Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(26): 40233-40251, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27244886

ABSTRACT

Several compounds bearing the indolinone chemical scaffold are known to possess anticancer properties. For example, the tyrosine kinase inhibitor sunitinib is an arylideneindolin-2-one compound. The chemical versatility associated with structural modifications of indolinone compounds underlies the potential to discover additional derivatives possessing anticancer properties. Previously synthesized 3-(2-oxoethylidene)indolin-2-one compounds, also known as supercinnamaldehyde (SCA) compounds in reference to the parent compound 1 [1-methyl-3(2-oxopropylidene)indolin-2-one], bear a nitrogen-linked α,ß-unsaturated carbonyl (Michael acceptor) moiety. Here we found that analogs bearing N-substituents, in particular compound 4 and 5 carrying an N-butyl and N-benzyl substituent, respectively, were strongly cytotoxic towards human HCT 116 colorectal and MCF-7 breast carcinoma cells. These compounds also displayed strong thioredoxin reductase (TrxR) inhibitory activity that was likely attributed to the electrophilicity of the Michael acceptor moiety. Their selectivity towards cellular TrxR inhibition over related antioxidant enzymes glutathione reductase (GR), thioredoxin (Trx) and glutathione peroxidase (GPx) was mediated through targeting of the selenocysteine (Sec) residue in the highly accessible C-terminal active site of TrxR. TrxR inhibition mediated by indolin-2-one compounds led to cellular Trx oxidation, increased oxidative stress and activation of apoptosis signal-regulating kinase 1 (ASK1). These events also led to activation of p38 and JNK mitogen-activated protein kinase (MAPK) signaling pathways, and cell death with apoptotic features of PARP cleavage and caspase 3 activation. In conclusion, these results suggest that indolin-2-one-based compounds specifically targeting TrxR may serve as novel drug leads for anticancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Indoles/chemistry , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Antioxidants/metabolism , Catalytic Domain , Cattle , Drug Design , Drug Screening Assays, Antitumor , Fibroblasts/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , HCT116 Cells , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Mice , Naphthoquinones/chemistry , Oxidative Stress , Protein Domains , Rats , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Selenocysteine/chemistry , Thioredoxins/metabolism
2.
Free Radic Biol Med ; 87: 125-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26119781

ABSTRACT

The thioredoxin (Trx) system is one major redox system in mammalian cells. One of its component, Trx, is involved in redox homeostasis and many cellular biological processes through participating in disulfide reduction, S-nitrosylation/S-denitrosylation reactions and protein-protein interactions. In this study, we report the identification of a novel interaction between cytosolic/nuclear Trx1 and apoptosis inducing factor (AIF), and the redox sensitivity and biological significance of the Trx-AIF interaction was characterized. Cytosolic Trx1 but not mitochondrial Trx2 was observed to interact with AIF under physiological conditions and Trx1's active site cysteines were crucial for the interaction. Under oxidative stress conditions, Trx-AIF interaction was disrupted. When the treated cells were allowed to recover from oxidative stress by means of removal of the oxidants, interaction between Trx1 and AIF was re-established time-dependently, which underpins the biological relevance of a Trx-dependent redox regulation of AIF-mediated cell death. Indeed, in times of oxidative stress, nuclear translocation of AIF was found to occur concurrently with perturbations to the Trx-AIF interaction. Once localized in the nucleus, reduced Trx1 hindered the interaction between AIF and DNA, thereby bringing about an attenuation of AIF-mediated DNA damage. In conclusion, characterization of the Trx-AIF interaction has led to an understanding of the effect of reduced Trx1 on possibly regulating AIF-dependent cell death through impeding AIF-mediated DNA damage. Importantly, identification of the novel interaction between Trx1 and AIF has provided opportunities to design and develop therapeutically relevant strategies that either promote or prevent this protein-protein interaction for the treatment of different disease states.


Subject(s)
Apoptosis Inducing Factor/metabolism , Oxidative Stress/genetics , Protein Interaction Maps/genetics , Thioredoxins/genetics , Animals , Apoptosis Inducing Factor/genetics , DNA Damage/genetics , HEK293 Cells , Humans , Mitochondria , Oxidation-Reduction , Thioredoxins/metabolism
3.
Antioxid Redox Signal ; 19(11): 1149-65, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23311917

ABSTRACT

AIMS: The role of thioredoxin reductase (TrxR) in tumorigenesis has made it an attractive anticancer target. A systematic approach for development of novel compounds as TrxR inhibitors is currently lacking. Structurally diversified TrxR inhibitors share in common electrophilic propensities for the sulfhydryl groups, among which include the Michael reaction acceptors containing an α,ß-unsaturated carbonyl moiety. We aimed to identify features among structurally diversified Michael acceptor-based compounds that would yield a strong TrxR inhibitory character. RESULTS: Structurally dissimilar Michael acceptor-based natural compounds such as isobutylamides, zerumbone, and shogaols (SGs) were found to possess a poor TrxR inhibitory activity, indicating that a sole Michael acceptor moiety was insufficient to produce TrxR inhibition. The 1,7-diphenyl-hept-3-en-5-one pharmacophore in 3-phenyl-3-SG, a novel SG analog that possessed comparable TrxR inhibitory and antiproliferative potencies as 6-SG, was modified to yield 1,5-diphenyl-pent-1-en-3-one (DPPen) and 1,3-diphenyl-pro-1-en-3-one (DPPro, also known as chalcone) pharmacophores. These Michael acceptor-centric pharmacophores, when substituted with the hydroxyl and fluorine groups, gave rise to analogs displaying a TrxR inhibitory character positively correlated to their antiproliferative potencies. Lead analogs 2,2'-diOH-5,5'-diF-DPPen and 2-OH-5-F-DPPro yielded a half-maximal TrxR inhibitory concentration of 9.1 and 10.5 µM, respectively, after 1-h incubation with recombinant rat TrxR, with the C-terminal selenocysteine residue found to be targeted. INNOVATION: Identification of Michael acceptor-centric pharmacophores among diversified compounds demonstrates that a systematic approach to discover and develop Michael acceptor-based TrxR inhibitors is feasible. CONCLUSION: A strong TrxR inhibitory character correlated to the antiproliferative potency is attributed to structural features that include an α,ß-unsaturated carbonyl moiety centered in a DPPen or DPPro pharmacophore bearing hydroxyl and fluorine substitutions.


Subject(s)
Antineoplastic Agents/pharmacology , Chalcone/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Amides/chemistry , Amides/pharmacology , Animals , Antineoplastic Agents/chemistry , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcone/analogs & derivatives , Chalcone/chemistry , Dose-Response Relationship, Drug , Glutathione Peroxidase/antagonists & inhibitors , Glutathione Peroxidase/metabolism , Glutathione Reductase/antagonists & inhibitors , Glutathione Reductase/metabolism , Humans , Models, Molecular , Protein Binding , Rats , Thioredoxin-Disulfide Reductase/chemistry , Thioredoxin-Disulfide Reductase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...