17 research outputs found

    Fotoquímica de compuestos heteroaromáticos tricíclicos en medios biomiméticos

    Full text link
    [EN] The use of biomimetic media such as serum albumins (SAs), a1-acid glycloproteins (AAGs), cyclodextrins (CDs) and sodium dodecyl sulfate (SDS) micelles is combined with photophysical techniques (fluorescence (F) and laser flash photolysis (LFP)) to investigate the issue of photosensitization by drugs or drug derivatives containing a triciclyc heteroaromatic chromophore, namely carbazole (CBZ) or phenothiazine (FTZ). The photobehavior of the methyl ester of carprofen (CPFMe) in the presence of bovine serum albumin (BSA) has been addressed first. The decay of triplet excited state of CPFMe in the presence of BSA at 1:2 (at 430 nm), needed two monoexponential terms and thus two triplet lifetimes for a satisfactory fitting, which revealed binding to two different sites within the protein. The shorter value of triplet lifetime was ascribed to CPFMe present in site I, based on the quenching of 3CPFMe* by the tryptophan (Trp) residue located in this pocket, through an electron transfer mechanism. No significant differences were found between (S)- and (R)-CPFMe. Within bovine a1-acid glycloprotein (BAAG), only one binding site was observed for both CPFMe enantiomers. Stereodifferentiation was observed in the triplet lifetimes in CPFMe@BAAG complexes, with the shorter lifetimes values for the (S)-enantiomer. CPFMe photobinding to BAAG was detected by following the changes in fluorescence of CPFMe@BAAG mixtures before and after irradiation. The process was more efficient for (S)-CPFMe. The proposed mechanism involves a reductive photodehalogenation from the triplet of (S)-CPFMe, giving rise to covalent adduct (S)-CBZMe-BAAG. As a way to circumvent the unfavorable thermodynamics of homolytic C-Cl cleavage through the triplet excited state, it has been proposed that the actual operating mechanism involves formation of triplet excimers. To deepen into the mechanism of CPFMe dehalogenation, two diastereomeric dyads based on CPF (CPF-CPF and CPF-CBZ) have been synthesized and irradiated. The self-quenching of the triplet excited states in both dyads is much faster than in CPFMe, and is related to formation of charge transfer species, clearly disfavored in non-polar solvents (THF). The trends observed in the triplet lifetimes, as well as the solvent effects on photoreactivity, are in full agreement with the mechanistic picture of a photoreductive dechlorination as operating mechanism in CPF-based systems. The photochemistry of the neuroleptic drug cyamemazine (CMZ) has been investigated in biomimetic media. The encapsulation process has been followed by fluorescence, as a hypsochromic shift of the band, concomitant with enhanced quantum yields and lifetimes. LFP revealed important changes associated with encapsulation, specifically more selective generation of the triplet excited state, which turns to be longer-lived. Light-induced oxidation of CMZ afforded the corresponding radical cation, which was trapped by oxygen to afford the N,S-dioxide. The reaction has been monitored by fluorescence, by the progressive appearance of a emission band at shorter wavelengths. The process resulted to be disfavored in the employed microenvironments (lipophilics). The slowest photooxidation rate corresponds to CMZ in SDS micelles and in AAGs, where the drug is located in a more hydrophobic domain, hardly accessible from the aqueous medium, and photoionization is nearly negligible. LFP of the anti-psychotic chlorpromazine (CPZ) and two phase I metabolites, CPZ-MD and CPZ-DD (mono and didemethylated) in the presence of increasing amounts of HSA, has allowed monitoring binding to the protein, from the enhancement of the Amax value of 3CPZ* (at 470 nm). The binding degree was higher for the parent drug, in agreement with its more hydrophobic character. The use of warfarin as site I displacement probe indicated that the three compounds only bind to this site. Marginal photobinding to HSA was observed in all cases, monitored by subtle changes in the fluorescence.[ES] Se ha planteado el uso de medios biomiméticos como albúminas séricas (ASs), a1-glicoproteinas ácidas (AGAs), ciclodextrinas (CDs) y micelas (SDS) combinado con técnicas fotofísicas (fluorescencia (F) y fotólisis de destello láser (LFP)) para investigar la fotosensibilización por fármacos que contienen un cromóforo carbazol (CBZ) o fenotiazina (FTZ). Se ha estudiado el fotocomportamiento del éster metílico del carprofeno (CPFMe) en presencia de albúmina sérica bovina (ASB). Las cinéticas de desaparición de los estados excitados triplete de CPFMe en presencia de ASB 1:2 (a 430 nm), reveló dos sitios de unión diferentes a la proteína. El tiempo menor se asoció al CPFMe presente en el sitio I, basado en la desactivación del 3CPFMe* por triptófano (Trp) alojado en ese sitio de la proteína a través de un mecanismo de transferencia electrónica (Te). Sin diferencias significantes entre (S)- y (R)-CPFMe. En a1-glicoproteina ácida bovina (AGAB), se observó un único sitio de unión para ambos enantiómeros de CPFMe con estereodiferenciación en los tiempos de triplete de los complejos CPFMe@AGAB, donde el más corto fue para el enantiómero (S). Se detectó fotounión de CPFMe a AGAB por los consiguientes cambios en los espectros de fluorescencia de las mezclas CPFMe@AGAB antes y después de irradiar. El proceso fue más eficiente para (S)-CPFMe. El mecanismo propuesto relaciona la fotodeshalogenación reductiva desde el estado excitado triplete de (S)-CPFMe, dando lugar al aducto covalente (S)-CBZMe-AGAB. Para evitar la termodinámica desfavorable de la ruptura homolítica C-Cl a través del estado excitado triplete, se ha propuesto que en el mecanismo real de reacción esté involucrada la formación de excímeros triplete. Para profundizar en el mecanismo de deshalogenación del CPFMe, se han sintetizado e irradiado dos diadas diastereoméricas basadas en CPF (CPF-CPF y CPF-CBZ). El "self-quenching" de los estados excitados triplete en ambas diadas es mucho más rápido que en CPFMe, y es relacionado con la formación de especies con separación de carga, claramente desfavorecido en disolventes apolares como tetrahidrofurano (THF). Los tiempos de vida de triplete, así como los efectos del disolvente en la fotorreactividad están de acuerdo con la teoría de una deshalogenación fotoreductiva como mecanismo en sistemas basados en CPF. Se ha investigado la fotoquímica del fármaco neuroléptico ciamemazina (CMZ) en medios biomiméticos. El proceso de encapsulación ha sido seguido por fluorescencia, como un desplazamiento hipsocrómico de la banda, así como un aumento de los rendimientos cuánticos y de los tiempos de vida. La FDL reveló importantes cambios asociados a la encapsulación, una generación del estado excitado triplete más selectiva, los cuales viven más tiempo. La fotooxidación de CMZ ocurre a través de fotoionización; el catión radical resultante es atrapado por oxígeno para formar su fotoproducto N,S-dióxido. La reacción ha sido monitorizada por fluorescencia, por la aparición progresiva de la banda de emisión a longitudes de onda más cortas. Éste proceso resultó ser desfavorecido en los microambentes utilizados (lipófilos). La velocidad de fotooxidación más lenta corresponde a CMZ en micelas SDS y en AGAs, donde el fármaco se encuentra en un dominio más hidrofóbico, difícilmente accesible desde el medio acuoso, donde la fotoionización es casi despreciable. La FDL del antipsicótico clorpromazina (CPZ) y dos de sus metabolitos de fase I CPZ-MD y CPZ-DD (mono y didesmetilado) en presencia de cantidades crecientes de albúmina sérica humana (ASH) ha permitido monitorizar la unión a proteína, por el aumento del valor de Amax del 3CPZ* (a 470 nm). El grado de unión fue mayor para el fármaco de origen, de acuerdo con su carácter más hidrofóbico. El uso de warfarina como sonda de desplazamiento del sitio I indicó que los tres compuestos sólo se unen a este sitio[CA] S'ha plantejat l'ús de mitjans biomimètics com albúmines sèriques (ASs), a1-glicoproteïnes àcides (AGAs), ciclodextrines (CDs) i micel·les de dodecil sulfat sòdic (SDS) combinat amb tècniques fotofísiques com a fluorescència (F) i fotòlisi de llampada làser (FLL) per a investigar la fotosensibilització per fàrmacs que contenen un cromòfor tipus carbazol (CBZ) o fenotiazina (FTZ). S'ha estudiat el comportament de l'èster metílic del carprofén (CPFMe) en presencia d'albúmina sèrica bovina (ASB). Les cinètiques de desaparició dels estats excitats triplet de CPFMe en presencia de ASB 1:2 (a 430 nm) van revelar dos llocs d'unió de CPFMe a la proteïna; el temps menor es va associar al lloc I a causa de la desactivació del 3CPFMe* per triptòfan (Trp) allotjat en eixe lloc de la proteïna a través d'un mecanisme de transferència electrònica (Te); no es van observar diferències significatives entre (S)- i (R)-CPFMe. En a1-glicoproteïna àcida bovina (AGAB), CPFMe es va encapsular en un únic lloc d'unió amb estereodiferenciació en els temps de vida de triplet dels complexos CPFMe@AGAB, amb un valor més curt per a l'enantiòmer (S). A més, es va detectar fotounió de CPFMe a AGAB pels consegüents canvis als espectres de fluorescència de les mescles CPFMe@AGAB abans i després d'irradiar. El procés va ser més eficient per a (S)-CPFMe. El mecanisme proposat relaciona la fotodeshalogenació reductiva des de l'estat excitat triplet de (S)-CPFMe i que forma el corresponent adducte (S)-CBZMe-AGAB. Per a evitar la termodinàmica desfavorida de la ruptura homolítica C-Cl a través del estat excitat triplet, s'ha proposat que al mecanisme real de reacció estigui involucrat la formació de excímers triplet. En aquest context, per aprofundir en el mecanisme de deshalogenació del CPFMe, s'ha sintetitzat e irradiat dos diades diastereomèriques (CPF-CPF i CPF-CBZ). El "self-quenching" dels estats excitats triplet de ambdues diades es molt més ràpid que en CPFMe, i es relaciona amb la formació d'espècies amb separació de càrrega, clarament desfavorit en dissolvents apolars com tetrahidrofurà (THF). Les tendències observades en els temps de vida de triplet, així com els efectes del dissolvent en la fotorreactivitat estan d'acord amb la teoria d'una deshalogenació fotoreductiva com a mecanisme en sistemes basats en CPF. S'ha investigat la fotoquímica del fàrmac neurolèptic ciamemazina (CMZ) en medis biomimètics. El procés d'encapsulació ha estat seguit per fluorescència, com un desplaçament hipsocròmic de la banda, així com un augment dels rendiments quàntics i dels temps de vida. La FLL va revelar canvis importants associats a l'encapsulació, específicament una generació de l'estat excitat triplet més selectiva i amb temps de vida més llarg. La fotooxidació de CMZ ocorre a través de fotoionització; el catió radical resultant es atrapat per oxigen per a formar el seu fotoproducte N,S-diòxid. La reacció ha estat monitoritzada per fluorescència, per l'aparició progressiva de la banda de emissió a longituds d'ona més curtes. Este procés resultà ser desfavorit als microambients utilitzats (lipofílics). La velocitat de fotooxidació més lenta correspon a CMZ en micel·les SDS i en AGAs, on el fàrmac està en un domini més hidrofòbic, difícilment accessible des del medi aquós i on la fotoionització és quasi menyspreable. La FLL de l'antipsicòtic clorpromazina (CPZ) i dos dels seus metabòlits CPZ-MD i CPZ-DD (mono i didesmetilat) en presència de quantitats creixents d'albúmina sèrica humana (ASH) ha permès monitoritzar la unió a proteïna, mitjançant l'augment del valor de Amax del 3CPZ* (a 470 nm). El grau d'unió va ser major per al fàrmac d'origen, d'acord amb el seu caràcter més hidrofòbic. L'ús de warfarina com a sonda de desplaçament del lloc I va indicar que els tres compostos només s'uneixen a eixe lloc. Es va observar una lleugera fotounió a ASHLimones Herrero, D. (2015). Fotoquímica de compuestos heteroaromáticos tricíclicos en medios biomiméticos [Tesis doctoral no publicada]. Universitat Politècnica de València. https://doi.org/10.4995/Thesis/10251/59392TESI

    Protein binding of lapatinib and its N- and O-dealkylated metabolites interrogated by fluorescence, ultrafast spectroscopy and molecular dynamics simulations

    Full text link
    [EN] Lapatinib (LAP) is an anticancer drug generally used to treat breast and lung cancer. It exhibits hypersensitivity reactions in addition to dermatological adverse effects and photosensitivity. Moreover, LAP binds to serum proteins and is readily biotransformed in humans, giving rise to several metabolites, such as N- and O-dealkylated products (N-LAP and O-LAP, respectively). In this context, the aim of the present work is to obtain key information on drug@protein complexation, the first step involved in a number of hypersensitivity reactions, by a combination of fluorescence, femtosecond transient absorption spectroscopy and molecular dynamics (MD) simulations. Following this approach, the behavior of LAP and its metabolites has been investigated in the presence of serum proteins, such as albumins and alpha(1)-acid glycoproteins (SAs and AGs, respectively) from human and bovine origin. Fluorescence results pointed to a higher affinity of LAP and its metabolites to human proteins; the highest one was found for LAP@HSA. This is associated to the coplanar orientation adopted by the furan and quinazoline rings of LAP, which favors emission from long-lived (up to the ns time-scale) locally-excited (LE) states, disfavoring population of intramolecular charge transfer (ICT) states. Moreover, the highly constrained environment provided by subdomain IB of HSA resulted in a frozen conformation of the ligand, contributing to fluorescence enhancement. Computational studies were clearly in line with the experimental observations, providing valuable insight into the nature of the binding sites and the conformational arrangement of the ligands inside the protein cavities. Besides, a good correlation was found between the calculated binding energies for each ligand@protein complex and the relative affinities observed in competition experiments.Financial support from the Spanish Government (RYC-201517737, CTQ2017-89416-R, SAF2016-75638-R ISCIII grants RETICS ARADyAL (RD16/0006/0004 and RD16/0006/0001), PI16/01877 and CPII16/00052), Conselleria d'Educacio Cultura i Esport (PROMETEO/2017/075), the Xunta de Galicia [ED431B 2018/04 and Centro singular de investigacion de Galicia accreditation 2019-2022 (ED431G 2019/03)] and the European Regional Development Fund is gratefully acknowledged.Andreu Ros, MI.; Lence, E.; González-Bello, C.; Mayorga, C.; Cuquerella Alabort, MC.; Vayá Pérez, I.; Miranda Alonso, MÁ. (2020). Protein binding of lapatinib and its N- and O-dealkylated metabolites interrogated by fluorescence, ultrafast spectroscopy and molecular dynamics simulations. Frontiers in Pharmacology. 11:1-11. https://doi.org/10.3389/fphar.2020.576495S11111Ahmed, S. S., Whritenour, J., Ahmed, M. M., Bibby, L., Darby, L., Wang, X. N., … Dickinson, A. M. (2019). Evaluation of a human in vitro skin test for predicting drug hypersensitivity reactions. Toxicology and Applied Pharmacology, 369, 39-48. doi:10.1016/j.taap.2019.02.005Andreu, I., Mayorga, C., & Miranda, M. A. (2010). Generation of reactive intermediates in photoallergic dermatitis. Current Opinion in Allergy & Clinical Immunology, 10(4), 303-308. doi:10.1097/aci.0b013e32833bc68cBlakely, K. M., Drucker, A. M., & Rosen, C. F. (2019). Drug-Induced Photosensitivity—An Update: Culprit Drugs, Prevention and Management. Drug Safety, 42(7), 827-847. doi:10.1007/s40264-019-00806-5Bteich, M. (2019). An overview of albumin and alpha-1-acid glycoprotein main characteristics: highlighting the roles of amino acids in binding kinetics and molecular interactions. Heliyon, 5(11), e02879. doi:10.1016/j.heliyon.2019.e02879Cosa, G. (2004). Photodegradation and photosensitization in pharmaceutical products: Assessing drug phototoxicity. Pure and Applied Chemistry, 76(2), 263-275. doi:10.1351/pac200476020263Curry, S., Mandelkow, H., Brick, P., & Franks, N. (1998). Crystal structure of human serum albumin complexed with fatty acid reveals an asymmetric distribution of binding sites. Nature Structural Biology, 5(9), 827-835. doi:10.1038/1869Friedman, M. D., Lacouture, M., & Dang, C. (2016). Dermatologic Adverse Events Associated With Use of Adjuvant Lapatinib in Combination With Paclitaxel and Trastuzumab for HER2-Positive Breast Cancer: A Case Series Analysis. Clinical Breast Cancer, 16(3), e69-e74. doi:10.1016/j.clbc.2015.11.001García-Lainez, G., Vayá, I., Marín, M. P., Miranda, M. A., & Andreu, I. (2020). In vitro assessment of the photo(geno)toxicity associated with Lapatinib, a Tyrosine Kinase inhibitor. Archives of Toxicology, 95(1), 169-178. doi:10.1007/s00204-020-02880-6Gonzalez, G., & Lage, A. (2007). Cancer Vaccines for Hormone/Growth Factor Immune Deprivation:A Feasible Approach for Cancer Treatment. Current Cancer Drug Targets, 7(3), 229-241. doi:10.2174/156800907780618310Götz, A. W., Williamson, M. J., Xu, D., Poole, D., Le Grand, S., & Walker, R. C. (2012). Routine Microsecond Molecular Dynamics Simulations with AMBER on GPUs. 1. Generalized Born. Journal of Chemical Theory and Computation, 8(5), 1542-1555. doi:10.1021/ct200909jHynes, N. E., & Lane, H. A. (2005). ERBB receptors and cancer: the complexity of targeted inhibitors. Nature Reviews Cancer, 5(5), 341-354. doi:10.1038/nrc1609Ishikawa, T., Kamide, R., & Niimura, M. (1994). Photoleukomelanodermatitis (Kobori) Induced by Afloqualone. The Journal of Dermatology, 21(6), 430-433. doi:10.1111/j.1346-8138.1994.tb01768.xJohnston, S. R. D., Hegg, R., Im, S.-A., Park, I. H., Burdaeva, O., Kurteva, G., … Gradishar, W. J. (2018). Phase III, Randomized Study of Dual Human Epidermal Growth Factor Receptor 2 (HER2) Blockade With Lapatinib Plus Trastuzumab in Combination With an Aromatase Inhibitor in Postmenopausal Women With HER2-Positive, Hormone Receptor–Positive Metastatic Breast Cancer: ALTERNATIVE. Journal of Clinical Oncology, 36(8), 741-748. doi:10.1200/jco.2017.74.7824Kabir, M. Z., Mukarram, A. K., Mohamad, S. B., Alias, Z., & Tayyab, S. (2016). Characterization of the binding of an anticancer drug, lapatinib to human serum albumin. Journal of Photochemistry and Photobiology B: Biology, 160, 229-239. doi:10.1016/j.jphotobiol.2016.04.005Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N., & Sternberg, M. J. E. (2015). The Phyre2 web portal for protein modeling, prediction and analysis. Nature Protocols, 10(6), 845-858. doi:10.1038/nprot.2015.053Krasner, J. (1972). Drug-Protein Interaction. Pediatric Clinics of North America, 19(1), 51-63. doi:10.1016/s0031-3955(16)32666-9Le Grand, S., Götz, A. W., & Walker, R. C. (2013). SPFP: Speed without compromise—A mixed precision model for GPU accelerated molecular dynamics simulations. Computer Physics Communications, 184(2), 374-380. doi:10.1016/j.cpc.2012.09.022Limones-Herrero, D., Palumbo, F., Vendrell-Criado, V., Andreu, I., Lence, E., González-Bello, C., … Jiménez, M. C. (2020). Investigation of metabolite-protein interactions by transient absorption spectroscopy and in silico methods. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 226, 117652. doi:10.1016/j.saa.2019.117652Limones-Herrero, D., Pérez-Ruiz, R., Lence, E., González-Bello, C., Miranda, M. A., & Jiménez, M. C. (2017). Mapping a protein recognition centre with chiral photoactive ligands. An integrated approach combining photophysics, reactivity, proteomics and molecular dynamics simulation studies. Chemical Science, 8(4), 2621-2628. doi:10.1039/c6sc04900aLin, N. U., Carey, L. A., Liu, M. C., Younger, J., Come, S. E., Ewend, M., … Winer, E. P. (2008). Phase II Trial of Lapatinib for Brain Metastases in Patients With Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer. Journal of Clinical Oncology, 26(12), 1993-1999. doi:10.1200/jco.2007.12.3588Lin, N. U., Diéras, V., Paul, D., Lossignol, D., Christodoulou, C., Stemmler, H.-J., … Winer, E. P. (2009). Multicenter Phase II Study of Lapatinib in Patients with Brain Metastases from HER2-Positive Breast Cancer. Clinical Cancer Research, 15(4), 1452-1459. doi:10.1158/1078-0432.ccr-08-1080MEDINA, P., & GOODIN, S. (2008). Lapatinib: A dual inhibitor of human epidermal growth factor receptor tyrosine kinases. Clinical Therapeutics, 30(8), 1426-1447. doi:10.1016/j.clinthera.2008.08.008Miller, B. R., McGee, T. D., Swails, J. M., Homeyer, N., Gohlke, H., & Roitberg, A. E. (2012). MMPBSA.py: An Efficient Program for End-State Free Energy Calculations. Journal of Chemical Theory and Computation, 8(9), 3314-3321. doi:10.1021/ct300418hMiranda, M. A., Boscaa, F., Vargas, F., & Canudas, N. (1994). PHOTOSENSITIZATION BY FENOFIBRATE. II. In vitro PHOTOTOXICITY OF THE MAJOR METABOLITES. Photochemistry and Photobiology, 59(2), 171-174. doi:10.1111/j.1751-1097.1994.tb05018.xMolins-Molina, O., Pérez-Ruiz, R., Lence, E., González-Bello, C., Miranda, M. A., & Jiménez, M. C. (2019). Photobinding of Triflusal to Human Serum Albumin Investigated by Fluorescence, Proteomic Analysis, and Computational Studies. Frontiers in Pharmacology, 10. doi:10.3389/fphar.2019.01028Montanaro, S., Lhiaubet-Vallet, V., Iesce, M., Previtera, L., & Miranda, M. A. (2008). A Mechanistic Study on the Phototoxicity of Atorvastatin: Singlet Oxygen Generation by a Phenanthrene-like Photoproduct. Chemical Research in Toxicology, 22(1), 173-178. doi:10.1021/tx800294zNicholson, R. ., Gee, J. M. ., & Harper, M. . (2001). EGFR and cancer prognosis. European Journal of Cancer, 37, 9-15. doi:10.1016/s0959-8049(01)00231-3Nuin, E., Pérez-Sala, D., Lhiaubet-Vallet, V., Andreu, I., & Miranda, M. A. (2016). Photosensitivity to Triflusal: Formation of a Photoadduct with Ubiquitin Demonstrated by Photophysical and Proteomic Techniques. Frontiers in Pharmacology, 7. doi:10.3389/fphar.2016.00277Pérez-Ruíz, R., Lence, E., Andreu, I., Limones-Herrero, D., González-Bello, C., Miranda, M. A., & Jiménez, M. C. (2017). A New Pathway for Protein Haptenation by β-Lactams. Chemistry - A European Journal, 23(56), 13986-13994. doi:10.1002/chem.201702643Peters, T. (1995). Ligand Binding by Albumin. All About Albumin, 76-132. doi:10.1016/b978-012552110-9/50005-2Pinheiro, S., & Curutchet, C. (2017). Can Förster Theory Describe Stereoselective Energy Transfer Dynamics in a Protein–Ligand Complex? The Journal of Physical Chemistry B, 121(10), 2265-2278. doi:10.1021/acs.jpcb.7b00217Ross, J. S., Gay, L. M., Wang, K., Ali, S. M., Chumsri, S., Elvin, J. A., … Stephens, P. J. (2016). NonamplificationERBB2genomic alterations in 5605 cases of recurrent and metastatic breast cancer: An emerging opportunity for anti-HER2 targeted therapies. Cancer, 122(17), 2654-2662. doi:10.1002/cncr.30102Salomon-Ferrer, R., Götz, A. W., Poole, D., Le Grand, S., & Walker, R. C. (2013). Routine Microsecond Molecular Dynamics Simulations with AMBER on GPUs. 2. Explicit Solvent Particle Mesh Ewald. Journal of Chemical Theory and Computation, 9(9), 3878-3888. doi:10.1021/ct400314ySchönfeld, D. L., Ravelli, R. B. G., Mueller, U., & Skerra, A. (2008). The 1.8-Å Crystal Structure of α1-Acid Glycoprotein (Orosomucoid) Solved by UV RIP Reveals the Broad Drug-Binding Activity of This Human Plasma Lipocalin. Journal of Molecular Biology, 384(2), 393-405. doi:10.1016/j.jmb.2008.09.020Schroeder, R., Stevens, C., & Sridhar, J. (2014). Small Molecule Tyrosine Kinase Inhibitors of ErbB2/HER2/Neu in the Treatment of Aggressive Breast Cancer. Molecules, 19(9), 15196-15212. doi:10.3390/molecules190915196Sekula, B., Zielinski, K., & Bujacz, A. (2013). Crystallographic studies of the complexes of bovine and equine serum albumin with 3,5-diiodosalicylic acid. International Journal of Biological Macromolecules, 60, 316-324. doi:10.1016/j.ijbiomac.2013.06.004Shen, G.-F., Liu, T.-T., Wang, Q., Jiang, M., & Shi, J.-H. (2015). Spectroscopic and molecular docking studies of binding interaction of gefitinib, lapatinib and sunitinib with bovine serum albumin (BSA). Journal of Photochemistry and Photobiology B: Biology, 153, 380-390. doi:10.1016/j.jphotobiol.2015.10.023Sigismund, S., Avanzato, D., & Lanzetti, L. (2017). Emerging functions of the EGFR in cancer. Molecular Oncology, 12(1), 3-20. doi:10.1002/1878-0261.12155Spector, N. L., Xia, W., Burris, H., Hurwitz, H., Dees, E. C., Dowlati, A., … Bacus, S. (2005). Study of the Biologic Effects of Lapatinib, a Reversible Inhibitor of ErbB1 and ErbB2 Tyrosine Kinases, on Tumor Growth and Survival Pathways in Patients With Advanced Malignancies. Journal of Clinical Oncology, 23(11), 2502-2512. doi:10.1200/jco.2005.12.157Thomas, R., & Weihua, Z. (2019). Rethink of EGFR in Cancer With Its Kinase Independent Function on Board. Frontiers in Oncology, 9. doi:10.3389/fonc.2019.00800Towles, J. K., Clark, R. N., Wahlin, M. D., Uttamsingh, V., Rettie, A. E., & Jackson, K. D. (2016). Cytochrome P450 3A4 and CYP3A5-Catalyzed Bioactivation of Lapatinib. Drug Metabolism and Disposition, 44(10), 1584-1597. doi:10.1124/dmd.116.070839Vargas, F., Canudas, N., Miranda, M. A., & Boscar, F. (1993). PHOTODEGRADATION AND in vitro PHOTOTOXICITY OF FENOFIBRATE, A PHOTOSENSITIZING ANTI-HYPEIUIPOPROTEINEMIC DRUG. Photochemistry and Photobiology, 58(4), 471-476. doi:10.1111/j.1751-1097.1993.tb04917.xVayá, I., Andreu, I., Lence, E., González‐Bello, C., Consuelo Cuquerella, M., Navarrete‐Miguel, M., … Miranda, M. A. (2020). Characterization of Locally Excited and Charge‐Transfer States of the Anticancer Drug Lapatinib by Ultrafast Spectroscopy and Computational Studies. Chemistry – A European Journal, 26(68), 15922-15930. doi:10.1002/chem.202001336Vayá, I., Andreu, I., Monje, V. T., Jiménez, M. C., & Miranda, M. A. (2015). Mechanistic Studies on the Photoallergy Mediated by Fenofibric Acid: Photoreactivity with Serum Albumins. Chemical Research in Toxicology, 29(1), 40-46. doi:10.1021/acs.chemrestox.5b00357Vayá, I., Lhiaubet-Vallet, V., Jiménez, M. C., & Miranda, M. A. (2014). Photoactive assemblies of organic compounds and biomolecules: drug–protein supramolecular systems. Chem. Soc. Rev., 43(12), 4102-4122. doi:10.1039/c3cs60413fWilson, J. N., Liu, W., Brown, A. S., & Landgraf, R. (2015). Binding-induced, turn-on fluorescence of the EGFR/ERBB kinase inhibitor, lapatinib. Organic & Biomolecular Chemistry, 13(17), 5006-5011. doi:10.1039/c5ob00239gZunszain, P. A., Ghuman, J., Komatsu, T., Tsuchida, E., & Curry, S. (2003). BMC Structural Biology, 3(1), 6. doi:10.1186/1472-6807-3-

    Investigation of metabolite-protein interactions by transient absorption spectroscopy and in silico methods

    Full text link
    [EN] Transient absorption spectroscopy in combination with in silico methods has been employed to study the interactions between human serum albumin (HSA) and the anti-psychotic agent chlorpromazine (CPZ) as well as its two demethylated metabolites (MCPZ and DCPZ). Thus, solutions containing CPZ, MCPZ or DCPZ and HSA (molar ligand:protein ratios between 1:0 and 1:3) were submitted to laser flash photolysis and the Delta A(max) value at lambda = 470 nm, corresponding to the triplet excited state, was monitored. In all cases, the protein-bound ligand exhibited higher Delta Amax values measured after the laser pulse and were also considerably longer-lived than the non-complexed forms. This is in agreement with an enhanced hydrophilicity of the metabolites, due to the replacement of methyl groups with H that led to a lower extent of protein binding. For the three compounds, laser flash photolysis displacement experiments using warfarin or ibuprofen indicated Sudlow site I as the main binding site. Docking and molecular dynamics simulation studies revealed that the binding mode of the two demethylated ligands with HSA would be remarkable different from CPZ, specially for DCPZ, which appears to come from the different ability of their terminal ammonium groups to stablish hydrogen bonding interactions with the negatively charged residues within the protein pocket (Glu153, Glu292) as well as to allocate the methyl groups in an apolar environment. DCPZ would be rotated 180 degrees in relation to CPZ locating the aromatic ring away from the Sudlow site I of HSA. (C) 2019 Elsevier B.V. All rights reserved.Financial support from Ministerio de Economia, Industria y Competitividad (CTQ2016-78875-P, SAF2016-75638-R, BES-2011-043706), Generalitat Valenciana (Prometeo 2017/075), Xunta de Galicia [Centro Singular de Investigacion de Galicia accreditation 2016-2019 (ED431G/09, ED431B 2018/04) and post-doctoral fellowship to E. L.] and European Union (European Regional Development Fund-ERDF) is gratefully acknowledged. I. A. holds a "Miguel Servet" contract (CP1116/00052) funded by the Carlos III Health Institute. We are grateful to the Centro de Supercomputacion de Galicia (CESGA) for computational facilities.Limones Herrero, D.; Palumbo, F.; Vendrell Criado, V.; Andreu Ros, MI.; Lence, E.; González-Bello, C.; Miranda Alonso, MÁ.... (2020). Investigation of metabolite-protein interactions by transient absorption spectroscopy and in silico methods. Spectrochimica Acta Part A Molecular and Biomolecular Spectroscopy. 226:1-8. https://doi.org/10.1016/j.saa.2019.117652S18226Yang, G. X., Li, X., & Snyder, M. (2012). Investigating metabolite–protein interactions: An overview of available techniques. Methods, 57(4), 459-466. doi:10.1016/j.ymeth.2012.06.013S. Hage, D., Anguizola, J., Barnaby, O., Jackson, A., J. Yoo, M., Papastavros, E., … Tong, Z. (2011). Characterization of Drug Interactions with Serum Proteins by Using High-Performance Affinity Chromatography. Current Drug Metabolism, 12(4), 313-328. doi:10.2174/138920011795202938Matsuda, R., Bi, C., Anguizola, J., Sobansky, M., Rodriguez, E., Vargas Badilla, J., … Hage, D. S. (2014). Studies of metabolite–protein interactions: A review. Journal of Chromatography B, 966, 48-58. doi:10.1016/j.jchromb.2013.11.043López-Muñoz, F., Alamo, C., cuenca, E., Shen, W., Clervoy, P., & Rubio, G. (2005). History of the Discovery and Clinical Introduction of Chlorpromazine. Annals of Clinical Psychiatry, 17(3), 113-135. doi:10.1080/10401230591002002Beckett, A. H., Beaven, M. A., & Robinson, A. E. (1963). Metabolism of chlorpromazine in humans. Biochemical Pharmacology, 12(8), 779-794. doi:10.1016/0006-2952(63)90108-4Chetty, M., Moodley, S. V., & Miller, R. (1994). Important Metabolites to Measure in Pharmacodynamic Studies of Chlorpromazine. Therapeutic Drug Monitoring, 16(1), 30-36. doi:10.1097/00007691-199402000-00004Hubbard, J. W., Midha, K. K., Hawes, E. M., McKAY, G., Marder, S. R., Aravagiri, M., & Korchinski, E. D. (1993). Metabolism of Phenothiazine and Butyrophenone Antipsychotic Drugs. British Journal of Psychiatry, 163(S22), 19-24. doi:10.1192/s0007125000292556García, C., Oyola, R., Piñero, L. E., Arce, R., Silva, J., & Sánchez, V. (2005). Substitution and Solvent Effects on the Photophysical Properties of Several Series of 10-Alkylated Phenothiazine Derivatives. The Journal of Physical Chemistry A, 109(15), 3360-3371. doi:10.1021/jp044530jNavaratnam, S., Parsons, B. J., Phillips, G. O., & Davies, A. K. (1978). Laser flash photolysis study of the photoionisation of chlorpromazine and promazine in solution. Journal of the Chemical Society, Faraday Transactions 1: Physical Chemistry in Condensed Phases, 74(0), 1811. doi:10.1039/f19787401811Palumbo, F., Garcia-Lainez, G., Limones-Herrero, D., Coloma, M. D., Escobar, J., Jiménez, M. C., … Andreu, I. (2016). Enhanced photo(geno)toxicity of demethylated chlorpromazine metabolites. Toxicology and Applied Pharmacology, 313, 131-137. doi:10.1016/j.taap.2016.10.024Garcia, C., Smith, G. A., McGimpsey, W. G., Kochevar, I. E., & Redmond, R. W. (1995). Mechanism and Solvent Dependence for Photoionization of Promazine and Chlorpromazine. Journal of the American Chemical Society, 117(44), 10871-10878. doi:10.1021/ja00149a010Nath, S., & Sapre, A. V. (2001). Photoinduced electron transfer from chloropromazine and promethazine to chloroalkanes accompanied by cleavage of C–Cl bond. Chemical Physics Letters, 344(1-2), 138-146. doi:10.1016/s0009-2614(01)00685-6Joshi, R., Ghanty, T. K., & Mukherjee, T. (2008). Reactions and structural investigation of chlorpromazine radical cation. Journal of Molecular Structure, 888(1-3), 401-408. doi:10.1016/j.molstruc.2008.01.025He, X. M., & Carter, D. C. (1992). Atomic structure and chemistry of human serum albumin. Nature, 358(6383), 209-215. doi:10.1038/358209a0Sharples, D. (1974). The binding of chlorpromazine to human serum albumin. Journal of Pharmacy and Pharmacology, 26(8), 640-641. doi:10.1111/j.2042-7158.1974.tb10679.xVerbeeck, R. K., Cardinal, J.-A., Hill, A. G., & Midha, K. K. (1983). Binding of phenothiazine neuroleptics to plasma proteins. Biochemical Pharmacology, 32(17), 2565-2570. doi:10.1016/0006-2952(83)90019-9Silva, D., Cortez, C. M., & Louro, S. R. W. (2004). Quenching of the intrinsic fluorescence of bovine serum albumin by chlorpromazine and hemin. Brazilian Journal of Medical and Biological Research, 37(7), 963-968. doi:10.1590/s0100-879x2004000700004Lázaro, E., Lowe, P. J., Briand, X., & Faller, B. (2008). New Approach To Measure Protein Binding Based on a Parallel Artificial Membrane Assay and Human Serum Albumin. Journal of Medicinal Chemistry, 51(7), 2009-2017. doi:10.1021/jm7012826Kaddurah-Daouk, R., Kristal, B. S., & Weinshilboum, R. M. (2008). Metabolomics: A Global Biochemical Approach to Drug Response and Disease. Annual Review of Pharmacology and Toxicology, 48(1), 653-683. doi:10.1146/annurev.pharmtox.48.113006.094715Korkuć, P., & Walther, D. (2015). Physicochemical characteristics of structurally determined metabolite-protein and drug-protein binding events with respect to binding specificity. Frontiers in Molecular Biosciences, 2. doi:10.3389/fmolb.2015.00051Ohnmacht, C. M., Chen, S., Tong, Z., & Hage, D. S. (2006). Studies by biointeraction chromatography of binding by phenytoin metabolites to human serum albumin. Journal of Chromatography B, 836(1-2), 83-91. doi:10.1016/j.jchromb.2006.03.043Roelofs, K. G., Wang, J., Sintim, H. O., & Lee, V. T. (2011). Differential radial capillary action of ligand assay for high-throughput detection of protein-metabolite interactions. Proceedings of the National Academy of Sciences, 108(37), 15528-15533. doi:10.1073/pnas.1018949108Jimenez, M., & Miranda, M. (2015). Triplet Excited States as a Source of Relevant (Bio)Chemical Information. Current Topics in Medicinal Chemistry, 14(23), 2734-2742. doi:10.2174/1568026614666141216100907Jiménez, M. C., Miranda, M. A., & Vayá, I. (2005). Triplet Excited States as Chiral Reporters for the Binding of Drugs to Transport Proteins. Journal of the American Chemical Society, 127(29), 10134-10135. doi:10.1021/ja0514489Vayá, I., Bueno, C. J., Jiménez, M. C., & Miranda, M. A. (2006). Use of Triplet Excited States for the Study of Drug Binding to Human and Bovine Serum Albumins. ChemMedChem, 1(9), 1015-1020. doi:10.1002/cmdc.200600061Vayá, I., Jiménez, M. C., & Miranda, M. A. (2008). Transient Absorption Spectroscopy for Determining Multiple Site Occupancy in Drug−Protein Conjugates. A Comparison between Human and Bovine Serum Albumins Using Flurbiprofen Methyl Ester as a Probe. The Journal of Physical Chemistry B, 112(9), 2694-2699. doi:10.1021/jp076960qPérez-Ruiz, R., Bueno, C. J., Jiménez, M. C., & Miranda, M. A. (2010). In situ Transient Absorption Spectroscopy to Assess Competition between Serum Albumin and Alpha-1-Acid Glycoprotein for Drug Transport. The Journal of Physical Chemistry Letters, 1(5), 829-833. doi:10.1021/jz1000227Nuin, E., Jiménez, M. C., Sastre, G., Andreu, I., & Miranda, M. A. (2013). Drug–Drug Interactions within Protein Cavities Probed by Triplet–Triplet Energy Transfer. The Journal of Physical Chemistry Letters, 4(10), 1603-1607. doi:10.1021/jz400640sAlonso, R., Yamaji, M., Jiménez, M. C., & Miranda, M. A. (2010). Enhanced Photostability of the Anthracene Chromophore in Aqueous Medium upon Protein Encapsulation. The Journal of Physical Chemistry B, 114(34), 11363-11369. doi:10.1021/jp104900rAlonso, R., Jiménez, M. C., & Miranda, M. A. (2011). Stereodifferentiation in the Compartmentalized Photooxidation of a Protein-Bound Anthracene. Organic Letters, 13(15), 3860-3863. doi:10.1021/ol201209hKitamura, K., Fujitani, K., Takahashi, K., Tanaka, Y., Hirako, S., Kotani, C., … Takegami, S. (2000). Synthesis of [N-13CH3] drugs (chlorpromazine, triflupromazine and promazine). Journal of Labelled Compounds and Radiopharmaceuticals, 43(9), 865-872. doi:10.1002/1099-1344(200008)43:93.0.co;2-eGhuman, J., Zunszain, P. A., Petitpas, I., Bhattacharya, A. A., Otagiri, M., & Curry, S. (2005). Structural Basis of the Drug-binding Specificity of Human Serum Albumin. Journal of Molecular Biology, 353(1), 38-52. doi:10.1016/j.jmb.2005.07.075Pérez-Ruiz, R., Molins-Molina, O., Lence, E., González-Bello, C., Miranda, M. A., & Jiménez, M. C. (2018). Photogeneration of Quinone Methides as Latent Electrophiles for Lysine Targeting. The Journal of Organic Chemistry, 83(21), 13019-13029. doi:10.1021/acs.joc.8b01559Roe, D. R., & Cheatham, T. E. (2013). PTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory Data. Journal of Chemical Theory and Computation, 9(7), 3084-3095. doi:10.1021/ct400341

    Enhanced photo(geno)toxicity of demethylated chlorpromazine metabolites

    Full text link
    Chlorpromazine (CPZ) is an anti-psychotic drug widely used to treat disorders such as schizophrenia or manic-depression. Unfortunately, CPZ exhibits undesirable side effects such as phototoxic and photoallergic reactions in humans. In general, the influence of drug metabolism on this type of reactions has not been previously considered in photosafety testing. Thus, the present work aims to investigate the possible photo(geno)toxic potential of drug metabolites, using CPZ as an established reference compound. In this case, the metabolites selected for the study are demethylchlorpromazine (DMCPZ), didemethylchlorpromazine (DDMCPZ) and chlorpromazine sulfoxide (CPZSO). The demethylated CPZ metabolites DMCPZ and DDMCPZ maintain identical chromophore to the parent drug. In this work, it has been found that the nature of the aminoalkyl side chain modulates the hydrophobicity and the photochemical properties (for instance, the excited state lifetimes), but it does not change the photoreactivity pattern, which is characterized by reductive photodehalogenation, triggered by homolytic carbon-chlorine bond cleavage with formation of highly reactive aryl radical intermediates. Accordingly, these metabolites are phototoxic to cells, as revealed by the 3T3 NRU assay; their photo-irritation factors are even higher than that of CPZ. The same trend is observed in photogenotoxicity studies, both with isolated and with cellular DNA, where DMCPZ and DDMCPZ are more active than CPZ itself. In summary, side-chain demethylation of CPZ, as a consequence of Phase I biotransformation, does not result a photodetoxification. Instead, it leads to metabolites that exhibit in an even enhanced photo(geno)toxicity.This work was supported by the MINECO (Grant: CTQ2013-47872), by Carlos III Institute of Health (Grants: RD12/0013/0009 and CP11/00154) and by the Generalitat Valenciana (Prometeo II/2013/005).Palumbo, F.; García Lainez, G.; Limones Herrero, D.; Coloma, MD.; Escobar, J.; Jiménez Molero, MC.; Miranda Alonso, MÁ.... (2016). Enhanced photo(geno)toxicity of demethylated chlorpromazine metabolites. Toxicology and Applied Pharmacology. 313:231-237. https://doi.org/10.1016/j.taap.2016.10.024S23123731

    Photo(geno)toxicity changes associated with hydroxylation of the aromatic chromophores during diclofenac metabolism

    Full text link
    [EN] Diclofenac (DCF) can cause adverse reactions such as gastrointestinal, renal and cardiovascular disorders; therefore, topical administration may be an attractive alternative to the management of local pain in order to avoid these side effects. However, previous studies have shown that DCF, in combination with sunlight, displays capability to induce photosensitivity disorders. In humans, DCF is biotransformed into hydroxylated metabolites at positions 4¿ and 5 (4¿OH-DCF and 5OH-DCF), and this chemical change produces non negligible alterations of the drug chromophore, resulting in a significant modification of its light-absorbing properties. In the present work, 5OH-DCF exhibited higher photo(geno)toxic potential than the parent drug, as shown by several in vitro assays (3T3 NRU phototoxicity, DNA ssb gel electrophoresis and COMET), whereas 4¿OH-DCF did not display significant photo(geno)toxicity. This could be associated, at least partially with their more efficient UV-light absorption by 5OH-DCF metabolite and with a higher photoreactivity. Interestingly, most of the cellular DNA damage photosensitized by DCF and 5OH-DCF was repaired by the cells after several hours, although this effect was not complete in the case of 5OH-DCF.This work was supported by the Carlos III Institute of Health (Grants: RD16/0006/0030, PI16/01877), by the MINECO (Grants: CTQ2013-47872, CTQ2016-78875), and by the Generalitat Valenciana (Prometeo 2017/075).García -Laínez, G.; Ana M Marínez-Reig; Limones Herrero, D.; Jiménez Molero, MC.; Miranda Alonso, MÁ.; Andreu Ros, MI. (2018). Photo(geno)toxicity changes associated with hydroxylation of the aromatic chromophores during diclofenac metabolism. Toxicology and Applied Pharmacology. 341:51-55. https://doi.org/10.1016/j.taap.2018.01.005S515534

    Identification of a common recognition center for a photoactive non-steroidal antiinflammatory drug in serum albumins of different species

    Full text link
    [EN] The non-steroidal anti-inflammatory drug (S)-carprofen (CPF) has been used as a photoactive probe to investigate the possible existence of a common recognition center in serum albumins (SAs) of different species. The methodology involves irradiation of the CPF/SA complexes, coupled with gel filtration chromatography or proteomic analysis of the photolysates, docking and molecular dynamics simulations. Photolysis of CPF/SA complexes at = 320 nm, and gel filtration chromatography, revealed that the protein fraction still contained the drug fluorophore, in agreement with covalent attachment of the photogenerated radical intermediate CBZ to SAs. After trypsin digestion and ESI-MS/MS, the incorporation of CBZ was detected at several positions in the different albumins. Remarkably, modifications at the IB/IIIA interface were observed in all cases (Tyr452 in HSA, RbSA and RtSA and Tyr451 in BSA, PSA and SSA). The molecular basis of this common recognition, studied by docking and molecular dynamics simulation studies on the corresponding non-covalent complexes, corroborated the experimentally observed covalent modifications. Our computational studies also revealed that the previously reported displacement of CPF by (S)-ibuprofen, a site II specific drug, would be due to an allosteric effect in site II, rather than a direct molecular displacement, as expected.Financial support from the Spanish Ministry of Economy and Competiveness (CTQ2016-78875-P, SAF2016-75638-R and BES-2014-069404), Generalitat Valenciana (PROMETEO2017/075), Conselleria de Cultura, Educacion e Ordenacion Universitaria (Centro singular de investigacion de Galicia accreditation 2016-2019, ED431G/09) and the European Regional Development Fund (ERDF) is acknowledged. This work was also supported by Instituto de Salud Carlos III (ISCIII) co-funded by Fondo Europeo de Desarrollo Regional FEDER for the Thematic Networks and Co-operative Research Centres: ARADyAL (RD16/0006/0030). EL thanks the Xunta de Galicia for his postdoctoral fellowship. We are also grateful to the Centro de Supercomputacion de Galicia (CESGA) for use of the Finis Terrae II supercomputer. The proteomic analysis was performed in the proteomics facility of SCSIE University of Valencia that belongs to ProteoRed PRB2-ISCIII and is supported by grant PT13/0001, of the PE I+D+I 2013-2016, funded by ISCIII and FEDER.Molins-Molina, O.; Lence, E.; Limones-Herrero, D.; González-Bello, C.; Miranda Alonso, MÁ.; Jiménez Molero, MC. (2019). Identification of a common recognition center for a photoactive non-steroidal antiinflammatory drug in serum albumins of different species. Organic Chemistry Frontiers. 6(1):99-109. https://doi.org/10.1039/c8qo01045eS9910961Limones-Herrero, D., Pérez-Ruiz, R., Lence, E., González-Bello, C., Miranda, M. A., & Jiménez, M. C. (2017). Mapping a protein recognition centre with chiral photoactive ligands. An integrated approach combining photophysics, reactivity, proteomics and molecular dynamics simulation studies. Chemical Science, 8(4), 2621-2628. doi:10.1039/c6sc04900aO’Brien, W. M., & Bagby, G. F. (1987). Carprofen: A New Nonsteroidal Antiinflammatory Drug Pharmacology, Clinical Efficacy and Adverse Effects. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy, 7(1), 16-24. doi:10.1002/j.1875-9114.1987.tb03500.xCurry, S. L., Cogar, S. M., & Cook, J. L. (2005). Nonsteroidal Antiinflammatory Drugs: A Review. Journal of the American Animal Hospital Association, 41(5), 298-309. doi:10.5326/0410298LEES, P., LANDONI, M. F., Giraudel, J., & TOUTAIN, P. L. (2004). Pharmacodynamics and pharmacokinetics of nonsteroidal anti-inflammatory drugs in species of veterinary interest. Journal of Veterinary Pharmacology and Therapeutics, 27(6), 479-490. doi:10.1111/j.1365-2885.2004.00617.xT. J. Peter , All about albumin: biochemistry, genetics and medical applications , Academic press , California , 1996He, X. M., & Carter, D. C. (1992). Atomic structure and chemistry of human serum albumin. Nature, 358(6383), 209-215. doi:10.1038/358209a0Kragh-Hansen, U., Chuang, V. T. G., & Otagiri, M. (2002). Practical Aspects of the Ligand-Binding and Enzymatic Properties of Human Serum Albumin. Biological and Pharmaceutical Bulletin, 25(6), 695-704. doi:10.1248/bpb.25.695Fasano, M., Curry, S., Terreno, E., Galliano, M., Fanali, G., Narciso, P., … Ascenzi, P. (2005). The extraordinary ligand binding properties of human serum albumin. IUBMB Life (International Union of Biochemistry and Molecular Biology: Life), 57(12), 787-796. doi:10.1080/15216540500404093Carter, D. C., & Ho, J. X. (1994). Structure of Serum Albumin. Advances in Protein Chemistry, 153-203. doi:10.1016/s0065-3233(08)60640-3Kosa, T., Maruyama, T., & Otagiri, M. (1997). Pharmaceutical Research, 14(11), 1607-1612. doi:10.1023/a:1012138604016Chang, C.-F., & Jeng, S.-R. (1995). Isolation and characterization of the female-specific protein (vitellogenin) in mature female hemolymph of the prawn Penaeus chinensis. Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology, 112(2), 257-263. doi:10.1016/0305-0491(95)00059-3Rahman, M. H., Maruyama, T., Okada, T., Yamasaki, K., & Otagiri, M. (1993). Study of interaction of carprofen and its enantiomers with human serum albumin—I. Biochemical Pharmacology, 46(10), 1721-1731. doi:10.1016/0006-2952(93)90576-iVayá, I., Pérez-Ruiz, R., Lhiaubet-Vallet, V., Jiménez, M. C., & Miranda, M. A. (2010). Drug–protein interactions assessed by fluorescence measurements in the real complexes and in model dyads. Chemical Physics Letters, 486(4-6), 147-153. doi:10.1016/j.cplett.2009.12.091Lhiaubet-Vallet, V., Boscá, F., & Miranda, M. A. (2006). Stereodifferentiating Drug−Biomolecule Interactions in the Triplet Excited State:  Studies on Supramolecular Carprofen/Protein Systems and on Carprofen−Tryptophan Model Dyads. The Journal of Physical Chemistry B, 111(2), 423-431. doi:10.1021/jp066968kRahman, M. H., Maruyama, T., Okada, T., Imai, T., & Otagiri, M. (1993). Study of interaction of carprofen and its enantiomers with human serum albumin—II. Biochemical Pharmacology, 46(10), 1733-1740. doi:10.1016/0006-2952(93)90577-jDivkovic, M., Pease, C. K., Gerberick, G. F., & Basketter, D. A. (2005). Hapten-protein binding: from theory to practical application in the in vitro prediction of skin sensitization. Contact Dermatitis, 53(4), 189-200. doi:10.1111/j.0105-1873.2005.00683.xJohannesson, G., Rosqvist, S., Lindh, C. H., Welinder, H., & Jönsson, B. A. G. (2001). Serum albumins are the major site for in vivo formation of hapten-carrier protein adducts in plasma from humans and guinea-pigs exposed to type-1 allergy inducing hexahydrophthalic anhydride. Clinical & Experimental Allergy, 31(7), 1021-1030. doi:10.1046/j.1365-2222.2001.01109.xLahoz, A., Hernández, D., Miranda, M. A., Pérez-Prieto, J., Morera, I. M., & Castell, J. V. (2001). Antibodies Directed to Drug Epitopes to Investigate the Structure of Drug−Protein Photoadducts. Recognition of a Common Photobound Substructure in Tiaprofenic Acid/Ketoprofen Cross-Photoreactivity. Chemical Research in Toxicology, 14(11), 1486-1491. doi:10.1021/tx0002482P. Jones , In vitro phototoxicity assays , in Principles and Practice of Skin Toxicology , ed. R. Chilcott and S. Price , John Wiley & Sons , 2008 , p. 169Merot, Y., Harms, M., & Saurat, J.-H. (1983). Photosensibilisation au carprofène (Imady®), un nouvel anti-inflammatoire non stéroïdien. Dermatology, 166(6), 301-307. doi:10.1159/000249894Roelandts, G., & Goh, C. L. (1986). Photosensitivity Associated with Carprofen. Dermatology, 172(1), 64-65. doi:10.1159/000249297Boscá, F., Marín, M. L., & Miranda, M. A. (2001). Photoreactivity of the Nonsteroidal Anti-inflammatory 2-Arylpropionic Acids with Photosensitizing Side Effects¶. Photochemistry and Photobiology, 74(5), 637. doi:10.1562/0031-8655(2001)0742.0.co;2Kerr, A. C., Muller, F., Ferguson, J., & Dawe, R. S. (2008). Occupational carprofen photoallergic contact dermatitis. British Journal of Dermatology, 159(6), 1303-1308. doi:10.1111/j.1365-2133.2008.08847.xMoser, J., Boscá, F., Lovell, W. W., Castell, J. V., Miranda, M. A., & Hye, A. (2000). Photobinding of carprofen to protein. Journal of Photochemistry and Photobiology B: Biology, 58(1), 13-19. doi:10.1016/s1011-1344(00)00115-9P.-L. Toutain , A.Ferran and A.Bousquet-Mélou , Species Differences in Pharmacokinetics and Pharmacodynamics , in Handbook of Experimental Pharmacology, Vol. 199, Comparative and Veterinary Pharmacology , ed. F. Cunningan , J. Elliot and P. Lees , Springer-Verlag , Berlin, Heidelberg , 2010Bosca, F., Encinas, S., Heelis, P. F., & Miranda, M. A. (1997). Photophysical and Photochemical Characterization of a Photosensitizing Drug:  A Combined Steady State Photolysis and Laser Flash Photolysis Study on Carprofen. Chemical Research in Toxicology, 10(7), 820-827. doi:10.1021/tx9700376Sekula, B., Ciesielska, A., Rytczak, P., Koziołkiewicz, M., & Bujacz, A. (2016). Structural evidence of the species-dependent albumin binding of the modified cyclic phosphatidic acid with cytotoxic properties. Bioscience Reports, 36(3). doi:10.1042/bsr20160089http://www.ccdc.cam.ac.uk/solutions/csd-discovery/components/gold/Sivertsen, A., Isaksson, J., Leiros, H.-K. S., Svenson, J., Svendsen, J.-S., & Brandsdal, B. O. (2014). Synthetic cationic antimicrobial peptides bind with their hydrophobic parts to drug site II of human serum albumin. BMC Structural Biology, 14(1). doi:10.1186/1472-6807-14-4Pérez-Ruíz, R., Lence, E., Andreu, I., Limones-Herrero, D., González-Bello, C., Miranda, M. A., & Jiménez, M. C. (2017). A New Pathway for Protein Haptenation by β-Lactams. Chemistry - A European Journal, 23(56), 13986-13994. doi:10.1002/chem.201702643Ghuman, J., Zunszain, P. A., Petitpas, I., Bhattacharya, A. A., Otagiri, M., & Curry, S. (2005). Structural Basis of the Drug-binding Specificity of Human Serum Albumin. Journal of Molecular Biology, 353(1), 38-52. doi:10.1016/j.jmb.2005.07.075Curry, S., Mandelkow, H., Brick, P., & Franks, N. (1998). Crystal structure of human serum albumin complexed with fatty acid reveals an asymmetric distribution of binding sites. Nature Structural Biology, 5(9), 827-835. doi:10.1038/1869Miller, B. R., McGee, T. D., Swails, J. M., Homeyer, N., Gohlke, H., & Roitberg, A. E. (2012). MMPBSA.py: An Efficient Program for End-State Free Energy Calculations. Journal of Chemical Theory and Computation, 8(9), 3314-3321. doi:10.1021/ct300418hWang, Z., Ho, J. X., Ruble, J. R., Rose, J., Rüker, F., Ellenburg, M., … Carter, D. C. (2013). Structural studies of several clinically important oncology drugs in complex with human serum albumin. Biochimica et Biophysica Acta (BBA) - General Subjects, 1830(12), 5356-5374. doi:10.1016/j.bbagen.2013.06.032Zunszain, P. A., Ghuman, J., Komatsu, T., Tsuchida, E., & Curry, S. (2003). BMC Structural Biology, 3(1), 6. doi:10.1186/1472-6807-3-6Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N., & Sternberg, M. J. E. (2015). The Phyre2 web portal for protein modeling, prediction and analysis. Nature Protocols, 10(6), 845-858. doi:10.1038/nprot.2015.053Vanquelef, E., Simon, S., Marquant, G., Garcia, E., Klimerak, G., Delepine, J. C., … Dupradeau, F.-Y. (2011). R.E.D. Server: a web service for deriving RESP and ESP charges and building force field libraries for new molecules and molecular fragments. Nucleic Acids Research, 39(suppl_2), W511-W517. doi:10.1093/nar/gkr288http://upjv.q4md-forcefieldtools.org/RED/Dupradeau, F.-Y., Pigache, A., Zaffran, T., Savineau, C., Lelong, R., Grivel, N., … Cieplak, P. (2010). The R.E.D. tools: advances in RESP and ESP charge derivation and force field library building. Physical Chemistry Chemical Physics, 12(28), 7821. doi:10.1039/c0cp00111bCornell, W. D., Cieplak, P., Bayly, C. I., Gould, I. R., Merz, K. M., Ferguson, D. M., … Kollman, P. A. (1995). A Second Generation Force Field for the Simulation of Proteins, Nucleic Acids, and Organic Molecules. Journal of the American Chemical Society, 117(19), 5179-5197. doi:10.1021/ja00124a002Case, D. A., Cheatham, T. E., Darden, T., Gohlke, H., Luo, R., Merz, K. M., … Woods, R. J. (2005). The Amber biomolecular simulation programs. Journal of Computational Chemistry, 26(16), 1668-1688. doi:10.1002/jcc.20290Wang, J., Wolf, R. M., Caldwell, J. W., Kollman, P. A., & Case, D. A. (2004). Development and testing of a general amber force field. Journal of Computational Chemistry, 25(9), 1157-1174. doi:10.1002/jcc.20035Wang, J., Wang, W., Kollman, P. A., & Case, D. A. (2006). Automatic atom type and bond type perception in molecular mechanical calculations. Journal of Molecular Graphics and Modelling, 25(2), 247-260. doi:10.1016/j.jmgm.2005.12.005Gordon, J. C., Myers, J. B., Folta, T., Shoja, V., Heath, L. S., & Onufriev, A. (2005). H++: a server for estimating pKas and adding missing hydrogens to macromolecules. Nucleic Acids Research, 33(Web Server), W368-W371. doi:10.1093/nar/gki464http://biophysics.cs.vt.edu/H++Götz, A. W., Williamson, M. J., Xu, D., Poole, D., Le Grand, S., & Walker, R. C. (2012). Routine Microsecond Molecular Dynamics Simulations with AMBER on GPUs. 1. Generalized Born. Journal of Chemical Theory and Computation, 8(5), 1542-1555. doi:10.1021/ct200909jLe Grand, S., Götz, A. W., & Walker, R. C. (2013). SPFP: Speed without compromise—A mixed precision model for GPU accelerated molecular dynamics simulations. Computer Physics Communications, 184(2), 374-380. doi:10.1016/j.cpc.2012.09.022Darden, T., York, D., & Pedersen, L. (1993). Particle mesh Ewald: AnN⋅log(N) method for Ewald sums in large systems. The Journal of Chemical Physics, 98(12), 10089-10092. doi:10.1063/1.464397Ryckaert, J.-P., Ciccotti, G., & Berendsen, H. J. . (1977). Numerical integration of the cartesian equations of motion of a system with constraints: molecular dynamics of n-alkanes. Journal of Computational Physics, 23(3), 327-341. doi:10.1016/0021-9991(77)90098-5W. L. DeLano , The PyMOL Molecular Graphics System , DeLano Scientific LLC , Palo Alto, CA, USA , 2008 . http://www.pymol.org/Roe, D. R., & Cheatham, T. E. (2013). PTRAJ and CPPTRAJ: Software for Processing and Analysis of Molecular Dynamics Trajectory Data. Journal of Chemical Theory and Computation, 9(7), 3084-3095. doi:10.1021/ct400341phttp://www.amber.utah.edu/AMBER-workshop/London-2015/pca

    Queridos usuarios

    No full text
    Treball Final de Grau en Comunicació Audiovisual. Codi: CA0932. Curs acadèmic: 2017-2018El presente trabajo de Final de Grado consiste en la producción y realización de un documental que habla sobre cómo las nuevas tecnologías comportan un cambio en la forma de relacionarse los jóvenes hoy en día. Un documental social que se aproxima a este fenómeno complejo y desconocido a través del cual se muestra que ocurren en los encuentros, interacciones y relaciones actuales.This final of degree project consists of producing and accomplishment of a documentary that it speaks on how the new technologies endure a change in the way of relating the young people nowadays. A social documentary that comes closer this phenomenon complex and known across which it appears that they happen in the meetings, interactions and current relations

    Bypassing the energy barrier of homolytic photodehalogenation in chloroaromatics through self-quenching

    Full text link
    Reductive photodehalogenation of chloroaromatics is assumed to proceed from the triplet excited state, although its energy is often insufficient to promote a clean homolytic C¿Cl cleavage. A clear-cut experimental proof is provided that correlates self-quenching of the directly observed triplet excited states of chlorocarbazole-based dyads 1 and 2 with photoreactivity via intramolecular charge transfer.Financial support from the Spanish Government (CTQ2010-14882, BES-2011-043706, JCI-2010-06204) and from the Generalitat Valenciana (Prometeo 2008/090 and GV/2012/041) is gratefully acknowledged.Limones Herrero, D.; Pérez Ruiz, R.; Jiménez Molero, MC.; Miranda Alonso, MÁ. (2013). Bypassing the energy barrier of homolytic photodehalogenation in chloroaromatics through self-quenching. Organic Letters. 15(6):1314-1317. doi:10.1021/ol400251sS1314131715

    Bypassing the Energy Barrier of Homolytic Photodehalogenation in Chloroaromatics through Self-Quenching

    No full text
    Reductive photodehalogenation of chloroaromatics is assumed to proceed from the triplet excited state, although its energy is often insufficient to promote a clean homolytic C–Cl cleavage. A clear-cut experimental proof is provided that correlates self-quenching of the directly observed triplet excited states of chlorocarbazole-based dyads <b>1</b> and <b>2</b> with photoreactivity via intramolecular charge transfer
    corecore