Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Analyst ; 148(3): 594-608, 2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36594590

RESUMO

The current study shows that Schiff base HL, (Z)-2,4-dibromo-6-(((piperidin-2-ylmethyl)imino)methyl)phenol, can be used successfully as a selective chemosensor for Zn(II) and Ni(II) among several competing cations in purely aqueous and semi-aqueous media. Under UV light in methanol-water (9 : 1) HEPES buffer, the receptor gives its response by changing its color to cyan color in the presence of Zn(II) and to bluish cyan color in the presence of Ni(II). Surprisingly, the chemosensor can only reliably identify Zn(II) in a hundred percent aqueous medium by changing its color to light yellow. UV and fluorescence studies in both aqueous and semi-aqueous media are used to further investigate this Zn(II) and Ni(II) recognition phenomenon. The high values of the host-guest binding constants, obtained by electronic and fluorescence titration, ensure that a strong bond exists between HL and Ni(II)/Zn(II). As anticipated, two highly luminescent mononuclear, crystalline compounds, complexes 1 and 2, have been developed by a separate reaction of HL and Zn(II)/Ni(II), and the high luminous properties are due to the occurrence of Chelation Enhanced Fluorescence (CHEF). According to the single crystal structure, the asymmetric units of both complexes consist of two deprotonated chemosensor units and one Zn(II)/Ni(II), leading to the formation of an octahedral complex. For Ni(II) and Zn(II) sensing, the predicted LOD is in the nanomolar range. Both complexes 1 and 2 are fluorescence active and studies to check their ATP detection ability, but intriguingly, only complex 2 is capable of detecting ATP in a fully aqueous solution. Finally, the live cell imaging study validates the two sensors' biosensing functionality.

2.
J Nanobiotechnology ; 21(1): 28, 2023 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-36694259

RESUMO

Hepatocellular carcinoma (HCC) is a leading cause of death globally. Even though the progressive invention of some very potent therapeutics has been seen, the success is limited due to the chemotherapeutic resistance and recurrence in HCC. Advanced targeted treatment options like immunotherapy, molecular therapy or surface-engineered nanotherapeutics could offer the benefits here owing to drug resistance over tumor heterogenicity. We have developed tumor-sensing phosphorothioate and amino-modified aptamer (AS1411)-conjugated stealth nanoliposomes, encapsulating with apigenin for precise and significant biodistribution of apigenin into the target tumor to exploit maximum bio-therapeutic assistances. The stable aptamer functionalized PEGylated nanoliposomes (Apt-NLCs) had an average vesicle size of 100-150 nm, a smooth surface, and an intact lamellarity, as ensured by DLS, FESEM, AFM, and Cryo-TEM. This study has specified in vitro process of optimum drug (apigenin) extrusion into the cancer cells by nucleolin receptor-mediated cellular internalization when delivered through modified AS1411 functionalized PEGylated nanoliposomes and ensured irreversible DNA damage in HCC. Significant improvement in cancer cell apoptosis in animal models, due to reduced clearance and higher intratumor drug accumulation along with almost nominal toxic effect in liver, strongly supports the therapeutic potential of aptamer-conjugated PEGylated nanoliposomes compared to the nonconjugated formulations in HCC. The study has established a robust superiority of modified AS1411 functionalized PEGylated nanoliposomes as an alternative drug delivery approach with momentous reduction of HCC tumor incidences.


Assuntos
Aptâmeros de Nucleotídeos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Ratos , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Apigenina/farmacologia , Apigenina/uso terapêutico , Distribuição Tecidual , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Sistemas de Liberação de Medicamentos , Oligodesoxirribonucleotídeos , Polietilenoglicóis/uso terapêutico , Linhagem Celular Tumoral
3.
Int J Biol Macromol ; 275(Pt 2): 133716, 2024 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-38977049

RESUMO

In the present study, one mononuclear Cu(II) [CuL(SCN)] (1) and one mononuclear Co(II) [CoLN3] (2) complexes, with a Schiff base ligand (HL) formed by condensation of 2-picolylamine and salicylaldehyde, have been successfully developed and structurally characterized. The square planer geometry of both complexes is fulfilled by the coordination of one deprotonated ligand and one ancillary ligand SCN-(1) or N3-(2) to the metal centre. Binding affinities of both complexes with deoxyribonucleic acid (DNA) and human serum albumin (HSA) are investigated using several biophysical and spectroscopic techniques. High values of the macromolecule-complex binding constants and other results confirm the effectiveness of both complexes towards binding with DNA and HSA. The determined values of the thermodynamic parameters support spontaneous interactions of both complexes with HSA, while fluorescence displacement and DNA melting studies establish groove-binding interactions with DNA for both complexes 1 and 2. The molecular modelling study validates the experimental findings. Both complexes are subjected to an MTT test establishing the anticancer property of complex 1 with lower risk to normal cells, confirmed by the IC50 values of the complex for HeLa cancer cells and HEK normal cells. Finally, a nuclear staining analysis reveals that the complexes have caused apoptotic cell death.

4.
ACS Omega ; 8(1): 636-647, 2023 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-36643564

RESUMO

A new quercetin-based iron(III) cationic complex [Fe(Qr)Cl(H2O)(MeO)] (complex 1) is created in the current study by condensation of quercetin with ferric chloride in the presence of Et3N. Comprehensive spectroscopic analysis and conductometric measurement are used to pinpoint complex 1. The generated complex's +3-oxidation state has been verified by electron paramagnetic resonance (EPR) research. Density functional theory analysis was used to structurally optimize the structure of complex 1. Before biomedical use, a variety of biophysical studies are implemented to evaluate the binding capacity of complex 1 with DNA and human serum albumin (HSA) protein. The findings of the electronic titration between complex 1 and DNA, as well as the stunning fall in the fluorescence intensities of the HSA and EtBr-DNA/DAPI-DNA domain after complex 1 is gradually added, give us confidence that complex 1 has a strong affinity for both macromolecules. It is interesting to note that the displacement experiment confirms partial intercalation as well as the groove binding mechanism of the title complex with DNA. The time-dependent fluorescence analysis indicates that after interaction with complex 1, HSA will exhibit static quenching. The thermodynamic parameter values in the HSA-complex 1 interaction provide evidence for the hydrophobicity-induced pathway leading to spontaneous protein-complex 1 interaction. The two macromolecules' configurations are verified to be preserved when they are associated with complex 1, and this is done via circular dichroism spectral titration. The molecular docking investigation, which is a theoretical experiment, provides complete support for the experimental findings. The potential of the investigated complex to be an anticancer drug has been examined by employing the MTT assay technique, which is carried out on HeLa cancer cell lines and HEK-293 normal cell lines. The MTT assay results validate the ability of complex 1 to display significant anticancer properties. Finally, by using the AO/PI staining approach, the apoptotic-induced cell-killing mechanism as well as the detection of cell morphological changes has been confirmed.

5.
Heliyon ; 9(12): e22712, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38125469

RESUMO

In the current study, one new quercetin-based Zn(II) complex [Zn(Qr)(CNNCN)(H2O)2] (Complex 1) which is developed by condensation of quercetin with ZnCl2 in the presence of NaN(CN)2 and Cu(II) complex [Cu(Qr)N3(CH3OH)(H2O)] (complex 2) which is developed by the condensation reaction of quercetin and CuCl2 in presence of NaN3, are thoroughly examined in relation to their use in biomedicine. The results of several spectroscopic studied confirm the structure of both the complexes and the Density Functional Theory (DFT) study helps to optimize the structure of complex 1 and 2. After completion of the identification process, DNA and Human Serum Albumin (HSA) binding efficacy of both the investigated complexes are performed by implementing a long range of biophysical studies and a thorough analysis of the results unveils that complex 1 has better interaction efficacy with the macromolecules than complex 2. The binding efficacy of complex 1 is comparatively higher towards both macromolecules because of its pure groove binding mode during interaction with DNA and the presence of an extra H-bond during connection with HSA. The experimental host-guest binding results is fully validated by molecular docking study. Interestingly complex 1 shows better antioxidant properties than complex 2, as well as quercetin, and it has strong anticancer property with minimal damage to normal cells, which is proved by the MTT assay study. Better DNA and HSA binding efficacy of 1 may be the reason for the better anticancer property of complex 1.

6.
J Biomol Struct Dyn ; 40(24): 14188-14203, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34842505

RESUMO

In the present work we have developed one mononuclear Zn(II) complex [Zn(L)(H2O)] (Complex 1) by utilizing a tetracoordinated ligand H2L, formed by simple condensation of 2, 2 dimethyl 1,3 diamino propane and 3- ethoxy salicylaldehyde and one newly designed mononuclear Co (III) complex [Co(L)(L1)] (complex 2) by utilizing (H2L) and 3- ethoxy salicylaldehyde(HL1) as an ancillary ligand. The newly developed complex 2 have been spectroscopically characterized. An interesting phenomenon has been noticed that in presence of ancillary ligand, the solubility in buffer solution and the thermal stability of complex 2 comparatively increases than 1. To check the effect of ancillary ligand, present in complex 2 towards the DNA and HSA binding efficacy, both the complexes have been taken into consideration to inspect their binding potentiality with the macromolecules. The 'on', 'off' fluorescence changes in presence of DNA and HSA, the binding constant values, obtained from electronic spectral titration, iodide induced quenching, competitive binding assay, circular dichroism (CD) spectral titration, time resolved fluorescence experiment unambiguously assure the better binding efficacy of complex 2 with the signal of minor groove binding mode with DNA along with no significant conformational changes of the macromolecules. The strong and spontaneous binding of complex 2 with CT-DNA is further supported by the Isothermal Titration Calorimetry (ITC) study. Furthermore TDDFT calculation of DNA with and without complex 2 significantly authorize the formation of complex 2-DNA adduct during the association. Finally Molecular Docking study properly verifies the experimental findings and provides justified explanation behinds experimental findings.


Assuntos
DNA , Zinco , Simulação de Acoplamento Molecular , Espectrometria de Fluorescência , Ligantes , DNA/química , Dicroísmo Circular
7.
ACS Omega ; 6(5): 3659-3674, 2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33585746

RESUMO

Two fluorescence active bromoaniline-based Schiff base chemosensors, namely, (E)-4-bromo-2-(((4-bromophenyl)imino)methyl)phenol (HL1 ) and (E)-2-(((4-bromophenyl)imino)methyl)phenol (HL2 ), have been employed for the selective and notable detection of Cu2+ and Zn2+ ions, respectively, with the simultaneous formation of two new metal complexes [Cu(L1)2] (1) and [Zn(L2)2] (2). X-ray single crystal analyses indicate that complexes 1 and 2 are tetra-coordinated systems with substantial CH...π/π...π stacking interactions in the solid-state crystal structures. These two complexes are exploited for the next step detection of Al3+ and Hg2+ where complex 2 exhibits impressive results via turn-off fluorescence quenching in (DMSO/H2O) HEPES buffer medium. The sensing phenomena are optimized by UV-vis spectral analyses as well as theoretical calculations (density functional theory and time-dependent density functional theory). The combined detection phenomena of the ligand (HL2 ) and complex 2 are exclusively utilized for the first time to construct a molecular memory device, intensifying their multisensoric properties. Furthermore, the DNA- and human serum albumin (HSA)-binding efficacies of these two complexes are examined by adopting electronic and fluorometric titration methods. Complex 2 shows a higher DNA-binding ability in comparison with complex 1, whereas in the case of HSA, the reverse situation is observed. Finally, the binding modes of both the complexes with DNA and HSA have been investigated through molecular docking studies, suggesting good agreement with the experimental results.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA