2,910 results on '"Auranofin"'
Search Results
2. Gold(I) ion and the phosphine ligand are necessary for the anti-Toxoplasma gondii activity of auranofin.
- Author
-
Ma, C, Tirtorahardjo, J, Schweizer, S, Zhang, J, Fang, Z, Xing, L, Xu, M, Herman, D, Kleinman, M, McCullough, B, Barrios, A, and Andrade, Rosa
- Subjects
Toxoplasma ,auranofin ,drug discovery ,gold ,repurposing ,Humans ,Auranofin ,Gold ,Toxoplasma ,Ligands ,Aurothioglucose ,Arthritis ,Rheumatoid ,Gold Sodium Thiomalate ,Toxoplasmosis ,Antiparasitic Agents ,Phosphines - Abstract
Auranofin, an FDA-approved drug for rheumatoid arthritis, has emerged as a promising antiparasitic medication in recent years. The gold(I) ion in auranofin is postulated to be responsible for its antiparasitic activity. Notably, aurothiomalate and aurothioglucose also contain gold(I), and, like auranofin, they were previously used to treat rheumatoid arthritis. Whether they have antiparasitic activity remains to be elucidated. Herein, we demonstrated that auranofin and similar derivatives, but not aurothiomalate and aurothioglucose, inhibited the growth of Toxoplasma gondii in vitro. We found that auranofin affected the T. gondii biological cycle (lytic cycle) by inhibiting T. gondiis invasion and triggering its egress from the host cell. However, auranofin could not prevent parasite replication once T. gondii resided within the host. Auranofin treatment induced apoptosis in T. gondii parasites, as demonstrated by its reduced size and elevated phosphatidylserine externalization (PS). Notably, the gold from auranofin enters the cytoplasm of T. gondii, as demonstrated by scanning transmission electron microscopy-energy dispersive X-ray spectroscopy (STEM-EDS) and Inductively Coupled Plasma-Mass Spectrometry (ICP-MS).IMPORTANCEToxoplasmosis, caused by Toxoplasma gondii, is a devastating disease affecting the brain and the eyes, frequently affecting immunocompromised individuals. Approximately 60 million people in the United States are already infected with T. gondii, representing a population at-risk of developing toxoplasmosis. Recent advances in treating cancer, autoimmune diseases, and organ transplants have contributed to this at-risk populations exponential growth. Paradoxically, treatments for toxoplasmosis have remained the same for more than 60 years, relying on medications well-known for their bone marrow toxicity and allergic reactions. Discovering new therapies is a priority, and repurposing FDA-approved drugs is an alternative approach to speed up drug discovery. Herein, we report the effect of auranofin, an FDA-approved drug, on the biological cycle of T. gondii and how both the phosphine ligand and the gold molecule determine the anti-parasitic activity of auranofin and other gold compounds. Our studies would contribute to the pipeline of candidate anti-T. gondii agents.
- Published
- 2024
3. Sirolimus and Auranofin in Treating Patients With Advanced or Recurrent Non-Small Cell Lung Cancer or Small Cell Lung Cancer
- Author
-
National Cancer Institute (NCI)
- Published
- 2024
4. Gold complexes: a new frontier in the battle against lung cancer.
- Author
-
Hanif, Aqsa, Al-Rawi, Sawsan S., Ibrahim, Ahmad H., Salman, Abbas Washeel, Zafar, Ayesha, Iqbal, Muhammad Adnan, Batool, Sana, and Abbas, Ghazanfar
- Abstract
Lung cancer is the second leading cause of the mortality related to the cancer. So, it is very necessary to explore the novel strategies to eradicate it. Currently, gold based medicinal compounds have emerged as remarkable anticancer agents and expressed strong potential against the lung cancer cell lines. This review provides a comprehensive overview of the history, advancements, and recent state of gold complexes in the treatment of the lung cancer. We discuss the biological evaluation in relation with the chemical structures of numerous gold complexes, including those featuring thiosemicarbazone,
N -heterocyclic imine,N -heterocyclic carbenes, steroidyl NHC, CAACs, carbamates, and diphosphanes as ligands. Gold complexes’ cytotoxicity has been assessed and contrasted with that of standard drugs such as auranofin and cisplatin, with a special focus on IC50 values for evaluating potency. This review targets to deliver a detailed understanding of the potential of the gold complexes in lung cancer therapies, paving way for the future research and clinical applications. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
5. On the binding of auranofin to Prdx6 and its potential role in cancer cell sensitivity to treatment.
- Author
-
Inague, Alex, Nakahata, Douglas H., Viviani, Lucas G., Alegria, Thiago G.P., Lima, Rodrigo S., Iijima, Thais S., Netto, Luís Eduardo S., Angeli, José Pedro F., Miyamoto, Sayuri, and de Paiva, Raphael E.F.
- Subjects
- *
AURANOFIN , *GOLD compounds , *CYTOTOXINS , *CELL anatomy , *MASS spectrometry - Abstract
In this study, we demonstrate that ferroptosis is a component of the cell death mechanism induced by auranofin in HT-1080 cells, in contrast to the gold(III) compounds [Au(phen)Cl 2 ]PF 6 and [Au(bnpy)Cl 2 ]. Additionally, we identify a potential role of Prdx6 in modulating the sensitivity of A-375 cells to auranofin treatment, whereas the gold(III) compounds evaluated here exhibit Prdx6-independent cytotoxicity. Finally, using mass spectrometry, we show that auranofin binds selectively to the catalytic Cys47 residue of Prdx6 in vitro under acidic conditions. No binding was observed with the C47S mutant or at neutral pH. [Display omitted] • Ferroptosis and Prdx6 role in protection were evaluated for three gold compounds. • The Au(I) compound auranofin induced ferroptosis in HT-1080 cells, while Au(III) compounds did not. • Knockout of Prdx6 in A-375 cells led to higher sensitivity, only in the case of auranofin treatment. • Auranofin binds to Prdx6 in vitro at Cys47, in acidic pH. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Oxidative Stress Mechanism by Gold Compounds: A Close Look at Total ROS Increase and the Inhibition of Antioxidant Enzymes.
- Author
-
Castro, Jennyfer, Clauss, Gustavo, Fontes, Josielle V., Oliveira, Laiane S., and Abbehausen, Camilla
- Subjects
- *
GLUTATHIONE reductase , *GOLD compounds , *THIOREDOXIN , *OXIDATIVE stress , *CHEMICAL structure - Abstract
The antitumor activity of various gold compounds is a promising field of investigation, attracting researchers seeking potential clinical candidates. To advance this research, they explore the complex mechanisms of action of these compounds. Since the discovery of the strong inhibition of thioredoxin reductase by auranofin, the primary mechanism explored has been the inhibition of this enzyme. This inhibition disrupts the redox balance in cells, promoting oxidative stress and triggering cell death. In this review, we analyzed studies from the past decade that measured cellular ROS increase and examined the coordination structures of gold compounds. We also correlate ROS increase with the inhibition of redox‐regulating enzymes, thioredoxin reductase, and glutathione reductase, to elucidate the relationship between these cellular effects and chemical structures. Our data compilation reveals that different structures exhibit varying efficacy: some significantly increase ROS production and inhibit thioredoxin reductase or glutathione reductase, while others elevate ROS levels without affecting these target enzymes, suggesting alternative mechanisms of action. This review consolidates critical evidence, enhancing our understanding of the mechanisms by which these gold complexes act and providing valuable insights for developing new therapeutic strategies against tumor cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Multifaceted Proteome Analysis at Solubility, Redox, and Expression Dimensions for Target Identification.
- Author
-
Saei, Amir A., Lundin, Albin, Lyu, Hezheng, Gharibi, Hassan, Luo, Huqiao, Teppo, Jaakko, Zhang, Xuepei, Gaetani, Massimiliano, Végvári, Ákos, Holmdahl, Rikard, Gygi, Steven P., and Zubarev, Roman A.
- Subjects
- *
BIOLOGICAL systems , *DENDRITIC cells , *MASS spectrometry , *INTERFERONS , *AURANOFIN , *PROTEOMICS , *TYPE I interferons , *INTERFERON receptors - Abstract
Multifaceted interrogation of the proteome deepens the system‐wide understanding of biological systems; however, mapping the redox changes in the proteome has so far been significantly more challenging than expression and solubility/stability analyses. Here, the first high‐throughput redox proteomics approach integrated with expression analysis (REX) is devised and combined with the Proteome Integral Solubility Alteration (PISA) assay. The whole PISA‐REX experiment with up to four biological replicates can be multiplexed into a single tandem mass tag TMTpro set. For benchmarking this compact tool, HCT116 cells treated with auranofin are analyzed, showing great improvement compared with previous studies. PISA‐REX is then applied to study proteome remodeling upon stimulation of human monocytes by interferon α (IFN‐α). Applying this tool to study the proteome changes in plasmacytoid dendritic cells (pDCs) isolated from wild‐type versus Ncf1‐mutant mice treated with interferon α, shows that NCF1 deficiency enhances the STAT1 pathway and modulates the expression, solubility, and redox state of interferon‐induced proteins. Providing comprehensive multifaceted information on the proteome, the compact PISA‐REX has the potential to become an industry standard in proteomics and to open new windows into the biology of health and disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Auranofin is active against Histoplasma capsulatum and reduces the expression of virulence-related genes.
- Author
-
de Abreu Almeida, Marcos, Baeza, Lilian Cristiane, Silva, Leandro B. R., Bernardes-Engemann, Andréa Reis, Almeida-Silva, Fernando, Coelho, Rowena Alves, de Andrade, Iara Bastos, Corrêa-Junior, Dario, Frases, Susana, Zancopé-Oliveira, Rosely Maria, Alanio, Alexandre, Taborda, Carlos Pelleschi, and Almeida-Paes, Rodrigo
- Subjects
- *
TENEBRIO molitor , *DRUG efficacy , *YEAST fungi , *AURANOFIN , *MYCOSES , *HISTOPLASMOSIS - Abstract
Background: Auranofin is an approved anti-rheumatic drug that has a broad-range inhibitory action against several microorganisms, including human pathogenic fungi. The auranofin activity against Histoplasma capsulatum, the dimorphic fungus that causes histoplasmosis, has not been properly addressed. Since there are few therapeutic options for this life-threatening systemic mycosis, this study evaluated the effects of auranofin on H. capsulatum growth and expression of virulence factors. Methodology/principal findings: Minimal inhibitory and fungicidal concentrations (MIC and MFC, respectively) of auranofin against 15 H. capsulatum strains with distinct genetic backgrounds were determined using the yeast form of the fungus and a microdilution protocol. Auranofin activity was also assessed on a macrophage model of infection and on a Tenebrio molitor invertebrate animal model. Expression of virulence-related genes was compared between auranofin treated and untreated H. capsulatum yeast cells using a quantitative PCR assay. Auranofin affected the growth of different strains of H. capsulatum, with MIC and MFC values ranging from 1.25 to 5.0 μM and from 2.5 to >10 μM, respectively. Auranofin was able to kill intracellular H. capsulatum yeast cells and conferred protection against the fungus in the experimental animal model of infection. Moreover, the expression of catalase A, HSP70, superoxide dismutase, thioredoxin reductase, serine proteinase, cytochrome C peroxidase, histone 2B, formamidase, metallopeptidase, Y20 and YPS3 proteins were reduced after six hours of auranofin treatment. CONCLUSIONS/SIGNIFICANCE: Auranofin is fungicidal against H. capsulatum and reduces the expression of several virulence-related genes, which makes this anti-rheumatic drug a good candidate for new medicines against histoplasmosis. Author summary: Histoplasmosis is a serious disease caused by the fungus Histoplasma capsulatum, which can be life-threatening and has limited treatment options. In this study, we investigated the potential of auranofin, a drug originally approved for treating rheumatoid arthritis, to combat H. capsulatum. We tested auranofin against different strains of the fungus and found that it effectively inhibited fungal growth. This drug was also effective in killing the fungus within cells from the immune system and provided protection in an experimental animal model. Furthermore, auranofin reduced the expression of several genes associated with the fungus's ability to cause disease. These genes are involved in critical functions such as detoxifying harmful substances and maintaining cellular structures. By suppressing these genes, auranofin hampers the fungus's ability to survive and cause infection. These findings suggest that auranofin could be repurposed as a treatment for histoplasmosis, offering a new avenue for therapy against this challenging infection. This research is significant because it highlights the potential of existing drugs to treat other diseases, broadening our arsenal of available treatments and contributing to better healthcare solutions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Auranofin as a Novel Anticancer Drug for Anaplastic Thyroid Cancer.
- Author
-
An, Seung-Chan, Jun, Hak Hoon, Kim, Kyeong Mi, Kim, Issac, Choi, Sujin, Yeo, Hyunjeong, Lee, Soonchul, and An, Hyun-Ju
- Subjects
- *
ANAPLASTIC thyroid cancer , *CANCER chemotherapy , *CANCER stem cells , *SUBCUTANEOUS injections , *DRUG repositioning - Abstract
Background/Objectives: Anaplastic thyroid cancer (ATC) is an aggressive and rare cancer with a poor prognosis, and traditional therapies have limited efficacy. This study investigates drug repositioning, focusing on auranofin, a gold-based drug originally used for rheumatoid arthritis, as a potential treatment for ATC. Methods: Auranofin was identified from an FDA-approved drug library and tested on two thyroid cancer cell lines, 8505C and FRO. Antitumor efficacy was evaluated through gene and protein expression analysis using Western blot, FACS, and mRNA sequencing. In vivo experiments were conducted using subcutaneous injections in nude mice to confirm the anticancer effects of auranofin. Results: Auranofin induced reactive oxygen species (ROS) production and apoptosis, leading to a dose-dependent reduction in cell viability, G1/S phase cell cycle arrest, and altered expression of regulatory proteins. It also inhibited cancer stem cell activity and suppressed epithelial–mesenchymal transition. mRNA sequencing revealed significant changes in the extracellular matrix–receptor interaction pathway, supported by Western blot results. In vivo xenograft models demonstrated strong antitumor activity. Conclusions: Auranofin shows promise as a repurposed therapeutic agent for ATC, effectively inhibiting cell proliferation, reducing metastasis, and promoting apoptosis. These findings suggest that auranofin could play a key role in future ATC treatment strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Structure, stability, and dynamics of inclusion complexes formed from auranofin derivatives and hydroxypropyl-β-cyclodextrin.
- Author
-
Dos Santos, Hélio F. and Anconi, Cleber P. A.
- Abstract
The inclusion compounds of auranofin (AF) and its iodide derivative (AF-I) with HP-β-CD were recently identified and characterized experimentally. In the present work, classical molecular dynamics and the quantum computational GFN2-xTB method were applied to investigate the inclusion processes. As a result, both approaches identified AF-I@HP-β-CD as the most favourable system, as observed experimentally. The higher stability of AF-I@HP-β-CD was explained by entropy and solvation factors, with the GFN2-xTB method providing a stability constant (logK
1:1 ) in good agreement with the experimental results: 0.21–1.21 for AF@HP-β-CD and 1.31–2.33 for AF-I@HP-β-CD (the experimental values are 1.48 and 2.52, respectively). The preferred inclusion mode for AF-I@HP-β-CD has a triethylphosphine (-PEt3 ) group pointed towards the head portion of the HP-β-CD where the HP groups are attached (labelled P2). The P2 mode showed short contacts between -CH2 CH3 groups (-PEt3 ) and -H-3 only (inside the CD cavity), which is also supported by ROESY experiments. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
11. Synergistic Dual Targeting of Thioredoxin and Glutathione Systems Irrespective of p53 in Glioblastoma Stem Cells.
- Author
-
Jamali, Fatemeh, Lan, Katherine, Daniel, Paul, Petrecca, Kevin, Sabri, Siham, and Abdulkarim, Bassam
- Subjects
REACTIVE oxygen species ,DRUG repositioning ,AURANOFIN ,CYTOTOXINS ,BRAIN cancer - Abstract
Glioblastoma (GBM) is an incurable primary brain cancer characterized by increased reactive oxygen species (ROS) production. The redox-sensitive tumor suppressor gene TP53, wild-type (wt) for 70% of patients, regulates redox homeostasis. Glioblastoma stem cells (GSCs) increase thioredoxin (Trx) and glutathione (GSH) antioxidant systems as survival redox-adaptive mechanisms to maintain ROS below the cytotoxic threshold. Auranofin, an FDA-approved anti-rheumatoid drug, inhibits thioredoxin reductase 1 (TrxR1). L-buthionine sulfoximine (L-BSO) and the natural product piperlongumine (PPL) inhibit the GSH system. We evaluated the cytotoxic effects of Auranofin alone and in combination with L-BSO or PPL in GBM cell lines and GSCs with a known TP53 status. The Cancer Genome Atlas/GBM analysis revealed a significant positive correlation between wtp53 and TrxR1 expression in GBM. Auranofin induced ROS-dependent cytotoxicity within a micromolar range in GSCs. Auranofin decreased TrxR1 expression, AKT (Ser-473) phosphorylation, and increased p53, p21, and PARP-1 apoptotic cleavage in wtp53-GSCs, while mutant-p53 was decreased in a mutant-p53 GSC line. Additionally, p53-knockdown in a wtp53-GSC line decreased TrxR1 expression and significantly increased sensitivity to Auranofin, suggesting the role of wtp53 as a negative redox-sensitive mechanism in response to Auranofin in GSCs. The combination of Auranofin and L-BSO synergistically increased ROS, decreased IC50s, and induced long-term cytotoxicity irrespective of p53 in GBM cell lines and GSCs. Intriguingly, Auranofin increased the expression of glutathione S-transferase pi-1 (GSTP-1), a target of PPL. Combining Auranofin with PPL synergistically decreased IC50s to a nanomolar range in GSCs, supporting the potential to repurpose Auranofin and PPL in GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Targeting the redox vulnerability of acute myeloid leukaemia cells with a combination of auranofin and vitamin C.
- Author
-
Hei, Zhiliang, Yang, Shujun, Ouyang, Guifang, Hanna, Jolimar, Lepoivre, Michel, Huynh, Tony, Aguinaga, Lorea, Cassinat, Bruno, Maslah, Nabih, Bourge, Mickaël, Golinelli‐Cohen, Marie‐Pierre, Guittet, Olivier, Vallières, Cindy, Vernis, Laurence, Fenaux, Pierre, and Huang, Meng‐Er
- Subjects
- *
ACUTE myeloid leukemia , *VITAMIN C , *CORD blood , *REACTIVE oxygen species , *MYELOID cells - Abstract
Summary: Acute myeloid leukaemia (AML) is a heterogeneous disease characterized by complex molecular and cytogenetic abnormalities. Pro‐oxidant cellular redox status is a common hallmark of AML cells, providing a rationale for redox‐based anticancer strategy. We previously discovered that auranofin (AUF), initially used for the treatment of rheumatoid arthritis and repositioned for its anticancer activity, can synergize with a pharmacological concentration of vitamin C (VC) against breast cancer cell line models. In this study, we observed that this drug combination synergistically and efficiently killed cells of leukaemic cell lines established from different myeloid subtypes. In addition to an induced elevation of reactive oxygen species and ATP depletion, a rapid dephosphorylation of 4E‐BP1 and p70S6K, together with a strong inhibition of protein synthesis were early events in response to AUF/VC treatment, suggesting their implication in AUF/VC‐induced cytotoxicity. Importantly, a study on 22 primary AML specimens from various AML subtypes showed that AUF/VC combinations at pharmacologically achievable concentrations were effective to eradicate primary leukaemic CD34+ cells from the majority of these samples, while being less toxic to normal cord blood CD34+ cells. Our findings indicate that targeting the redox vulnerability of AML with AUF/VC combinations could present a potential anti‐AML therapeutic approach. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Auranofin Suppresses the Growth of Canine Mammary Tumour Cells and Induces Apoptosis via the PI3K/AKT Pathway.
- Author
-
Lin, Zhaoyan, Chen, Rong, Wang, Jiao, Zheng, Yu, He, Zixuan, Yan, Ye, Zhang, Linxi, Huang, Xiaohong, and Zhang, Hong
- Subjects
- *
PI3K/AKT pathway , *FEMALE dogs , *GOLD compounds , *MAMMARY glands , *CELL lines - Abstract
Canine mammary gland tumour (CMT) is the most common spontaneous tumour in intact female dogs and often exhibits metastases. Auranofin (AF) is a gold complex used for treating rheumatism. The excellent anti‐tumour ability of AF has been demonstrated in various types of human and canine tumours. In this study, five CMT cell lines (CIPp, CMT‐7364, CHMp, CIPm and CTBp) and three CMT primary cells (G7894, L1883 and L6783) were used to explore the anti‐tumour effect of AF on CMT. Two CMT cell lines (CIPp and CMT‐7364) were used to search the underlying mechanism of the effect of AF on CMT. The results showed that AF inhibited the growth, migration, invasion, and colony formation abilities of CMT cells. Additionally, the growth of CMT in a 3D cell culture model was effectively suppressed by AF. Furthermore, AF induced cell apoptosis of CMT cells via the PI3K/AKT pathway. In conclusion, AF effectively induces CMT apoptosis by regulating the PI3K/AKT pathway, indicating that AF should be explored as a potential CMT treatment in future studies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Auranofin-loaded PLGA nanoparticles alleviate cognitive deficit induced by streptozotocin in rats model: modulation of oxidative stress, neuroinflammatory markers, and neurotransmitters.
- Author
-
Kushawaha, Shiv Kumar, Ashawat, Mahendra Singh, and Baldi, Ashish
- Abstract
Alzheimer's disease remains an unsolved neurological puzzle with no cure. Current therapies offer only symptomatic relief, hindered by limited uptake through the blood–brain barrier. Auranofin, an FDA-approved compound, exhibits potent antioxidative and anti-inflammatory properties targeting brain disorders. Yet, its oral bioavailability challenge prompts the exploration of nanoformulation-based solutions enhancing blood–brain barrier penetrability. The study aimed to investigate the neuroprotective potential of auranofin nanoparticles in streptozotocin-induced AD rats. Auranofin-containing polylactic-co-glycolic acid nanoparticles were formulated by the multiple emulsion solvent evaporation method. Characterization was done by determining entrapment efficiency, particle size distribution, surface charge, and morphology. An in vivo study was performed by administering streptozotocin (3 mg/kg/i.c.v., days 1 and 3), auranofin (5 and 10 mg/kg), auranofin nanoparticles (2.5 and 5 mg/kg), and donepezil (2 mg/kg) for 14 days orally. Behavioral deficits were evaluated using the open field test, Morris water maze, objective recognition test, change in oxidative stress levels, and AD markers in the brain. Following the decapitation of the rats, the brains were excised to isolate the hippocampus. Subsequent analyses included the quantification of biochemical and neuroinflammatory markers, as well as the assessment of neurotransmitter levels. The characterization of auranofin nanoparticles showed an entrapment efficiency of 98%, an average particle size of 101.5 ± 10.3 nm, a surface charge of 27.5 ± 5.10 mV, and a polydispersity index of 0.438 ± 0.12. In vivo, administration of auranofin and auranofin nanoparticles significantly reversed streptozotocin-induced cognitive deficits, biochemical alteration, neuroinflammatory markers, and neurotransmitter levels. The present finding suggests that auranofin nanoparticles have more significant neuroprotective potential than auranofin alone. The therapeutic efficacy may be attributed to its antioxidant and anti-inflammatory properties, as well as its positive neuromodulatory effects. Therefore, our findings suggest that it could be a promising candidate for Alzheimer's disease therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Pro-Oxidant Auranofin and Glutathione-Depleting Combination Unveils Synergistic Lethality in Glioblastoma Cells with Aberrant Epidermal Growth Factor Receptor Expression.
- Author
-
Martinez-Jaramillo, Elvis, Jamali, Fatemeh, Abdalbari, Farah H., Abdulkarim, Bassam, Jean-Claude, Bertrand J., Telleria, Carlos M., and Sabri, Siham
- Subjects
- *
GLUTATHIONE , *PROTEINS , *COMBINATION drug therapy , *IN vitro studies , *GLIOMAS , *RESEARCH funding , *ANTIRHEUMATIC agents , *CELL lines , *GENE expression , *REACTIVE oxygen species , *CYTOTOXINS , *EPIDERMAL growth factor receptors , *DRUG resistance - Abstract
Simple Summary: Glioblastoma (GBM) is a fatal brain cancer. Over 50% of GBM tumors overexpress EGFR, a protein involved in tumor progression and drug resistance. The thioredoxin and glutathione antioxidant systems counteract oxidative stress, leading to drug resistance in GBM. In this study, we aimed to investigate the impact of EGFR overexpression on the response to the thioredoxin inhibitor and arthritis medication, auranofin and glutathione inhibitor L-buthionine-sulfoximine (L-BSO). Using EGFR-negative and two derived EGFR-overexpressing GBM cells, we validated the hypothesis according to which the auranofin and L-BSO combination reach the lethal threshold of oxidative stress. Auranofin increased oxidative stress, DNA damage, and cell killing while decreasing EGFR. Auranofin and L-BSO combination induced further cell killing, revealing the vulnerability of EGFR in response to co-targeting the thioredoxin and glutathione systems. Our findings unravel the significance of impairing antioxidant defense in EGFR-driven cancer and repurposing auranofin and L-BSO treatment to improve the dismal survival of patients with GBM. Glioblastoma (GBM) is the most prevalent and advanced malignant primary brain tumor in adults. GBM frequently harbors epidermal growth factor receptor (EGFR) wild-type (EGFRwt) gene amplification and/or EGFRvIII activating mutation. EGFR-driven GBM relies on the thioredoxin (Trx) and/or glutathione (GSH) antioxidant systems to withstand the excessive production of reactive oxygen species (ROS). The impact of EGFRwt or EGFRvIII overexpression on the response to a Trx/GSH co-targeting strategy is unknown. In this study, we investigated Trx/GSH co-targeting in the context of EGFR overexpression in GBM. Auranofin is a thioredoxin reductase (TrxR) inhibitor, FDA-approved for rheumatoid arthritis. L-buthionine-sulfoximine (L-BSO) inhibits GSH synthesis by targeting the glutamate–cysteine ligase catalytic (GCLC) enzyme subunit. We analyzed the mechanisms of cytotoxicity of auranofin and the interaction between auranofin and L-BSO in U87MG, U87/EGFRwt, and U87/EGFRvIII GBM isogenic GBM cell lines. ROS-dependent effects were assessed using the antioxidant N-acetylsteine. We show that auranofin decreased TrxR1 activity and increased ROS. Auranofin decreased cell vitality and colony formation and increased protein polyubiquitination through ROS-dependent mechanisms, suggesting the role of ROS in auranofin-induced cytotoxicity in the three cell lines. ROS-dependent PARP-1 cleavage was associated with EGFRvIII downregulation in U87/EGFRvIII cells. Remarkably, the auranofin and L-BSO combination induced the significant depletion of intracellular GSH and synergistic cytotoxicity regardless of EGFR overexpression. Nevertheless, molecular mechanisms associated with cytotoxicity were modulated to a different extent among the three cell lines. U87/EGFRvIII exhibited the most prominent ROS increase, P-AKT(Ser-473), and AKT decrease along with drastic EGFRvIII downregulation. U87/EGFRwt and U87/EGFRvIII displayed lower basal intracellular GSH levels and synergistic ROS-dependent DNA damage compared to U87MG cells. Our study provides evidence for ROS-dependent synergistic cytotoxicity of auranofin and L-BSO combination in GBM in vitro. Unraveling the sensitivity of EGFR-overexpressing cells to auranofin alone, and synergistic auranofin and L-BSO combination, supports the rationale to repurpose this promising pro-oxidant treatment strategy in GBM. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. The Many Lives of Auranofin: How an Old Anti-Rheumatic Agent May Become a Promising Antimicrobial Drug.
- Author
-
Coscione, Francesca, Zineddu, Stefano, Vitali, Valentina, Fondi, Marco, Messori, Luigi, and Perrin, Elena
- Subjects
TREATMENT effectiveness ,ANTIRHEUMATIC agents ,DRUG repositioning ,ANTI-infective agents ,DRUG resistance in microorganisms - Abstract
Auranofin (AF) is a gold-based compound with a well-known pharmacological and toxicological profile, currently used in the treatment of some severe forms of rheumatoid arthritis. Over the last twenty years, AF has also been repurposed as antiviral, antitumor, and antibacterial drug. In this review we focused on the antibacterial properties of AF, specifically researching the minimal inhibitory concentrations (MIC) of AF in both mono- and diderm bacteria reported so far in literature. AF proves to be highly effective against monoderm bacteria, while diderm are far less susceptible, probably due to the outer membrane barrier. We also reported the current mechanistic hypotheses concerning the antimicrobial properties of AF, although a conclusive description of its antibacterial mode of action is not yet available. Even if its mechanism of action has not been fully elucidated yet and further studies are required to optimize its delivery strategy, AF deserves additional investigation because of its unique mode of action and high efficacy against a wide range of pathogens, which could lead to potential applications in fighting antimicrobial resistance and improving therapeutic outcomes in infectious diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Auranofin and reactive oxygen species inhibit protein synthesis and regulate the level of the PLK1 protein in Ewing sarcoma cells.
- Author
-
Haight, Joseph A., Koppenhafer, Stacia L., Geary, Elizabeth L., and Gordon, David J.
- Subjects
EWING'S sarcoma ,REACTIVE oxygen species ,PROTEIN synthesis ,ONCOGENIC proteins ,AURANOFIN ,REDUCTASE inhibitors ,CELL cycle proteins - Abstract
Novel therapeutic approaches are needed for the treatment of Ewing sarcoma tumors. We previously identified that Ewing sarcoma cell lines are sensitive to drugs that inhibit protein translation. However, translational and therapeutic approaches to inhibit protein synthesis in tumors are limited. In this work, we identified that reactive oxygen species, which are generated by a wide range of chemotherapy and other drugs, inhibit protein synthesis and reduce the level of critical proteins that support tumorigenesis in Ewing sarcoma cells. In particular, we identified that both hydrogen peroxide and auranofin, an inhibitor of thioredoxin reductase and regulator of oxidative stress and reactive oxygen species, activate the repressor of protein translation 4E-BP1 and reduce the levels of the oncogenic proteins RRM2 and PLK1 in Ewing and other sarcoma cell lines. These results provide novel insight into the mechanism of how ROSinducing drugs target cancer cells via inhibition of protein translation and identify a mechanistic link between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Comparison of the mechanism of antimicrobial action of the gold(I) compound auranofin in Gram-positive and Gram-negative bacteria
- Author
-
Laísa Quadros Barsé, Agnes Ulfig, Marharyta Varatnitskaya, Melissa Vázquez-Hernández, Jihyun Yoo, Astrid M. Imann, Natalie Lupilov, Marina Fischer, Katja Becker, Julia E. Bandow, and Lars I. Leichert
- Subjects
auranofin ,gold ,drug repurposing ,thioredoxin reductase ,proteomics ,Escherichia coli ,Microbiology ,QR1-502 - Abstract
ABSTRACT While highly effective at killing Gram-positive bacteria, auranofin lacks significant activity against Gram-negative species for reasons that largely remain unclear. Here, we aimed to elucidate the molecular mechanisms underlying the low susceptibility of the Gram-negative model organism Escherichia coli to auranofin when compared to the Gram-positive model organism Bacillus subtilis. The proteome response of E. coli exposed to auranofin suggests a combination of inactivation of thiol-containing enzymes and the induction of systemic oxidative stress. Susceptibility tests in E. coli mutants lacking proteins upregulated upon auranofin treatment suggested that none of them are directly involved in E. coli’s high tolerance to auranofin. E. coli cells lacking the efflux pump component TolC were more sensitive to auranofin treatment, but not to an extent that would fully explain the observed difference in susceptibility of Gram-positive and Gram-negative organisms. We thus tested whether E. coli’s thioredoxin reductase (TrxB) is inherently less sensitive to auranofin than TrxB from B. subtilis, which was not the case. However, E. coli strains lacking the low-molecular-weight thiol glutathione, but not glutathione reductase, showed a high susceptibility to auranofin. Bacterial cells expressing the genetically encoded redox probe roGFP2 allowed us to observe the oxidation of cellular protein thiols in situ. Based on our findings, we hypothesize that auranofin leads to a global disturbance in the cellular thiol redox homeostasis in bacteria, but Gram-negative bacteria are inherently more resistant due to the presence of drug export systems and high cellular concentrations of glutathione.IMPORTANCEAuranofin is an FDA-approved drug for the treatment of rheumatoid arthritis. However, it has also high antibacterial activity, in particular against Gram-positive organisms. In the current antibiotics crisis, this would make it an ideal candidate for drug repurposing. However, its much lower activity against Gram-negative organisms prevents its broad-spectrum application. Here we show that, on the level of the presumed target, there is no difference in susceptibility between Gram-negative and Gram-positive species: thioredoxin reductases from both Escherichia coli and Bacillus subtilis are equally inhibited by auranofin. In both species, auranofin treatment leads to oxidative protein modification on a systemic level, as monitored by proteomics and the genetically encoded redox probe roGFP2. The single largest contributor to E. coli’s relative resistance to auranofin seems to be the low-molecular-weight thiol glutathione, which is absent in B. subtilis and other Gram-positive species.
- Published
- 2024
- Full Text
- View/download PDF
19. Multifaceted Proteome Analysis at Solubility, Redox, and Expression Dimensions for Target Identification
- Author
-
Amir A. Saei, Albin Lundin, Hezheng Lyu, Hassan Gharibi, Huqiao Luo, Jaakko Teppo, Xuepei Zhang, Massimiliano Gaetani, Ákos Végvári, Rikard Holmdahl, Steven P. Gygi, and Roman A. Zubarev
- Subjects
auranofin ,immunology ,interferon ,lupus ,mass spectrometry ,oxidation and reduction ,Science - Abstract
Abstract Multifaceted interrogation of the proteome deepens the system‐wide understanding of biological systems; however, mapping the redox changes in the proteome has so far been significantly more challenging than expression and solubility/stability analyses. Here, the first high‐throughput redox proteomics approach integrated with expression analysis (REX) is devised and combined with the Proteome Integral Solubility Alteration (PISA) assay. The whole PISA‐REX experiment with up to four biological replicates can be multiplexed into a single tandem mass tag TMTpro set. For benchmarking this compact tool, HCT116 cells treated with auranofin are analyzed, showing great improvement compared with previous studies. PISA‐REX is then applied to study proteome remodeling upon stimulation of human monocytes by interferon α (IFN‐α). Applying this tool to study the proteome changes in plasmacytoid dendritic cells (pDCs) isolated from wild‐type versus Ncf1‐mutant mice treated with interferon α, shows that NCF1 deficiency enhances the STAT1 pathway and modulates the expression, solubility, and redox state of interferon‐induced proteins. Providing comprehensive multifaceted information on the proteome, the compact PISA‐REX has the potential to become an industry standard in proteomics and to open new windows into the biology of health and disease.
- Published
- 2024
- Full Text
- View/download PDF
20. The rheumatoid arthritis drug auranofin exerts potent anti-lymphoma effect by stimulating TXNRD-mediated ROS generation and inhibition of energy metabolism
- Author
-
Mengqi Yang, Jiaxin Liu, Jianan Li, Shijun Wen, Yumin Hu, Wenhua Lu, Jinyun Liu, Peng Huang, and Panpan Liu
- Subjects
Lymphoma ,Auranofin ,TXNRD ,ROS ,ATP ,Immune checkpoint ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Since the survival of lymphoma patients who experience disease progression or relapse remains very poor, new therapeutic approaches and effective drugs are urgently needed. Here we show that auranofin (AF), an anti-rheumatoid drug thought to inhibit thioredoxin reductases (TXNRDs) as its mechanism of action, exhibited potent activity against multiple cancer types, especially effective against B cell lymphoma. Surprisingly, a knockdown of TXNRD1 and TXNRD2 did not cause significant cytotoxicity, suggesting that abrogation of TXNRD enzyme per se was insufficient to cause cancer cell death. Further mechanistic study showed that the interaction of AF with TXNRD could convert this antioxidant enzyme to a ROS-generating molecule via disrupting its electron transport, leading to a leak of electrons that interact with molecular oxygen to form superoxide. AF also suppressed energy metabolism by inhibiting both mitochondria complex II and the glycolytic enzyme GAPDH, leading to a significant depletion of ATP and inhibition of cancer growth in vitro and in vivo. Importantly, we found that the AF-mediated ROS stress could induce PD-L1 expression, revealing an unwanted effect of AF in causing immune suppression. We further showed that a combination of AF with anti-PD-1 antibody could enhance the anticancer activity in a syngeneic immune-competent mouse B-cell lymphoma model. Our study suggests that AF could be a potential drug for lymphoma treatment, and its combination with immune checkpoint inhibitors would be a logical strategy to increase the therapeutic activity.
- Published
- 2024
- Full Text
- View/download PDF
21. Auranofin loaded silk fibroin nanoparticles for colorectal cancer treatment
- Author
-
Pérez-Lloret, Marta, Reidy, Eileen, Lozano-Pérez, Antonio Abel, Marchal, Juan A., Lens, Piet N. L., Ryan, Aideen E., and Erxleben, Andrea
- Published
- 2024
- Full Text
- View/download PDF
22. Hypomorphic NOTCH1 Expression Alters Cardiomyocyte Cellular Architecture in Hypoplastic Left Heart Syndrome
- Subjects
Heart diseases ,Genetic disorders ,Stem cells ,Hypoplastic left heart syndrome ,Auranofin ,Physical fitness ,Health - Abstract
2024 OCT 19 (NewsRx) -- By a News Reporter-Staff News Editor at Obesity, Fitness & Wellness Week -- According to news reporting based on a preprint abstract, our journalists obtained [...]
- Published
- 2024
23. Findings from Sun Yat-sen University Update Knowledge of Apoptosis (Novel Gold-based Complex Gc7 Suppresses Cancer Cell Proliferation Via Impacting Energy Metabolism Mediated By Mitochondria)
- Subjects
Auranofin ,Cancer ,Apoptosis ,Physical fitness ,Health - Abstract
2024 OCT 12 (NewsRx) -- By a News Reporter-Staff News Editor at Obesity, Fitness & Wellness Week -- Researchers detail new data in Cellular Physiology - Apoptosis. According to news [...]
- Published
- 2024
24. Recharacterization of RSL3 reveals that the selenoproteome is a druggable target in colorectal cancer (Updated August 28, 2024)
- Subjects
Cell death ,Colorectal cancer ,Auranofin ,Physical fitness ,Health - Abstract
2024 SEP 14 (NewsRx) -- By a News Reporter-Staff News Editor at Obesity, Fitness & Wellness Week -- According to news reporting based on a preprint abstract, our journalists obtained [...]
- Published
- 2024
25. Auranofin inhibition of thioredoxin reductase sensitizes lung neuroendocrine tumor cells (NETs) and small cell lung cancer (SCLC) cells to sorafenib as well as inhibiting SCLC xenograft growth
- Author
-
Spenser S. Johnson, Dijie Liu, Jordan T. Ewald, Claudia Robles-Planells, Casey Pulliam, Keegan A. Christensen, Khaliunaa Bayanbold, Brian R. Wels, Shane R. Solst, M. Sue O’Dorisio, Yusuf Menda, Douglas R. Spitz, and Melissa A. Fath
- Subjects
auranofin ,sorafenib ,small cell lung cancer ,thioredoxin reductase ,lung neuroendocrine tumor ,dms273 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - 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.
- Published
- 2024
- Full Text
- View/download PDF
26. The rheumatoid arthritis drug auranofin lowers leptin levels and exerts antidiabetic effects in obese mice.
- Author
-
Cox, Aaron, Masschelin, Peter, Saha, Pradip, Felix, Jessica, Sharp, Robert, Lian, Zeqin, Xia, Yan, Chernis, Natasha, Bader, David, Kim, Kang, Li, Xin, Yoshino, Jun, Li, Gang, Sun, Zheng, Wu, Huaizhu, Coarfa, Cristian, Moore, David, Klein, Samuel, Sun, Kai, and Hartig, Sean
- Subjects
auranofin ,inflammation ,insulin sensitivity ,leptin reduction ,obesity ,Animals ,Mice ,Mice ,Obese ,Hypoglycemic Agents ,Auranofin ,Diabetes Mellitus ,Type 2 ,Arthritis ,Rheumatoid ,Obesity - Abstract
Low-grade, sustained inflammation in white adipose tissue (WAT) characterizes obesity and coincides with type 2 diabetes mellitus (T2DM). However, pharmacological targeting of inflammation lacks durable therapeutic effects in insulin-resistant conditions. Through a computational screen, we discovered that the FDA-approved rheumatoid arthritis drug auranofin improved insulin sensitivity and normalized obesity-associated abnormalities, including hepatic steatosis and hyperinsulinemia in mouse models of T2DM. We also discovered that auranofin accumulation in WAT depleted inflammatory responses to a high-fat diet without altering body composition in obese wild-type mice. Surprisingly, elevated leptin levels and blunted beta-adrenergic receptor activity achieved by leptin receptor deletion abolished the antidiabetic effects of auranofin. These experiments also revealed that the metabolic benefits of leptin reduction were superior to immune impacts of auranofin in WAT. Our studies uncover important metabolic properties of anti-inflammatory treatments and contribute to the notion that leptin reduction in the periphery can be accomplished to treat obesity and T2DM.
- Published
- 2022
27. FDA-approved disulfiram as a novel treatment for aggressive leukemia.
- Author
-
Karsa, Mawar, Xiao, Lin, Ronca, Emma, Bongers, Angelika, Spurling, Dayna, Karsa, Ayu, Cantilena, Sandra, Mariana, Anna, Failes, Tim W., Arndt, Greg M., Cheung, Laurence C., Kotecha, Rishi S., Sutton, Rosemary, Lock, Richard B., Williams, Owen, de Boer, Jasper, Haber, Michelle, Norris, Murray D., Henderson, Michelle J., and Somers, Klaartje
- Subjects
- *
CYTARABINE , *DISULFIRAM , *DRUG side effects , *ACUTE leukemia , *LEUKEMIA , *ACUTE myeloid leukemia , *CANCER cell growth - Abstract
Acute leukemia continues to be a major cause of death from disease worldwide and current chemotherapeutic agents are associated with significant morbidity in survivors. While better and safer treatments for acute leukemia are urgently needed, standard drug development pipelines are lengthy and drug repurposing therefore provides a promising approach. Our previous evaluation of FDA-approved drugs for their antileukemic activity identified disulfiram, used for the treatment of alcoholism, as a candidate hit compound. This study assessed the biological effects of disulfiram on leukemia cells and evaluated its potential as a treatment strategy. We found that disulfiram inhibits the viability of a diverse panel of acute lymphoblastic and myeloid leukemia cell lines (n = 16) and patient-derived xenograft cells from patients with poor outcome and treatment-resistant disease (n = 15). The drug induced oxidative stress and apoptosis in leukemia cells within hours of treatment and was able to potentiate the effects of daunorubicin, etoposide, topotecan, cytarabine, and mitoxantrone chemotherapy. Upon combining disulfiram with auranofin, a drug approved for the treatment of rheumatoid arthritis that was previously shown to exert antileukemic effects, strong and consistent synergy was observed across a diverse panel of acute leukemia cell lines, the mechanism of which was based on enhanced ROS induction. Acute leukemia cells were more sensitive to the cytotoxic activity of disulfiram than solid cancer cell lines and non-malignant cells. While disulfiram is currently under investigation in clinical trials for solid cancers, this study provides evidence for the potential of disulfiram for acute leukemia treatment. Key messages: Disulfiram induces rapid apoptosis in leukemia cells by boosting oxidative stress. Disulfiram inhibits leukemia cell growth more potently than solid cancer cell growth. Disulfiram can enhance the antileukemic efficacy of chemotherapies. Disulfiram strongly synergises with auranofin in killing acute leukemia cells by ROS induction. We propose testing of disulfiram in clinical trial for patients with acute leukemia. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Thioredoxin Reductase Inhibitor Suppresses the Local Progression of Rhabdomyosarcoma With PDX Models.
- Author
-
HIDEYUKI KINOSHITA, SEIKO KINOSHITA, HIROTO KAMODA, YOKO HAGIWARA, SEIJI OHTORI, and TSUKASA YONEMOTO
- Abstract
Background/Aim: Chemoresistance in rhabdomyosarcoma (RMS) is associated with poor survival, necessitating the development of novel anticancer drugs. Auranofin (AUR), an anti-rheumatic drug, is a thioredoxin reductase (TXNRD) inhibitor with anticancer properties. Although patient-derived xenograft (PDX) models are essential for studying cancer biology, reports on sarcomas using the PDX model are scarce because of their rarity. This study aimed to investigate the effectiveness of AUR treatment in RMS using a PDX model to evaluate its impact on local progression. Materials and Methods: A 20-year-old woman who was diagnosed with alveolar RMS was used to generate the PDX model. RMS PDX tumors were implanted in nude mice and divided into non-treated (vehicle) and treated (AUR) groups. Tumor volume and weight were evaluated, and immunohistochemical staining was performed to evaluate local progression of the sarcoma. The relationship between the TXNRD-1 expression and survival probability of patients with RMS was evaluated using publicly available expression cohorts. Results: AUR significantly suppressed RMS tumor progression over time. It also significantly suppressed the tumor size and weight at the time of excision. Histological evaluation showed that AUR induced oxidative stress in the PDX mouse models and inhibited the local progression of RMS by inducing apoptosis. High TXNRD-1 expression was found to be a negative prognostic factor for overall survival in patients with RMS. Conclusion: AURinduced inhibition of TXNRDs can significantly impede the local progression of RMS through the oxidative stressapoptosis pathway as demonstrated in PDX models. Thus, targeting TXNRD inhibition may be a promising therapeutic strategy for the treatment of RMS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Auranofin and reactive oxygen species inhibit protein synthesis and regulate the level of the PLK1 protein in Ewing sarcoma cells
- Author
-
Joseph A. Haight, Stacia L. Koppenhafer, Elizabeth L. Geary, and David J. Gordon
- Subjects
reactive oxygen species ,protein synthesis ,auranofin ,4E-BP1 ,RRM2 ,plk1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Novel therapeutic approaches are needed for the treatment of Ewing sarcoma tumors. We previously identified that Ewing sarcoma cell lines are sensitive to drugs that inhibit protein translation. However, translational and therapeutic approaches to inhibit protein synthesis in tumors are limited. In this work, we identified that reactive oxygen species, which are generated by a wide range of chemotherapy and other drugs, inhibit protein synthesis and reduce the level of critical proteins that support tumorigenesis in Ewing sarcoma cells. In particular, we identified that both hydrogen peroxide and auranofin, an inhibitor of thioredoxin reductase and regulator of oxidative stress and reactive oxygen species, activate the repressor of protein translation 4E-BP1 and reduce the levels of the oncogenic proteins RRM2 and PLK1 in Ewing and other sarcoma cell lines. These results provide novel insight into the mechanism of how ROS-inducing drugs target cancer cells via inhibition of protein translation and identify a mechanistic link between ROS and the DNA replication (RRM2) and cell cycle regulatory (PLK1) pathways.
- Published
- 2024
- Full Text
- View/download PDF
30. A CD Study of a Structure-Based Selection of N-Heterocyclic Bis-Carbene Gold(I) Complexes as Potential Ligands of the G-Quadruplex-Forming Human Telomeric hTel23 Sequence
- Author
-
Maria Marzano, Filippo Prencipe, Pietro Delre, Giuseppe Felice Mangiatordi, Gabriele Travagliante, Luisa Ronga, Gennaro Piccialli, Michele Saviano, Stefano D’Errico, Diego Tesauro, and Giorgia Oliviero
- Subjects
auranofin ,cancer ,chemotherapy ,N-heterocyclic carbene gold(I) complexes ,G-quadruplex ,CD spectroscopy ,Organic chemistry ,QD241-441 - Abstract
Herein, we report the structure-based selection via molecular docking of four N-heterocyclic bis-carbene gold(I) complexes, whose potential as ligands for the hTel23 G-quadruplex structure has been investigated using circular dichroism (CD) spectroscopy, CD melting, and polyacrylamide gel electrophoresis (PAGE). The complex containing a bis(1,2,3,4,6,7,8,9-octahydro-11H-11λ3-pyridazino[1,2-a]indazol-11-yl) scaffold induces a transition from the hybrid (3 + 1) topology to a prevalent parallel G-quadruplex conformation, whereas the complex featuring a bis(2-(2-acetamidoethyl)-3λ3-imidazo[1,5-a]pyridin-3(2H)-yl) moiety disrupted the original G-quadruplex structure. These results deserve particular attention in light of the recent findings on the pathological involvements of G-quadruplexes in neurodegenerative diseases.
- Published
- 2024
- Full Text
- View/download PDF
31. Auranofin as a Novel Anticancer Drug for Anaplastic Thyroid Cancer
- Author
-
Seung-Chan An, Hak Hoon Jun, Kyeong Mi Kim, Issac Kim, Sujin Choi, Hyunjeong Yeo, Soonchul Lee, and Hyun-Ju An
- Subjects
anaplastic thyroid cancer ,auranofin ,chemotherapy ,drug repositioning ,Medicine ,Pharmacy and materia medica ,RS1-441 - Abstract
Background/Objectives: Anaplastic thyroid cancer (ATC) is an aggressive and rare cancer with a poor prognosis, and traditional therapies have limited efficacy. This study investigates drug repositioning, focusing on auranofin, a gold-based drug originally used for rheumatoid arthritis, as a potential treatment for ATC. Methods: Auranofin was identified from an FDA-approved drug library and tested on two thyroid cancer cell lines, 8505C and FRO. Antitumor efficacy was evaluated through gene and protein expression analysis using Western blot, FACS, and mRNA sequencing. In vivo experiments were conducted using subcutaneous injections in nude mice to confirm the anticancer effects of auranofin. Results: Auranofin induced reactive oxygen species (ROS) production and apoptosis, leading to a dose-dependent reduction in cell viability, G1/S phase cell cycle arrest, and altered expression of regulatory proteins. It also inhibited cancer stem cell activity and suppressed epithelial–mesenchymal transition. mRNA sequencing revealed significant changes in the extracellular matrix–receptor interaction pathway, supported by Western blot results. In vivo xenograft models demonstrated strong antitumor activity. Conclusions: Auranofin shows promise as a repurposed therapeutic agent for ATC, effectively inhibiting cell proliferation, reducing metastasis, and promoting apoptosis. These findings suggest that auranofin could play a key role in future ATC treatment strategies.
- Published
- 2024
- Full Text
- View/download PDF
32. Synergistic Dual Targeting of Thioredoxin and Glutathione Systems Irrespective of p53 in Glioblastoma Stem Cells
- Author
-
Fatemeh Jamali, Katherine Lan, Paul Daniel, Kevin Petrecca, Siham Sabri, and Bassam Abdulkarim
- Subjects
Glioblastoma ,Glioblastoma stem cells (GSCs) ,auranofin ,thioredoxin reductase ,oxidative stress ,glutathione (GSH) ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Glioblastoma (GBM) is an incurable primary brain cancer characterized by increased reactive oxygen species (ROS) production. The redox-sensitive tumor suppressor gene TP53, wild-type (wt) for 70% of patients, regulates redox homeostasis. Glioblastoma stem cells (GSCs) increase thioredoxin (Trx) and glutathione (GSH) antioxidant systems as survival redox-adaptive mechanisms to maintain ROS below the cytotoxic threshold. Auranofin, an FDA-approved anti-rheumatoid drug, inhibits thioredoxin reductase 1 (TrxR1). L-buthionine sulfoximine (L-BSO) and the natural product piperlongumine (PPL) inhibit the GSH system. We evaluated the cytotoxic effects of Auranofin alone and in combination with L-BSO or PPL in GBM cell lines and GSCs with a known TP53 status. The Cancer Genome Atlas/GBM analysis revealed a significant positive correlation between wtp53 and TrxR1 expression in GBM. Auranofin induced ROS-dependent cytotoxicity within a micromolar range in GSCs. Auranofin decreased TrxR1 expression, AKT (Ser-473) phosphorylation, and increased p53, p21, and PARP-1 apoptotic cleavage in wtp53-GSCs, while mutant-p53 was decreased in a mutant-p53 GSC line. Additionally, p53-knockdown in a wtp53-GSC line decreased TrxR1 expression and significantly increased sensitivity to Auranofin, suggesting the role of wtp53 as a negative redox-sensitive mechanism in response to Auranofin in GSCs. The combination of Auranofin and L-BSO synergistically increased ROS, decreased IC50s, and induced long-term cytotoxicity irrespective of p53 in GBM cell lines and GSCs. Intriguingly, Auranofin increased the expression of glutathione S-transferase pi-1 (GSTP-1), a target of PPL. Combining Auranofin with PPL synergistically decreased IC50s to a nanomolar range in GSCs, supporting the potential to repurpose Auranofin and PPL in GBM.
- Published
- 2024
- Full Text
- View/download PDF
33. Auranofin for Giardia Protozoa
- Published
- 2022
34. Dual targeting of KDM1A and antioxidants is an effective anticancer strategy
- Subjects
Antioxidants ,Auranofin ,Cancer -- Care and treatment ,Physical fitness ,Health - Abstract
2024 JUL 6 (NewsRx) -- By a News Reporter-Staff News Editor at Obesity, Fitness & Wellness Week -- According to news reporting based on a preprint abstract, our journalists obtained [...]
- Published
- 2024
35. Auranofin sensitizes breast cancer cells to paclitaxel mediated cell death via regulating FOXO3/Nrf2/Keap1 signaling pathway.
- Author
-
Deepika, N., Rajendra Prasad, N., and Radhiga, T.
- Subjects
- *
PACLITAXEL , *NUCLEAR factor E2 related factor , *CELL death , *METASTATIC breast cancer , *AURANOFIN , *CANCER cells , *BREAST cancer , *CELLULAR signal transduction - Abstract
Nuclear factor erythroid 2‐related factor 2 (Nrf2) is a transcriptional factor which acts as a regulator for cellular oxidative stress, and tightly regulated by Kelch‐like ECH‐associated protein 1 (Keap1). In this study, we found that auranofin and paclitaxel combination treatment increased TUNEL positive apoptotic cells and enhanced the DNA damage marker γ‐H2AX in MCF‐7 and MDA‐MB‐231 breast cancer cells. The immunoblotting analysis revealed the combination of auranofin and paclitaxel significantly increased the FOXO3 expression in a concentration dependent manner. Further we observed that auranofin and paclitaxel treatment prevents the translocation of Nrf2 in a concentration dependent manner. The increased FOXO3 expression might be involved in the cytoplasmic degradation of Nrf1‐Keap1 complex. Further, the molecular docking results confirm auranofin act as the agonist for Foxo3. Therefore, the present results suggest that auranofin sensitize the breast cancer cells to paclitaxel via regulating FOXO3/Nrf2/Keap1signaling pathway. Significance Statement: The present results show that auranofin enhances paclitaxel induced TUNEL positive apoptotic cells. Further, auranofin treatment increases FOXO3 which in turn inhibit the dissociation of Nrf2 and Keap1 in both MDA‐MB‐231 and MCF‐7 cell lines. Therefore, the present results clearly illustrate the chemosensitizing property of auranofin via regulating FOXO3/Nrf2/Keap1 signaling pathway in the breast cancer cells. Auranofin is already a Food and Drug administration approved drug and it could be considered for the clinical applications after preclinical experimentation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
36. Auranofin sensitizes breast cancer cells to paclitaxel chemotherapy by disturbing the cellular redox system.
- Author
-
Natarajan, Deepika, Prasad, N. Rajendra, Sudharsan, M., Bharathiraja, Pradhapsingh, and Lakra, Deepa Swati
- Subjects
- *
CELL death , *OXIDATION-reduction reaction , *AURANOFIN , *METASTATIC breast cancer , *CANCER cells , *BREAST cancer , *PACLITAXEL - Abstract
The intrinsic redox status of cancer cells limits the efficacy of chemotherapeutic drugs. Auranofin, a Food and Drug Administration‐approved gold‐containing compound, documented with effective pharmacokinetics and safety profiles in humans, has recently been repurposed for anticancer activity. This study examined the paclitaxel‐sensitizing effect of auranofin by targeting redox balance in the MDA‐MB‐231 and MCF‐7 breast cancer cell lines. Auranofin treatment depletes the activities of superoxide dismutase, catalase, and glutathione peroxidase and alters the redox ratio in the breast cancer cell lines. Furthermore, it has been noticed that auranofin augmented paclitaxel‐mediated cytotoxicity in a concentration‐dependent manner in both MDA‐MB‐231 and MCF‐7 cell lines. Moreover, auranofin increased the levels of intracellular reactive oxygen species (observed using 2, 7‐diacetyl dichlorofluorescein diacetate staining) and subsequently altered the mitochondrial membrane potential (rhodamine‐123 staining) in a concentration‐dependent manner. Further, the expression of apoptotic marker p21 was found to be higher in auranofin plus paclitaxel‐treated breast cancer cells compared to paclitaxel‐alone treatment. Thus, the present results illustrate the chemosensitizing property of auranofin in MDA‐MB‐231 and MCF‐7 breast cancer cell lines via oxidative metabolism. Therefore, auranofin could be considered a chemosensitizing agent during cancer chemotherapy. Significance statement: In this study, auranofin, a Food and Drug Administration‐approved drug, has been repurposed for anticancer activity. The results illustrate that auranofin improved paclitaxel‐mediated cytotoxicity in breast cancer cell lines. The auranofin treatment altered the intracellular redox balance in the breast cancer cells, which contributes to paclitaxel‐mediated cell cycle arrest and subsequent apoptotic cell death. Thus, the present results illustrate the chemosensitizing property of auranofin in breast cancer cell lines via oxidative metabolism. Therefore, auranofin could be considered a chemosensitizing agent during cancer chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
37. Direct inhibition of dioxygenases TET1 by the rheumatoid arthritis drug auranofin selectively induces cancer cell death in T-ALL
- Author
-
Long Chen, Anqi Ren, Yuan Zhao, Hangyu Chen, Qifang Wu, Mengzhu Zheng, Zijian Zhang, Tongcun Zhang, Wu Zhong, Jian Lin, and Haichuan Zhu
- Subjects
DNA methylation ,TET1 ,Auranofin ,5hmC ,c-Myc ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract T-cell acute lymphoblastic leukemia (T-ALL) is a type of hematologic tumor with malignant proliferation of hematopoietic progenitor cells. However, traditional clinical treatment of T-ALL included chemotherapy and stem cell transplantation always lead to recurrence and poor prognosis, thus new therapeutic targets and drugs are urgently needed for T-ALL treatment. In this study, we showed that TET1 (ten-eleven translocation 1), a key participant of DNA epigenetic control, which catalyzes the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) to modulate gene expression, was highly upregulated in human T-ALL and negatively correlated with the prognosis of patients. Knockdown of TET1 suppressed T-ALL growth and progression, suggesting that TET1 inhibition maybe an effective way to fight T-ALL via DNA epigenetic modulation. Combining structure-guided virtual screening and cell-based high-throughput screening of FDA-approved drug library, we discovered that auranofin, a gold-containing compound, is a potent TET1 inhibitor. Auranofin inhibited the catalytic activity of TET1 through competitive binding to its substrates binding pocket and thus downregulated the genomic level of 5hmC marks and particularly epigenetically reprogramed the expression of oncogene c-Myc in T-ALL in TET1-dependent manner and resulted in suppression of T-ALL in vitro and in vivo. These results revealed that TET1 is a potential therapeutic target in human T-ALL and elucidated the mechanism that TET1 inhibitor auranofin suppressed T-ALL through the TET1/5hmC/c-Myc signaling pathway. Our work thus not only provided mechanism insights for T-ALL treatment, but also discovered potential small molecule therapeutics for T-ALL.
- Published
- 2023
- Full Text
- View/download PDF
38. The Many Lives of Auranofin: How an Old Anti-Rheumatic Agent May Become a Promising Antimicrobial Drug
- Author
-
Francesca Coscione, Stefano Zineddu, Valentina Vitali, Marco Fondi, Luigi Messori, and Elena Perrin
- Subjects
auranofin ,drug repurposing ,thioredoxin reductase ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Auranofin (AF) is a gold-based compound with a well-known pharmacological and toxicological profile, currently used in the treatment of some severe forms of rheumatoid arthritis. Over the last twenty years, AF has also been repurposed as antiviral, antitumor, and antibacterial drug. In this review we focused on the antibacterial properties of AF, specifically researching the minimal inhibitory concentrations (MIC) of AF in both mono- and diderm bacteria reported so far in literature. AF proves to be highly effective against monoderm bacteria, while diderm are far less susceptible, probably due to the outer membrane barrier. We also reported the current mechanistic hypotheses concerning the antimicrobial properties of AF, although a conclusive description of its antibacterial mode of action is not yet available. Even if its mechanism of action has not been fully elucidated yet and further studies are required to optimize its delivery strategy, AF deserves additional investigation because of its unique mode of action and high efficacy against a wide range of pathogens, which could lead to potential applications in fighting antimicrobial resistance and improving therapeutic outcomes in infectious diseases.
- Published
- 2024
- Full Text
- View/download PDF
39. Gold(I) ion and the phosphine ligand are necessary for the anti-Toxoplasma gondii activity of auranofin
- Author
-
C. I. Ma, J. A. Tirtorahardjo, S. S. Schweizer, J. Zhang, Z. Fang, L. Xing, M. Xu, D. A. Herman, M. T. Kleinman, B. S. McCullough, A. M. Barrios, and R. M. Andrade
- Subjects
auranofin ,Toxoplasma ,repurposing ,drug discovery ,gold ,Microbiology ,QR1-502 - Abstract
ABSTRACTAuranofin, an FDA-approved drug for rheumatoid arthritis, has emerged as a promising antiparasitic medication in recent years. The gold(I) ion in auranofin is postulated to be responsible for its antiparasitic activity. Notably, aurothiomalate and aurothioglucose also contain gold(I), and, like auranofin, they were previously used to treat rheumatoid arthritis. Whether they have antiparasitic activity remains to be elucidated. Herein, we demonstrated that auranofin and similar derivatives, but not aurothiomalate and aurothioglucose, inhibited the growth of Toxoplasma gondii in vitro. We found that auranofin affected the T. gondii biological cycle (lytic cycle) by inhibiting T. gondii’s invasion and triggering its egress from the host cell. However, auranofin could not prevent parasite replication once T. gondii resided within the host. Auranofin treatment induced apoptosis in T. gondii parasites, as demonstrated by its reduced size and elevated phosphatidylserine externalization (PS). Notably, the gold from auranofin enters the cytoplasm of T. gondii, as demonstrated by scanning transmission electron microscopy-energy dispersive X-ray spectroscopy (STEM-EDS) and Inductively Coupled Plasma-Mass Spectrometry (ICP-MS).IMPORTANCEToxoplasmosis, caused by Toxoplasma gondii, is a devastating disease affecting the brain and the eyes, frequently affecting immunocompromised individuals. Approximately 60 million people in the United States are already infected with T. gondii, representing a population at-risk of developing toxoplasmosis. Recent advances in treating cancer, autoimmune diseases, and organ transplants have contributed to this at-risk population's exponential growth. Paradoxically, treatments for toxoplasmosis have remained the same for more than 60 years, relying on medications well-known for their bone marrow toxicity and allergic reactions. Discovering new therapies is a priority, and repurposing FDA-approved drugs is an alternative approach to speed up drug discovery. Herein, we report the effect of auranofin, an FDA-approved drug, on the biological cycle of T. gondii and how both the phosphine ligand and the gold molecule determine the anti-parasitic activity of auranofin and other gold compounds. Our studies would contribute to the pipeline of candidate anti-T. gondii agents.
- Published
- 2024
- Full Text
- View/download PDF
40. Direct inhibition of dioxygenases TET1 by the rheumatoid arthritis drug auranofin selectively induces cancer cell death in T-ALL.
- Author
-
Chen, Long, Ren, Anqi, Zhao, Yuan, Chen, Hangyu, Wu, Qifang, Zheng, Mengzhu, Zhang, Zijian, Zhang, Tongcun, Zhong, Wu, Lin, Jian, and Zhu, Haichuan
- Subjects
- *
AURANOFIN , *CELL death , *CANCER cells , *DIOXYGENASES , *SMALL molecules - Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a type of hematologic tumor with malignant proliferation of hematopoietic progenitor cells. However, traditional clinical treatment of T-ALL included chemotherapy and stem cell transplantation always lead to recurrence and poor prognosis, thus new therapeutic targets and drugs are urgently needed for T-ALL treatment. In this study, we showed that TET1 (ten-eleven translocation 1), a key participant of DNA epigenetic control, which catalyzes the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) to modulate gene expression, was highly upregulated in human T-ALL and negatively correlated with the prognosis of patients. Knockdown of TET1 suppressed T-ALL growth and progression, suggesting that TET1 inhibition maybe an effective way to fight T-ALL via DNA epigenetic modulation. Combining structure-guided virtual screening and cell-based high-throughput screening of FDA-approved drug library, we discovered that auranofin, a gold-containing compound, is a potent TET1 inhibitor. Auranofin inhibited the catalytic activity of TET1 through competitive binding to its substrates binding pocket and thus downregulated the genomic level of 5hmC marks and particularly epigenetically reprogramed the expression of oncogene c-Myc in T-ALL in TET1-dependent manner and resulted in suppression of T-ALL in vitro and in vivo. These results revealed that TET1 is a potential therapeutic target in human T-ALL and elucidated the mechanism that TET1 inhibitor auranofin suppressed T-ALL through the TET1/5hmC/c-Myc signaling pathway. Our work thus not only provided mechanism insights for T-ALL treatment, but also discovered potential small molecule therapeutics for T-ALL. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
41. Auranofin Induces Lethality Driven by Reactive Oxygen Species in High-Grade Serous Ovarian Cancer Cells.
- Author
-
Abdalbari, Farah H., Martinez-Jaramillo, Elvis, Forgie, Benjamin N., Tran, Estelle, Zorychta, Edith, Goyeneche, Alicia A., Sabri, Siham, and Telleria, Carlos M.
- Subjects
- *
GLUTATHIONE , *ACETYLCYSTEINE , *PLATINUM compounds , *DRUG repositioning , *OVARIAN tumors , *ANTINEOPLASTIC agents , *APOPTOSIS , *ANTIRHEUMATIC agents , *CELL survival , *RESEARCH funding , *TUMORS , *REACTIVE oxygen species , *CELL lines , *OXIDOREDUCTASES , *DNA damage , *CELL surface antigens , *TUMOR grading , *IMMUNODIAGNOSIS , *PHARMACODYNAMICS , *CHEMICAL inhibitors - Abstract
Simple Summary: High-grade serous ovarian cancer (HGSOC) is the most prevalent type of ovarian cancer, accounting for 70% of ovarian cancer deaths. This is primarily due to the development of resistance against standard platinum-based chemotherapy. Several drugs are currently undergoing repurposing against ovarian cancer, including auranofin (AF), an anti-rheumatoid agent. The mechanism of action of AF has been studied in various cancers, however, there have been fewer studies on the effects of AF in HGSOC. In this study, we explore the mechanisms of action of AF in human HGSOC cells that are sensitive or resistant to platinum. We demonstrate the various cytotoxic effects of AF in HGSOC via the targeting of multiple pathways, suggesting the potential use of AF in a long-term consolidation therapy against this disease. High-grade serous ovarian cancer (HGSOC) accounts for 70% of ovarian cancer cases, and the survival rate remains remarkably low due to the lack of effective long-term consolidation therapies. Clinical remission can be temporarily induced by platinum-based chemotherapy, but death subsequently results from the extensive growth of a platinum-resistant component of the tumor. This work explores a novel treatment against HGSOC using the gold complex auranofin (AF). AF primarily functions as a pro-oxidant by inhibiting thioredoxin reductase (TrxR), an antioxidant enzyme overexpressed in ovarian cancer. We investigated the effect of AF on TrxR activity and the various mechanisms of cytotoxicity using HGSOC cells that are clinically sensitive or resistant to platinum. In addition, we studied the interaction between AF and another pro-oxidant, L-buthionine sulfoximine (L-BSO), an anti-glutathione (GSH) compound. We demonstrated that AF potently inhibited TrxR activity and reduced the vitality and viability of HGSOC cells regardless of their sensitivities to platinum. We showed that AF induces the accumulation of reactive oxygen species (ROS), triggers the depolarization of the mitochondrial membrane, and kills HGSOC cells by inducing apoptosis. Notably, AF-induced cell death was abrogated by the ROS-scavenger N-acetyl cysteine (NAC). In addition, the lethality of AF was associated with the activation of caspases-3/7 and the generation of DNA damage, effects that were also prevented by the presence of NAC. Finally, when AF and L-BSO were combined, we observed synergistic lethality against HGSOC cells, which was mediated by a further increase in ROS and a decrease in the levels of the antioxidant GSH. In summary, our results support the concept that AF can be used alone or in combination with L-BSO to kill HGSOC cells regardless of their sensitivity to platinum, suggesting that the depletion of antioxidants is an efficient strategy to mitigate the course of this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
42. Antiprotozoal activity of auranofin on Trypanosoma cruzi, Leishmania tropica and Toxoplasma gondii: in vitro and ex vivo study.
- Author
-
Yıldırım, Ahmet, Özbilgin, Ahmet, and Yereli, Kor
- Subjects
TRYPANOSOMA cruzi ,AURANOFIN ,TOXOPLASMA gondii ,CHAGAS' disease ,LEISHMANIA ,DRUG efficacy ,TRICHOMONIASIS ,ISCHEMIC colitis - Abstract
Background Three obligate intracellular protozoan parasite species, which are responsible for significant morbidity and mortality and settle in macrophage cells, affect more than one-half of the world's population, namely, Trypanosoma cruzi, Leishmania tropica and Toxoplasma gondii , which are causative agents of Chagas disease, leishmaniasis and toxoplasmosis, respectively. In the current study, it was aimed to investigate the in vitro and ex vivo antiprotozoal activity of auranofin on T. cruzi, L. tropica and T. gondii. Methods The in vitro drug efficacy (IC
50 ) of auranofin was investigated by haemocytometry and the CellTiter-Glo assay methods and the ex vivo drug efficacy (IC50 ) by light microscopic examination of Giemsa-stained slides. Also, the cytotoxic activity (CC50 ) of auranofin was examined by the CellTiter-Glo assay. The selectivity index (SI) was calculated for auranofin. Results According to IC50 , CC50 and SI data, auranofin did not exhibit cytotoxic activity on Vero cells, but exhibited antiprotozoal activity on epimastigotes and intracellular amastigotes of T. cruzi , promastigotes and intracellular amastigotes of L. tropica and intracellular tachyzoites of T. gondii (p<0.05). Conclusions The detection antiprotozoal activity of auranofin on T. cruzi, L. tropica and T. gondii according to the IC50 , CC50 and SI values is considered an important and promising development. This is significant because auranofin may be an effective alternative treatment for Chagas disease, leishmaniasis and toxoplasmosis in the future. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
43. Thioredoxin Reductase and Organometallic Complexes: A Pivotal System to Tackle Multidrug Resistant Tumors?
- Author
-
Salmain, Michèle, Gaschard, Marie, Baroud, Milad, Lepeltier, Elise, Jaouen, Gérard, Passirani, Catherine, and Vessières, Anne
- Subjects
- *
ORGANOMETALLIC compounds , *OXIDOREDUCTASES , *TAMOXIFEN , *TUMORS , *DRUG resistance in cancer cells , *ENZYME inhibitors , *CYTOTOXINS , *OXIDATION-reduction reaction , *PHARMACODYNAMICS - Abstract
Simple Summary: The identification of biological targets is an essential step in deciphering the mechanism of action of anticancer drugs. In this review, we chose to study the relationship between the inhibition of thioredoxin reductase (TrxR), a key enzyme in maintaining the redox balance of cells, and the cytotoxic effects of two groups of organometallic complexes. The first group is essentially composed of Au(I) and Au(III) complexes and the second one comprises metallocifens (organometallic complexes derived from tamoxifen). The results show that these two groups interact differently with TrxR at the molecular level. Even if the contribution of TrxR inhibition to the cytotoxicity of complexes is clearly established for many of them, the number of complexes for which TrxR inhibition plays a predominant role appears quite limited. Eventually, the antiproliferative activity of most of the complexes appears to stem from the interaction with several targets, a favorable strategy to tackle MDR tumors. Cancers classified as multidrug-resistant (MDR) are a family of diseases with poor prognosis despite access to increasingly sophisticated treatments. Several mechanisms explain these resistances involving both tumor cells and their microenvironment. It is now recognized that a multi-targeting approach offers a promising strategy to treat these MDR tumors. Inhibition of thioredoxin reductase (TrxR), a key enzyme in maintaining redox balance in cells, is a well-identified target for this approach. Auranofin was the first inorganic gold complex to be described as a powerful inhibitor of TrxR. In this review, we will first recall the main results obtained with this metallodrug. Then, we will focus on organometallic complexes reported as TrxR inhibitors. These include gold(I), gold(III) complexes and metallocifens, i.e., organometallic complexes of Fe and Os derived from tamoxifen. In these families of complexes, similarities and differences in the molecular mechanisms of TrxR inhibition will be highlighted. Finally, the possible relationship between TrxR inhibition and cytotoxicity will be discussed and put into perspective with their mode of action. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
44. Inhibition of selenoprotein synthesis is not the mechanism by which auranofin inhibits growth of Clostridioides difficile.
- Author
-
Johnstone, Michael A., Holman, Matthew A., and Self, William T.
- Subjects
- *
CLOSTRIDIOIDES difficile , *AURANOFIN , *DRUG repositioning , *ANTIRHEUMATIC agents , *TREATMENT failure , *SELENOPROTEINS - Abstract
Clostridioides difficile infections (CDIs) are responsible for a significant number of antibiotic-associated diarrheal cases. The standard-of-care antibiotics for C. difficile are limited to fidaxomicin and vancomycin, with the recently obsolete metronidazole recommended if both are unavailable. No new antimicrobials have been approved for CDI since fidaxomicin in 2011, despite varying rates of treatment failure among all standard-of-care drugs. Drug repurposing is a rational strategy to generate new antimicrobials out of existing therapeutics approved for other indications. Auranofin is a gold-containing anti-rheumatic drug with antimicrobial activity against C. difficile and other microbes. In a previous report, our group hypothesized that inhibition of selenoprotein biosynthesis was auranofin's primary mechanism of action against C. difficile. However, in this study, we discovered that C. difficile mutants lacking selenoproteins are still just as sensitive to auranofin as their respective wild-type strains. Moreover, we found that selenite supplementation dampens the activity of auranofin against C. difficile regardless of the presence of selenoproteins, suggesting that selenite's neutralization of auranofin is not because of compensation for a chemically induced selenium deficiency. Our results clarify the findings of our original study and may aid drug repurposing efforts in discovering the compound's true mechanism of action against C. difficile. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
45. Auranofin Modulates Thioredoxin Reductase/Nrf2 Signaling in Peripheral Immune Cells and the CNS in a Mouse Model of Relapsing–Remitting EAE.
- Author
-
Al-Kharashi, Layla A., Al-Harbi, Naif O., Ahmad, Sheikh F., Attia, Sabry M., Algahtani, Mohammad M., Ibrahim, Khalid E., Bakheet, Saleh A., Alanazi, Mohammed M., Alqarni, Saleh A., Alsanea, Sary, and Nadeem, Ahmed
- Subjects
NUCLEAR factor E2 related factor ,THIOREDOXIN ,INFLAMMATORY mediators ,AUTOIMMUNE diseases ,LABORATORY mice ,NITRIC-oxide synthases ,NF-kappa B - Abstract
Multiple sclerosis (MS) is one of the most prevalent chronic inflammatory autoimmune diseases. It causes the demyelination of neurons and the subsequent degeneration of the central nervous system (CNS). The infiltration of leukocytes of both myeloid and lymphoid origins from the systemic circulation into the CNS triggers autoimmune reactions through the release of multiple mediators. These mediators include oxidants, pro-inflammatory cytokines, and chemokines which ultimately cause the characteristic plaques observed in MS. Thioredoxin reductase (TrxR) and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling plays a crucial role in the regulation of inflammation by modulating the transcription of antioxidants and the suppression of inflammatory cytokines. The gold compound auranofin (AFN) is known to activate Nrf2 through the inhibition of TrxR; however, the effects of this compound have not been explored in a mouse model of relapsing–remitting MS (RRMS). Therefore, this study explored the influence of AFN on clinical features, TrxR/Nrf2 signaling [heme oxygenase 1 (HO-1), superoxide dismutase 1 (SOD-1)] and oxidative/inflammatory mediators [IL-6, IL-17A, inducible nitric oxide synthase (iNOS), myeloperoxidase (MPO), nitrotyrosine] in peripheral immune cells and the CNS of mice with the RR type of EAE. Our results showed an increase in TrxR activity and a decrease in Nrf2 signaling in SJL/J mice with RR-EAE. The treatment with AFN caused the amelioration of the clinical features of RR-EAE through the elevation of Nrf2 signaling and the subsequent upregulation of the levels of antioxidants as well as the downregulation of oxidative/pro-inflammatory mediators in peripheral immune cells and the CNS. These data suggest that AFN may be beneficial in the treatment of RRMS. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
46. Ferroptosis: A New Way to Die
- Author
-
Schor, Jacob
- Subjects
Auranofin ,Amino acids ,Apoptosis ,Health - Abstract
Understanding how things die is important for doctors who want to keep their patients alive. In 1972, the word apoptosis was invented to describe the specific form of cell death [...]
- Published
- 2024
47. Auranofin Resistance in Toxoplasma gondii Decreases the Accumulation of Reactive Oxygen Species but Does Not Target Parasite Thioredoxin Reductase
- Author
-
I., Christopher, Tirtorahardjo, James A, Jan, Sharon, Schweizer, Sakura S, Rosario, Shawn AC, Du, Yanmiao, Zhang, Jerry J, Morrissette, Naomi S, and Andrade, Rosa M
- Subjects
Microbiology ,Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Genetics ,Biodefense ,Foodborne Illness ,Infectious Diseases ,Emerging Infectious Diseases ,Antimicrobial Resistance ,Animals ,Auranofin ,Parasites ,Reactive Oxygen Species ,Thioredoxin-Disulfide Reductase ,Toxoplasma ,gold ,repurposing ,anti-parasitic ,resistance ,redox ,auranofin ,superoxide ,Biochemistry and Cell Biology ,Medical microbiology - Abstract
Auranofin, a reprofiled FDA-approved drug originally designed to treat rheumatoid arthritis, has emerged as a promising anti-parasitic drug. It induces the accumulation of reactive oxygen species (ROS) in parasites, including Toxoplasma gondii. We generated auranofin resistant T. gondii lines through chemical mutagenesis to identify the molecular target of this drug. Resistant clones were confirmed with a competition assay using wild-type T. gondii expressing yellow fluorescence protein (YFP) as a reference strain. The predicted auranofin target, thioredoxin reductase, was not mutated in any of our resistant lines. Subsequent whole genomic sequencing analysis (WGS) did not reveal a consensus resistance locus, although many have point mutations in genes encoding redox-relevant proteins such as superoxide dismutase (TgSOD2) and ribonucleotide reductase. We investigated the SOD2 L201P mutation and found that it was not sufficient to confer resistance when introduced into wild-type parasites. Resistant clones accumulated less ROS than their wild type counterparts. Our results demonstrate that resistance to auranofin in T. gondii enhances its ability to abate oxidative stress through diverse mechanisms. This evidence supports a hypothesized mechanism of auranofin anti-parasitic activity as disruption of redox homeostasis.
- Published
- 2021
48. Auranofin Resistance in Toxoplasma gondii Decreases the Accumulation of Reactive Oxygen Species but Does Not Target Parasite Thioredoxin Reductase.
- Author
-
Ma, Christopher I, Tirtorahardjo, James A, Jan, Sharon, Schweizer, Sakura S, Rosario, Shawn AC, Du, Yanmiao, Zhang, Jerry J, Morrissette, Naomi S, and Andrade, Rosa M
- Subjects
Animals ,Parasites ,Toxoplasma ,Reactive Oxygen Species ,Auranofin ,Thioredoxin-Disulfide Reductase ,anti-parasitic ,auranofin ,gold ,redox ,repurposing ,resistance ,superoxide ,Biochemistry and Cell Biology ,Microbiology - Abstract
Auranofin, a reprofiled FDA-approved drug originally designed to treat rheumatoid arthritis, has emerged as a promising anti-parasitic drug. It induces the accumulation of reactive oxygen species (ROS) in parasites, including Toxoplasma gondii. We generated auranofin resistant T. gondii lines through chemical mutagenesis to identify the molecular target of this drug. Resistant clones were confirmed with a competition assay using wild-type T. gondii expressing yellow fluorescence protein (YFP) as a reference strain. The predicted auranofin target, thioredoxin reductase, was not mutated in any of our resistant lines. Subsequent whole genomic sequencing analysis (WGS) did not reveal a consensus resistance locus, although many have point mutations in genes encoding redox-relevant proteins such as superoxide dismutase (TgSOD2) and ribonucleotide reductase. We investigated the SOD2 L201P mutation and found that it was not sufficient to confer resistance when introduced into wild-type parasites. Resistant clones accumulated less ROS than their wild type counterparts. Our results demonstrate that resistance to auranofin in T. gondii enhances its ability to abate oxidative stress through diverse mechanisms. This evidence supports a hypothesized mechanism of auranofin anti-parasitic activity as disruption of redox homeostasis.
- Published
- 2021
49. A Proof-of-concept Clinical Trial Assessing the Safety of the Coordinated Undermining of Survival Paths by 9 Repurposed Drugs Combined With Metronomic Temozolomide (CUSP9v3 Treatment Protocol) for Recurrent Glioblastoma
- Author
-
Reliable Cancer Therapies, Anticancer Fund, Belgium, and Marc-Eric Halatsch, Prof. Dr.
- Published
- 2021
50. Auranofin targets UBA1 and enhances UBA1 activity by facilitating ubiquitin trans-thioesterification to E2 ubiquitin-conjugating enzymes.
- Author
-
Yan, Wenjing, Zhong, Yongwang, Hu, Xin, Xu, Tuan, Zhang, Yinghua, Kales, Stephen, Qu, Yanyan, Talley, Daniel C., Baljinnyam, Bolormaa, LeClair, Christopher A., Simeonov, Anton, Polster, Brian M., Huang, Ruili, Ye, Yihong, Rai, Ganesha, Henderson, Mark J., Tao, Dingyin, and Fang, Shengyun
- Subjects
UBIQUITIN-conjugating enzymes ,AURANOFIN ,UBIQUITIN ,UBIQUITINATION ,ALZHEIMER'S disease ,RHEUMATOID arthritis - Abstract
UBA1 is the primary E1 ubiquitin-activating enzyme responsible for generation of activated ubiquitin required for ubiquitination, a process that regulates stability and function of numerous proteins. Decreased or insufficient ubiquitination can cause or drive aging and many diseases. Therefore, a small-molecule enhancing UBA1 activity could have broad therapeutic potential. Here we report that auranofin, a drug approved for the treatment of rheumatoid arthritis, is a potent UBA1 activity enhancer. Auranofin binds to the UBA1's ubiquitin fold domain and conjugates to Cys1039 residue. The binding enhances UBA1 interactions with at least 20 different E2 ubiquitin-conjugating enzymes, facilitating ubiquitin charging to E2 and increasing the activities of seven representative E3s in vitro. Auranofin promotes ubiquitination and degradation of misfolded ER proteins during ER-associated degradation in cells at low nanomolar concentrations. It also facilitates outer mitochondrial membrane-associated degradation. These findings suggest that auranofin can serve as a much-needed tool for UBA1 research and therapeutic exploration. Decreased activity of the E1 ubiquitin-activating enzyme UBA1 can contribute to aging and diseases like Alzheimer's and VEXAS syndrome. Here, the authors found that auranofin, a rheumatoid arthritis drug, can significantly boost UBA1 activity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.