Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 341
Filtrar
1.
Acta Pharmacol Sin ; 43(1): 229-239, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33767381

RESUMO

Androgen receptor (AR), a ligand-activated transcription factor, is a master regulator in the development and progress of prostate cancer (PCa). A major challenge for the clinically used AR antagonists is the rapid emergence of resistance induced by the mutations at AR ligand binding domain (LBD), and therefore the discovery of novel anti-AR therapeutics that can combat mutation-induced resistance is quite demanding. Therein, blocking the interaction between AR and DNA represents an innovative strategy. However, the hits confirmed targeting on it so far are all structurally based on a sole chemical scaffold. In this study, an integrated docking-based virtual screening (VS) strategy based on the crystal structure of the DNA binding domain (DBD) of AR was conducted to search for novel AR antagonists with new scaffolds and 2-(2-butyl-1,3-dioxoisoindoline-5-carboxamido)-4,5-dimethoxybenzoicacid (Cpd39) was identified as a potential hit, which was competent to block the binding of AR DBD to DNA and showed decent potency against AR transcriptional activity. Furthermore, Cpd39 was safe and capable of effectively inhibiting the proliferation of PCa cell lines (i.e., LNCaP, PC3, DU145, and 22RV1) and reducing the expression of the genes regulated by not only the full-length AR but also the splice variant AR-V7. The novel AR DBD-ARE blocker Cpd39 could serve as a starting point for the development of new therapeutics for castration-resistant PCa.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , DNA/antagonistas & inibidores , Descoberta de Drogas , Simulação de Acoplamento Molecular , Receptores Androgênicos/metabolismo , Antagonistas de Receptores de Andrógenos/química , Sítios de Ligação/efeitos dos fármacos , DNA/química , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Estrutura Molecular , Receptores Androgênicos/química , Relação Estrutura-Atividade
2.
ChemMedChem ; 16(23): 3576-3587, 2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34524728

RESUMO

The NRF2 transcription factor is a key regulator in cellular oxidative stress response, and acts as a tumor suppressor. Aberrant activation of NRF2 has been implicated in promoting chemo-resistance, tumor growth, and metastasis by activating its downstream target genes. Hence, inhibition of NRF2 promises to be an attractive therapeutic strategy to suppress cell proliferation and enhance cell apoptosis in cancer. Direct targeting of NRF2 with small-molecules to discover protein-DNA interaction inhibitors is challenging as it is a largely intrinsically disordered protein. To discover molecules that bind to NRF2 at the DNA binding interface, we performed an NMR-based fragment screen against its DNA-binding domain. We discovered several weakly binding fragment hits that bind to a region overlapping with the DNA binding site. Using SAR by catalogue we developed an initial structure-activity relationship for the most interesting initial hit series. By combining NMR chemical shift perturbations and data-driven docking, binding poses which agreed with NMR information and the observed SAR were elucidated. The herein discovered NRF2 hits and proposed binding modes form the basis for future structure-based optimization campaigns on this important but to date 'undrugged' cancer driver.


Assuntos
DNA/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Ligação Proteica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Sítios de Ligação , DNA/metabolismo , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Fator 2 Relacionado a NF-E2/química , Fator 2 Relacionado a NF-E2/metabolismo , Ressonância Magnética Nuclear Biomolecular , Domínios Proteicos , Relação Estrutura-Atividade
3.
Expert Opin Drug Metab Toxicol ; 16(3): 195-207, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32067518

RESUMO

Introduction: The worldwide incidence of central nervous system (CNS) primary tumors is increasing. Most of the chemotherapeutic agents used for treating these cancer types induce DNA damage, and their activity is affected by the functional status of repair systems involved in the detection or correction of DNA lesions. Unfortunately, treatment of malignant high-grade tumors is still an unmet medical need.Areas covered: We summarize the action mechanisms of the main DNA inhibitors used for the treatment of brain tumors. In addition, studies on new agents or drug combinations investigated for this indication are reviewed, focusing our attention on clinical trials that in the last 3 years have been completed, terminated or are still recruiting patients.Expert opinion: Much still needs to be done to render aggressive CNS tumors curable or at least to transform them from lethal to chronic diseases, as it is possible for other cancer types. Drugs with improved penetration in the CNS, toxicity profile, and activity against primary and recurrent tumors are eagerly needed. Targeted agents with innovative mechanisms of action and ability to harness the cells of the tumor microenvironment against cancer cells represent a promising approach for improving the clinical outcome of CNS tumors.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Dano ao DNA/efeitos dos fármacos , Inibidores da Topoisomerase/administração & dosagem , Animais , Antimetabólitos Antineoplásicos/metabolismo , Antineoplásicos/metabolismo , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/metabolismo , DNA/antagonistas & inibidores , DNA/metabolismo , Dano ao DNA/fisiologia , Humanos , Inibidores da Topoisomerase/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/fisiologia
4.
Molecules ; 25(5)2020 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-32106590

RESUMO

3,6-Diethynyl-9,10-diethoxyphenanthrene (4) was synthesized from phenanthrene and employed in the synthesis of the binuclear gold(I) alkynyl complexes (R3P)Au(C≡C-3-[C14H6-9,10-diethoxy]-6-C≡C)Au(PR3) (R = Ph (5a), Cy (5b)). The diyne 4 and complexes 5a and 5b were characterized by NMR spectroscopy, mass spectrometry, and elemental analysis. UV-Vis spectroscopy studies of the metal complexes and precursor diyne show strong p à p* transitions in the near UV region that red shift by ca. 50 nm upon coordination at the gold centers. The emission spectrum of 4 shows an intense fluorescence band centered at 420 nm which red shifts, slightly upon coordination of 4 to gold. Binding studies of 4, 5a, and 5b against calf thymus DNA were carried out, revealing that 4, 5a, and 5b have >40% stronger binding affinities than the commonly used intercalating agent ethidium bromide. The molecular docking scores of 4, 5a, and 5b with B-DNA suggest a similar trend in behavior to that observed in the DNA-binding study. Unlike the ligand 4, promising anticancer properties for 5a and 5b were observed against several cell lines; the DNA binding capability of the precursor alkyne was maintained, and its anticancer efficacy enhanced by the gold centers. Such phenanthrenyl complexes could be promising candidates in certain biological applications because the two components (phenanthrenyl bridge and metal centers) can be altered independently to improve the targeting of the complex, as well as the biological and physicochemical properties.


Assuntos
Antineoplásicos/química , Complexos de Coordenação/farmacologia , DNA/química , Neoplasias/tratamento farmacológico , Alcinos/síntese química , Alcinos/química , Alcinos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação/síntese química , Complexos de Coordenação/química , DNA/antagonistas & inibidores , Ensaios de Seleção de Medicamentos Antitumorais , Etídio/química , Ouro/química , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Simulação de Acoplamento Molecular , Neoplasias/química
5.
ACS Appl Mater Interfaces ; 11(50): 46604-46613, 2019 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-31763811

RESUMO

Antisense oligonucleotide (ASO)-induced cellular signaling pathway alteration is evolving as a promising therapeutic strategy for improving antitumor chemotherapy. However, the inherent instability and inefficiency of ASO delivery remain major hurdles for its application. Herein, we developed a self-assembled DNA nanosponge (DNS) for adsorption and clearance of intracellular miR-21. The densely packed DNA nanostructure is able to provide large amounts of repeated ASO copies for efficient capturing of miR-21 and inhibiting the miRNAs function in mammalian cells. The cell apoptosis-related protein expression (Bcl-2, Bax, and cleaved caspase-3/9) can be obviously interrupted with the delivery of DNS. Besides, we have shown that the DNS can efficiently carry Dox for chemotherapy and inducing tumor cell (MCF-7) apoptosis meanwhile has little affect to normal cells (Hs578 Bst). These polymeric DNS systems mimic the natural RNA circle-based miRNA sponges and have potential to be applied for specific and efficient regulation of gene expression in tumor cells for synergistic antitumor chemotherapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , DNA/genética , MicroRNAs/genética , Oligonucleotídeos Antissenso/farmacologia , Adsorção/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caspase 3/genética , Citoplasma/genética , DNA/antagonistas & inibidores , Doxorrubicina/química , Doxorrubicina/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Camundongos , MicroRNAs/antagonistas & inibidores , Nanopartículas/química , Oligonucleotídeos Antissenso/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Circular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Chem Biodivers ; 16(11): e1900315, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31532059

RESUMO

Here, we report the synthesis and characterization of four new aroyl-hydrazone derivatives L1 -L4 , and their structural as well as biological activities have been explored. In addition to docking with bovine serum albumin (BSA) and duplex DNA, the experimental results demonstrate the effective binding of L1 -L4 with BSA protein and calf thymus DNA (ct-DNA) which is in agreement with the docking results. Further biological activities of L1 -L4 have been examined through molecular docking with different proteins which are involved in the propagation of viral or cancer diseases. L1 shows best binding affinity with influenza A virus polymerase PB2 subunit (2VY7) with binding energy -11.42 kcal/mol and inhibition constant 4.23 nm, whereas L2 strongly bind with the hepatitis C virus NS5B polymerase (2WCX) with binding energy -10.47 kcal/mol and inhibition constant 21.06 nm. Ligand L3 binds strongly with TGF-beta receptor 1 (3FAA) and L4 with cancer-related EphA2 protein kinases (1MQB) with binding energy -10.61 kcal/mol, -10.02 kcal/mol and inhibition constant 16.67 nm and 45.41 nm, respectively. The binding energies of L1 -L4 are comparable with binding energies of their proven inhibitors. L1 , L3 and L4 can be considered as both 3FAA and 1MQB dual targeting anticancer agents, while L1 and L3 are both 2VY7 and 2WCX dual targeting antiviral agents. On the other side, L2 and L4 target only one virus related target (2WCX). Furthermore, the geometry optimizations of L1 -L4 were performed via density functional theory (DFT). Moreover, all four ligands (L1 -L4 ) were characterized by NMR, FT-IR, ESI-MS, elemental analysis and their molecular structures were validated by single crystal X-ray diffraction studies.


Assuntos
Antineoplásicos/farmacologia , Antivirais/farmacologia , DNA/antagonistas & inibidores , Desenho de Fármacos , Hidrazonas/farmacologia , Simulação de Acoplamento Molecular , Soroalbumina Bovina/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antivirais/síntese química , Antivirais/química , Bovinos , Proliferação de Células/efeitos dos fármacos , Cristalografia por Raios X , DNA/química , Teoria da Densidade Funcional , Ensaios de Seleção de Medicamentos Antitumorais , Hepacivirus/efeitos dos fármacos , Hidrazonas/síntese química , Hidrazonas/química , Vírus da Influenza A/efeitos dos fármacos , Ligantes , Testes de Sensibilidade Microbiana , Estrutura Molecular , Soroalbumina Bovina/química
7.
J Pharmacol Exp Ther ; 371(2): 320-326, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31455631

RESUMO

CC-115, a triazole-containing compound, is a dual mammalian target of rapamycin (mTOR)/DNA-dependent protein kinase (DNA-PK) inhibitor currently in clinical trials. To develop this compound further, we investigated factors that may affect cellular response to CC-115. Previously, fatty acid synthase (FASN) was shown to upregulate DNA-PK activity and contribute to drug resistance; therefore, we hypothesized that FASN may affect cellular response to CC-115. Instead, however, we showed that CC-115 is a substrate of ATP-binding cassette G2 (ABCG2), a member of the ATP-binding cassette transporter superfamily, and that expression of ABCG2, not FASN, affects the potency of CC-115. ABCG2 overexpression significantly increases resistance to CC-115. Inhibiting ABCG2 function, using small-molecule inhibitors, sensitizes cancer cells to CC-115. We also found that CC-115 may be a substrate of ABCB1, another known ABC protein that contributes to drug resistance. These findings suggest that expression of ABC transporters, including ABCB1 and ABCG2, may affect the outcome in clinical trials testing CC-115. Additionally, the data indicate that ABC transporters may be used as markers for future precision use of CC-115. SIGNIFICANCE STATEMENT: In this article, we report our findings on the potential mechanism of resistance to CC-115, a dual inhibitor of mTOR and DNA-PK currently in clinical trials. We show that CC-115 is a substrate of ABCG2 and can be recognized by ABCB1, which contributes to CC-115 resistance. These findings provide novel information and potential guidance on future clinical testing of CC-115.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Resistência a Medicamentos/efeitos dos fármacos , Proteínas de Neoplasias/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Pirazinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Triazóis/farmacologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Ensaios Clínicos como Assunto/métodos , DNA/antagonistas & inibidores , DNA/metabolismo , Relação Dose-Resposta a Droga , Resistência a Medicamentos/fisiologia , Células HEK293 , Humanos , Células MCF-7 , Fatores de Risco , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/fisiologia
8.
Bull Exp Biol Med ; 164(2): 247-251, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29177902

RESUMO

Culturing of bone marrow cells in serum-free RPMI-1640 medium for 24 h was accompanied by a decrease in the rate of [3H]-thymidine incorporation into DNA. Addition of native apolipoprotein A-I (apoA-I) or plasma LDL and HDL to the culture medium increased this parameter. In contrast to native apoA-I, its modified form decelerated DNA synthesis in bone marrow cells. A similar inhibitory effect of modified protein was observed in cultures of human embryonic kidney cells (HEK293) and in rapidly proliferating mouse macrophage cell line ANA-1. The only exclusion was human myeloid cell line U937: neither native nor modified apoA-I affected DNA synthesis in these cells. Thus, the regulatory effects of apoA-I are tissue-specific; this protein can produce either stimulatory or inhibitory effect on DNA biosynthesis in cells depending on its conformation.


Assuntos
Apolipoproteína A-I/farmacologia , DNA/biossíntese , Lipoproteínas HDL/farmacologia , Lipoproteínas LDL/farmacologia , Lipoproteínas VLDL/farmacologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Linhagem Celular , DNA/agonistas , DNA/antagonistas & inibidores , Células HEK293 , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Especificidade de Órgãos , Ratos , Ratos Wistar , Timidina/metabolismo , Trítio , Células U937
9.
J Microbiol Biotechnol ; 27(11): 2070-2073, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-28910865

RESUMO

We have discovered a novel chemical compound, (E)-3-(furan-2-yl)-N-(4-sulfamoylphenyl) acrylamide, that suppresses the enzymatic activities of SARS coronavirus helicase. To determine the inhibitory effect, ATP hydrolysis and double-stranded DNA unwinding assays were performed in the presence of various concentrations of the compound. Through these assays, we obtained IC50 values of 2.09 ± 0.30 µM (ATP hydrolysis) and 13.2 ± 0.9 µM (DNA unwinding), respectively. Moreover, we found that the compound did not have any significant cytotoxicity when 40 µM of it was used. Our results showed that the compound might be useful to be developed as an inhibitor against SARS coronavirus.


Assuntos
Antivirais/antagonistas & inibidores , DNA Helicases/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/enzimologia , Trifosfato de Adenosina , Antivirais/química , Antivirais/isolamento & purificação , Linhagem Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA/antagonistas & inibidores , Inibidores Enzimáticos/química , Inibidores Enzimáticos/isolamento & purificação , Humanos , Hidrólise , Concentração Inibidora 50
10.
Biometals ; 30(3): 321-334, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28303361

RESUMO

Three ruthenium(II) phosphine/diimine/picolinate complexes were selected aimed at investigating anticancer activity against several cancer cell lines and the capacity of inhibiting the supercoiled DNA relaxation mediated by human topoisomerase IB (Top 1). The structure-lipophilicity relationship in membrane permeability using the Caco-2 cells have also been evaluated in this study. SCAR 5 was found to present 45 times more cytotoxicity against breast cancer cell when compared to cisplatin. SCAR 4 and 5 were both found to be capable of inhibiting the supercoiled DNA relaxation mediated by Top 1. Interaction studies showed that SCAR 4 and 5 can bind to DNA through electrostatic interactions while SCAR 6 is able to bind covalently to DNA. The complexes SCAR were found to interact differently with bovine serum albumin (BSA) suggesting hydrophobic interactions with albumin. The permeability of all complexes was seen to be dependent on their lipophilicity. SCAR 4 and 5 exhibited high membrane permeability (P app  > 10 × 10-6 cm·s-1) in the presence of BSA. The complexes may pass through Caco-2 monolayer via passive diffusion mechanism and our results suggest that lipophilicity and interaction with BSA may influence the complexes permeation. In conclusion, we demonstrated that complexes have powerful pharmacological activity, with different results for each complex depending on the combination of their ligands.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , DNA Topoisomerases Tipo I/metabolismo , Compostos Organometálicos/farmacologia , Rutênio/farmacologia , Inibidores da Topoisomerase/administração & dosagem , Inibidores da Topoisomerase/farmacologia , Administração Oral , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Sítios de Ligação/efeitos dos fármacos , Bovinos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA/antagonistas & inibidores , DNA/química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/síntese química , Compostos Organometálicos/química , Rutênio/administração & dosagem , Rutênio/química , Soroalbumina Bovina/antagonistas & inibidores , Soroalbumina Bovina/química , Relação Estrutura-Atividade , Inibidores da Topoisomerase/síntese química , Inibidores da Topoisomerase/química
11.
DNA Repair (Amst) ; 44: 33-41, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27311543

RESUMO

DNA interstrand crosslinks (ICLs) covalently join the two strands of a DNA duplex and block essential processes such as DNA replication and transcription. Several important anti-tumor drugs such as cisplatin and nitrogen mustards exert their cytotoxicity by forming ICLs. However, multiple complex pathways repair ICLs and these are thought to contribute to the development of resistance towards ICL-inducing agents. While the understanding of many aspects of ICL repair is still rudimentary, studies in recent years have provided significant insights into the pathways of ICL repair. In this perspective we review the recent advances made in elucidating the mechanisms of ICL repair with a focus on the role of TLS polymerases. We describe the emerging models for how these enzymes contribute to and are regulated in ICL repair, discuss the key open questions and examine the implications for this pathway in anti-cancer therapy.


Assuntos
Reparo do DNA , DNA de Neoplasias/genética , DNA Polimerase Dirigida por DNA/genética , DNA/genética , Neoplasias/genética , Animais , Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Reagentes de Ligações Cruzadas/uso terapêutico , DNA/antagonistas & inibidores , DNA/metabolismo , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/antagonistas & inibidores , DNA de Neoplasias/metabolismo , DNA Polimerase Dirigida por DNA/classificação , DNA Polimerase Dirigida por DNA/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Mecloretamina/uso terapêutico , Neoplasias/enzimologia , Neoplasias/patologia , Neoplasias/terapia , Transcrição Gênica/efeitos dos fármacos , Xenopus
12.
Biosci Biotechnol Biochem ; 80(2): 313-7, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26338495

RESUMO

Myeloperoxidase (MPO)-generated halogenating molecules, such as hypochlorous acid and hypobromous acid (HOBr), in inflammatory regions are postulated to contribute to disease progression. In this study, we showed that ergothioneine (EGT), derived from an edible mushroom, inhibited MPO activity as well as the formation of 8-bromo-2'-deoxyguanosine in vitro. The HOBr scavenging effect of EGT is higher than those of ascorbic acid and glutathione. We initially observed that the administration of Coprinus comatus, an edible mushroom containing a high amount of EGT, inhibited the UV-B-induced inflammatory responses and DNA halogenation, suggesting that EGT is a promising anti-inflammatory agent from mushrooms.


Assuntos
Agaricales/química , Anti-Inflamatórios/farmacologia , DNA/antagonistas & inibidores , Ergotioneína/farmacologia , Peroxidase/antagonistas & inibidores , Animais , Anti-Inflamatórios/isolamento & purificação , Ácido Ascórbico/farmacologia , Bromatos/antagonistas & inibidores , Bromatos/metabolismo , DNA/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/antagonistas & inibidores , Desoxiguanosina/biossíntese , Ergotioneína/isolamento & purificação , Glutationa/farmacologia , Halogenação/efeitos dos fármacos , Ácido Hipocloroso/antagonistas & inibidores , Ácido Hipocloroso/metabolismo , Inflamação/tratamento farmacológico , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Pelados , Peroxidase/metabolismo , Raios Ultravioleta/efeitos adversos
13.
Org Biomol Chem ; 13(10): 2866-9, 2015 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-25581012

RESUMO

Cucurbiturils (CB6 and CB7) were shown to inhibit the enzymatically catalyzed restriction of plasmids and linear DNA. This effect can be inverted by supramolecular masking of the macrocycles through competitive complexation with polyamines. These experiments provide supramolecular control of biocatalytic processes.


Assuntos
DNA/antagonistas & inibidores , DNA/química , Endonucleases/química , Inibidores Enzimáticos/farmacologia , Compostos Macrocíclicos/química , Animais , Ligação Competitiva , Catálise , Bovinos , DNA Super-Helicoidal/química , Inibidores Enzimáticos/química , Concentração de Íons de Hidrogênio , Hidrólise , Cinética , Peptídeos/química , Plasmídeos/metabolismo , Poliaminas/química
14.
Med Chem ; 11(4): 373-82, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25524547

RESUMO

We tested the antiproliferative activity and mechanism of the action of several novel aminoacridine derivatives. Six different cancer cell lines were used to evaluate the potential cytotoxic effect of eleven aminoacridine-based molecules. A standard MTT assay was used for cell bioavailability analysis. Additionally, the potential cytotoxic effect of the tested compounds on non-cancer cells was investigated in rat skeletal muscle myotubes (L6) and in bovine aortic smooth muscle cells. In order to investigate whether the DNA binding activity of tested compounds correlated with their cytotoxic effect, circular dichroism (CD) measurement and DNA T4 ligase assay were performed. Finally, the potential mutagenic activity of the lead compound 5 was investigated. The cytotoxic effect of compound 5 in cancer cells was obtained in lower concentrations than the well-known: 9- aminoacridine based drug, amsacrine. The lead compound binds to DNA, but in a different mode than the parent molecules. Additionally, compound 5 was not cytotoxic in the effective range of concentrations in non-cancer cells. In identical concentrations, the parent compound (9-aminoacridine) and amsacrine were extremely toxic for both types of these normal cells. Finally, based on CD measurement and T4 ligase assay, it was confirmed that 5 binds to DNA but in different from the parent compounds manner. Important to mention, that compound 5 might have increased mutagenic activity which must be verified in vivo. Based on these in vitro results, we conclude that 5 is a more potent and more selective antiprolifirative compound than amsacrine. Compound 5 was also more effective in HepG2 and P-12 cells. Thus, 5 is suitable for future in vivo biological evaluation and its structure might be used as a basis for developing novel anticancer drugs.


Assuntos
Aminoacridinas/síntese química , Antineoplásicos/síntese química , Substâncias Intercalantes/farmacologia , Aminoacridinas/farmacologia , Amsacrina/química , Amsacrina/toxicidade , Animais , Antineoplásicos/farmacologia , Bovinos , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA/antagonistas & inibidores , DNA/química , DNA Ligase Dependente de ATP , DNA Ligases/química , Humanos , Substâncias Intercalantes/química , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Especificidade de Órgãos , Ratos , Relação Estrutura-Atividade
15.
Med Chem ; 11(4): 342-53, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25494808

RESUMO

A series of 1,3,6-triphenylpyrazolo[3,4-b]pyridin-4-one derivatives was designed, synthesized and evaluated for cytotoxic activity in A375 human melanoma and human erythroleukemia (HEL) cells. The new pyrazolopyridones displayed comparable activities to the antitumor compound etoposide. The inhibitory effect of compounds 17, 18, 27 and 32 against topoisomerase II-mediated cleavage activities was measured finding good correlation with the results obtained from MTS assay. Docking studies into bacterial topoisomerase II (DNA Gyrase), topoisomerase IIα and topoisomerase IIß binding sites in the DNA binding interface were performed.


Assuntos
Antineoplásicos/síntese química , Proteínas de Ligação a DNA/antagonistas & inibidores , Pirazóis/síntese química , Piridonas/síntese química , Inibidores da Topoisomerase II/síntese química , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/antagonistas & inibidores , DNA/química , DNA Topoisomerases Tipo II/química , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Etoposídeo/farmacologia , Humanos , Simulação de Acoplamento Molecular , Pirazóis/farmacologia , Piridonas/farmacologia , Relação Estrutura-Atividade , Inibidores da Topoisomerase II/farmacologia
16.
Bioorg Med Chem ; 22(7): 1983-92, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24630693

RESUMO

Increasing the affinity of diamidines for AT-rich regions of DNA has long been an important goal of medicinal chemists who wanted to improve the antiparasitic and antifungal properties of that class of derivatives. In recent years it was demonstrated that diamidines could interfere with many other biomolecular targets including ion channels as well as enzymes and modulate some RNA-protein, DNA-protein, and protein-protein interactions. It is therefore not surprising that diamidines now emerge as novel potential drug candidates for the treatment of various diseases, i.a. neurodegenerative disorders, acidosis-related pathological conditions, hypertension, thrombosis, type 2 diabetes, myotonic dystrophy, and cancers. A summary of the most striking results obtained to date in those domains is presented is this review.


Assuntos
Amidinas/farmacologia , Amidinas/uso terapêutico , Amidinas/química , Animais , DNA/antagonistas & inibidores , Diabetes Mellitus Tipo 2/tratamento farmacológico , Enzimas/metabolismo , Humanos , Hipertensão/tratamento farmacológico , Canais Iônicos/antagonistas & inibidores , Distrofia Miotônica/tratamento farmacológico , Neoplasias/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Proteínas/antagonistas & inibidores , RNA/antagonistas & inibidores , Trombose/tratamento farmacológico
17.
Cancer Chemother Pharmacol ; 73(2): 363-71, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24297683

RESUMO

PURPOSE: Maximum tolerated dose, safety, pharmacokinetics, and pharmacodynamics were assessed in this phase 1 study of PNT2258, a BCL-2-targeted liposomal formulation of a 24-base DNA oligonucleotide called PNT100. METHODS: Patients with malignant solid tumors were assigned sequentially to one of ten dose-escalation cohorts of PNT2258 at 1, 2, 4, 8, 16, 32, 64, 85, 113, and 150 mg/m(2) administered intravenously on days 1 through 5 of each 21-day cycle. Pharmacokinetics were determined on days 1 and 5 of the first cycle. Lymphocyte and platelets concentrations were measured for evidence of BCL2-targeted effect. CT scans were used to identify radiologic evidence of anti-tumor effect. RESULTS: Twenty-two subjects received PNT2258, and the maximum tolerated dose for PNT2258 was not reached. Doses at or above 32 mg/m(2) resulted in exposure to PNT2258 above the exposure level required for anti-tumor activity in preclinical xenograft testing of 22,377 ng h/ml (PK analysis 2012). Fatigue was the most commonly reported adverse event. Dose-limiting toxicity, manifesting as a transient increase in aspartate aminotransferase, occurred at 150 mg/m(2), the highest dose tested. Four subjects, two each with diagnosis of non-small-cell lung cancer and sarcoma, treated at doses of 64 mg/m(2) or higher, remained on study for 5-8 cycles. CONCLUSIONS: PNT2258 was safe and well tolerated at the doses tested up to 150 mg/m(2). Exposure to PNT2258 resulted in clinically manageable decreases in lymphocyte and platelet concentrations.


Assuntos
Neoplasias/tratamento farmacológico , Oligodesoxirribonucleotídeos/administração & dosagem , Oligonucleotídeos/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , DNA/antagonistas & inibidores , Feminino , Humanos , Lipossomos/administração & dosagem , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Oligonucleotídeos/efeitos adversos , Oligonucleotídeos/farmacocinética , Resultado do Tratamento
18.
J Med Chem ; 56(5): 1830-42, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23414143

RESUMO

Some new psoralen derivatives were synthesized and evaluated as inhibitors of NF-κB/DNA interaction, with the aim to investigate the structural determinants required to inhibit this interaction. Starting from molecular docking studies, several possible protein binding sites were proposed and several three-dimensional quantitative structure-activity relationship (3D-QSAR) models were built using the docked poses of 29 (the most active psoralen in the series) as templates for alignment of the inhibitors. The selected best model was validated through the prediction of the activity of 17 novel compounds. All the experimental data agreed with the computational experiments, supporting the reliability of the computational approach. The hypothesis about the interaction with NF-κB was also supported by surface plasmon resonance based assays using compound 29. All the collected data allowed the identification of compound 29 as a potential candidate for the development of pharmaceutical strategies against the inflammatory phenotype of cystic fibrosis.


Assuntos
Furocumarinas/farmacologia , Anti-Inflamatórios/síntese química , Anti-Inflamatórios/farmacologia , Sítios de Ligação , Fibrose Cística/tratamento farmacológico , DNA/antagonistas & inibidores , DNA/metabolismo , Furocumarinas/síntese química , Concentração Inibidora 50 , Simulação de Dinâmica Molecular , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Ligação Proteica , Relação Quantitativa Estrutura-Atividade , Ressonância de Plasmônio de Superfície
19.
Oncol Rep ; 29(4): 1275-84, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23381931

RESUMO

The prognosis for malignant melanoma is poor; therefore, new diagnostic methods and treatment strategies are urgently needed. Phosphodiesterase 2 (PDE2) is one of 21 phosphodiesterases, which are divided into 11 families (PDE1-PDE11). PDE2 hydrolyzes cyclic AMP (cAMP) and cyclic GMP (cGMP), and its binding to cGMP enhances the hydrolysis of cAMP. We previously reported the expression of PDE1, PDE3 and PDE5 in human malignant melanoma cells. However, the expression of PDE2 in these cells has not been investigated. Herein, we examined the expression of PDE2A and its role in human oral malignant melanoma PMP cells. Sequencing of RT-PCR products revealed that PDE2A2 was the only variant expressed in PMP cells. Four point mutations were detected; one missense mutation at nucleotide position 734 (from C to T) resulted in the substitution of threonine with isoleucine at amino acid position 214. The other three were silent mutations. An in vitro migration assay and a terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay revealed that suppressing PDE2 activity with its specific inhibitor, erythro-9-(2-hydroxy-3-nonyl)-adenine (EHNA), had no impact on cell motility or apoptosis. Furthermore, the cytotoxicity of EHNA, assessed using a trypan blue exclusion assay, was negligible. On the other hand, assessment of cell proliferation by BrdU incorporation and cell cycle analysis by flow cytometry revealed that EHNA treatment inhibited DNA synthesis and increased the percentage of G2/M-arrested cells. Furthermore, cyclin A mRNA expression was downregulated, while cyclin E mRNA expression was upregulated in EHNA-treated cells. Our results demonstrated that the PDE2A2 variant carrying point mutations is expressed in PMP cells and may affect cell cycle progression by modulating cyclin A expression. Thus, PDE2A2 is a possible new molecular target for the treatment of malignant melanoma.


Assuntos
Adenina/análogos & derivados , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , DNA/biossíntese , Melanoma/metabolismo , Neoplasias Bucais/metabolismo , Adenina/farmacologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/antagonistas & inibidores , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Ciclina A/genética , Ciclina A/metabolismo , DNA/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/patologia , Mutação
20.
Leuk Res ; 36(12): 1536-40, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22954607

RESUMO

S-allylthio-6-mercaptopurine and its ribose derivative were tested for anti-leukemic activity, using a human- mouse B-CLL model. The novel prodrugs contain two components, a purine analog, which interferes with DNA synthesis, and an S-allylthio, readily engaging in thiol-disulfide exchange reactions. The latter component targets the redox homeostasis which is more sensitive in leukemic cells, than in normal B-cells. Upon administration, the prodrug permeates cells, instantly reacts with free thiol, forming S-allyl mixed disulfides and releasing purine. Several cycles of thiol-disulfide exchange reactions occur, thus extending the duration of the prodrug effects. The concerted action of 2 components, as compared with purine alone, boosted in vitro apoptotis in B-CLL cells from 10% to 38%, and decreased in vivo engraftment of B-CLL from 30% to 0.7%.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Linfócitos B/efeitos dos fármacos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Mercaptopurina/análogos & derivados , Mercaptopurina/farmacologia , Pró-Fármacos/farmacologia , Ácidos Sulfínicos/farmacologia , Compostos Alílicos , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/metabolismo , Apoptose/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/patologia , Permeabilidade da Membrana Celular , DNA/antagonistas & inibidores , DNA/biossíntese , Modelos Animais de Doenças , Dissulfetos/química , Dissulfetos/metabolismo , Sinergismo Farmacológico , Feminino , Humanos , Leucemia Linfocítica Crônica de Células B/metabolismo , Leucemia Linfocítica Crônica de Células B/patologia , Mercaptopurina/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Oxirredução , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismo , Ácidos Sulfínicos/química , Ácidos Sulfínicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA