Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Int J Mol Sci ; 25(13)2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-39000166

RESUMO

Pyridoxal-S-methyl-isothiosemicarbazone (PLITSC) is a member of an important group of ligands characterized by different complexation modes to various transition metals. In this contribution, a new complex containing two differently protonated PLITSC ligands ([Fe(PLITSC-H)(PLITSC)]SO4)∙2.5H2O was obtained. The crystal structure was solved by the X-ray analysis and used further for the optimization at B3LYP/6-311++G(d,p)(H,C,N,O,S)/def2-TZVP(Fe) level of theory. Changes in the interaction strength and bond distance due to protonation were observed upon examination by the Quantum Theory of Atoms in Molecules. The protein binding affinity of [Fe(PLITSC-H)(PLITSC)]SO4 towards transport proteins (Bovine Serum Albumin (BSA) and Human Serum Albumin (HSA)) was investigated by the spectrofluorimetric titration and molecular docking. The interactions with the active pocket containing fluorescent amino acids were examined in detail, which explained the fluorescence quenching. The interactions between complex and DNA were followed by the ethidium-bromide displacement titration and molecular docking. The binding along the minor groove was the dominant process involving complex in the proximity of DNA.


Assuntos
DNA , Simulação de Acoplamento Molecular , Ligação Proteica , Soroalbumina Bovina , Ligantes , DNA/química , DNA/metabolismo , Humanos , Soroalbumina Bovina/química , Soroalbumina Bovina/metabolismo , Cristalografia por Raios X , Tiossemicarbazonas/química , Tiossemicarbazonas/metabolismo , Bovinos , Complexos de Coordenação/química , Complexos de Coordenação/metabolismo , Animais , Prótons , Compostos Férricos/química , Albumina Sérica Humana/química , Albumina Sérica Humana/metabolismo , Sítios de Ligação , Ferro/química , Ferro/metabolismo
2.
ACS Chem Neurosci ; 15(7): 1432-1455, 2024 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-38477556

RESUMO

Alzheimer's disease (AD) is the most prevalent cause of dementia characterized by a progressive cognitive decline. Addressing neuroinflammation represents a promising therapeutic avenue to treat AD; however, the development of effective antineuroinflammatory compounds is often hindered by their limited blood-brain barrier (BBB) permeability. Consequently, there is an urgent need for accurate, preclinical AD patient-specific BBB models to facilitate the early identification of immunomodulatory drugs capable of efficiently crossing the human AD BBB. This study presents a unique approach to BBB drug permeability screening as it utilizes the familial AD patient-derived induced brain endothelial-like cell (iBEC)-based model, which exhibits increased disease relevance and serves as an improved BBB drug permeability assessment tool when compared to traditionally employed in vitro models. To demonstrate its utility as a small molecule drug candidate screening platform, we investigated the effects of diacetylbis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(atsm)) and a library of metal bis(thiosemicarbazone) complexes─a class of compounds exhibiting antineuroinflammatory therapeutic potential in neurodegenerative disorders. By evaluating the toxicity, cellular accumulation, and permeability of those compounds in the AD patient-derived iBEC, we have identified 3,4-hexanedione bis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(dtsm)) as a candidate with good transport across the AD BBB. Furthermore, we have developed a multiplex approach where AD patient-derived iBEC were combined with immune modulators TNFα and IFNγ to establish an in vitro model representing the characteristic neuroinflammatory phenotype at the patient's BBB. Here, we observed that treatment with CuII(dtsm) not only reduced the expression of proinflammatory cytokine genes but also reversed the detrimental effects of TNFα and IFNγ on the integrity and function of the AD iBEC monolayer. This suggests a novel pathway through which copper bis(thiosemicarbazone) complexes may exert neurotherapeutic effects on AD by mitigating BBB neuroinflammation and related BBB integrity impairment. Together, the presented model provides an effective and easily scalable in vitro BBB platform for screening AD drug candidates. Its improved translational potential makes it a valuable tool for advancing the development of metal-based compounds aimed at modulating neuroinflammation in AD.


Assuntos
Doença de Alzheimer , Tiossemicarbazonas , Humanos , Barreira Hematoencefálica/metabolismo , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Cobre/metabolismo , Doenças Neuroinflamatórias , Tiossemicarbazonas/farmacologia , Tiossemicarbazonas/metabolismo , Tiossemicarbazonas/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo
3.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36142627

RESUMO

Neuroinflammation has a major role in several brain disorders including Alzheimer's disease (AD), yet at present there are no effective anti-neuroinflammatory therapeutics available. Copper(II) complexes of bis(thiosemicarbazones) (CuII(gtsm) and CuII(atsm)) have broad therapeutic actions in preclinical models of neurodegeneration, with CuII(atsm) demonstrating beneficial outcomes on neuroinflammatory markers in vitro and in vivo. These findings suggest that copper(II) complexes could be harnessed as a new approach to modulate immune function in neurodegenerative diseases. In this study, we examined the anti-neuroinflammatory action of several low-molecular-weight, charge-neutral and lipophilic copper(II) complexes. Our analysis revealed that one compound, a thiosemicarbazone-pyridylhydrazone copper(II) complex (CuL5), delivered copper into cells in vitro and increased the concentration of copper in the brain in vivo. In a primary murine microglia culture, CuL5 was shown to decrease secretion of pro-inflammatory cytokine macrophage chemoattractant protein 1 (MCP-1) and expression of tumor necrosis factor alpha (Tnf), increase expression of metallothionein (Mt1), and modulate expression of Alzheimer's disease-associated risk genes, Trem2 and Cd33. CuL5 also improved the phagocytic function of microglia in vitro. In 5xFAD model AD mice, treatment with CuL5 led to an improved performance in a spatial working memory test, while, interestingly, increased accumulation of amyloid plaques in treated mice. These findings demonstrate that CuL5 can induce anti-neuroinflammatory effects in vitro and provide selective benefit in vivo. The outcomes provide further support for the development of copper-based compounds to modulate neuroinflammation in brain diseases.


Assuntos
Doença de Alzheimer , Tiossemicarbazonas , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Animais , Fatores Quimiotáticos/metabolismo , Complexos de Coordenação , Cobre/metabolismo , Modelos Animais de Doenças , Glicoproteínas de Membrana/metabolismo , Metalotioneína/metabolismo , Camundongos , Microglia/metabolismo , Receptores Imunológicos/metabolismo , Tiossemicarbazonas/metabolismo , Tiossemicarbazonas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
4.
Molecules ; 26(9)2021 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-34063080

RESUMO

Copper(II) complexes of thiosemicarbazones (TSCs) often exhibit anticancer properties, and their pharmacokinetic behavior can be affected by their interaction with blood transport proteins. Interaction of copper(II) complexes of an {N,N,S} donor α-N-pyridyl TSC (Triapine) and an {O,N,S} donor 2-hydroxybenzaldehyde TSC (STSC) with human serum albumin (HSA) was investigated by UV-visible and electron paramagnetic resonance spectroscopy at physiological pH. Asp-Ala-His-Lys and the monodentate N-methylimidazole were also applied as binding models. Conditional formation constants were determined for the ternary copper(II)-TSC complexes formed with HSA, DAHK, and N-methylimidazole based on the spectral changes of both charge transfer and d-d bands. The neutral N-methylimidazole displays a similar binding affinity to both TSC complexes. The partially negatively charged tetrapeptide binds stronger to the positively charged Triapine complex in comparison to the neutral STSC complex, while the opposite trend was observed for HSA, which demonstrates the limitations of the use of simple ligands to model the protein binding. The studied TSC complexes are able to bind to HSA in a fast process, and the conditional constants suggest that their binding strength is only weak-to-moderate.


Assuntos
Cobre/metabolismo , Albumina Sérica Humana/metabolismo , Tiossemicarbazonas/metabolismo , Anisotropia , Simulação por Computador , Humanos , Oligopeptídeos/química , Oxirredução , Ligação Proteica , Piridinas/química , Soluções , Espectrofotometria Ultravioleta , Tiossemicarbazonas/química , Fatores de Tempo
5.
ChemistryOpen ; 10(4): 486-492, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33908707

RESUMO

The Cu(II)-diacetyl-bis (N4-methylthiosemicarbazone) complex (ATSM-Cu(II)) has been suggested as a promising positron emission tomography (PET) agent for hypoxia imaging. Several in-vivo studies have shown its potential to detect hypoxic tumors. However, its uptake mechanism and its specificity to various cancer cell lines have been less studied. Herein, we tested ATSM-Cu(II) toxicity, uptake, and reduction, using four different cell types: (1) mouse breast cancer cells (DA-3), (2) human embryonic kidney cells (HEK-293), (3) breast cancer cells (MCF-7), and (4) cervical cancer cells (Hela) under normoxic and hypoxic conditions. We showed that ATSM-Cu(II) is toxic to breast cancer cells under normoxic and hypoxic conditions; however, it is not toxic to normal HEK-293 non-cancer cells. We showed that the Cu(I) content in breast cancer cell after treatment with ATSM-Cu(II) under hypoxic conditions is higher than in normal cells, despite that the uptake of ATSM-Cu(II) is a bit higher in normal cells than in breast cancer cells. This study suggests that the redox potential of ATSM-Cu(II) is higher in breast cancer cells than in normal cells; thus, its toxicity to cancer cells is increased.


Assuntos
Hipóxia/metabolismo , Compostos Organometálicos/metabolismo , Tiossemicarbazonas/metabolismo , Animais , Linhagem Celular Tumoral , Complexos de Coordenação , Radioisótopos de Cobre/química , Radioisótopos de Cobre/metabolismo , Transportador de Cobre 1/metabolismo , Células HEK293 , Humanos , Camundongos , Compostos Organometálicos/química , Compostos Organometálicos/toxicidade , Oxirredução , Tiossemicarbazonas/química , Tiossemicarbazonas/toxicidade
6.
Inorg Chem ; 59(23): 17109-17122, 2020 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-33231439

RESUMO

Metal complexes have numerous applications in the current era, particularly in the field of pharmaceutical chemistry and catalysis. A novel synthetic approach for the same is always a beneficial addition to the literature. Henceforth, for the first time, we report the formation of three new Pd(II) complexes through the Michael addition pathway. Three chromone-based thiosemicarbazone ligands (SVSL1-SVSL3) and Pd(II) complexes (1-3) were synthesized and characterized by analytical and spectroscopic tools. The Michael addition pathway for the formation of complexes was confirmed by spectroscopic studies. Distorted square planar structure of complex 2 was confirmed by single-crystal X-ray diffraction. Complexes 1-3 were subjected to DNA- and BSA-binding studies. The complex with cyclohexyl substituent on the terminal N of thiosemicarbazone (3) showed the highest binding efficacy toward these biomolecules, which was further understood through molecular docking studies. The anticancer potential of these complexes was studied preliminarily by using MTT assay in cancer and normal cell lines along with the benchmark drugs (cisplatin, carboplatin, and gemcitabine). It was found that complex 3 was highly toxic toward MDA-MB-231 and AsPC-1 cancer cells with IC50 values of 0.5 and 0.9 µM, respectively, and was more efficient than the standard drugs. The programmed cell death mechanism of the complexes in MDA-MB-231 cancer cells was confirmed. Furthermore, the complexes induced apoptosis via ROS-mediated mitochondrial signaling pathway. Conveniently, all the complexes showed less toxicity (≥50 µM) against MCF-10a normal cell line. Molecular docking studies were performed with VEGFR2, EGFR, and SARS-CoV-2 main protease to illustrate the binding efficiency of the complexes with these receptors. To our surprise, binding potential of the complexes with SARS-CoV-2 main protease was higher than that with chloroquine and hydroxychloroquine.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Complexos de Coordenação/farmacologia , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , SARS-CoV-2/enzimologia , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Cromonas/síntese química , Cromonas/metabolismo , Cromonas/farmacologia , Complexos de Coordenação/síntese química , Complexos de Coordenação/metabolismo , Proteases 3C de Coronavírus/metabolismo , DNA/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/metabolismo , Humanos , Substâncias Intercalantes/síntese química , Substâncias Intercalantes/metabolismo , Substâncias Intercalantes/farmacologia , Ligantes , Simulação de Acoplamento Molecular , Paládio/química , Ligação Proteica , Tiossemicarbazonas/síntese química , Tiossemicarbazonas/metabolismo , Tiossemicarbazonas/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
J Med Chem ; 63(22): 13719-13732, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33190481

RESUMO

COTI-2 is a novel anticancer thiosemicarbazone in phase I clinical trial. However, the effects of metal complexation (a main characteristic of thiosemicarbazones) and acquired resistance mechanisms are widely unknown. Therefore, in this study, the copper and iron complexes of COTI-2 were synthesized and evaluated for their anticancer activity and impact on drug resistance in comparison to metal-free thiosemicarbazones. Investigations using Triapine-resistant SW480/Tria and newly established COTI-2-resistant SW480/Coti cells revealed distinct structure-activity relationships. SW480/Coti cells were found to overexpress ABCC1, and COTI-2 being a substrate for this efflux pump. This was unexpected, as ABCC1 has strong selectivity for glutathione adducts. The recognition by ABCC1 could be explained by the reduction kinetics of a ternary Cu-COTI-2 complex with glutathione. Thus, only thiosemicarbazones forming stable, nonreducible copper(II)-glutathione adducts are recognized and, in turn, effluxed by ABCC1. This reveals a crucial connection between copper complex chemistry, glutathione interaction, and the resistance profile of clinically relevant thiosemicarbazones.


Assuntos
Aminoquinolinas/metabolismo , Cobre/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Glutationa/metabolismo , Líquido Intracelular/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Tiossemicarbazonas/metabolismo , Aminoquinolinas/química , Aminoquinolinas/farmacologia , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cobre/química , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glutationa/química , Humanos , Líquido Intracelular/efeitos dos fármacos , Tiossemicarbazonas/química , Tiossemicarbazonas/farmacologia , Difração de Raios X
8.
Molecules ; 25(21)2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33126761

RESUMO

Dipeptidyl peptidase-4 (DPP-4) inhibition has been recognized as a promising approach to develop safe and potent antidiabetic agents for the management of type 2 diabetes. In this context, new thiosemicarbazones (2a-o) were prepared efficiently by the reaction of aromatic aldehydes with 4-[4-(1H-pyrazol-1-yl)phenyl]thiosemicarbazide (1), which was obtained via the reaction of 4-(1H-pyrazol-1-yl)phenyl isothiocyanate with hydrazine hydrate. Compounds 2a-o were evaluated for their DPP-4 inhibitory effects based on a convenient fluorescence-based assay. 4-[4-(1H-pyrazol-1-yl)phenyl]-1-(4-bromobenzylidene)thiosemicarbazide (2f) was identified as the most effective DPP-4 inhibitor in this series with an IC50 value of 1.266 ± 0.264 nM when compared with sitagliptin (IC50 = 4.380 ± 0.319 nM). MTT test was carried out to assess the cytotoxic effects of compounds 2a-o on NIH/3T3 mouse embryonic fibroblast (normal) cell line. According to cytotoxicity assay, compound 2f showed cytotoxicity towards NIH/3T3 cell line with an IC50 value higher than 500 µM pointing out its favourable safety profile. Molecular docking studies indicated that compound 2f presented π-π interactions with Arg358 and Tyr666 via pyrazole scaffold and 4-bromophenyl substituent, respectively. Overall, in vitro and in silico studies put emphasis on that compound 2f attracts a great notice as a drug-like DPP-4 inhibitor for further antidiabetic research.


Assuntos
Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/síntese química , Inibidores da Dipeptidil Peptidase IV/farmacologia , Desenho de Fármacos , Pirazóis/química , Tiossemicarbazonas/síntese química , Tiossemicarbazonas/farmacologia , Técnicas de Química Sintética , Dipeptidil Peptidase 4/química , Inibidores da Dipeptidil Peptidase IV/química , Inibidores da Dipeptidil Peptidase IV/metabolismo , Simulação de Acoplamento Molecular , Conformação Proteica , Tiossemicarbazonas/química , Tiossemicarbazonas/metabolismo
9.
Sci Rep ; 9(1): 16006, 2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31690816

RESUMO

Cardiovascular progenitor cells (CPCs) derived from human pluripotent stem cells (hPSCs) are proposed to be invaluable cell sources for experimental and clinical studies. This wide range of applications necessitates large-scale production of CPCs in an in vitro culture system, which enables both expansion and maintenance of these cells. In this study, we aimed to develop a defined and efficient culture medium that uses signaling factors for large-scale expansion of early CPCs, called cardiogenic mesodermal cells (CMCs), which were derived from hPSCs. Chemical screening resulted in a medium that contained a reproducible combination of three factors (A83-01, bFGF, and CHIR99021) that generated 1014 CMCs after 10 passages without the propensity for tumorigenicity. Expanded CMCs retained their gene expression pattern, chromosomal stability, and differentiation tendency through several passages and showed both the safety and possible cardio-protective potentials when transplanted into the infarcted rat myocardium. These CMCs were efficiently cryopreserved for an extended period of time. This culture medium could be used for both adherent and suspension culture conditions, for which the latter is required for large-scale CMC production. Taken together, hPSC-derived CMCs exhibited self-renewal capacity in our simple, reproducible, and defined medium. These cells might ultimately be potential, promising cell sources for cardiovascular studies.


Assuntos
Sistema Cardiovascular/citologia , Meios de Cultura/metabolismo , Células-Tronco Pluripotentes/citologia , Animais , Sistema Cardiovascular/metabolismo , Diferenciação Celular , Proliferação de Células , Meios de Cultura/química , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Masculino , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/terapia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/transplante , Pirazóis/metabolismo , Piridinas/metabolismo , Pirimidinas/metabolismo , Ratos , Ratos Wistar , Tiossemicarbazonas/metabolismo
10.
J Photochem Photobiol B ; 199: 111585, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31450131

RESUMO

Thiosemicarbazone derivatives are known for their broad biological activity including their antitumor potency. The aim of the current study was to examine the effect of a novel series of non-toxic iron chelators on the accumulation of protoporphyrin IX after external 5-aminolevulonic acid administration. From this series we selected one the most promising derivative which causes a pronounced increase in the concentration of protoporphyrin IX. The increase of the photosensitizer concentration is necessary for the trigger the efficient therapeutic effect of the photodynamic reaction. For selected compound 2 we performed an examination of a panel of the genes that are involved in the heme biosynthesis and degradation. Results indicated the crucial roles of ferrochelatase and heme oxygenase in the described processes. Surprisingly, there was a strict dependence on the type of the tested cell line. A decrease in the expression of the two aforementioned enzymes after incubation with compound 2 and 5-aminolevulonic acid is a commonly known fact and we detected this trend for the MCF-7 and HCT 116 cell lines. However, we noticed the upregulation of the tested targets for the Hs683 cells. These unconventional results prompted us to do a more in-depth analysis of the described processes. In conclusion, we found that compound 2 is a novel, highly effective booster of photodynamic therapy that has prospective applications.


Assuntos
Antineoplásicos/síntese química , Ferro/química , Fármacos Fotossensibilizantes/síntese química , Protoporfirinas/química , Tiossemicarbazonas/metabolismo , Células A549 , Ácido Aminolevulínico/química , Ácido Aminolevulínico/metabolismo , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , Ferroquelatase/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Heme Oxigenase (Desciclizante)/metabolismo , Humanos , Células MCF-7 , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Protoporfirinas/farmacologia , Tiossemicarbazonas/síntese química
11.
Metallomics ; 11(5): 994-1004, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31011727

RESUMO

Thiosemicarbazones (TSCs) are a class of strong metal ion ligands, which are currently being investigated for several applications, such as anticancer treatment. In addition to these ligands only, which exert their activity upon interaction with metal ions in cells, preformed metal-TSC complexes are also widely studied, predominantly with the essential metal ions iron, copper and zinc. Currently, it is unclear what the active species are, which complexes are present and what are their biological targets. Herein, we study the complexes of copper(ii), zinc(ii) and iron(ii) with three TSCs, PT, 3-AP (triapine) and Dp44mT, (latter two are currently in clinical trials), concerning their reactivity with glutathione (GSH) and Zn7-metallothionein (Zn7MT-1, 2 and 3). These two cysteine-containing molecules can have a major impact on metal-TSC complexes because they are abundant in the cytosol and nucleus, they are strong metal ligands and have the potential to reduce Cu(ii) and Fe(iii). Our results indicate that Fe(ii)-TSC is stable in the presence of typical cytosolic concentrations of GSH and Zn7MT. In contrast, all three Cu(ii)-TSCs react rapidly due to the reduction of Cu(ii) to Cu(i), which is then transferred to MT. This suggests that Cu(ii)-TSCs are rapidly dissociated in a cytosolic-type environment and the catalytic generation of reactive oxygen species by Cu(ii)-TSCs is stopped. Moreover, in the case Cu(ii)-Dp44mT, transmetallation with Zn(ii) from MT occurs. The reaction of Zn(ii)-TSCs is ligand dependent, from predominant dissociation for PT and 3-AP, to very little dissociation of Zn(ii)-Dp44mT2. These results indicate that GSH and Zn7MT may be important factors in the fate of Cu(ii)- and Zn(ii)-TSCs. In particular, for Cu, its chemistry is complex, and these reactions may also occur for other families of Cu-complexes used in cancer treatment or for other applications.


Assuntos
Cobre/metabolismo , Glutationa/metabolismo , Ferro/metabolismo , Metalotioneína/metabolismo , Tiossemicarbazonas/metabolismo , Zinco/metabolismo , Concentração de Íons de Hidrogênio , Ligantes , Espécies Reativas de Oxigênio/metabolismo , Espectrofotometria Ultravioleta , Tiossemicarbazonas/química
12.
Anal Bioanal Chem ; 411(11): 2383-2394, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30820631

RESUMO

Solid-phase microextraction (SPME) is an alternative method to dialysis and ultrafiltration for the determination of plasma protein binding (PPB) of drugs. It is particularly advantageous for complicated analytes where standard methods are not applicable. Di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) is a lead compound of novel thiosemicarbazone anti-cancer drugs, which entered clinical trials in 2016. However, this agent exhibited non-specific binding on filtration membranes and had intrinsic chelation activity, which precluded standard PPB methods. In this study, using a simple and fast procedure, we prepared novel SPME fibers for extraction of DpC based on a metal-free, silicon string support, covered with C18 sorbent. Reproducibility of the preparation process was demonstrated by the percent relative standard deviation (RSD) of ≤ 9.2% of the amount of DpC extracted from PBS by several independently prepared fibers. The SPME procedure was optimized by evaluating extraction and desorption time profiles. Suitability of the optimized protocol was verified by examining reproducibility, linearity, and recovery of DpC extracted from PBS or plasma. All samples extracted by SPME were analyzed using an optimized and validated UHPLC-MS/MS method. The developed procedure was applied to the in vitro determination of PPB of DpC at two clinically relevant concentrations (500 and 1000 ng/mL). These studies showed that DpC is highly bound to plasma proteins (PPB ≥ 88%) and this did not differ significantly between both concentrations tested. This investigation provides novel data in the applicability of SPME for the determination of PPB of chelators, as well as useful information for the clinical development of DpC. Graphical abstract.


Assuntos
Antineoplásicos/metabolismo , Proteínas Sanguíneas/metabolismo , Piridinas/metabolismo , Microextração em Fase Sólida/instrumentação , Tiossemicarbazonas/metabolismo , Adsorção , Animais , Bovinos , Cromatografia Líquida de Alta Pressão/métodos , Desenho de Equipamento , Ligação Proteica , Ratos , Silício/química , Microextração em Fase Sólida/métodos , Espectrometria de Massas em Tandem/métodos
13.
Eur J Med Chem ; 164: 654-664, 2019 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-30641446

RESUMO

Although many N-heterocyclic thiosemicarbazone copper complexes have been proposed as potential anticancer agents, little is known about their intracellular localization in cells. In the present study, we synthesized two fluorescent N-heterocyclic thiosemicarbazone copper complexes, ([CuII(L)(Br)] 1 and [CuII2CuI(L)2(Br)3] 2, where HL is (E)-N,N-dimethyl-2-(quinolin-8-ylmethylene)hydrazinecarbothioamide), to assess their intracellular distribution. Our fluorescence studies demonstrated that complex 1 showed an intense emission band at ca. 510 nm (λex = 405 nm) similar to that of complex 2, albeit with about four times lower emission intensity. Both copper complexes showed significantly greater cytotoxicity toward several tumor cell-types with better IC50 (0.27-0.91 µM) than the HL ligand and cisplatin. Scratching wound healing assay and invasion assay were performed, revealing that the copper complexes have good antimetastatic activity. Confocal fluorescence imaging allowed ascertaining that complex 2 was primarily localized to mitochondria. Further studies revealed that the anticancer mechanisms of complex 2 might involve the mitochondrial-mediated apoptotic pathway, probably caused by the reducing mitochondrial membrane potential and induction of ROS (reactive oxygen species) production. Furthermore, complex 2 exhibited promising cytostatic effects in a three-dimensional HeLa spheroid model.


Assuntos
Antineoplásicos/química , Mitocôndrias/metabolismo , Metástase Neoplásica/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Tiossemicarbazonas/uso terapêutico , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fluorescência , Células HeLa , Humanos , Metástase Neoplásica/diagnóstico por imagem , Compostos Organometálicos/metabolismo , Compostos Organometálicos/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Tiossemicarbazonas/metabolismo , Tiossemicarbazonas/toxicidade
15.
Anal Bioanal Chem ; 410(9): 2343-2361, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29476231

RESUMO

Clinical failure of novel drugs is often related to their rapid metabolism and excretion. This highlights the importance of elucidation of their pharmacokinetic profile already at the preclinical stage of drug development. Triapine, the most prominent representative of α-N-heterocyclic thiosemicarbazones, was investigated in more than 30 clinical phase I/II trials, but the results against solid tumors were disappointing. Recent investigations from our group suggested that this is, at least partially, based on the fast metabolism and excretion. In order to establish more detailed structure/activity/metabolism relationships, herein a panel of 10 different Triapine derivatives was investigated for their metabolic pathways. From the biological point of view, the panel consists of terminally dimethylated thiosemicarbazones with nanomolar IC50 values, derivatives with micromolar cytotoxicities comparable to Triapine and a completely inactive representative. To study the oxidative metabolism, a purely instrumental approach based on electrochemistry/mass spectrometry was applied and the results were compared to the data obtained from microsomal incubations. Overall, the investigated thiosemicarbazones underwent the phase I metabolic reactions dehydrogenation, hydroxylation, oxidative desulfuration (to semicarbazone and amidrazone) and demethylation. Notably, dehydrogenation resulted in a ring-closure reaction with formation of thiadiazoles. Although strong differences between the metabolic pathways of the different thiosemicarbazones were observed, they could not be directly correlated to their cytotoxicities. Finally, the metabolic pathways for the most cytotoxic compound were elucidated also in tissues collected from drug-treated mice, confirming the data obtained by electrochemical oxidation and microsomes. In addition, the in vivo experiments revealed a very fast metabolism and excretion of the compound. Graphical abstract Structure/activity/metabolisation relationships for 10 anticancer thiosemicarbazones were established using electrochemical oxidation coupled to mass spectrometry (EC-MS) and human liver microsomes analyzed by LC-MS.


Assuntos
Redes e Vias Metabólicas , Piridinas/metabolismo , Tiossemicarbazonas/metabolismo , Animais , Humanos , Hidroxilação , Rim/metabolismo , Fígado/metabolismo , Camundongos Endogâmicos BALB C , Microssomos Hepáticos/metabolismo , Oxirredução , Piridinas/análise , Piridinas/sangue , Piridinas/urina , Tiossemicarbazonas/análise , Tiossemicarbazonas/sangue , Tiossemicarbazonas/urina
16.
J Inorg Biochem ; 181: 11-17, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29353085

RESUMO

The tetranuclear Pt complex (PtL)4 (where L2- is the anion derived from para-isopropyl thiosemicarbazone) was first described in A.G. Quiroga et al., J. Med. Chem. 41, 1998, 1399-1408. (PtL)4 manifests antiproliferative properties toward various cancer cell lines being a promising anticancer drug candidate. Yet, details of its reactivity with biomolecules have not been elucidated. To this end, we investigated the reactions of (PtL)4 with a few model proteins, i.e. bovine pancreatic ribonuclease (RNase A), cytochrome c (Cyt c) and hen egg white lysozyme (Lysozyme), through electrospray ionization mass spectrometry and other biophysical methods. A rich reactivity of (PtL)4 with the above-mentioned model proteins is observed, leading to the formation of numerous metallodrug-protein adducts. The tetranuclear complex breaks down and various fragments bind proteins up to high metal/protein ratios; this typically results into very complicated mass spectral patterns. However, some of the main mass peaks could be assigned in the case of the Lysozyme adduct. In addition, crystallographic data were obtained for the (PtL)4/Lysozyme and (PtL)4/RNase A adducts pointing at His side chains as the primary binding sites for monometallic Pt fragments. Notably, a few selected features of the interactions observed in the (PtL)4/protein adducts were reproduced by reacting (PtL)4 with a small molecule, i.e. N-methylimidazole. In conclusion, the present study confirms the prodrug nature of the tetraplatinum complex, clarifies one possible pathway for its activation through cluster disassembly and allows initial identification of adducts formed with a representative protein.


Assuntos
Citocromos c/metabolismo , Modelos Moleculares , Muramidase/metabolismo , Platina/metabolismo , Ribonuclease Pancreático/metabolismo , Tiossemicarbazonas/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Proteínas Aviárias/química , Proteínas Aviárias/metabolismo , Sítios de Ligação , Bovinos , Quelantes/química , Quelantes/metabolismo , Galinhas , Complexos de Coordenação/química , Complexos de Coordenação/metabolismo , Cristalografia por Raios X , Citocromos c/química , Cavalos , Ligantes , Conformação Molecular , Estrutura Molecular , Peso Molecular , Muramidase/química , Platina/química , Ribonuclease Pancreático/química , Tiossemicarbazonas/química
17.
Analyst ; 142(17): 3165-3176, 2017 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-28745337

RESUMO

α-N-Heterocyclic thiosemicarbazones are among the most promising ribonucleotide reductase inhibitors identified so far. Triapine, the most prominent representative of this class of substances, has been investigated in multiple phase I and II clinical trials. With regard to clinical practice, Triapine showed activity against hematological diseases, but ineffectiveness against a variety of solid tumors. However, the reasons are still vague and the amount of ADME (absorption, distribution, metabolism and excretion) data for Triapine available in the literature is very limited. Therefore, different analytical tools were used to investigate the metabolism of Triapine including electrochemical oxidations, liver microsomes and in vivo samples from mice. The main metabolic reactions, observed by all three methods, were dehydrogenation and hydroxylations, confirming that electrochemistry, as a purely instrumental approach, can be applied for the simulation of metabolic pathways. The dehydrogenated metabolite M1 was identified as a thiadiazole ring-closed oxidation product of Triapine. From a biological point of view, M1, as a key metabolite, is of interest since the crucial chemical property of α-N-heterocyclic thiosemicarbazones to bind metal ions is lost and cytotoxicity studies showed no anticancer activity of M1. The in vivo data of the urine samples revealed very high levels of the metabolites and Triapine itself already 15 min after treatment. This clearly indicates that Triapine is rapidly metabolised and excreted, which represents an important step forward to understand the possible reason for the inefficiency of Triapine against solid tumors.


Assuntos
Antineoplásicos/metabolismo , Piridinas/metabolismo , Tiossemicarbazonas/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Microssomos Hepáticos/metabolismo , Oxirredução
18.
Br J Cancer ; 116(8): 1057-1064, 2017 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-28324887

RESUMO

BACKGROUND: Hypoxia imaging is a promising tool for targeted therapy but the links between imaging features and underlying molecular characteristics of the tumour have not been investigated. The aim of this study was to compare hypoxia biomarkers and gene expression in oropharyngeal squamous cell carcinoma (OPSCC) diagnostic biopsies with hypoxia imaged with 64Cu-ATSM PET/CT. METHODS: 64Cu-ATSM imaging, molecular and clinical data were obtained for 15 patients. Primary tumour SUVmax, tumour to muscle ratio (TMR) and hypoxic volume were tested for association with reported hypoxia gene signatures in diagnostic biopsies. A putative gene signature for hypoxia in OPSCCs (hypoxic volume-associated gene signature (HVS)) was derived. RESULTS: Hypoxic volume was significantly associated with a reported hypoxia gene signature (rho=0.57, P=0.045), but SUVmax and TMR were not. Immunohistochemical staining with the hypoxia marker carbonic anhydrase 9 (CA9) was associated with a gene expression hypoxia response (rho=0.63, P=0.01). Sixteen genes were positively and five genes negatively associated with hypoxic volume (adjusted P<0.1; eight genes had adjusted P<0.05; HVS). This signature was associated with inferior 3-year progression-free survival (HR=1.5 (1.0-2.2), P=0.047) in an independent patient cohort. CONCLUSIONS: 64Cu-ATSM-defined hypoxic volume was associated with underlying hypoxia gene expression response. A 21-gene signature derived from hypoxic volume from patients with OPSCCs in our study may be linked to progression-free survival.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/patologia , Hipóxia/patologia , Neoplasias Orofaríngeas/patologia , Transcriptoma , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/genética , Radioisótopos de Cobre/metabolismo , Feminino , Humanos , Hipóxia/diagnóstico por imagem , Hipóxia/genética , Processamento de Imagem Assistida por Computador/métodos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias Orofaríngeas/diagnóstico por imagem , Neoplasias Orofaríngeas/genética , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Prognóstico , Compostos Radiofarmacêuticos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tiossemicarbazonas/metabolismo
19.
Mol Pharmacol ; 91(6): 567-575, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28320780

RESUMO

Small-molecule restoration of wild-type structure and function to mutant p53 (so-called mutant reactivation) is a highly sought-after goal in cancer drug development. We previously discovered that small-molecule zinc chelators called zinc metallochaperones (ZMCs) reactivate mutant p53 by restoring zinc binding to zinc-deficient p53 mutants. The lead compound identified from the NCI-60 human tumor cell lines screen, NSC319726 (ZMC1), belongs to the thiosemicarbazone (TSC) class of metal ion chelators that bind iron, copper, magnesium, zinc, and other transition metals. Here, we have investigated the other TSCs, NSC319725 and NSC328784, identified in the same screen, as well as the more well studied TSC, 3-AP (Triapine), to determine whether they function as ZMCs. We measured the zinc Kd zinc ionophore activity, ability to restore zinc to purified p53 DNA binding domain (DBD), and ability to restore site-specific DNA binding to purified R175H-DBD in vitro. We tested all four TSCs in a number of cell-based assays to examine mutant p53 reactivation and the generation of reactive oxygen species (ROS). We found that NSC319725 and NSC328784 behave similarly to ZMC1 in both biophysical and cell-based assays and are heretofore named ZMC2 (NSC319725) and ZMC3 (NSC328784). 3-AP generates a ROS signal similar to ZMC1-3, but it fails to function as a ZMC both in vitro and in cells and ultimately does not reactivate p53. These findings indicate that not all TSCs function as ZMCs, and much of their activity can be predicted by their affinity for zinc.


Assuntos
Inibidores do Crescimento/metabolismo , Metalochaperonas/metabolismo , Mutação/fisiologia , Tiossemicarbazonas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Zinco/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Relação Dose-Resposta a Droga , Inibidores do Crescimento/farmacologia , Humanos , Mutação/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA