Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 63(10): e202317901, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38088924

RESUMEN

Rising antimicrobial resistance (AMR) and lack of innovation in the antibiotic pipeline necessitate novel approaches to discovering new drugs. Metal complexes have proven to be promising antimicrobial compounds, but the number of studied compounds is still low compared to the millions of organic molecules investigated so far. Lately, machine learning (ML) has emerged as a valuable tool for guiding the design of small organic molecules, potentially even in low-data scenarios. For the first time, we extend the application of ML to the discovery of metal-based medicines. Utilising 288 modularly synthesized ruthenium arene Schiff-base complexes and their antibacterial properties, a series of ML models were trained. The models perform well and are used to predict the activity of 54 new compounds. These displayed a 5.7x higher hit-rate (53.7 %) against methicillin-resistant Staphylococcus aureus (MRSA) compared to the original library (9.4 %), demonstrating that ML can be applied to improve the success-rates in the search of new metalloantibiotics. This work paves the way for more ambitious applications of ML in the field of metal-based drug discovery.


Asunto(s)
Complejos de Coordinación , Staphylococcus aureus Resistente a Meticilina , Rutenio , Rutenio/farmacología , Pruebas de Sensibilidad Microbiana , Antibacterianos/farmacología , Complejos de Coordinación/farmacología
2.
J Am Chem Soc ; 145(11): 6453-6461, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36881731

RESUMEN

Nitroreductases (NTRs) constitute an important class of oxidoreductase enzymes that have evolved to metabolize nitro-containing compounds. Their unique characteristics have spurred an array of potential uses in medicinal chemistry, chemical biology, and bioengineering toward harnessing nitro caging groups and constructing NTR variants for niche applications. Inspired by how they carry out enzymatic reduction via a cascade of hydride transfer reactions, we sought to develop a synthetic small-molecule NTR system based on transfer hydrogenation mediated by transition metal complexes harnessing native cofactors. We report the first water-stable Ru-arene complex capable of selectively and fully reducing nitroaromatics into anilines in a biocompatible buffered aqueous environment using formate as the hydride source. We further demonstrated its application to activate nitro-caged sulfanilamide prodrug in formate-abundant bacteria, specifically pathogenic methicillin-resistant Staphylococcus aureus. This proof of concept paves the way for a new targeted antibacterial chemotherapeutic approach leveraging on redox-active metal complexes for prodrug activation via bioinspired nitroreduction.


Asunto(s)
Complejos de Coordinación , Staphylococcus aureus Resistente a Meticilina , Profármacos , Profármacos/farmacología , Staphylococcus aureus Resistente a Meticilina/metabolismo , Complejos de Coordinación/farmacología , Bacterias/metabolismo , Nitrocompuestos/química , Nitrorreductasas/metabolismo , Formiatos
3.
Angew Chem Int Ed Engl ; 62(50): e202310040, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-37621226

RESUMEN

Antimicrobial resistance, caused by persistent adaptation and growing resistance of pathogenic bacteria to overprescribed antibiotics, poses one of the most serious and urgent threats to global public health. The limited pipeline of experimental antibiotics in development further exacerbates this looming crisis and new drugs with alternative modes of action are needed to tackle evolving pathogenic adaptation. Transition metal complexes can replenish this diminishing stockpile of drug candidates by providing compounds with unique properties that are not easily accessible using pure organic scaffolds. We spotlight four emerging strategies to harness these unique properties to develop new targeted antibacterial agents.


Asunto(s)
Complejos de Coordinación , Elementos de Transición , Antibacterianos/farmacología , Bacterias , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico
4.
Inorg Chem ; 61(26): 10167-10181, 2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35713376

RESUMEN

Indolo[3,2-d][1]benzazepines (paullones), indolo[3,2-d][2]benzazepines, and indolo[2,3-d][2]benzazepines (latonduines) are isomeric scaffolds of current medicinal interest. Herein, we prepared a small library of novel indolo[3,2-d][2]benzazepine-derived ligands HL1-HL4 and copper(II) complexes 1-4. All compounds were characterized by spectroscopic methods (1H and 13C NMR, UV-vis, IR) and electrospray ionization (ESI) mass spectrometry, while complexes 2 and 3, in addition, by X-ray crystallography. Their purity was confirmed by HPLC coupled with high-resolution ESI mass spectrometry and/or elemental analysis. The stability of compounds in aqueous solutions in the presence of DMSO was confirmed by 1H NMR and UV-vis spectroscopy measurements. The compounds revealed high antiproliferative activity in vitro in the breast cancer cell line MDA-MB-231 and hepatocellular carcinoma cell line LM3 in the low micromolar to nanomolar concentration range. Important structure-activity relationships were deduced from the comparison of anticancer activities of HL1-HL4 and 1-4 with those of structurally similar paullone-derived (HL5-HL7 and 5-7) and latonduine-derived scaffolds (HL8-HL11 and 8-11). The high anticancer activity of the lead drug candidate 4 was linked to reactive oxygen species and endoplasmic reticulum stress induction, which were confirmed by fluorescent microscopy and Western blot analysis.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Antineoplásicos/química , Antineoplásicos/farmacología , Benzazepinas/química , Línea Celular Tumoral , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Cobre/química , Cobre/farmacología , Cristalografía por Rayos X , Ligandos , Relación Estructura-Actividad
5.
Proc Natl Acad Sci U S A ; 116(28): 13943-13951, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31221747

RESUMEN

Cisplatin [cis-diamminedichloroplatinum(II) (cis-DDP)] is one of the most successful anticancer agents effective against a wide range of solid tumors. However, its use is restricted by side effects and/or by intrinsic or acquired drug resistance. Here, we probed the role of glutathione transferase (GST) P1-1, an antiapoptotic protein often overexpressed in drug-resistant tumors, as a cis-DDP-binding protein. Our results show that cis-DDP is not a substrate for the glutathione (GSH) transferase activity of GST P1-1. Instead, GST P1-1 sequesters and inactivates cisplatin with the aid of 2 solvent-accessible cysteines, resulting in protein subunits cross-linking, while maintaining its GSH-conjugation activity. Furthermore, it is well known that GST P1-1 binding to the c-Jun N-terminal kinase (JNK) inhibits JNK phosphorylation, which is required for downstream apoptosis signaling. Thus, in turn, GST P1-1 overexpression and Pt-induced subunit cross-linking could modulate JNK apoptotic signaling, further confirming the role of GST P1-1 as an antiapoptotic protein.


Asunto(s)
Cisplatino/química , Gutatión-S-Transferasa pi/química , Proteínas Quinasas JNK Activadas por Mitógenos/química , Neoplasias/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutatión/química , Gutatión-S-Transferasa pi/genética , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Neoplasias/genética , Fosforilación , Unión Proteica/efectos de los fármacos , Conformación Proteica , Transducción de Señal/efectos de los fármacos
6.
Inorg Chem ; 60(3): 1823-1831, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33464875

RESUMEN

A common challenge in Pt(IV) prodrug design is the limited repertoire of linkers available to connect the Pt(IV) scaffold with the bioactive payload. The commonly employed linkers are either too stable, leading to a linker artifact on the payload upon release, or too unstable, leading to premature release. In this study, we report the synthesis of a new class of Pt(IV) prodrugs using masked self-immolative 4-aminobenzyl linkers for controlled and traceless codrug delivery. Upon reduction of self-immolative Pt(IV) prodrugs, the detached axial ligands undergo decarboxylation and 1,6-elimination for payload release. Introduction of self-immolative linkers conferred good aqueous stability to the Pt(IV) codrug complex. Investigation revealed that efficient 1,6-elimination could be attributed to stabilization of the p-aza-quinone-methide intermediate. In particular, the self-immolative Pt(IV) prodrugs with cinnamate and coumarin derivatives were more potent than the coadministration of cisplatin with an unconjugated cinnamate or coumarin payload in vitro.


Asunto(s)
Antineoplásicos/química , Cisplatino/química , Compuestos Organoplatinos/química , Profármacos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Humanos , Estructura Molecular , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/farmacología , Profármacos/síntesis química , Profármacos/farmacología
7.
Angew Chem Int Ed Engl ; 60(17): 9264-9269, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33290628

RESUMEN

Mitochondria have emerged as important targets for cisplatin in cancer therapy. Apart from cisplatin, anticancer Pt complexes based on similar scaffolds have also been developed to target mitochondria. Yet cellular processing of cisplatin or these mitochondria-targeting Pt analogues remained unexplored, largely due to a lack of tools capable of probing these Pt drugs within an intracellular environment. We developed the first mitochondria-targeted fluorescent probe for real-time monitoring of Pt accumulation in mitochondria. We applied the probe to investigate mitochondria as cellular targets for Pt drug complexes and uncovered two distinct pathways whereby these Pt complexes could be delivered to mitochondria after cell entry.


Asunto(s)
Antineoplásicos/metabolismo , Cisplatino/metabolismo , Colorantes Fluorescentes/metabolismo , Mitocondrias/química , Antineoplásicos/química , Cisplatino/química , Colorantes Fluorescentes/química , Células HeLa , Humanos , Mitocondrias/metabolismo , Imagen Óptica , Espectrometría de Fluorescencia
8.
Angew Chem Int Ed Engl ; 60(24): 13405-13413, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33755286

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by an aberrant metabolic phenotype with high metastatic capacity, resulting in poor patient prognoses and low survival rates. We designed a series of novel AuIII cyclometalated prodrugs of energy-disrupting Type II antidiabetic drugs namely, metformin and phenformin. Prodrug activation and release of the metformin ligand was achieved by tuning the cyclometalated AuIII fragment. The lead complex 3met was 6000-fold more cytotoxic compared to uncoordinated metformin and significantly reduced tumor burden in mice with aggressive breast cancers with lymphocytic infiltration into tumor tissues. These effects was ascribed to 3met interfering with energy production in TNBCs and inhibiting associated pro-survival responses to induce deadly metabolic catastrophe.


Asunto(s)
Antineoplásicos/metabolismo , Metformina/metabolismo , Profármacos/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/química , Evaluación Preclínica de Medicamentos , Metabolismo Energético/efectos de los fármacos , Oro/química , Humanos , Metformina/química , Ratones , Conformación Molecular , Fenformina/química , Fenformina/metabolismo , Profármacos/química , Profármacos/farmacología , Profármacos/uso terapéutico , Trasplante Heterólogo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
9.
Angew Chem Int Ed Engl ; 59(24): 9314-9318, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32141662

RESUMEN

The abundance and evolving pathogenic behavior of bacterial microorganisms give rise to antibiotic tolerance and resistance which pose a danger to global public health. New therapeutic strategies are needed to keep pace with this growing threat. We propose a novel approach for targeting bacteria by harnessing formate, a cell metabolite found only in particular bacterial species, to activate an antibacterial prodrug and selectively inhibit their growth. This strategy is premised on transfer hydrogenation reaction on a biorthogonal substrate utilizing native formate as the hydride source as a means of uncaging an antibacterial prodrug. Using coordination-directed 3-component assembly to prepare a library of 768 unique Ru-Arene Schiff-base complexes, we identified several candidates that efficiently reduced sulfonyl azide functional group in the presence of formate. This strategy paves the way for a new approach of targeted antibacterial therapy by exploiting unique bacterial metabolites.


Asunto(s)
Antibacterianos/metabolismo , Formiatos/metabolismo , Profármacos/metabolismo , Rutenio/química , Antibacterianos/farmacología , Catálisis , Hidrogenación , Bases de Schiff/química
10.
Angew Chem Int Ed Engl ; 59(43): 19070-19078, 2020 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-32716112

RESUMEN

Immunogenic cell death (ICD) is a rare immunostimulatory form of cell death that can improve the clinical outcomes of chemo-immunotherapeutic combination regimens through the establishment of a long-term cancer immunity. None of the clinically used DNA-binding PtII complexes is considered a Type II ICD inducer. We generated a series of PtII -carbene complexes by applying minor structural alterations to the scaffold of a Type II ICD inducer Pt-NHC and compared their efficiency in triggering ICD-related cellular responses and phagocytosis. We successfully identified PlatinER, a novel highly potent PtII candidate with superior ICD properties. Crucially, the magnitude of ICD-associated phagocytosis induced upon exposure of cancer cells to Pt complexes was dependent on the levels of ER-localized reactive oxygen species (ROS) generation, which underpins their mechanisms of action and provides a feasible approach for the design of more effective Type II ICD inducers.


Asunto(s)
Antineoplásicos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Compuestos Organoplatinos/farmacología , Antineoplásicos/química , Espectroscopía de Resonancia Magnética con Carbono-13 , Línea Celular Tumoral , Factor 2 Eucariótico de Iniciación/metabolismo , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Compuestos Organoplatinos/química , Fagocitosis/efectos de los fármacos , Espectroscopía de Protones por Resonancia Magnética , Especies Reactivas de Oxígeno/metabolismo , Espectrometría de Masa por Ionización de Electrospray , eIF-2 Quinasa/metabolismo
11.
Carbon N Y ; 50(4): 1625-1634, 2019 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-31105316

RESUMEN

Carbon nanotubes (CNTs) have emerged as promising drug delivery systems particularly for cancer therapy, due to their abilities to overcome some of the challenges faced by cancer treatment, namely non-specificity, poor permeability into tumour tissues, and poor stability of anticancer drugs. Encapsulation of anticancer agents inside CNTs provides protection from external deactivating agents. However, the open ends of the CNTs leave the encapsulated drugs exposed to the environment and eventually their uncontrolled release before reaching the desired target. In this study, we report the successful encapsulation of cisplatin, a FDA-approved chemotherapeutic drug, into multi-walled carbon nanotubes and the capping at the ends with functionalised gold nanoparticles to achieve a "carbon nanotube bottle" structure. In this proof-of-concept study, these caps did not prevent the encapsulation of drug in the inner space of CNTs; on the contrary, we achieved higher drug loading inside the nanotubes in comparison with data reported in literature. In addition, we demonstrated that encapsulated cisplatin could be delivered in living cells under physiological conditions to exert its pharmacological action.

12.
Angew Chem Int Ed Engl ; 58(1): 164-167, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30407697

RESUMEN

The PtIV prodrug strategy has emerged as an excellent alternative to tackle the problems associated with conventional PtII drug therapy. However, there is a lack of tools to study how this new class of PtIV drugs are processed at the cellular level. Herein, we report the first ratiometric probe for cisplatin detection and use it to investigate PtIV anticancer complexes in biological systems. The probe was able to distinguish between cisplatin and its PtIV derivatives, allowing us to probe the intracellular reduction of PtIV prodrug complexes. The correlation between the amount of active PtII species available after intracellular reduction of PtIV complexes and their cytotoxicity and the role glutathione plays in the reduction of PtIV complexes were investigated.


Asunto(s)
Cisplatino/química , Colorantes Fluorescentes/uso terapéutico , Platino (Metal)/química , Profármacos/uso terapéutico , Humanos
13.
Angew Chem Int Ed Engl ; 58(24): 8109-8114, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-30945417

RESUMEN

A novel and highly efficient dual-targeting platform was designed to ensure targeted in vivo delivery of dual-action PtIV prodrugs. The dual targeting was established by liposomal encapsulation of PtIV complexes, thereby utilizing the enhanced permeability and retention (EPR) effect as the first stage of targeting to attain a high accumulation of the drug-loaded liposomes in the tumor. After the release of the PtIV prodrug inside cancer cells, a second stage of targeting directed a portion of the PtIV prodrugs to the mitochondria. Upon intracellular reduction, these PtIV prodrugs released two bioactive molecules, acting both on the mitochondrial and on the nuclear DNA. Our PtIV system showed excellent activity in vitro and in vivo, characterized by a cytotoxicity in a low micromolar range and complete tumor remission, respectively. Notably, marked in vivo activity was accompanied by reduced kidney toxicity, highlighting the unique therapeutic potential of our novel dual-targeting dual-action platform.


Asunto(s)
Antineoplásicos/uso terapéutico , Riñón/efectos de los fármacos , Platino (Metal)/uso terapéutico , Antineoplásicos/farmacología , Humanos , Platino (Metal)/farmacología
14.
Chemistry ; 24(8): 1870-1876, 2018 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-29047166

RESUMEN

A new pre-assembled ratiometric sensing platform was constructed from a coumarin donor and a rhodamine acceptor designed for through-bond energy transfer (TBET). A phenylacetylene linker was installed to disrupt the planarity of the extended conjugated system but retaining the efficient energy transfer between the donor and acceptor motifs. To demonstrate its versatility as a sensing platform, we conjugated recognition motifs through amide coupling reactions to yield two TBET chemosensors capable of sensing either endogenously produced NO and ClO- . Both probes possessed high selectivity for their analytes, exhibited good stability under physiological conditions, and performed well as bioimaging probes in living cells.

15.
Mol Pharm ; 15(8): 3020-3031, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29979603

RESUMEN

Current anticancer drug discovery efforts focus on the identification of first-in-class compounds with a mode-of-action distinct from conventional DNA-targeting agents for chemotherapy. An emerging trend is the identification of endoplasmic reticulum (ER) targeting compounds that induce ER stress in cancer cells, leading to cell death. However, a limited pool of such compounds has been identified to date, and there are limited studies done on such compounds to allow for the rational design of ER stress-inducing agents. In our present study, we present a series of highly cytotoxic, ER stress-inducing Ru(II)-arene Schiff-Base (RAS) complexes, bearing iminoquinoline chelate ligands. We demonstrate that by structural modification to the iminoquinoline ligand, we could tune its π-acidity and influence reactive oxygen species (ROS) induction, switching between a ROS-mediated ER stress pathway activation and one that is not mediated by ROS induction. Our current study adds to the available ER stress inducers and shows how structural tuning could be used as a means to modulate the mode-of-action of such compounds.


Asunto(s)
Antineoplásicos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Compuestos Organometálicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Neoplasias/patología , Compuestos Organometálicos/química , Compuestos Organometálicos/uso terapéutico , Quinolinas/química , Quinolinas/farmacología , Quinolinas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Rutenio/química , Rutenio/farmacología , Bases de Schiff/química , Bases de Schiff/farmacología , Bases de Schiff/uso terapéutico , Relación Estructura-Actividad
16.
Inorg Chem ; 57(17): 10702-10717, 2018 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-30106571

RESUMEN

With the aim of enhancing the biological activity of ruthenium-nitrosyl complexes, new compounds with four equatorially bound indazole ligands, namely, trans-[RuCl(Hind)4(NO)]Cl2·H2O ([3]Cl2·H2O) and trans-[RuOH(Hind)4(NO)]Cl2·H2O ([4]Cl2·H2O), have been prepared from trans-[Ru(NO2)2(Hind)4] ([2]). When the pH-dependent solution behavior of [3]Cl2·H2O and [4]Cl2·H2O was studied, two new complexes with two deprotonated indazole ligands were isolated, namely [RuCl(ind)2(Hind)2(NO)] ([5]) and [RuOH(ind)2(Hind)2(NO)] ([6]). All prepared compounds were comprehensively characterized by spectroscopic (IR, UV-vis, 1H NMR) techniques. Compound [2], as well as [3]Cl2·2(CH3)2CO, [4]Cl2·2(CH3)2CO, and [5]·0.8CH2Cl2, the latter three obtained by recrystallization of the first isolated compounds (hydrates or anhydrous species) from acetone and dichloromethane, respectively, were studied by X-ray diffraction methods. The photoinduced release of NO in [3]Cl2 and [4]Cl2 was investigated by cyclic voltammetry and resulting paramagnetic NO species were detected by EPR spectroscopy. The quantum yields of NO release were calculated and found to be low (3-6%), which could be explained by NO dissociation and recombination dynamics, assessed by femtosecond pump-probe spectroscopy. The geometry and electronic parameters of Ru species formed upon NO release were identified by DFT calculations. The complexes [3]Cl2 and [4]Cl2 showed considerable antiproliferative activity in human cancer cell lines with IC50 values in low micromolar or submicromolar concentration range and are suitable for further development as potential anticancer drugs. p53-dependence of Ru-NO complexes [3]Cl2 and [4]Cl2 was studied and p53-independent mode of action was confirmed. The effects of NO release on the cytotoxicity of the complexes with or without light irradiation were investigated using NO scavenger carboxy-PTIO.


Asunto(s)
Indazoles/química , Óxido Nítrico/química , Óxidos de Nitrógeno , Compuestos Organometálicos , Rutenio , Antineoplásicos/química , Antineoplásicos/farmacología , Western Blotting , Supervivencia Celular , Cisplatino/farmacología , Estabilidad de Medicamentos , Electroquímica , Células HCT116 , Humanos , Concentración 50 Inhibidora , Ligandos , Modelos Moleculares , Óxidos de Nitrógeno/química , Óxidos de Nitrógeno/farmacología , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Teoría Cuántica , Rutenio/química , Rutenio/farmacología , Agua/química , Difracción de Rayos X
17.
Genes Cells ; 21(3): 226-40, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26891792

RESUMEN

Doxorubicin is a widely used chemotherapeutic agent, but its utility is limited by cellular resistance and off-target effects. To understand the molecular mechanisms regulating chemotherapeutic responses to doxorubicin, we previously carried out a genomewide search of doxorubicin-resistance genes in Schizosaccharomyces pombe fission yeast and showed that these genes are organized into networks that counteract doxorubicin cytotoxicity. Here, we describe the identification of a subgroup of doxorubicin-resistance genes that, when disrupted, leads to reduced tolerance to exogenous calcium. Unexpectedly, we observed a suppressive effect of calcium on doxorubicin cytotoxicity, where concurrent calcium and doxorubicin treatment resulted in significantly higher cell survival compared with cells treated with doxorubicin alone. Conversely, inhibitors of voltage-gated calcium channels enhanced doxorubicin cytotoxicity in the mutants. Consistent with these observations in fission yeast, calcium also suppressed doxorubicin cytotoxicity in human breast cancer cells. Further epistasis analyses in yeast showed that this suppression of doxorubicin toxicity by calcium was synergistically dependent on Rav1 and Vph2, two regulators of vacuolar-ATPase assembly; this suggests potential modulation of the calcium-doxorubicin interaction by fluctuating proton concentrations within the cellular environment. Thus, the modulatory effects of drugs or diet on calcium concentrations should be considered in doxorubicin treatment regimes.


Asunto(s)
Calcio/farmacología , Doxorrubicina/toxicidad , Inhibidores de Topoisomerasa II/toxicidad , Bloqueadores de los Canales de Calcio/farmacología , Supervivencia Celular , Farmacorresistencia Fúngica/genética , Genes Fúngicos , Humanos , Células MCF-7 , Schizosaccharomyces/efectos de los fármacos , Schizosaccharomyces/metabolismo
18.
Nucleic Acids Res ; 43(11): 5284-96, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-25916851

RESUMEN

Platinum-based anticancer drugs act therapeutically by forming DNA adducts, but suffer from severe toxicity and resistance problems, which have not been overcome in spite of decades of research. And yet defined chromatin targets have generally not been considered in the drug development process. Here we designed novel platinum-intercalator species to target a highly deformed DNA site near the nucleosome center. Between two seemingly similar structural isomers, we find a striking difference in DNA site selectivity in vitro, which comes about from stereochemical constraints that limit the reactivity of the trans isomer to special DNA sequence elements while still allowing the cis isomer to efficiently form adducts at internal sites in the nucleosome core. This gives the potential for controlling nucleosome site targeting in vivo, which would engender sensitivity to epigenetic distinctions and in particular cell type/status-dependent differences in nucleosome positioning. Moreover, while both compounds yield very similar DNA-adduct structures and display antitumor cell activity rivalling that of cisplatin, the cis isomer, relative to the trans, has a much more rapid cytotoxic effect and distinct impact on cell function. The novel stereochemical principles for controlling DNA site selectivity we discovered could aid in the design of improved site discriminating agents.


Asunto(s)
Antineoplásicos/química , Sustancias Intercalantes/química , Naftalimidas/química , Nucleosomas/química , Compuestos Organoplatinos/química , Antineoplásicos/toxicidad , Línea Celular Tumoral , Aductos de ADN/análisis , Humanos , Sustancias Intercalantes/toxicidad , Naftalimidas/toxicidad , Compuestos Organoplatinos/toxicidad , Estereoisomerismo
19.
Mol Pharm ; 13(7): 2543-54, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27174050

RESUMEN

p53 is a key tumor suppressor gene involved in key cellular processes and implicated in cancer therapy. However, it is inactivated in more than 50% of all cancers due to mutation or overexpression of its negative regulators. This leads to drug resistance and poor chemotherapeutic outcome as most clinical drugs act via a p53-dependent mechanism of action. An attractive strategy to circumvent this resistance would be to identify new anticancer drugs that act via p53-independent mode of action. In the present study, we identified 9 Ru (II)-Arene Schiff-base (RAS) complexes able to induce p53-independent cytotoxicity and discuss structural features that are required for their p53-independent activity. Increasing hydrophobicity led to an increase in cellular accumulation in cells with a corresponding increase in efficacy. We further showed that all nine complexes demonstrated p53-independent activity. This was despite significant differences in their physicochemical properties, suggesting that the iminoquinoline ligand, a common structural feature for all the complexes, is required for the p53-independent activity.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacología , Rutenio/química , Bases de Schiff/química , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Células HCT116 , Humanos , Estructura Molecular , Relación Estructura-Actividad
20.
Anal Chem ; 87(20): 10292-8, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26382664

RESUMEN

The salient optical properties of highly luminescent semiconductor nanocrystals render them ideal fluorophores for clinical diagnostics, therapeutics, and highly sensitive biochip applications. Microfluidic systems allow miniaturization and integration of multiple biochemical processes in a single device and do not require sophisticated diagnostic tools. Herein, we describe a microfluidic system that integrates RNA extraction, reverse transcription to cDNA, amplification and detection within one integrated device to detect histidine decarboxylase (HDC) gene directly from human white blood cells samples. When anisotropic semiconductor nanorods (NRs) were used as the fluorescent probes, the detection limit was found to be 0.4 ng of total RNA, which was much lower than that obtained using spherical quantum dots (QDs) or organic dyes. This was attributed to the large action cross-section of NRs and their high probability of target capture in a pull-down detection scheme. The combination of large scale integrated microfluidics with highly fluorescent semiconductor NRs may find widespread utility in point-of-care devices and multitarget diagnostics.


Asunto(s)
Histidina Descarboxilasa/genética , Técnicas Analíticas Microfluídicas/instrumentación , Nanotubos/química , ARN/análisis , ARN/sangre , Semiconductores , Anisotropía , Colorantes Fluorescentes/química , Humanos , Puntos Cuánticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA