., Garcia-Fernandez, L.F., Francesch, A., Trigili, C., Barasoain, I., Andreu, J.M., Diaz, J.F., and San Feliciano, A. (2012). Synthesis and antimitotic and tubulin interaction profiles of novel pinacol derivatives of podophyllotoxins. J Med Chem 55, 6724-6737.es_ES
dc.description.abstract(25) Trigili, C., Pera, B., Barbazanges, M., Cossy, J., Meyer, C., Pineda, O., Rodriguez-Escrich, C., Urpi, F., Vilarrasa, J., Diaz, J.F., et al. (2011). Mechanism of action of the cytotoxic macrolides amphidinolide X and J. Chembiochem 12, 1027-1030.es_ES
dc.language.isospaes_ES
dc.publisherCSIC - Centro de Investigaciones Biológicas Margarita Salas (CIB)es_ES
dc.publisherUniversidad Complutense de Madrides_ES
dc.rightsopenAccesses_ES
dc.subjectMicrotubuloses_ES
dc.subjectActinaes_ES
dc.subjectAgentes cuya diana son microtubuloses_ES
dc.subjectAntitumorales_ES
dc.subjectResistencia a la quimoterapiaes_ES
dc.subjectEstabilizantes de microtúbuloses_ES
dc.titleEstudio y caracterización del mecanismo de acción bioquímico y celular de compuestos antitumorales dirigidos contra el citoesqueletoes_ES
dc.typetesis doctorales_ES
dc.description.peerreviewedPeer reviewedes_ES
dc.contributor.funderMinisterio de Ciencia e Innovación (España)es_ES
dc.relation.csices_ES
oprm.item.hasRevisionno ko 0 false*
dc.identifier.funderhttp://dx.doi.org/10.13039/501100004837es_ES
dc.type.coarhttp://purl.org/coar/resource_type/c_db06es_ES
item.openairetypetesis doctoral-
item.grantfulltextopen-
item.cerifentitytypePublications-
item.openairecristypehttp://purl.org/coar/resource_type/c_18cf-
item.fulltextWith Fulltext-
item.languageiso639-1es-
Aparece en las colecciones: (CIB) Tesis
Ficheros en este ítem:
Fichero Descripción Tamaño Formato
Tesis_Chiara_Trigili_2013.pdf6,66 MBAdobe PDFVista previa
Visualizar/Abrir
Show simple item record

CORE Recommender

Page view(s)

678
checked on 23-abr-2024

Download(s)

899
checked on 23-abr-2024

Google ScholarTM

Check


NOTA: Los ítems de Digital.CSIC están protegidos por copyright, con todos los derechos reservados, a menos que se indique lo contrario.