Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 432
Filter
1.
Cancer Biol Ther ; 25(1): 2382524, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39054566

ABSTRACT

Thioredoxin Reductase (TrxR) functions to recycle thioredoxin (Trx) during hydroperoxide metabolism mediated by peroxiredoxins and is currently being targeted using the FDA-approved anti-rheumatic drug, auranofin (AF), to selectively sensitize cancer cells to therapy. AF treatment decreased TrxR activity and clonogenic survival in small cell lung cancer (SCLC) cell lines (DMS273 and DMS53) as well as the H727 atypical lung carcinoid cell line. AF treatment also significantly sensitized DMS273 and H727 cell lines in vitro to sorafenib, an FDA-approved multi-kinase inhibitor that depleted intracellular glutathione (GSH). The pharmacokinetic, pharmacodynamic, and safety profile of AF was examined in nude mice with DMS273 xenografts administered AF intraperitoneally at 2 mg/kg or 4 mg/kg (IP) once (QD) or twice daily (BID) for 1-5 d. Plasma levels of AF were 10-20 µM (determined by mass spectrometry of gold), and the optimal inhibition of TrxR activity was obtained at 4 mg/kg once daily, with no effect on glutathione peroxidase 1 activity. This AF treatment extended for 14 d, inhibited TrxR (>75%), and resulted in a significant prolongation of median overall survival from 19 to 23 d (p = .04, N = 30 controls, 28 AF). In this experiment, there were no observed changes in animal bodyweight, complete blood counts (CBCs), bone marrow toxicity, blood urea nitrogen, or creatinine. These results support the hypothesis that AF effectively inhibits TrxR both in vitro and in vivo in SCLC, sensitizes NETs and SCLC to sorafenib, and could be repurposed as an adjuvant therapy with targeted agents that induce disruptions in thiol metabolism.


Subject(s)
Auranofin , Lung Neoplasms , Phenylurea Compounds , Small Cell Lung Carcinoma , Sorafenib , Thioredoxin-Disulfide Reductase , Xenograft Model Antitumor Assays , Auranofin/pharmacology , Auranofin/therapeutic use , Animals , Sorafenib/pharmacology , Sorafenib/therapeutic use , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Small Cell Lung Carcinoma/metabolism , Humans , Mice , Cell Line, Tumor , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/metabolism , Mice, Nude , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Niacinamide/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Chem Biol Interact ; 399: 111137, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-38977166

ABSTRACT

Aberrant activation of thioredoxin reductase (TrxR) is correlated with tumor occurrence and progression, suggesting that TrxR inhibitors can be used as antitumor agents. In this study, we evaluated the anticancer efficacy of eupalinilides B on colorectal cancer cells. Eupalinilides B primarily targeted the conserved selenocysteine 498 residues in TrxR. Besides, it inhibited the enzyme activity in an irreversible manner. After eupalinilides B was used to pharmacologically inhibit TrxR, reactive oxygen species accumulated, and the intracellular redox balance was broken, finally causing oxidative stress-induced tumor cell apoptosis. Significantly, eupalinilides B treatment inhibited in vivo tumor growth. Targeting TrxR by eupalinilides B reveals the new mechanism underlying eupalinilides B and provides insight in developing eupalinilides B as the candidate antitumor chemotherapeutic agent for the treatment of cancer.


Subject(s)
Antineoplastic Agents , Apoptosis , Colorectal Neoplasms , Reactive Oxygen Species , Thioredoxin-Disulfide Reductase , Apoptosis/drug effects , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Humans , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mice, Inbred BALB C , Mice, Nude , Oxidative Stress/drug effects
3.
Biomaterials ; 311: 122705, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39047537

ABSTRACT

Overexpression of thioredoxin reductase (TXNRD) plays crucial role in tumorigenesis. Therefore, designing TXNRD inhibitors is a promising strategy for targeted anticancer drug development. However, poor selectivity has always been a challenge, resulting in unavoidable toxicity in clinic. Herein we demonstrate a strategy to develop highly selective chiral metal complexes-based TXNRD inhibitors. By manipulating the conformation of two distinct weakly interacting groups, we optimize the compatibility between the drug and the electrophilic group within the active site of TXNRD to enhance their non-covalent interaction, thus effectively avoids the poor selectivity deriving from covalent drug interaction, on the basis of ensuring the strong inhibition. Detailed experimental and computational results demonstrate that the chiral isomeric drugs bind to the active site of TXNRD, and the interaction strength is well modulated by chirality. Especially, the meso-configuration, in which the two large sterically hindered active groups are positioned on opposite sides of the drug, exhibits the highest number of non-covalent interactions and most effective inhibition on TXNRD. Taken together, this work not only provides a novel approach for developing highly selective proteinase inhibitors, but also sheds light on possible underlying mechanisms for future application.


Subject(s)
Enzyme Inhibitors , Thioredoxin-Disulfide Reductase , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Humans , Catalytic Domain , Stereoisomerism , Molecular Docking Simulation
4.
J Med Chem ; 67(13): 11138-11151, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38951717

ABSTRACT

Using a multigram-scalable synthesis, we obtained nine dinuclear complexes based on nonendogenous iron(I) centers and featuring variable aminocarbyne and P-ligands. One compound from the series (FEACYP) emerged for its strong cytotoxicity in vitro against four human cancer cell lines, surpassing the activity of cisplatin by 3-6 times in three cell lines, with an average selectivity index of 6.2 compared to noncancerous HEK293 cells. FEACYP demonstrated outstanding water solubility (15 g/L) and stability in physiological-like solutions. It confirmed its superior antiproliferative activity when tested in 3D spheroids of human pancreatic cancer cells and showed a capacity to inhibit thioredoxin reductase (TrxR) similar to auranofin. In vivo treatment of murine LLC carcinoma with FEACYP (8 mg kg-1 dose) led to excellent tumor growth suppression (88%) on day 15, with no signs of systemic toxicity and only limited body weight loss.


Subject(s)
Adamantane , Antineoplastic Agents , Solubility , Humans , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mice , Adamantane/pharmacology , Adamantane/analogs & derivatives , Adamantane/chemistry , Adamantane/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Structure-Activity Relationship , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Iron/chemistry , Iron/metabolism , Water/chemistry , Drug Screening Assays, Antitumor , Phosphines/chemistry , Phosphines/pharmacology , Drug Stability , HEK293 Cells , Organophosphorus Compounds
5.
Biomater Sci ; 12(15): 3918-3932, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38939985

ABSTRACT

The thioredoxin system is involved in cancer development and therefore is a promising target for cancer chemotherapy. Thioredoxin reductase (TrxR) is a key component of the thioredoxin (Trx) system, and is overexpressed in many cancers to inhibit apoptosis-related proteins. Alternatively, inhibition of thioredoxin reductase and upregulation of apoptosis factors provide a therapeutic strategy for anti-tumor treatment. In this study, an ultrasound-activatable meso-organosilica nanomedicine was prepared by integrating chloroquine (CQ) into hollow mesoporous organosilica (CQ@MOS). The meso-organosilica nanomedicine can inhibit the activity of thioredoxin reductase, elevate cellular reactive oxygen species (ROS) levels, upregulate the pro-apoptotic factors in the c-Jun N-terminal kinase (JNK) apoptosis pathway and induce autophagy inhibition, further resulting in mitochondrial membrane potential (MMP) depolarization and cellular ATP content decrease, ultimately causing significant damage to tumor cells. Moreover, CQ@MOS can efficiently deliver chloroquine into cancer cells and promote an enhanced sonodynamic effect for effective anti-tumor chemotherapy and sonodynamic therapy. This study may enlighten us on a new anti-tumor strategy and suggest its promising applications in cancer treatments.


Subject(s)
Antineoplastic Agents , Apoptosis , Nanomedicine , Thioredoxin-Disulfide Reductase , Humans , Apoptosis/drug effects , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Up-Regulation/drug effects , Reactive Oxygen Species/metabolism , Chloroquine/pharmacology , Chloroquine/chemistry , Organosilicon Compounds/chemistry , Organosilicon Compounds/pharmacology , Animals , Cell Line, Tumor , Membrane Potential, Mitochondrial/drug effects , Ultrasonic Therapy
6.
ACS Infect Dis ; 10(5): 1753-1766, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38606463

ABSTRACT

The antibacterial activity of silver species is well-established; however, their mechanism of action has not been adequately explored. Furthermore, issues of low-molecular silver compounds with cytotoxicity, stability, and solubility hamper their progress to drug leads. We have investigated silver N-heterocyclic carbene (NHC) halido complexes [(NHC)AgX, X = Cl, Br, and I] as a promising new type of antibacterial silver organometallics. Spectroscopic studies and conductometry established a higher stability for the complexes with iodide ligands, and nephelometry indicated that the complexes could be administered in solutions with physiological chloride levels. The complexes showed a broad spectrum of strong activity against pathogenic Gram-negative bacteria. However, there was no significant activity against Gram-positive strains. Further studies clarified that tryptone and yeast extract, as components of the culture media, were responsible for this lack of activity. The reduction of biofilm formation and a strong inhibition of both glutathione and thioredoxin reductases with IC50 values in the nanomolar range were confirmed for selected compounds. In addition to their improved physicochemical properties, the compounds with iodide ligands did not display cytotoxic effects, unlike the other silver complexes. In summary, silver NHC complexes with iodide secondary ligands represent a useful scaffold for nontoxic silver organometallics with improved physicochemical properties and a distinct mechanism of action that is based on inhibition of thioredoxin and glutathione reductases.


Subject(s)
Anti-Bacterial Agents , Glutathione Reductase , Gram-Negative Bacteria , Silver , Thioredoxin-Disulfide Reductase , Humans , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glutathione Reductase/antagonists & inhibitors , Glutathione Reductase/metabolism , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Microbial Sensitivity Tests , Silver/chemistry , Silver/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
7.
Bioorg Chem ; 122: 105711, 2022 05.
Article in English | MEDLINE | ID: mdl-35247807

ABSTRACT

Overexpression of thioredoxin reductase (TrxR) has been linked to tumorigenesis and phenotypic maintenance of malignant tumors. Thus, targeting TrxR with natural molecules is a promising strategy for developing anticancer drugs. Sinomenine is a naturally occurring alkaloid isolated from Sinomenium acutum. The drug, Zhengqing Fengtongning made from sinomenine, has been universally applied in rheumatoid arthritis treatment in China as well as other Asian countries for decades. Recently, increasing evidence indicates that sinomenine appears to be a promising therapeutic agent against various cancer cells. However, the exact mechanism underlying the anticancer activity of sinomenine remains unclear. In this study, we identified sinomenine as a kind of new inhibitor for TrxR. Pharmacological inhibition of TrxR by sinomenine results in the decrease of thiols content, increases the levels of reactive oxygen species, and finally facilitates oxidative stress-mediated cancer cell apoptosis. It is vital that knockdown in TrxR1 by shRNA can increase cell sensitivity to sinomenine. Treatment with sinomenine in vivo leads to a decrease in TrxR activity and tumor growth, and an increase in apoptosis. Our findings provide a novel action mechanism related to sinomenine and presents an insight on how to develop sinomenine as a chemotherapeutic agent for cancer therapy.


Subject(s)
Morphinans , Thioredoxin-Disulfide Reductase , Uterine Cervical Neoplasms , Antirheumatic Agents , Apoptosis , Drug Repositioning , Female , Humans , Morphinans/pharmacology , Reactive Oxygen Species , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Uterine Cervical Neoplasms/drug therapy
8.
Eur J Med Chem ; 227: 113897, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34649064

ABSTRACT

Microtubule target agents (MTAs) are widely-used clinical anti-cancer drugs for decades, but the acquired drug resistance severely restricted their application. Thioredoxin reductases (TrxR) was reported to be overexpressed in most tumors and closely related to high risk of cancer recurrence and drug resistance, making it a potential target for anticancer drug discovery. Multi-target-directed ligands (MTDLs) by a single molecule provide a logical and alternative approach to drug combinations. In this work, based on the structure-activity relationships obtained in our previous study, some structure modifications were performed. On one hand, the retained skeleton structure of MTAs endowed its tubulin polymerization inhibition activity, on the other hand, the selenium-containing structure and α,ß-unsaturated ketone moiety endowed the TrxR inhibition activity. As results, the newly obtained compounds exhibited superior anti-proliferative activities towards various human cancer cells and drug-resistance cells, and displayed high selectivity towards various human normal cells. The mechanism study revealed that the dual effect of cell cycle arrest triggered by targeting tubulin and the abnormal accumulation of ROS caused by TrxR inhibition eventually lead to cell apoptosis. Notably, compared with the MTA agents CA-4P, and the TrxR inhibitor Ethaselen, the optimized compound 14c, which served as dual-targeting inhibitor of tubulin and TrxR, exerted greatly improved in vivo anti-tumor activity. In summary, 14c deserved further consideration for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Chalcones/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Tubulin Modulators/pharmacology , Tubulin/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Indoles/chemistry , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Structure-Activity Relationship , Thioredoxin-Disulfide Reductase/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry
9.
J Nat Prod ; 84(12): 3161-3168, 2021 12 24.
Article in English | MEDLINE | ID: mdl-34806369

ABSTRACT

The natural products piperlongumine (1) and ligustrazine (2) have been reported to exert antiproliferative effects against various types of cancer cells by up-regulating the level of reactive oxidative species (ROS). However, the moderate activities of 1 and 2 limit their application. To improve their potential antitumor activity, novel piperlongumine/ligustrazine derivatives were designed and prepared, and their potential pharmacological effects were determined in vitro and in vivo. Among the derivatives obtained, 11 exerted more prominent inhibitory activities against proliferation of drug-sensitive/-resistant cancer cells with lower IC50 values than 1. Particularly, the IC50 value of 11 against drug-resistant Bel-7402/5-FU cells was 0.9 µM, which was about 9-fold better than that of 1 (IC50 value of 8.4 µM). Mechanistic studies showed that 11 demonstrated thioredoxin reductase (TrxR) inhibitory activity, increase of ROS levels, decrease of mitochondrial transmembrane potential levels, and occurrence of DNA damage and autophagy, in a dose-dependent manner, via regulation of DNA damage protein H2AX and autophagy-associated proteins LC3, beclin-1, and p62 in drug-resistant Bel-7402/5-FU cells. Finally, compound 11 at 5 mg/kg displayed potent antitumor activity in vivo with tumor suppression of 76% (w/w). Taken together, compound 11 may represent a promising candidate drug for the chemotherapy of drug-resistant hepatocellular carcinoma and warrant more intensive study.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Dioxolanes/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Liver Neoplasms/drug therapy , Pyrazines/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Carcinoma, Hepatocellular/pathology , Dioxolanes/chemistry , Humans , Liver Neoplasms/pathology , Pyrazines/chemistry
10.
J Nat Prod ; 84(11): 2961-2970, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34752085

ABSTRACT

The brevetoxins, neurotoxins produced by Karenia brevis, the Florida red tide dinoflagellate, effect fish and wildlife mortalities and adverse public health and economic impacts during recurrent blooms. Knowledge of the biochemical consequences of toxin production for K. brevis could provide insights into an endogenous role of the toxins, yet this aspect has not been thoroughly explored. In addition to neurotoxicity, the most abundant of the brevetoxins, PbTx-2, inhibits mammalian thioredoxin reductase (TrxR). The thioredoxin system, composed of the enzymes TrxR and thioredoxin (Trx), is present in all living organisms and is responsible in part for maintaining cellular redox homeostasis. Herein, we describe the cloning, expression, and semisynthesis of the selenoprotein TrxR from K. brevis (KbTrxR) and reductase activity toward a variety of substrates. Unlike mammalian TrxR, KbTrxR reduces oxidized glutathione (GSSG). We further demonstrate that PbTx-2 is an inhibitor of KbTrxR. Covalent adducts between KbTrxR and rat TrxR were detected by mass spectrometry. While both enzymes are adducted at or near the catalytic centers, the specific residues are distinct. Biochemical differences reported for high and low toxin producing strains of K. brevis are consistent with the inhibition of KbTrxR and suggest that PbTx-2 is an endogenous regulator of this critical enzyme.


Subject(s)
Dinoflagellida/metabolism , Marine Toxins/pharmacology , Neurotoxins/pharmacology , Oxocins/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
11.
Anticancer Res ; 41(10): 4947-4955, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34593442

ABSTRACT

BACKGROUND/AIM: Auranofin (AUR), a thioredoxin reductase (TXNRD) inhibitor, shows anticancer activity against several cancers. This study investigated the effects of AUR on the local progression and pulmonary metastasis of osteosarcoma (OS). MATERIALS AND METHODS: Publicly available expression cohorts were analysed to study the relationship between TXNRD-2 expression and the survival of patients with OS. The murine OS cell line LM8 was stimulated with AUR. Cell viability, apoptosis-related protein levels, caspase activity, and wound healing were analysed. Tumor progression and pulmonary metastasis were investigated in C3H mice implanted with LM8 cells. RESULTS: High-level expression of TXNRD-2 represented a negative prognostic factor for metastasis and overall survival in patients with OS. AUR induced apoptosis of OS cells via the oxidative stress-MAPK-Caspase 3 pathway, and suppressed the migration of OS cells. AUR inhibited the pulmonary metastasis of OS, but not local progression. CONCLUSION: AUR represents a potential therapeutic drug for suppressing pulmonary metastasis of OS.


Subject(s)
Auranofin/pharmacology , Bone Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Osteosarcoma/drug therapy , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Antirheumatic Agents/pharmacology , Apoptosis , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Movement , Cell Proliferation , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred C3H , Osteosarcoma/metabolism , Osteosarcoma/pathology , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
J Med Chem ; 64(21): 16132-16146, 2021 11 11.
Article in English | MEDLINE | ID: mdl-34704769

ABSTRACT

Upregulation of the selenoprotein thioredoxin reductase (TrxR) is of pathological significance in maintaining tumor phenotypes. Thus, TrxR inhibitors are promising cancer therapeutic agents. We prepared different amino-substituted phenylarsine oxides and evaluated their cytotoxicity and inhibition of TrxR. Compared with our reported p-substituted molecule (8), the o-substituted molecule (10) shows improved efficacy (nearly a fourfold increase) to kill leukemia HL-60 cells. Although the compounds 8 and 10 display similar potency to inhibit the purified TrxR, the o-substitution 10 exhibits higher potency than the p-substitution 8 to inhibit the cellular TrxR activity. Molecular docking results demonstrate the favorable weak interactions of the o-amino group with the TrxR C-terminal active site. Efficient inhibition of TrxR consequently induces the oxidative stress-mediated apoptosis of cancer cells. Silence of the TrxR expression sensitizes the cells to the arsenic compound treatment, further supporting the critical involvement of TrxR in the cellular actions of compound 10.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenicals/pharmacology , Leukemia/drug therapy , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Apoptosis/drug effects , Arsenicals/chemistry , HL-60 Cells , Humans , Molecular Docking Simulation , Molecular Structure , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
13.
Arch Biochem Biophys ; 711: 109028, 2021 10 30.
Article in English | MEDLINE | ID: mdl-34509463

ABSTRACT

Elevated expression of thioredoxin reductase (TrxR) is associated with the tumorigenesis and resistance to cancer chemoradiotherapy, highlighting the potential of TrxR inhibitors as anticancer drugs. Deoxyelephantopin (DET) is the major active ingredient of Elephantopus scaber and reveals potent anticancer activity. However, the potential mechanism of action and the cellular target of DET are still unknown. Here, we found that DET primarily targets the Sec residue of TrxR and irreversibly prohibits enzyme activity. Suppression of TrxR by DET leads to accumulation of reactive oxygen species and dysregulation in intracellular redox balance, eventually inducing cancer cell apoptosis mediated by oxidative stress. Noticeably, down-regulation of TrxR1 by shRNA increases cell sensitivity to DET. Collectively, targeting of TrxR1 by DET uncovers a novel mechanism of action in DET and deepens the understanding of developing DET as a potential chemotherapeutic agent for treating cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Lactones/pharmacology , Sesquiterpenes/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Antineoplastic Agents, Phytogenic/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Lactones/metabolism , Molecular Docking Simulation , Oxidative Stress/drug effects , Protein Binding , Rats , Reactive Oxygen Species/metabolism , Sesquiterpenes/metabolism , Thioredoxin-Disulfide Reductase/metabolism
14.
Chem Commun (Camb) ; 57(59): 7296-7299, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34223569

ABSTRACT

A chlorine e6 (Ce6) and curcumin (Cur) based self-delivery nanomedicine (CeCu) is prepared for chemotherapy sensitized photodynamic therapy (PDT). The chemotherapeutic agent of Cur could inhibit the TrxR activity to destroy the cellular ROS-defence system for enhanced PDT, which provides synergistic effects for tumor precision therapy in consideration of the unfavorable tumor microenvironments.


Subject(s)
Nanomedicine , Photochemotherapy/methods , Animals , Cell Line , Cell Survival/drug effects , Chlorophyllides , Curcumin/chemistry , Curcumin/metabolism , Curcumin/pharmacology , Humans , Mice , Microscopy, Confocal , Neoplasms/drug therapy , Neoplasms/pathology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyrins/chemistry , Porphyrins/metabolism , Porphyrins/pharmacology , Reactive Oxygen Species/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Transplantation, Heterologous , Tumor Microenvironment
15.
ChemMedChem ; 16(22): 3402-3409, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34268875

ABSTRACT

A series of (NHC)Au(I)Cl monocarbene complexes and their gold(III) analogues (NHC)Au(III)Cl3 were prepared and investigated as antibacterial agents and inhibitors of bacterial TrxR. The complexes showed stronger antibacterial effects against the Gram-positive MRSA and E. faecium strains than against several Gram-negative bacteria. All complexes were efficient inhibitors of bacterial thioredoxin reductase, indicating that inhibition of this enzyme might be involved in their mechanism of action. The efficacy of gold(I) and gold(III) analogues was comparable in most of the assays. The cytotoxicity of the gold NHC compounds against cancer and human cells was overall weaker than the activity against the Gram-positive bacteria, suggesting that their optimization as antibacterials warrants further investigation.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Gold/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Heterocyclic Compounds/pharmacology , Methane/analogs & derivatives , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Gold/chemistry , Gram-Negative Bacteria/enzymology , Gram-Positive Bacteria/enzymology , Heterocyclic Compounds/chemistry , Humans , Methane/chemistry , Methane/pharmacology , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Thioredoxin-Disulfide Reductase/metabolism
16.
Molecules ; 26(14)2021 Jul 15.
Article in English | MEDLINE | ID: mdl-34299558

ABSTRACT

A series of ruthenium(II) complexes with N-heterocyclic carbene (NHC) ligands of the general type (arene)(NHC)Ru(II)X2 (where X = halide) was prepared, characterized, and evaluated as antibacterial agents in comparison to the respective metal free benzimidazolium cations. The ruthenium(II) NHC complexes generally triggered stronger bacterial growth inhibition than the metal free benzimidazolium cations. The effects were much stronger against Gram-positive bacteria (Bacillus subtilis and Staphylococcus aureus) than against Gram-negative bacteria (Escherichia coli, Acinetobacter baumannii, Pseudomonas aeruginosa), and all complexes were inactive against the fungus Candida albicans. Moderate inhibition of bacterial thioredoxin reductase was confirmed for selected complexes, indicating that inhibition of this enzyme might be a contributing factor to the antibacterial effects.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/enzymology , Ruthenium/chemistry , Ruthenium/pharmacology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Bacteria/drug effects , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Bacterial Proteins/antagonists & inhibitors , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Methane/analogs & derivatives , Methane/chemistry , Methane/pharmacology , Models, Molecular
17.
J Med Chem ; 64(10): 6608-6620, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33974434

ABSTRACT

Trichomonas vaginalis causes the most common, nonviral sexually transmitted infection. Only metronidazole (Mz) and tinidazole are approved for treating trichomoniasis, yet resistance is a clinical problem. The gold(I) complex, auranofin, is active against T. vaginalis and other protozoa but has significant human toxicity. In a systematic structure-activity exploration, we show here that diversification of gold(I) complexes, particularly as halides with simple C1-C3 trialkyl phosphines or as bistrialkyl phosphine complexes, can markedly improve potency against T. vaginalis and selectivity over human cells compared to that of the existing antirheumatic gold(I) drugs. All gold(I) complexes inhibited the two most abundant isoforms of the presumed target enzyme, thioredoxin reductase, but a subset of compounds were markedly more active against live T. vaginalis than the enzyme, suggesting that alternative targets exist. Furthermore, all tested gold(I) complexes acted independently of Mz and were able to overcome Mz resistance, making them candidates for the treatment of Mz-refractory trichomoniasis.


Subject(s)
Antiprotozoal Agents/chemistry , Coordination Complexes/chemistry , Gold/chemistry , Phosphines/chemistry , Animals , Antiprotozoal Agents/metabolism , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Cell Survival/drug effects , Coordination Complexes/metabolism , Coordination Complexes/pharmacology , Coordination Complexes/therapeutic use , Disease Models, Animal , Drug Resistance/drug effects , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Parasitic Sensitivity Tests , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Structure-Activity Relationship , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxin-Disulfide Reductase/metabolism , Trichomonas Infections/drug therapy , Trichomonas Infections/parasitology , Trichomonas vaginalis/drug effects , Trophozoites/drug effects
18.
ACS Appl Mater Interfaces ; 13(18): 21108-21118, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33942607

ABSTRACT

Chronic lymphocytic leukemia (CLL) is still incurable by conventional chemotherapy due to the resistance to apoptosis. We have previously found that a peptide-capped gold cluster (Au25Sv9) can target on the aberrant oxidative stress in CLL cells to specially inhibit thioredoxin reductase (TrxR) activity, resulting in significant apoptosis. However, the required doses of the gold cluster for inducing apoptosis are high, restricting its potential for further applications. Notably, the most recent studies suggested that CLL cells overexpressed antiapoptotic BCL-2 protein to prevent chemotherapy-induced apoptosis, indicating that BCL-2 could be a promising target for CLL therapy. Regrettably, the nonmitochondrial-targeted Au25Sv9 has little effect on BCL-2. In this study, we successfully screened a modified BADBH3 peptide (B1P) that could antagonize BCL-2 protein in CLL cells. We found that B1P could effectively sensitize MEC-1 cells to a subliminal dose of Au25Sv9. To simplify the treatment regimen, we directly fabricated a gold cluster capped with the B1P peptides by one-step synthesis to integrate the BCL-2 antagonistic activity into the gold the cluster, named BGC. We already found that low doses of BGC could significantly induce more apoptosis in MEC-1 cells than equivalent doses of the Au25Sv9 cluster or B1P peptide alone. Mechanistically, in addition to the inherent inhibitory effect of gold clusters on TrxR activity, BGC could bind to BCL-2 on mitochondria and activate the BCL-2 family-mediated mitochondrial apoptosis cascade more effectively. These results demonstrated that antagonizing the overexpressed BCL-2 in CLL cells, together with inhibiting TrxR simultaneously by a single gold cluster, is a promising strategy for the treatment of CLL cells. This study will provide a paradigm and reference for the development of functionalized gold clusters with rationally designed peptides, and opens up a new opportunity for the treatment of CLL in clinical settings.


Subject(s)
Antineoplastic Agents/pharmacology , Gold/chemistry , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Peptides/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Amino Acid Sequence , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/enzymology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Peptides/chemistry , Proto-Oncogene Proteins c-bcl-2/chemistry , Reactive Oxygen Species/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
19.
ACS Nano ; 15(5): 9176-9185, 2021 05 25.
Article in English | MEDLINE | ID: mdl-33939413

ABSTRACT

Gold complexes have been recognized as potential anticancer agents against various kinds of diseases due to their inherent suppressions of antioxidant thioredoxin reductase (TrxR) activity. Herein, a powerful aggregation-induced emission luminogen (AIEgen), TBP-Au, was designed and synthesized by integrating an anticancer Au(I) moiety with an AIE-active photosensitizer (TBP), in which both the production and consumption routes of reactive oxygen species (ROS) were elaborately considered simultaneously to boost the anticancer efficacy. It has been demonstrated that TBP-Au could realize superior two-photon fluorescence imaging in tumor tissues with high resolution and deep penetration as well as long-term imaging in live animals due to its AIE property. In addition, the introduction of a special Au(I) moiety could tune the organelle specificity and efficiently facilitate the ROS-determined photodynamic therapy (PDT). More impressively, TBP-Au could efficiently eliminate cancer cells under light irradiation through the preconceived synergetic approaches from the PDT and the effective suppression of TrxR, demonstrating that TBP-Au holds great potential for precise cancer theranostics.


Subject(s)
Gold , Neoplasms/drug therapy , Photochemotherapy , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Animals , Antioxidants , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use
20.
Apoptosis ; 26(5-6): 253-260, 2021 06.
Article in English | MEDLINE | ID: mdl-33860865

ABSTRACT

Tuberous sclerosis, angiomyolipoma and lymphangioleiomyomatosis are a group of diseases characterized by mutation in tuberous sclerosis genes (TSC 1-2). TSC mutation leads to continuous activation of the mTOR pathway that requires adaptation to increased ATP requirement. With limited treatment options, there is an increasing demand to identify novel therapeutic targets and to understand the correlations between mTOR pathway activation and the lack of cell death in the presence of TSC mutation. In the current study, we demonstrate deregulation of p53 controlled and mitochondria associated cell death processes. The study also reveals that treatment of TSC mutant cells with the drug candidate Proxison combined with reduced concentration of rapamycin can increase production of reactive oxygen species (ROS), can modify miRNA expression pattern associated with p53 regulation and can reduce cell viability.


Subject(s)
Apoptosis/genetics , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Tuberous Sclerosis Complex 1 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/genetics , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Flavonoids/pharmacology , Humans , MicroRNAs/genetics , Mitochondria/metabolism , Mutation , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Tumor Suppressor Protein p53/genetics
SELECTION OF CITATIONS
SEARCH DETAIL