Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
J Cancer Res Clin Oncol ; 150(7): 353, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012375

ABSTRACT

This article presents an in-depth exploration of the roles of Telomere Repeat-binding Factors 1 and 2 (TRF1 and TRF2), and the shelterin complex, in the context of cancer biology. It emphasizes their emerging significance as potential biomarkers and targets for therapeutic intervention. Central to the shelterin complex, TRF1 and TRF2 are crucial in maintaining telomere integrity and genomic stability, their dysregulation often being a hallmark of cancerous cells. The article delves into the diagnostic and prognostic capabilities of TRF1 and TRF2 across various cancer types, highlighting their sensitivity and specificity. Furthermore, it reviews current strides in drug discovery targeting the shelterin complex, detailing specific compounds and their modes of action. The review candidly addresses the challenges in developing therapies aimed at the shelterin complex, including drug resistance, off-target effects, and issues in drug delivery. By synthesizing recent research findings, the article sheds light on the intricate relationship between telomere biology and cancer development. It underscores the urgency for continued research to navigate the existing challenges and fully leverage the therapeutic potential of TRF1, TRF2, and the shelterin complex in the realm of cancer treatment.


Subject(s)
Neoplasms , Telomere , Telomeric Repeat Binding Protein 1 , Telomeric Repeat Binding Protein 2 , Humans , Neoplasms/genetics , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/therapy , Telomeric Repeat Binding Protein 2/metabolism , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 1/genetics , Telomere/metabolism , Molecular Targeted Therapy/methods , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Shelterin Complex , Telomere-Binding Proteins
2.
Drug Discov Today ; 29(8): 104056, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38844065

ABSTRACT

As a global health challenge, cancer prompts continuous exploration for innovative therapies that are also based on new targets. One promising avenue is targeting the shelterin protein complex, a safeguard for telomeres crucial in preventing DNA damage. The role of shelterin in modulating ataxia-telangiectasia mutated (ATM) and ataxia-telangiectasia and Rad3-related (ATR) kinases, key players in the DNA damage response (DDR), establishes its significance in cancer cells. Disrupting these defence mechanisms of shelterins, especially in cancer cells, renders telomeres vulnerable, potentially leading to genomic instability and hindering cancer cell survival. In this review, we outline recent approaches exploring shelterins as potential anticancer targets, highlighting the prospect of developing selective molecules to exploit telomere vulnerabilities toward new innovative cancer treatments.

4.
J Enzyme Inhib Med Chem ; 39(1): 2302920, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38221785

ABSTRACT

Human DNA topoisomerases are essential for crucial cellular processes, including DNA replication, transcription, chromatin condensation, and maintenance of its structure. One of the significant strategies employed in cancer treatment involves the inhibition of a specific type of topoisomerase, known as topoisomerase II (Topo II). Carbazole derivatives, recognised for their varied biological activities, have recently become a significant focus in oncological research. This study assesses the efficacy of three symmetrically substituted carbazole derivatives: 2,7-Di(2-furyl)-9H-carbazole (27a), 3,6-Di(2-furyl)-9H-carbazole (36a), and 3,6-Di(2-thienyl)-9H-carbazole (36b) - as anticancer agents. Among investigated carbazole derivatives, compound 3,6-di(2-furyl)-9H-carbazole bearing two furan moieties emerged as a novel catalytic inhibitor of Topo II. Notably, 3,6-di(2-furyl)-9H-carbazole effectively selectively inhibited the relaxation and decatenation activities of Topo IIα, with minimal effects on the IIß isoform. These findings underscore the potential of compound 3,6-Di(2-furyl)-9H-carbazole as a promising lead candidate warranting further investigation in the realm of anticancer drug development.


Subject(s)
Antineoplastic Agents , Topoisomerase II Inhibitors , Humans , Topoisomerase II Inhibitors/pharmacology , Topoisomerase II Inhibitors/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Carbazoles/pharmacology , Carbazoles/chemistry , DNA Topoisomerases, Type II , Apoptosis
5.
Chemistry ; 29(55): e202300970, 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37332024

ABSTRACT

In this work, we present studies on relatively new and still not well-explored potential anticancer targets which are shelterin proteins, in particular the TRF1 protein can be blocked by in silico designed "peptidomimetic" molecules. TRF1 interacts directly with the TIN2 protein, and this protein-protein interaction is crucial for the proper functioning of telomere, which could be blocked by our novel modified peptide molecules. Our chemotherapeutic approach is based on assumption that modulation of TRF1-TIN2 interaction may be more harmful for cancer cells as cancer telomeres are more fragile than in normal cells. We have shown in vitro within SPR experiments that our modified peptide PEP1 molecule interacts with TRF1, presumably at the site originally occupied by the TIN2 protein. Disturbance of the shelterin complex by studied molecule may not in short term lead to cytotoxic effects, however blocking TRF1-TIN2 resulted in cellular senescence in cellular breast cancer lines used as a cancer model. Thus, our compounds appeared useful as starting model compounds for precise blockage of TRF proteins.


Subject(s)
Shelterin Complex , Telomeric Repeat Binding Protein 2 , Telomeric Repeat Binding Protein 1/chemistry , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomere/metabolism , Peptides/pharmacology
6.
Antioxidants (Basel) ; 12(5)2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37237882

ABSTRACT

Mangiferin is a strong antioxidant that presents a wide range of biological activities. The aim of this study was to evaluate, for the first time, the influence of mangiferin on tyrosinase, an enzyme responsible for melanin synthesis and the unwanted browning process of food. The research included both the kinetics and molecular interactions between tyrosinase and mangiferin. The research proved that mangiferin inhibits tyrosinase activity in a dose-dependent manner with IC50 290 +/- 6.04 µM, which was found comparable with the standard kojic acid (IC50 217.45 +/- 2.54 µM). The mechanism of inhibition was described as mixed inhibition. The interaction between tyrosinase enzyme and mangiferin was confirmed with capillary electrophoresis (CE). The analysis indicated the formation of two main, and four less significant complexes. These results have also been supported by the molecular docking studies. It was indicated that mangiferin binds to tyrosinase, similarly to L-DOPA molecule, both in the active center and peripheral site. As it was presented in molecular docking studies, mangiferin and L-DOPA molecules can interact in a similar way with surrounding amino acid residues of tyrosinase. Additionally, hydroxyl groups of mangiferin may interact with amino acids on the tyrosinase external surface causing non-specific interaction.

7.
J Cancer Res Clin Oncol ; 149(10): 8131-8141, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37031434

ABSTRACT

The mechanisms of antigen processing and presentation play a crucial role in the recognition and targeting of cancer cells by the immune system. Cancer cells can evade the immune system by downregulating or losing the expression of the proteins recognized by the immune cells as antigens, creating an immunosuppressive microenvironment, and altering their ability to process and present antigens. This review focuses on the mechanisms of cancer immune evasion with a specific emphasis on the role of antigen presentation machinery. The study of the immunopeptidome, or peptidomics, has provided insights into the mechanisms of cancer immune evasion and has potential applications in cancer diagnosis and treatment. Additionally, manipulating the epigenetic landscape of cancer cells plays a critical role in suppressing the immune response against cancer. Targeting these mechanisms through the use of HDACis, DNMTis, and combination therapies has the potential to improve the efficacy of cancer immunotherapy. However, further research is needed to fully understand the mechanisms of action and optimal use of these therapies in the clinical setting.


Subject(s)
Antigen Presentation , Neoplasms , Humans , Neoplasms/therapy , Immunotherapy , Tumor Microenvironment
8.
ACS Nano ; 16(12): 20577-20588, 2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36475617

ABSTRACT

Blue phase liquid crystals (BPLCs) are chiral mesophases with 3D order, which makes them a promising template for doping nanoparticles (NPs), yielding tunable nanomaterials attractive for microlasers and numerous microsensor applications. However, doping NPs to BPLCs causes BP lattice extension, which translates to elongation of operating wavelengths of light reflection. Here, it is demonstrated that small (2.4 nm diameter) achiral gold (Au) NPs decorated with designed LC-like ligands can enhance the chiral twist of BPLCs (i.e., reduce cell size of the single BP unit up to ∼14% and ∼7% for BPI and BPII, respectively), translating to a blue-shift of Bragg reflection. Doping NPs also significantly increases the thermal stability of BPs from 5.5 °C (for undoped BPLC) up to 22.8 °C (for doped BPLC). In line with our expectations, both effects are saturated, and their magnitude depends on the concentration of investigated nanodopants as well the BP phase type. Our research highlights the critical role of functionalization of Au NPs on the phase sequence of BPLCs. We show that inappropriate selection of surface ligands can destabilize BPs. Our BPLC and Au NPs are photochemically stable and exhibit great miscibility, preventing NP aggregation in the BPLC matrix over the long term. We believe that our findings will improve the fabrication of advanced nanomaterials into 3D periodic soft photonic structures.

9.
ACS Nano ; 16(11): 18472-18482, 2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36342742

ABSTRACT

Circularly polarized luminescent (CPL) films with high dissymmetry factors hold great potential for optoelectronic applications. Herein, we propose a strategy for achieving strongly dissymetric CPL in nanocomposite films based on chirality induction and energy transfer to semiconductor nanocrystals. First, focusing on a purely organic system, aggregation-induced emission (AIE) and CPL activity of organic liquid crystals (LCs) forming helical nanofilaments was detected, featuring green emission with high dissymmetry factors glum ∼ 10-2. The handedness of helical filaments, and thus the sign of CPL, was controlled via minute amounts of a small chiral organic dopant. Second, nanocomposite films were fabricated by incorporating InP/ZnS semiconductor quantum dots (QDs) into the LC matrix, which induced the chiral assembly of QDs and endowed them with chiroptical properties. Due to the spectral matching of the components, energy transfer (ET) from LC to QDs was possible enabling a convenient way of tuning CPL wavelengths by varying the LC/QD ratio. As obtained, composite films exhibited absolute glum values up to ∼10-2 and thermally on/off switchable luminescence. Overall, we demonstrate the induction of chiroptical properties by the assembly of nonchiral building QDs on the chiral organic template and energy transfer from organic films to QDs, representing a simple and versatile approach to tune the CPL activity of organic materials.

10.
Cell Death Dis ; 13(11): 1005, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36437244

ABSTRACT

Telomerase reactivation is one of the hallmarks of cancer, which plays an important role in cellular immortalization and the development and progression of the tumor. Chemical telomerase inhibitors have been shown to trigger replicative senescence and apoptotic cell death both in vitro and in vivo. Due to its upregulation in various cancers, telomerase is considered a potential target in cancer therapy. In this study, we identified potent, small-molecule telomerase inhibitors using a telomerase repeat amplification protocol assay. The results of the assay are the first evidence of telomerase inhibition by anthraquinone derivatives that do not exhibit G-quadruplex-stabilizing properties. The stability of telomerase in the presence of its inhibitor was evaluated under nearly physiological conditions using a cellular thermal shift assay. Our data showed that the compound induced aggregation of the catalytic subunit (hTERT) of human telomerase, and molecular studies confirmed the binding of the hit compound with the active site of the enzyme. The ability of new derivatives to activate DNA double-strand breaks (DSBs) was determined by high-resolution microscopy and flow cytometry in tumor cell lines differing in telomere elongation mechanism. The compounds triggered DSBs in TERT-positive A549 and H460 lung cancer cell lines, but not in TERT-negative NHBE normal human bronchial epithelial and ALT-positive U2OS osteosarcoma cell lines, which indicates that the induction of DSBs was dependent on telomerase inhibition. The observed DNA damage activated DNA damage response pathways involving ATM/Chk2 and ATR/Chk1 cascades. Additionally, the compounds induced apoptotic cell death through extrinsic and intrinsic pathways in lung cancer cells. Taken together, our study demonstrated that anthraquinone derivatives can be further developed into novel telomerase-related anticancer agents.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Telomerase , Humans , Telomere , DNA Damage , Anthraquinones/pharmacology , Cell Line, Tumor
11.
Biophys Chem ; 291: 106909, 2022 12.
Article in English | MEDLINE | ID: mdl-36330853

ABSTRACT

Low solubility of reactants or products in aqueous solutions can result in the enzymatic catalytic reactions that can occur in non-aqueous solutions. In current study we investigated aqueous solutions containing different organic solvents / deep eutectic solvents (DESs) that can influence the protease enzyme's activity, structural, and thermal stabilities. Retroviral aspartic protease enzyme is responsible for the cleavage of the polypeptide precursors into mature viral components, a very crucial step for virus life cycle. In molecular dynamic simulations (MDS), the complex of the protease enzyme with Darunavirwas found highly stable in urea aqueous solution compared to when with the ethylene glycol (EG) or glycerol solvents. Particularly, in different organic solvents the presence of Darunavir induced protein-protein interactions within the protease homodimer. For the systems with EG or glycerol solvents, the flap domains of the enzyme formed an "open" conformation which lead to a weak binding affinity with the drug. Conserved D25 and G27 residues among this family of the aspartic protease enzymes made a stable binding with Darunavir in the urea systems. Unfolding of the protease dimer was initiated due to self-aggregation for the EG or glycerol organic solvents, which formed an "open" conformation for the flap domains. On the contrary lack of such clustering in urea solvent, the protease showed conventional structural folding in the presence or absence of the drug molecule. These novel findings may help to better understand the protease enzymes, which could be controlled by deep eutectic solvents.


Subject(s)
Ethylene Glycol , Glycerol , Darunavir , Solvents/chemistry , Urea/pharmacology , Urea/chemistry , Peptide Hydrolases
12.
Article in English | MEDLINE | ID: mdl-36305423

ABSTRACT

The development of plasmonic nanomaterials with chiral geometry has drawn extensive attention owing to their practical implications in chiral catalysis, chiral metamaterials, or enantioselective biosensing and medicine. However, due to the lack of effective synthesis methods of hydrophobic nanoparticles (NPs) showing intrinsic, plasmonic chirality, their applications are currently limited to aqueous systems. In this work, we resolve the problem of achieving hydrophobic Au NPs with intrinsic chirality by efficient phase transfer of water-soluble NPs using low molecular weight, liquid crystal-like ligands. We confirmed that, after the phase transfer, Au NPs preserve strong, far-field circular dichroism (CD) signals, attesting their chiral geometry. The universality of the method is exemplified by using different types of NPs and ligands. We further highlight the potential of the proposed approach to realize chiral plasmonic, inorganic/organic nanocomposites with block copolymers, liquid crystals, and compounds forming physical gels. All soft matter composites sustain plasmonic CD signals with electron microscopies confirming well-dispersed nanoinclusions. The developed methodology allows us to expand the portfolio of plasmonic NPs with intrinsic structural chirality, thereby broadening the scope of their applications toward soft-matter based systems.

13.
J Cell Mol Med ; 26(14): 3950-3964, 2022 07.
Article in English | MEDLINE | ID: mdl-35701366

ABSTRACT

The acridanone derivative 5-dimethylaminopropylamino-8-hydroxytriazoloacridinone (C-1305) has been described as a potent inhibitor of cancer cell growth. Its mechanism of action in in vitro conditions was attributed, among others, to its ability to bind and stabilize the microtubule network and subsequently exhibit its tumour-suppressive effects in synergy with paclitaxel (PTX). Therefore, the objective of the present study was to analyse the effects of the combined treatment of C-1305 and PTX in vivo. In addition, considering the results of previous genomic analyses, particular attention was given to the effects of this treatment on tumour angiogenesis. Treatment with C-1305 revealed antitumor effect in A549 lung cancer cells, and combined treatment with PTX showed tendency to anticancer activity in HCT116 colon cancer xenografts. It also improved tumour blood perfusion in both tumour models. The plasma level of CCL2 was increased and that of PDGF was decreased after combined treatment with C-1305 and PTX. The experimental results showed that the levels of FGF1, TGF-ß and Ang-4 decreased, whereas the levels of ERK1/2 and Akt phosphorylation increased in HCT116 tumour tissue following combined treatment with both drugs. The results of in vitro capillary-like structure formation assay demonstrated the inhibiting effect of C-1305 on this process. Although previous in vitro and in vivo studies suggested a positive effect of C-1305 on cancer cells, combined treatment of HCT116 human colon and A549 lung cancer cells with both PTX and C-1305 in vivo showed that the antitumor activity was restricted and associated with the modulation of tumour angiogenesis.


Subject(s)
Colonic Neoplasms , Lung Neoplasms , Acridines , Apoptosis , Colonic Neoplasms/drug therapy , Heterografts , Humans , Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Triazoles
14.
Chem Commun (Camb) ; 58(53): 7364-7367, 2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35621065

ABSTRACT

Liquid crystalline (LC) dimers formed helical nanofilaments depending on the parity of the alkyl linker, revealing an unusual odd-even effect. Molecular dynamics simulations were used to investigate the observed tendency. Elongation of the linker translates to an increase of the pitch of the helices, which allows achieving tuneable helical assemblies of Au nanoparticles doped to the LC matrix. The impact of the tuneable pitch of helices on the chiral optical properties of composites was investigated with full-wave simulations based on the T-matrix method.


Subject(s)
Liquid Crystals , Metal Nanoparticles , Gold , Liquid Crystals/chemistry , Molecular Dynamics Simulation , Polymers/chemistry
15.
Int J Mol Sci ; 23(9)2022 May 05.
Article in English | MEDLINE | ID: mdl-35563554

ABSTRACT

The molecular mechanisms of telomerase reverse transcriptase (TERT) upregulation in breast cancer (BC) are complex. We compared genetic variability within TERT and telomere length with the clinical data of patients with BC. Additionally, we assessed the expression of the TERT, MYC, TP53 and SP1 genes in BC patients and in BC organoids (3D cell cultures obtained from breast cancer tissues). We observed the same correlation in the blood of BC patients and in BC organoids between the expression of TERT and TP53. Only in BC patients was a correlation found between the expression of the TERT and MYC genes and between TP53 and MYC. We found associations between TERT genotypes (rs2735940 and rs10069690) and TP53 expression and telomere length. BC patients with the TT genotype rs2735940 have a shorter telomere length, but patients with A allele rs10069690 have a longer telomere length. BC patients with a short allele VNTR-MNS16A showed higher expression of the SP1 and had a longer telomere. Our results bring new insight into the regulation of TERT, MYC, TP53 and SP1 gene expression related to TERT genetic variability and telomere length. Our study also showed for the first time a similar relationship in the expression of the above genes in BC patients and in BC organoids. These findings suggest that TERT genetic variability, expression and telomere length might be useful biomarkers for BC, but their prognostic value may vary depending on the clinical parameters of BC patients and tumor aggressiveness.


Subject(s)
Breast Neoplasms , Telomerase , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Genes, myc , Humans , Polymorphism, Single Nucleotide , Sp1 Transcription Factor/genetics , Telomerase/genetics , Telomere/pathology , Tumor Suppressor Protein p53/genetics
16.
Eur J Med Chem ; 238: 114453, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35609396

ABSTRACT

Mitochondrial targeting plays an important role in anticancer therapy. The Mn(III)-promoted cyclization of 5-(1H-indol-3-yl)-3-oxopentanoic acid allow to obtain novel substituted carbazole derivatives that can act as mitochondria-disruptive agents. The starting materials used for the synthesis of these new aminocarbazoles are oxopentanoate derivatives of tryptophan. The scope and limitation of this method of synthesis are determined by a series of experiments. The prepared carbazole derivatives are screened for their in vitro anticancer activity against a broad panel of human cancer cells and normal cell lines. Among the tested compounds, the most active ones are examined further against human colon cancer cells (HCT-116) and human bone osteosarcoma (U-2 OS), in complex in vitro cellular assays, including studies on cell cycle distribution, intracellular compartmentalization, antimigratory properties, mitochondrial generation of reactive oxygen species, DNA damage, and type of cellular death. The results reveal that the synthesized compounds display potent oxidative activity inducing massive accumulation of DNA double-strand breaks, which lead to a parallel change in the assembly of mitochondria causing their dysfunction. These findings provide new leads for the treatment of colon cancer and osteosarcoma.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Colonic Neoplasms , Osteosarcoma , Apoptosis , Bone Neoplasms/metabolism , Carbazoles , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Humans , Mitochondria/metabolism , Osteosarcoma/metabolism , Reactive Oxygen Species/metabolism , Tryptophan/pharmacology
17.
Sci Rep ; 12(1): 3703, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260633

ABSTRACT

Lung cancer is considered to account for approximately one-fifth of all malignant tumor-related deaths worldwide and is therefore one of the most lethal malignancies. Pyrazole scaffold possesses a wide range of biological and pharmacological activities, which play important roles in medicinal chemistry. The present study reports the synthesis and in vitro biological characterization of nine pyrazoles derived from chalcones as potential anticancer agents for non-small cell lung cancer A-549, H226, and H460 cell lines. Most of the compounds efficiently inhibited the growth of all the tested cancer cell lines at micromolar concentrations. One of the most active compounds (PCH-1) was further evaluated for its effect on cell cycle distribution, apoptosis, migration, epithelial-mesenchymal transition, and oxidative stress. Furthermore, studies on the mechanism of action revealed that PCH-1 disrupts microtubule assembly, leading to cancer cell death. Molecular modeling studies confirmed the potent interaction of PCH-1 with the vinblastine binding site on tubulin. Overall, this study provides novel opportunities to identify anticancer agents in the pyrazole series.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Chalcone , Chalcones , Lung Neoplasms , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation , Chalcone/pharmacology , Chalcones/pharmacology , Drug Screening Assays, Antitumor , Humans , Lung Neoplasms/drug therapy , Pyrazoles/therapeutic use , Structure-Activity Relationship , Tubulin/metabolism
18.
PLoS Comput Biol ; 17(10): e1009454, 2021 10.
Article in English | MEDLINE | ID: mdl-34613958

ABSTRACT

The current surge in bacterial multi-drug resistance (MDR) is one of the largest challenges to public health, threatening to render ineffective many therapies we rely on for treatment of serious infections. Understanding different factors that contribute to MDR is hence crucial from the global "one health" perspective. In this contribution, we focus on the prototypical broad-selectivity proton-coupled antiporter EmrE, one of the smallest known ligand transporters that confers resistance to aromatic cations in a number of clinically relevant species. As an asymmetric homodimer undergoing an "alternating access" protomer-swap conformational change, it serves as a model for the mechanistic understanding of more complex drug transporters. Here, we present a free energy and solvent accessibility analysis that indicates the presence of two complementary ligand translocation pathways that remain operative in a broad range of conditions. Our simulations show a previously undescribed desolvated apo state and anticorrelated accessibility in the ligand-bound state, explaining on a structural level why EmrE does not disrupt the pH gradient through futile proton transfer. By comparing the behavior of a number of model charged and/or aromatic ligands, we also explain the origin of selectivity of EmrE towards a broad class of aromatic cations. Finally, we explore unbiased pathways of ligand entry and exit to identify correlated structural changes implicated in ligand binding and release, as well as characterize key intermediates of occupancy changes.


Subject(s)
Antiporters , Escherichia coli Proteins , Ion Transport/physiology , Antiporters/chemistry , Antiporters/genetics , Antiporters/metabolism , Computational Biology , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Ligands , Molecular Dynamics Simulation , Protons , Thermodynamics
19.
Nanomaterials (Basel) ; 11(9)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34578613

ABSTRACT

Self-assembly of metal nanoparticles has applications in the fabrication of optically active materials. Here, we introduce a facile strategy for the fabrication of films of binary nanoparticle assemblies. Dynamic control over the configuration of gold nanorods and nanospheres is achieved via the melting of bound and unbound fractions of liquid-crystal-like nanoparticle ligands. This approach provides a route for the preparation of hierarchical nanoparticle superstructures with applications in reversibly switchable, visible-range plasmonic technologies.

20.
Int J Mol Sci ; 22(9)2021 May 06.
Article in English | MEDLINE | ID: mdl-34066491

ABSTRACT

Poor efficiency of chemotherapeutics in the eradication of Cancer Stem Cells (CSCs) has been driving the search for more active and specific compounds. In this work, we show how cell density-dependent stage culture profiles can be used in drug development workflows to achieve more robust drug activity (IC50 and EC50) results. Using flow cytometry and light microscopy, we characterized the cytological stage profiles of the HL-60-, A-549-, and HEK-293-derived sublines with a focus on their primitive cell content. We then used a range of cytotoxic substances-C-123, bortezomib, idarubicin, C-1305, doxorubicin, DMSO, and ethanol-to highlight typical density-related issues accompanying drug activity determination. We also showed that drug EC50 and selectivity indices normalized to primitive cell content are more accurate activity measurements. We tested our approach by calculating the corrected selectivity index of a novel chemotherapeutic candidate, C-123. Overall, our study highlights the usefulness of accounting for primitive cell fractions in the assessment of drug efficiency.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Count , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Humans , Inhibitory Concentration 50 , Neoplasm Staging , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...