Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
J Chem Phys ; 158(4): 045101, 2023 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-36725513

RESUMEN

Triplex DNA structure has potential therapeutic application in inhibiting the expression of genes involved in cancer and other diseases. As a DNA-targeting antitumor and antibiotic drug, coralyne shows a remarkable binding propensity to triplex over canonical duplex and thus can modulate the stability of triplex structure, providing a prospective gene targeting strategy. Much less is known, however, about coralyne-binding interactions with triplex. By combining multiple steady-state spectroscopy with ultrafast fluorescence spectroscopy, we have investigated the binding behaviors of coralyne with typical triplexes. Upon binding with a G-containing triplex, the fluorescence of coralyne is markedly quenched owing to the photoinduced electron transfer (PET) of coralyne with the G base. Systematic studies show that the PET rates are sensitive to the binding configuration and local microenvironment, from which the coexisting binding modes of monomeric (full and partial) intercalation and aggregate stacking along the sugar-phosphate backbone are distinguished and their respective contributions are determined. It shows that coralyne has preferences for monomeric intercalation within CGG triplex and pure TAT triplex, whereas CGC+ triplex adopts mainly backbone binding of coralyne aggregates due to charge repulsion, revealing the sequence-specific binding selectivity. The triplex-DNA-induced aggregation of coralyne could be used as a probe for recognizing the water content in local DNA structures. The strong π-π stacking of intercalated coralyne monomer with base-triplets plays an important role in stabilizing the triplex structure. These results provide mechanistic insights for understanding the remarkable propensity of coralyne in selective binding to triplex DNA and shed light on the prospective applications of coralyne-triplex targeted anti-gene therapeutics.


Asunto(s)
ADN , Espectrometría de Fluorescencia , Desnaturalización de Ácido Nucleico , Conformación de Ácido Nucleico , ADN/química
2.
J Am Chem Soc ; 143(36): 14738-14747, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34467764

RESUMEN

Oxidative stress produces a variety of radicals in DNA, including pyrimidine nucleobase radicals. The nitrogen-centered DNA radical 2'-deoxycytidin-N4-yl radical (dC·) plays a role in DNA damage mediated by one electron oxidants, such as HOCl and ionizing radiation. However, the reactivity of dC· is not well understood. To reduce this knowledge gap, we photochemically generated dC· from a nitrophenyl oxime nucleoside and within chemically synthesized oligonucleotides from the same precursor. dC· formation is confirmed by transient UV-absorption spectroscopy in laser flash photolysis (LFP) experiments. LFP and duplex DNA cleavage experiments indicate that dC· oxidizes dG. Transient formation of the dG radical cation (dG+•) is observed in LFP experiments. Oxidation of the opposing dG in DNA results in hole transfer when the opposing dG is part of a dGGG sequence. The sequence dependence is attributed to a competition between rapid proton transfer from dG+• to the opposing dC anion formed and hole transfer. Enhanced hole transfer when less acidic O6-methyl-2'-deoxyguanosine is opposite dC· supports this proposal. dC· produces tandem lesions in sequences containing thymidine at the 5'-position by abstracting a hydrogen atom from the thymine methyl group. The corresponding thymidine peroxyl radical completes tandem lesion formation by reacting with the 5'-adjacent nucleotide. As dC· is reduced to dC, its role in the process is traceless and is only detectable because of the ability to independently generate it from a stable precursor. These experiments reveal that dC· oxidizes neighboring nucleotides, resulting in deleterious tandem lesions and hole transfer in appropriate sequences.


Asunto(s)
Daño del ADN/efectos de los fármacos , ADN/efectos de los fármacos , Desoxicitidina/química , Radicales Libres/química , ADN/química , Desoxicitidina/análogos & derivados , Desoxicitidina/efectos de la radiación , Desoxiguanosina/química , Oximas/química , Oximas/efectos de la radiación , Fotólisis , Rayos Ultravioleta
3.
Nucleic Acids Res ; 47(22): 11514-11526, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31724721

RESUMEN

Phosphorothioate (PS) modifications naturally appear in bacteria and archaea genome and are widely used as antisense strategy in gene therapy. But the chemical effects of PS introduction as a redox active site into DNA (S-DNA) is still poorly understood. Herein, we perform time-resolved spectroscopy to examine the underlying mechanisms and dynamics of the PS oxidation by potent radicals in free model, in dinucleotide, and in S-oligomer. The crucial sulphur-centered hemi-bonded intermediates -P-S∴S-P- were observed and found to play critical roles leading to the stable adducts of -P-S-S-P-, which are backbone DNA lesion products. Moreover, the oxidation of the PS moiety in dinucleotides d[GPSG], d[APSA], d[GPSA], d[APSG] and in S-oligomers was monitored in real-time, showing that PS oxidation can compete with adenine but not with guanine. Significantly, hole transfer process from A+• to PS and concomitant -P-S∴S-P- formation was observed, demonstrating the base-to-backbone hole transfer unique to S-DNA, which is different from the normally adopted backbone-to-base hole transfer in native DNA. These findings reveal the distinct backbone lesion pathway brought by the PS modification and also imply an alternative -P-S∴S-P-/-P-S-S-P- pathway accounting for the interesting protective role of PS as an oxidation sacrifice in bacterial genome.


Asunto(s)
Bacterias/genética , ADN Bacteriano/química , Oligonucleótidos Fosforotioatos/química , Azufre/química , Genoma Bacteriano/genética , Conformación de Ácido Nucleico , Oxidación-Reducción , Análisis Espectral
4.
J Phys Chem A ; 124(29): 6076-6083, 2020 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-32585092

RESUMEN

Proton transfer is regarded as a fundamental process in chemical reactions of DNA molecules and continues to be an active research theme due to the connection with charge transport and oxidation damage of DNA. For the guanine radical cation (G•+) derived from one-electron oxidation, experiments suggest a facile proton transfer within the G•+:C base pair, and a rapid deprotonation from N1 in free base or single-strand DNA. To address the deprotonation mechanism, we perform a thorough investigation on deprotonation of G•+ in free G base by combining density functional theory (DFT) and laser flash photolysis spectroscopy. Experimentally, kinetics of deprotonation is monitored at temperatures varying from 280 to 298 K, from which the activation energy of 15.1 ± 1.5 kJ/mol is determined for the first time. Theoretically, four solvation models incorporating explicit waters and the polarized continuum model (PCM), i.e., 3H2O-PCM, 4H2O-PCM, 5H2O-PCM, and 7H2O-PCM models are used to calculate deprotonation potential energy profile, and the barriers of 5.5, 13.4, 14.4, and 13.7 kJ/mol are obtained, respectively. It is shown that at least four explicit waters are required for properly simulating the deprotonation reaction, where the participation of protonated water cluster plays key roles in facilitating the proton release from G•+.


Asunto(s)
Guanina/química , Protones , Daño del ADN , Guanina/metabolismo , Modelos Moleculares , Conformación Molecular , Oxidación-Reducción , Teoría Cuántica
5.
J Chem Phys ; 152(3): 035101, 2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-31968979

RESUMEN

One-electron oxidation of adenine (A) leads initially to the formation of adenine radical cation (A•+). Subsequent deprotonation of A•+ can provoke deoxyribonucleic acid (DNA) damage, which further causes senescence, cancer formation, and even cell death. However, compared with considerable reports on A•+ reactions in free deoxyadenosine (dA) and duplex DNA, studies in non-B-form DNA that play critical biological roles are rare at present. It is thus of vital importance to explore non-B-form DNA, among which the triplex is an emerging topic. Herein, we investigate the deprotonation behavior of A•+ in the TAT triplex with continuous A bases by time-resolved laser flash photolysis. The rate constants for the one-oxidation of triplex 8.4 × 108 M-1 s-1 and A•+ deprotonation 1.3 × 107 s-1 are obtained. The kinetic isotope effect of A•+ deprotonation in the TAT triplex is 1.8, which is characteristic of a direct release of the proton into the solvent similar to free base dA. It is thus elucidated that the A•+ proton bound with the third strand is most likely to be released into the solvent because of the weaker Hoogsteen H-bonding interaction and the presence of the highly mobile hydration waters within the third strand. Additionally, it is confirmed through Fourier transform infrared spectroscopy that the deprotonation of A•+ results in the dissociation of the third strand and disruption of the secondary structure of the triplex. These results provide valuable kinetic data and in-depth mechanistic insights for understanding the adenine oxidative DNA damage in the triplex.


Asunto(s)
Adenina/química , ADN/química , Electrones , Timina/química , Enlace de Hidrógeno , Oxidación-Reducción
6.
Angew Chem Int Ed Engl ; 59(32): 13406-13413, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32365264

RESUMEN

Guanine radicals are important reactive intermediates in DNA damage. Hydroxyl radical (HO. ) has long been believed to react with 2'-deoxyguanosine (dG) generating 2'-deoxyguanosin-N1-yl radical (dG(N1-H). ) via addition to the nucleobase π-system and subsequent dehydration. This basic tenet was challenged by an alternative mechanism, in which the major reaction of HO. with dG was proposed to involve hydrogen atom abstraction from the N2-amine. The 2'-deoxyguanosin-N2-yl radical (dG(N2-H). ) formed was proposed to rapidly tautomerize to dG(N1-H). . We report the first independent generation of dG(N2-H). in high yield via photolysis of 1. dG(N2-H). is directly observed upon nanosecond laser flash photolysis (LFP) of 1. The absorption spectrum of dG(N2-H). is corroborated by DFT studies, and anti- and syn-dG(N2-H). are resolved for the first time. The LFP experiments showed no evidence for tautomerization of dG(N2-H). to dG(N1-H). within hundreds of microseconds. This observation suggests that the generation of dG(N1-H). via dG(N2-H). following hydrogen atom abstraction from dG is unlikely to be a major pathway when HO. reacts with dG.


Asunto(s)
Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Radicales Libres/análisis , Desoxiguanosina/efectos de la radiación , Radicales Libres/química , Radical Hidroxilo/química , Fotólisis , Espectrofotometría Ultravioleta , Rayos Ultravioleta
7.
J Am Chem Soc ; 141(5): 1970-1979, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30624927

RESUMEN

Radical cations of nucleobases are key intermediates causing genome mutation, among which cytosine C•+ is of growing importance because the ensuing cytosine oxidation causes GC → AT transversions in DNA replication. Although the chemistry and biology of steady-state C oxidation products have been characterized, time-resolved study of initial degradation pathways of C•+ is still at the preliminary stage. Herein, we choose i-motif, a unique C-quadruplex structure composed of hemiprotonated base pairs C(H)+:C, to examine C•+ degradation in a DNA surrounding without interference of G bases. Comprehensive time-resolved spectroscopy were performed to track C•+ dynamics in i-motif and in free base dC. The competing pathways of deprotonation (1.4 × 107 s-1), tautomerization (8.8 × 104 s-1), and hydration (5.3 × 103 s-1) are differentiated, and their rate constants are determined for the first time, underlining the strong reactivity of C•+. Distinct pathway is observed in i-motif compared with dC, showing the prominent features of C•+ hydration forming C(5OH)• and C(6OH)•. By further experiments of pH-dependence, comparison with single strand, and with Ag+ mediated i-motif, the mechanisms of C•+ degradation in i-motif are disclosed. The hydrogen-bonding within C(H)+:C plays a significant role in guiding the reaction flux, by blocking the tautomerization of C(-H)• and reversing the equilibrium from C(-H)• to C•+. The C radicals in i-motif thus retain more cation character, and are mainly subject to hydration leading to lesion products that can induce disruption of i-motif structure and affect its critical roles in gene-regulation.


Asunto(s)
Citosina/química , ADN/química , Desoxicitidina/química , Cationes/química , Radicales Libres/química , Enlace de Hidrógeno , Conformación de Ácido Nucleico
8.
Chemphyschem ; 20(5): 757-765, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30702794

RESUMEN

Azabases are intriguing DNA and RNA analogues and have been used as effective antiviral and anticancer medicines. However, photosensitivity of these drugs has also been reported. Here, pH-controlled intersystem crossing (ISC) process of 9H 8-azaadenine (8-AA) in aqueous solution is reported. Broadband transient absorption measurements reveal that the hydrogen atom at N9 position can greatly affect ISC of 8-AA and ISC is more favorable when 8-AA is in its neutral form in aqueous solution. The initial excited ππ* (S2 ) state evolves through ultrafast internal conversion (IC) (4.2 ps) to the lower-lying nπ* state (S1 ), which further stands as a door way state for ISC with a time constant of 160 ps. The triplet state has a lifetime of 6.1 µs. On the other hand, deprotonation at N9 position promotes the IC from the ππ* (S2 ) state to the ground state (S0 ) and the lifetime of the S2 state is determined to be 10 ps. The experimental results are further supported by time-dependent density functional theory (TDDFT) calculations. Singlet oxygen generation yield is measured to be 13.8 % for the neutral 8-AA while the deprotonated one exhibit much lower yield (<2 %), implying that this compound could be a potential pH-sensitized photodynamic therapy agent.

9.
Angew Chem Int Ed Engl ; 58(36): 12580-12584, 2019 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-31310447

RESUMEN

Chlorosilanes are versatile reagents in organic synthesis and material science. A mild pathway is now reported for the quantitative conversion of hydrosilanes to silyl chlorides under visible-light irradiation using neutral eosin Y as a hydrogen-atom-transfer photocatalyst and dichloromethane as a chlorinating agent. Stepwise chlorination of di- and trihydrosilanes was achieved in a highly selective fashion assisted by continuous-flow micro-tubing reactors. The ability to access silyl radicals using photocatalytic Si-H activation promoted by eosin Y offers new perspectives for the synthesis of valuable silicon reagents in a convenient and green manner.

10.
Phys Chem Chem Phys ; 20(19): 13598-13606, 2018 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-29736516

RESUMEN

Herein, mechanistic aspects of the photogeneration and quenching of guanine radical cation through one-electron oxidation of the G-quadruplex of G2T2G2TGTG2T2G2 (TBA) sequence were investigated by a combined quantum mechanical/molecular mechanical (QM/MM) approach at the CASPT2//CASSCF/AMBER level of theory. Herein, one electron promotion of the oxygen lone pair of the photo-excited photosensitizer peroxydisulfate to its O-O σ* orbital was first demonstrated to become tunable through the varied reduction ability of the G base in the presence or absence of interbase hydrogen bonding, thereby dynamically controlling the deprotonation site in G-quadruplex TBA. The quenching of G radical cation mediated by the formation of SO42-via photoinduced electron transfer can be triggered effectively by the deprotonation reaction of free proton rather than that of the hydrogen-bonded proton in G-G (G-quartet) and G-T (loop) aqueous surrounding. By calculating the deprotonation paths for the G radical cation, the deprotonation reactions in G-quadruplex TBA were verified to proceed predominantly along the site of imino proton (N1-H) in the loop moiety; this showed the coexisting occurrence of amino (N2-H) deprotonation in the G-quartet part. The mechanistic features discussed in this study represent significant advances in the understanding of DNA radical chemistry.


Asunto(s)
ADN/química , G-Cuádruplex , Guanina/química , Fármacos Fotosensibilizantes/química , Transporte de Electrón , Enlace de Hidrógeno , Fenómenos Mecánicos , Modelos Moleculares , Conformación Molecular , Oxidación-Reducción , Protones , Termodinámica , Agua/química
11.
Int J Mol Sci ; 19(4)2018 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-29617273

RESUMEN

Recognition of unusual left-handed Z-DNA by specific binding of small molecules is crucial for understanding biological functions in which this particular structure participates. Recent investigations indicate that zinc cationic porphyrin (ZnTMPyP4) is promising as a probe for recognizing Z-DNA due to its characteristic chiroptical properties upon binding with Z-DNA. However, binding mechanisms of the ZnTMPyP4/Z-DNA complex remain unclear. By employing time-resolved UV-visible absorption spectroscopy in conjunction with induced circular dichroism (ICD), UV-vis, and fluorescence measurements, we examined the binding interactions of ZnTMPyP4 towards B-DNA and Z-DNA. For the ZnTMPyP4/Z-DNA complex, two coexisting binding modes were identified as the electrostatic interaction between pyridyl groups and phosphate backbones, and the major groove binding by zinc(II) coordinating with the exposed guanine N7. The respective contribution of each mode is assessed, allowing a complete scenario of binding modes revealed for the ZnTMPyP4/Z-DNA. These interaction modes are quite different from those (intercalation and partial intercalation modes) for the ZnTMPyP4/B-DNA complex, thereby resulting in explicit differentiation between B-DNA and Z-DNA. Additionally, the binding interactions of planar TMPyP4 to DNA were also investigated as a comparison. It is shown that without available virtual orbitals to coordinate, TMPyP4 binds with Z-DNA solely in the intercalation mode, as with B-DNA, and the intercalation results in a structural transition from Z-DNA to B-ZNA. These results provide mechanistic insights for understanding ZnTMPyP4 as a probe of recognizing Z-DNA and afford a possible strategy for designing new porphyrin derivatives with available virtual orbitals for the discrimination of B-DNA and Z-DNA.


Asunto(s)
ADN/química , Metaloporfirinas/química , Conformación de Ácido Nucleico , ADN/metabolismo , ADN Forma B/química , ADN Forma B/metabolismo , ADN de Forma Z/química , ADN de Forma Z/metabolismo , Metaloporfirinas/metabolismo , Porfirinas/química , Porfirinas/metabolismo , Unión Proteica , Análisis Espectral
12.
J Chem Phys ; 146(2): 025103, 2017 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-28088146

RESUMEN

5-Iodouracil (5-IU) can be integrated into DNA and acts as a UV sensitive chromophore suitable for probing DNA structure and DNA-protein interactions based on the photochemical reactions of 5-IU. Here, we perform joint studies of time-resolved Fourier transform infrared (TR-FTIR) spectroscopy and ab initio calculations to examine the state-specific photochemical reaction mechanisms of the 5-IU. The fact that uracil (U) is observed in TR-FTIR spectra after 266 nm irradiation of 5-IU in acetonitrile and ascribed to the product of hydrogen abstraction by the uracil-5-yl radical (U·) provides experimental evidence for the C-I bond homolysis of 5-IU. The excited state potential energy curves are calculated with the complete active space second-order perturbation//complete active space self-consistent field method, from which a singlet predissociation mechanism is elucidated. It is shown that the initially populated 1(ππ*) state crosses with the repulsive 1(πσ*) or 1(nIσ*) state, through which 5-IU undergoes dissociation to the fragments of (U·) radical and iodine atom. In addition, the possibility of intersystem crossing (ISC) is evaluated based on the calculated vertical excitation energies. Although a probable ISC from 1(ππ*) state to 3(nOπ*) and then to the lowest triplet 3(ππ*) could occur in principal, there is little possibility for the excited state populations bifurcating to triplet manifold, given that the singlet state predissociation follows repulsive potential and should occur within dozens to hundreds of femtoseconds. Such low population of triplet states means that the contribution of triplet state to photoreactions of 5-IU should be quite minor. These results demonstrate clearly a physical picture of C-I bond homolysis of 5-IU and provide mechanistic illuminations to the interesting applications of 5-IU as photoprobes and in radiotherapy of cancer.


Asunto(s)
Uracilo/análogos & derivados , Modelos Químicos , Espectroscopía Infrarroja por Transformada de Fourier , Rayos Ultravioleta , Uracilo/química , Uracilo/efectos de la radiación
13.
Chemistry ; 22(28): 9676-86, 2016 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-27249665

RESUMEN

Construction of Gd(III) photosensitizers is important for designing theranostic agents owing to the unique properties arising from seven unpaired f electrons of the Gd(3+) ion. Combining these with the advantages of porpholactones with tunable NIR absorption, we herein report the synthesis of Gd(III) complexes Gd-1-4 (1, porphyrin; 2, porpholactone; 3 and 4, cis- and trans-porphodilactone, respectively) and investigated their function as singlet oxygen ((1) O2 ) photosensitizers. These Gd complexes displayed (1) O2 quantum yields (ΦΔ s) from 0.64-0.99 with the order Gd-1

14.
J Phys Chem A ; 120(27): 5016-22, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26859442

RESUMEN

The rich photo-oxidation pathways and products of terrylenediimide (TDI) with singlet oxygen ((1)O2) have been examined by powerful computational approaches. Potential energy profiles and product fluorescence properties are characterized. A variety of new products are unraveled and predicted to emit fluorescence at both visible and near-infrared ranges, which could open the possibility for interesting applications of using TDI as a fluorescence probe for the single-molecule detection of (1)O2 and designing multicolor photoconvertible fluorophores based on (1)O2 oxidation.

15.
J Am Chem Soc ; 137(1): 259-66, 2015 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-25506785

RESUMEN

Although numerous studies have been devoted to the charge transfer through double-stranded DNA (dsDNA), one of the major problems that hinder their potential applications in molecular electronics is the fast deprotonation of guanine cation (G(+•)) to form a neutral radical that can cause the termination of hole transfer. It is thus of critical importance to explore other DNA structures, among which G-quadruplexes are an emerging topic. By nanosecond laser flash photolysis, we report here the direct observation and findings of the unusual deprotonation behavior (loss of amino proton N2-H instead of imino proton N1-H) and slower (1-2 orders of magnitude) deprotonation rate of G(+•) within G-quadruplexes, compared to the case in the free base dG or dsDNA. Four G-quadruplexes AG3(T2AG3)3, (G4T4G4)2, (TG4T)4, and G2T2G2TGTG2T2G2 (TBA) are measured systematically to examine the relationship of deprotonation with the hydrogen-bonding surroundings. Combined with in depth kinetic isotope experiments and pKa analysis, mechanistic insights have been further achieved, showing that it should be the non-hydrogen-bonded free proton to be released during deprotonation in G-quadruplexes, which is the N2-H exposed to solvent for G bases in G-quartets or the free N1-H for G base in the loop. The slower N2-H deprotonation rate can thus ensure less interruption of the hole transfer. The unique deprotonation features observed here for G-quadruplexes open possibilities for their interesting applications as molecular electronic devices, while the elucidated mechanisms can provide illuminations for the rational design of G-quadruplex structures toward such applications and enrich the fundamental understandings of DNA radical chemistry.


Asunto(s)
ADN/química , G-Cuádruplex , Guanina/química , Protones , Cationes/química , Radicales Libres/química
16.
J Am Chem Soc ; 137(33): 10745-52, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26247480

RESUMEN

Learning nature's approach to modulate photophysical properties of NIR porphyrinoids by fine-tuning ß-substituents including the number and position, in a manner similar to naturally occurring chlorophylls, has the potential to circumvent the disadvantages of traditional "extended π-conjugation" strategy such as stability, molecular size, solubility, and undesirable π-π stacking. Here we show that such subtle structural changes in Pt(II) or Pd(II) cis/trans-porphodilactones (termed by cis/trans-Pt/Pd) influence photophysical properties of the lowest triplet excited states including phosphorescence, Stokes shifts, and even photosensitization ability in triplet-triplet annihilation reactions with rubrene. Prominently, the overall upconversion capability (η, η = ε·Φ(UC)) of Pd or Pt trans-complex is 10(4) times higher than that of cis-analogue. Nanosecond time-resolved infrared (TR-IR) spectroscopy experiments showed larger frequency shift of ν(C═O) bands (ca. 10 cm(-1)) of cis-complexes than those of trans-complexes in the triplet excited states. These spectral features, combining with TD-DFT calculations, suggest the strong electronic coupling between the lactone moieties and the main porphyrin chromophores and thus the importance of precisely positioning ß-substituents by mimicking chlorophylls, as an alternative to "extended π-conjugation", in designing NIR active porphyrinoids.


Asunto(s)
Biomimética/métodos , Diseño de Fármacos , Sustancias Luminiscentes/química , Metaloporfirinas/química , Modelos Moleculares , Conformación Molecular , Paladio/química , Fármacos Fotosensibilizantes/química , Platino (Metal)/química , Estereoisomerismo
17.
J Phys Chem A ; 118(39): 9105-12, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-24964272

RESUMEN

The potential energy profiles toward formation of cyclobutane pyrimidine dimers CPD and the physical quenching after UV excitation were explored for the dinucleotide thymine dinucleoside monophosphate (TpT) using density functional theory (ωB97XD) and the time-dependent density functional theory (TD-ωB97XD). The ωB97XD functional that includes empirical dispersion correction is shown to be an appropriate method to obtain rational results for the current large reaction system of TpT. Photophysical quenching is shown to be predominant over the photochemical CPD formation. Following the initial excitation to the (1)ππ* state, the underlying dark (1)nπ* state bifurcates the excited population to the prevailing IC to S0 and the small ISC to the long-lived triplet state T1 via T4 ((3)ππ*) state that has negligible energy gap with (1)nπ* state. Even for the reactive T1 state, two physical quenching pathways resulting in the conversion back to ground-state reactant via the T1/S0 crossing points are newly located, which are in strong competition with CPD formation. These results provide rationale for the recently observed nanosecond triplet decay rates in the single-stranded (dT)18 and inefficiency of deleterious CPD formation, which allow for a deeper understanding of DNA photostability.


Asunto(s)
ADN/química , Fosfatos de Dinucleósidos/química , Dímeros de Pirimidina/química , Simulación por Computador , Modelos Químicos , Procesos Fotoquímicos , Rayos Ultravioleta
18.
J Chem Phys ; 140(14): 141101, 2014 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-24735281

RESUMEN

We experimentally reconstructed the structure of the N2Ar van der Waals complex with the technique of laser-based channel-selected Coulomb explosion imaging. The internuclear distance between the N2 center of mass and the Ar atom, i.e., the length of the van der Waals bond, was determined to be 3.88 Å from the two-body explosion channels. The angle between the van der Waals bond and the N2 principal axis was determined to be 90° from the three-body explosion channels. The reconstructed structure was contrasted with our high level ab initio calculations. The agreement demonstrated the potential application of laser-based Coulomb explosion in imaging transient molecular structure, particularly for floppy van der Waals complexes, whose structures remain difficult to be determined by conventional spectroscopic methods.

19.
Photochem Photobiol ; 100(2): 368-379, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37792888

RESUMEN

The direct photoionization of DNA canonical bases under ultraviolet radiation is difficult due to the high ionization potentials. According to previous quantum chemical calculations, methylation can have great influence on the ionization potential. Are methylated nucleobases prone to photoionization and cause DNA damage? As an important epigenetic modification in transcription, expression, and regulation of genes, it is of great biological significance to explore the effect of methylation on base photoionization from the experimental perspective. Herein, we study the photoionization behavior of methylated purines 6 mA and 6mG at 266 nm using a nanosecond transient UV-Vis spectroscopy. The hydrated electron and methylated base radicals are observed, indicating the occurrence of photoionization for both 6mG and 6 mA. We measured one-photon ionization yields to be (5.0 ± 0.2) × 10-3 and (1.4 ± 0.2) × 10-3 for 6mG and 6 mA, respectively. These are higher than those of (dA)20 and (dA20 )·(dT20 ) previously reported, indicating that methylation significantly promotes base photoionization with a stronger effect than base stacking, consistent with calculations in literature. Given that the hydrated electrons and methylated base radicals from photoionization can trigger a cascade of deleterious reactions, the methylated purine bases may act as hotspots of DNA photoionization damage of living organisms.


Asunto(s)
Purinas , Rayos Ultravioleta , Purinas/química , ADN/química , Análisis Espectral
20.
J Phys Chem B ; 128(2): 576-584, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38189153

RESUMEN

Despite the increased interest of visible-light-absorbing compound Hypericin (Hyp) in photodiagnosis, photocatalysis, and photodynamic therapy (PDT) applications, a major obstacle still exists; i.e., the photoactivity is diminished due to the facile aggregation of Hyp in aqueous environment that induces excited-state quenching. Herein, we explore the excited-state property of Hyp bound to the DNA G-quadruplex by combining multiple steady-state and time-resolved spectroscopy. We find that the aggregation-induced quenching effect can be successfully prevented by appropriate G-quadruplex binders that disperse Hyp into monomer. The binding of Hyp/G-quadruplex is selective, however, exhibiting a preferential binding toward parallel G-quadruplexes (c-kit2, C14B1, STAT3, S50, and PS2.M), over antiparallel or hybrid G-quadruplex (Tel22, TBA). The excited-state property of Hyp is highly related to the binding behavior, showing a consistent trend that the better the Hyp/G-quadruplex binding, the longer the triplet 3Hyp* lifetime and the higher the efficiency to produce 1O2. For Hyp/c-kit2, the major binding mode is 5'-end stacking, which offers protection from collisional quenching reactions and ensures a stable photocycle of 3Hyp*-O2 energy transfer forming 1O2, leading to the highest 1O2 quantum yield (0.67) with superior photostability. These findings open possibilities of developing Hyp/G-quadruplex complex as a biocompatible photosensitizer for PDT applications, etc.


Asunto(s)
G-Cuádruplex , Perileno , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/química , Perileno/química , Antracenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA