1
|
Wang Y, Yuan H, Fang R, Zhang R, Wang WJ. Unveiling the cytotoxicity of a new gold(I) complex towards hepatocellular carcinoma by inhibiting TrxR activity. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1537-1548. [PMID: 39314165 DOI: 10.3724/abbs.2024155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Hepatocellular carcinoma (HCC), the predominant type of liver cancer, is an aggressive malignancy with limited therapeutic options. In this study, we assess a collection of newly designed gold(I) phosphine complexes. Remarkably, the compound GC002 exhibits the greatest toxicity to HCC cells and outperforms established medications, such as sorafenib and auranofin, in terms of antitumor efficacy. GC002 triggers irreversible necroptosis in HCC cells by increasing the intracellular accumulation of reactive oxygen species (ROS). Mechanistically, GC002 significantly suppresses the activity of thioredoxin reductase (TrxR), which plays a crucial role in regulating redox homeostasis and is often overexpressed in HCC by binding directly to the enzyme. Our in vivo xenograft study confirms that GC002 possesses remarkable antitumor activity against HCC without severe side effects. These findings not only highlight the novel mechanism of controlling necroptosis via TrxR and ROS but also identify GC002 as a promising candidate for the further development of antitumor agents targeting HCC.
Collapse
Affiliation(s)
- Yuan Wang
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
- The School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Haokun Yuan
- The Key Laboratory for Human Disease Gene Study of Sichuan Province and the Department of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ruiqin Fang
- The School of Life Science, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Ran Zhang
- Faculty of Science and Engineering, University of Groningen, Groningen, 9713 AV, the Netherlands
| | - Wei-Jia Wang
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361104, China
| |
Collapse
|
2
|
Hu Y, Zhu Y, Shi J, Wei X, Tang C, Guan X, Zhang W. Plasma Thioredoxin Reductase as a Potential Diagnostic Biomarker for Breast Cancer. Clin Breast Cancer 2024; 24:e464-e473.e3. [PMID: 38616444 DOI: 10.1016/j.clbc.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/03/2024] [Accepted: 03/10/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Early diagnosis of breast cancer is critical to the treatment and prognosis of breast cancer patients. Our aim is to explore more practical and effective diagnostic methods to facilitate early treatment and improve prognosis for breast cancer patients. MATERIALS AND METHODS The Mann-Whitney U test, receiver operating characteristic curve, Youden index, Chi-square test, and Fisher's exact test were used to determine whether plasma thioredoxin reductase (TrxR) could be used for the clinical diagnosis of breast cancer. The Wilcoxon signed-rank test was used to validate the prognostic potential of plasma TrxR activity assessment. RESULTS A total of 761 patients were included, including 537 cases of breast cancer and 224 cases of benign breast diseases. Plasma TrxR activity in the breast cancer group [8.0 (6.0, 9.45) U/mL] was significantly higher than that in the benign group [3.05 (1.20, 6.275) U/mL]. The diagnostic efficiency of TrxR for breast cancer was higher than that of other conventional breast cancer biomarkers, with an area under the curve of 0.821 (95% CI = 0.791-0.852). In addition, TrxR can be used in combination with conventional tumor markers to further improve the diagnostic efficiency. The optimal TrxR threshold for identifying benign and malignant diseases is 7.45 U/mL. We detected plasma TrxR activity and serum tumor markers before and after antitumor therapies in 333 breast cancer patients and found that their trends were basically the same, with a significant decrease in plasma TrxR activity after treatment. CONCLUSION Plasma TrxR activity can be used as a suitable biomarker for breast cancer diagnosis and efficacy assessment.
Collapse
Affiliation(s)
- Yixuan Hu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yinxing Zhu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China; Department of Radiation Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Junfeng Shi
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaowei Wei
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Cuiju Tang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China.
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Ferreira RR, Carvalho RV, Coelho LL, Gonzaga BMDS, Bonecini-Almeida MDG, Garzoni LR, Araujo-Jorge TC. Current Understanding of Human Polymorphism in Selenoprotein Genes: A Review of Its Significance as a Risk Biomarker. Int J Mol Sci 2024; 25:1402. [PMID: 38338681 PMCID: PMC10855570 DOI: 10.3390/ijms25031402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 02/12/2024] Open
Abstract
Selenium has been proven to influence several biological functions, showing to be an essential micronutrient. The functional studies demonstrated the benefits of a balanced selenium diet and how its deficiency is associated with diverse diseases, especially cancer and viral diseases. Selenium is an antioxidant, protecting the cells from damage, enhancing the immune system response, preventing cardiovascular diseases, and decreasing inflammation. Selenium can be found in its inorganic and organic forms, and its main form in the cells is the selenocysteine incorporated into selenoproteins. Twenty-five selenoproteins are currently known in the human genome: glutathione peroxidases, iodothyronine deiodinases, thioredoxin reductases, selenophosphate synthetase, and other selenoproteins. These proteins lead to the transport of selenium in the tissues, protect against oxidative damage, contribute to the stress of the endoplasmic reticulum, and control inflammation. Due to these functions, there has been growing interest in the influence of polymorphisms in selenoproteins in the last two decades. Selenoproteins' gene polymorphisms may influence protein structure and selenium concentration in plasma and its absorption and even impact the development and progression of certain diseases. This review aims to elucidate the role of selenoproteins and understand how their gene polymorphisms can influence the balance of physiological conditions. In this polymorphism review, we focused on the PubMed database, with only articles published in English between 2003 and 2023. The keywords used were "selenoprotein" and "polymorphism". Articles that did not approach the theme subject were excluded. Selenium and selenoproteins still have a long way to go in molecular studies, and several works demonstrated the importance of their polymorphisms as a risk biomarker for some diseases, especially cardiovascular and thyroid diseases, diabetes, and cancer.
Collapse
Affiliation(s)
- Roberto Rodrigues Ferreira
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Manguinhos, Pav. Cardoso Fontes, Sala 64, Rio de Janeiro 21040-360, Brazil; (R.V.C.); (L.L.C.); (B.M.d.S.G.); (L.R.G.)
| | - Regina Vieira Carvalho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Manguinhos, Pav. Cardoso Fontes, Sala 64, Rio de Janeiro 21040-360, Brazil; (R.V.C.); (L.L.C.); (B.M.d.S.G.); (L.R.G.)
| | - Laura Lacerda Coelho
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Manguinhos, Pav. Cardoso Fontes, Sala 64, Rio de Janeiro 21040-360, Brazil; (R.V.C.); (L.L.C.); (B.M.d.S.G.); (L.R.G.)
| | - Beatriz Matheus de Souza Gonzaga
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Manguinhos, Pav. Cardoso Fontes, Sala 64, Rio de Janeiro 21040-360, Brazil; (R.V.C.); (L.L.C.); (B.M.d.S.G.); (L.R.G.)
| | - Maria da Gloria Bonecini-Almeida
- Laboratory of Immunology and Immunogenetics, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Avenida Brasil 4365, Manguinhos, Rio de Janeiro 21040-360, Brazil;
| | - Luciana Ribeiro Garzoni
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Manguinhos, Pav. Cardoso Fontes, Sala 64, Rio de Janeiro 21040-360, Brazil; (R.V.C.); (L.L.C.); (B.M.d.S.G.); (L.R.G.)
| | - Tania C. Araujo-Jorge
- Laboratory of Innovations in Therapies, Education and Bioproducts, Oswaldo Cruz Institute (LITEB-IOC/Fiocruz), Oswaldo Cruz Foundation (Fiocruz), Avenida Brasil 4365, Manguinhos, Pav. Cardoso Fontes, Sala 64, Rio de Janeiro 21040-360, Brazil; (R.V.C.); (L.L.C.); (B.M.d.S.G.); (L.R.G.)
| |
Collapse
|
4
|
Maia LB, Maiti BK, Moura I, Moura JJG. Selenium-More than Just a Fortuitous Sulfur Substitute in Redox Biology. Molecules 2023; 29:120. [PMID: 38202704 PMCID: PMC10779653 DOI: 10.3390/molecules29010120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Living organisms use selenium mainly in the form of selenocysteine in the active site of oxidoreductases. Here, selenium's unique chemistry is believed to modulate the reaction mechanism and enhance the catalytic efficiency of specific enzymes in ways not achievable with a sulfur-containing cysteine. However, despite the fact that selenium/sulfur have different physicochemical properties, several selenoproteins have fully functional cysteine-containing homologues and some organisms do not use selenocysteine at all. In this review, selected selenocysteine-containing proteins will be discussed to showcase both situations: (i) selenium as an obligatory element for the protein's physiological function, and (ii) selenium presenting no clear advantage over sulfur (functional proteins with either selenium or sulfur). Selenium's physiological roles in antioxidant defence (to maintain cellular redox status/hinder oxidative stress), hormone metabolism, DNA synthesis, and repair (maintain genetic stability) will be also highlighted, as well as selenium's role in human health. Formate dehydrogenases, hydrogenases, glutathione peroxidases, thioredoxin reductases, and iodothyronine deiodinases will be herein featured.
Collapse
Affiliation(s)
- Luisa B. Maia
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology | NOVA FCT, 2829-516 Caparica, Portugal; (I.M.); (J.J.G.M.)
| | - Biplab K. Maiti
- Department of Chemistry, School of Sciences, Cluster University of Jammu, Canal Road, Jammu 180001, India
| | - Isabel Moura
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology | NOVA FCT, 2829-516 Caparica, Portugal; (I.M.); (J.J.G.M.)
| | - José J. G. Moura
- LAQV, REQUIMTE, Department of Chemistry, NOVA School of Science and Technology | NOVA FCT, 2829-516 Caparica, Portugal; (I.M.); (J.J.G.M.)
| |
Collapse
|
5
|
Yang C, Xue M, He Y, Yin H, Yang C, Zhong D, Zeng H, Zheng Y, Diao X. Absorption, Distribution, Metabolism, and Excretion of [ 14C]BS1801, a Selenium-Containing Drug Candidate, in Rats. Molecules 2023; 28:8102. [PMID: 38138590 PMCID: PMC10745422 DOI: 10.3390/molecules28248102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
BS1801 is a selenium-containing drug candidate with potential for treating liver and lung fibrosis. To fully elucidate the biotransformation of BS1801 in animals and provide sufficient preclinical drug metabolism data for human mass balance study, the metabolism of BS1801 in rats was investigated. We used radiolabeling techniques to investigate the mass balance, tissue distribution, and metabolite identification of BS1801 in Sprague-Dawley/Long-Evans rats after a single oral dose of 100 mg/kg (100 μCi/kg) [14C]BS1801: 1. The mean recovery of radioactive substances in urine and feces was 93.39% within 168 h postdose, and feces were the main excretion route. 2. Additionally, less than 1.00% of the dose was recovered from either urine or bile. 3. BS1801-related components were widely distributed throughout the body. 4. Fifteen metabolites were identified in rat plasma, urine, feces, and bile, and BS1801 was detected only in feces. 5. BS1801-M484, the methylation product obtained via a N-Se bond reduction in BS1801, was the most abundant drug-related component in plasma. The main metabolic pathways of BS1801 were reduction, amide hydrolysis, oxidation, and methylation. Overall, BS1801 was distributed throughout the body, and excreted mainly as an intact BS1801 form through feces. No differences were observed between male and female rats in distribution, metabolism, and excretion of BS1801.
Collapse
Affiliation(s)
- Cheng Yang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China;
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; (Y.H.); (C.Y.); (D.Z.)
| | - Mingzhen Xue
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Yifei He
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; (Y.H.); (C.Y.); (D.Z.)
| | - Hanwei Yin
- Shanghai Yuanxi Pharmaceutical Technology Co., Ltd., Shanghai 201203, China;
| | - Chen Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; (Y.H.); (C.Y.); (D.Z.)
| | - Dafang Zhong
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; (Y.H.); (C.Y.); (D.Z.)
| | - Huihui Zeng
- Shanghai Yuanxi Pharmaceutical Technology Co., Ltd., Shanghai 201203, China;
| | - Yuandong Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; (Y.H.); (C.Y.); (D.Z.)
| | - Xingxing Diao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China;
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; (Y.H.); (C.Y.); (D.Z.)
| |
Collapse
|
6
|
Ban Q, Chi W, Wang X, Wang S, Hai D, Zhao G, Zhao Q, Granato D, Huang X. (-)-Epigallocatechin-3-Gallate Attenuates the Adverse Reactions Triggered by Selenium Nanoparticles without Compromising Their Suppressing Effect on Peritoneal Carcinomatosis in Mice Bearing Hepatocarcinoma 22 Cells. Molecules 2023; 28:molecules28093904. [PMID: 37175313 PMCID: PMC10180376 DOI: 10.3390/molecules28093904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Increasing evidence shows that selenium and polyphenols are two types of the most reported compounds in tumor chemoprevention due to their remarkable antitumor activity and high safety profile. The cross-talk between polyphenols and selenium is a hot research topic, and the combination of polyphenols and selenium is a valuable strategy for fighting cancer. The current work investigated the combination anti-peritoneal carcinomatosis (PC) effect of selenium nanoparticles (SeNPs) and green tea (Camellia sinensis) polyphenol (-)-epigallocatechin-3-gallate (EGCG) in mice bearing murine hepatocarcinoma 22 (H22) cells. Results showed that SeNPs alone significantly inhibited cancer cell proliferation and extended the survival time of mice bearing H22 cells. Still, the potential therapeutic efficacy is accompanied by an approximately eighty percent diarrhea rate. When EGCG was combined with SeNPs, EGCG did not affect the tumor proliferation inhibition effect but eliminated diarrhea triggered by SeNPs. In addition, both the intracellular selectively accumulated EGCG without killing effect on cancer cells and the enhanced antioxidant enzyme levels in ascites after EGCG was delivered alone by intraperitoneal injection indicated that H22 cells were insensitive to EGCG. Moreover, EGCG could prevent SeNP-caused systemic oxidative damage by enhancing serum superoxide dismutase, glutathione, and glutathione peroxidase levels in healthy mice. Overall, we found that H22 cells are insensitive to EGCG, but combining EGCG with SeNPs could protect against SeNP-triggered diarrhea without compromising the suppressing efficacy of SeNPs on PC in mice bearing H22 cells and attenuate SeNP-caused systemic toxicity in healthy mice. These results suggest that EGCG could be employed as a promising candidate for preventing the adverse reactions of chemotherapy including chemotherapy-induced diarrhea and systemic toxicity in cancer individuals.
Collapse
Affiliation(s)
- Qiuyan Ban
- College of Horticulture, Henan Agricultural University, Zhengzhou 450002, China
| | - Wenjing Chi
- College of Horticulture, Henan Agricultural University, Zhengzhou 450002, China
| | - Xiaoxiao Wang
- College of Food Science & Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Shiqiong Wang
- College of Food Science & Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Dan Hai
- College of Food Science & Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Guangshan Zhao
- College of Food Science & Technology, Henan Agricultural University, Zhengzhou 450002, China
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiuyan Zhao
- College of Food Science & Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Daniel Granato
- Department of Biological Sciences, Faculty of Science and Engineering, University of Limerick, V94 T9PX Limerick, Ireland
| | - Xianqing Huang
- College of Food Science & Technology, Henan Agricultural University, Zhengzhou 450002, China
| |
Collapse
|
7
|
Shutkov IA, Okulova YN, Mazur DM, Melnichuk NA, Babkov DA, Sokolova EV, Spasov AA, Milaeva ER, Nazarov AA. New Organometallic Ru(II) Compounds with Lonidamine Motif as Antitumor Agents. Pharmaceutics 2023; 15:pharmaceutics15051366. [PMID: 37242608 DOI: 10.3390/pharmaceutics15051366] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The combination of one molecule of organic and metal-based fragments that exhibit antitumor activity is a modern approach in the search for new promising drugs. In this work, biologically active ligands based on lonidamine (a selective inhibitor of aerobic glycolysis used in clinical practice) were introduced into the structure of an antitumor organometallic ruthenium scaffold. Resistant to ligand exchange reactions, compounds were prepared by replacing labile ligands with stable ones. Moreover, cationic complexes containing two lonidamine-based ligands were obtained. Antiproliferative activity was studied in vitro by MTT assays. It was shown that the increase in the stability in ligand exchange reactions does not influence cytotoxicity. At the same time, the introduction of the second lonidamine fragment approximately doubles the cytotoxicity of studied complexes. The ability to induce apoptosis and caspase activation in tumour cell MCF7 was studied by employing flow cytometry.
Collapse
Affiliation(s)
- Ilya A Shutkov
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Yulia N Okulova
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Dmitrii M Mazur
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Nikolai A Melnichuk
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Denis A Babkov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39 Novorossiyskaya Street, 400087 Volgograd, Russia
| | - Elena V Sokolova
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39 Novorossiyskaya Street, 400087 Volgograd, Russia
| | - Alexander A Spasov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39 Novorossiyskaya Street, 400087 Volgograd, Russia
| | - Elena R Milaeva
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| | - Alexey A Nazarov
- Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, 119991 Moscow, Russia
| |
Collapse
|
8
|
Abstract
Significance: Thioredoxin (Trx) is a powerful antioxidant that reduces protein disulfides to maintain redox stability in cells and is involved in regulating multiple redox-dependent signaling pathways. Recent Advance: The current accumulation of findings suggests that Trx participates in signaling pathways that interact with various proteins to manipulate their dynamic regulation of structure and function. These network pathways are critical for cancer pathogenesis and therapy. Promising clinical advances have been presented by most anticancer agents targeting such signaling pathways. Critical Issues: We herein link the signaling pathways regulated by the Trx system to potential cancer therapeutic opportunities, focusing on the coordination and strengths of the Trx signaling pathways in apoptosis, ferroptosis, immunomodulation, and drug resistance. We also provide a mechanistic network for the exploitation of therapeutic small molecules targeting the Trx signaling pathways. Future Directions: As research data accumulate, future complex networks of Trx-related signaling pathways will gain in detail. In-depth exploration and establishment of these signaling pathways, including Trx upstream and downstream regulatory proteins, will be critical to advancing novel cancer therapeutics. Antioxid. Redox Signal. 38, 403-424.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zhengjia Zhao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | | | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China.,School of Chemistry and Chemical Engineering, Nanjing University of Science & Technology, Nanjing, China
| |
Collapse
|
9
|
Therapeutic strategies for liver diseases based on redox control systems. Biomed Pharmacother 2022; 156:113764. [DOI: 10.1016/j.biopha.2022.113764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
|
10
|
Koh WX, Fong JL, Goh PX, Gomez AP, Wong ZX, Leong WK. Photochemical reactions of dinuclear organometallic complexes with diphenyl dichalcogenides. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2022.122517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
11
|
Zhou J, Yu LZ, Fan YL, Guo CH, Lv XM, Zhou ZY, Huang HD, Miao DD, Zhang SP, Li XY, Zhao PP, Liu XP, Hu WH, Zhang C. Discovery of novel hydroxyamidine based indoleamine 2,3-dioxygenase 1 (IDO1) and thioredoxin reductase 1 (TrxR1) dual inhibitors. Eur J Med Chem 2022; 245:114860. [DOI: 10.1016/j.ejmech.2022.114860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/08/2022] [Accepted: 10/16/2022] [Indexed: 11/26/2022]
|
12
|
Xu Q, Zhang J, Zhao Z, Chu Y, Fang J. Revealing PACMA 31 as a new chemical type TrxR inhibitor to promote cancer cell apoptosis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119323. [PMID: 35793738 DOI: 10.1016/j.bbamcr.2022.119323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/05/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022]
Abstract
Thioredoxin reductase (TrxR) is a pivotal regulator of redox homeostasis, while dysregulation of redox homeostasis is a hallmark for cancer cells. Thus, there is considerable potential to inhibit the aberrantly upregulated TrxR in cancer cells to discover selective cancer therapeutic agents. Nevertheless, the structural types of TrxR inhibitors presented currently are still relatively limited. We herein report that PACMA 31, previously reported to inhibit protein disulfide isomerase (PDI), is a potent TrxR inhibitor. PACMA 31 possesses a pharmacophore scaffold that is structurally different from the announced TrxR inhibitors and exhibits effective cytotoxicity against cervical cancer cells. Our results reveal that PACMA 31 selectively inhibits TrxR over the related glutathione reductase (GR) and in the presence of reduced glutathione (GSH). Further studies with mutant enzyme and molecular docking suggest that the propynamide fragment of PACMA 31 interacts covalently with the selenocysteine residue of TrxR. Moreover, PACMA 31 effectively and selectively curbs TrxR activity in cells and further stimulates the production of reactive oxygen species (ROS) at low micromolar concentrations, which in turn triggers the accumulation of oxidized thioredoxin (Trx) and GSSG in cells. Follow-up studies demonstrate that PACMA 31 targets TrxR in cells to induce oxidative stress-mediated cancer cell apoptosis. Our results provide a new structural type of TrxR inhibitor that may serve as a useful probe for investigating the biology of TrxR-implicated pathways, and uncover a new target of PACMA 31 that contributes to it becoming a candidate for cancer treatment.
Collapse
Affiliation(s)
- Qianhe Xu
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China..
| | - Zhengjia Zhao
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Yajun Chu
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Jianguo Fang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China..
| |
Collapse
|
13
|
Chakraborty S, Sircar E, Bhattacharyya C, Choudhuri A, Mishra A, Dutta S, Bhatta S, Sachin K, Sengupta R. S-Denitrosylation: A Crosstalk between Glutathione and Redoxin Systems. Antioxidants (Basel) 2022; 11:1921. [PMID: 36290644 PMCID: PMC9598160 DOI: 10.3390/antiox11101921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 08/27/2023] Open
Abstract
S-nitrosylation of proteins occurs as a consequence of the derivatization of cysteine thiols with nitric oxide (NO) and is often associated with diseases and protein malfunction. Aberrant S-nitrosylation, in addition to other genetic and epigenetic factors, has gained rapid importance as a prime cause of various metabolic, respiratory, and cardiac disorders, with a major emphasis on cancer and neurodegeneration. The S-nitrosoproteome, a term used to collectively refer to the diverse and dynamic repertoire of S-nitrosylated proteins, is relatively less explored in the field of redox biochemistry, in contrast to other covalently modified versions of the same set of proteins. Advancing research is gradually unveiling the enormous clinical importance of S-nitrosylation in the etiology of diseases and is opening up new avenues of prompt diagnosis that harness this phenomenon. Ever since the discovery of the two robust and highly conserved S-nitrosoglutathione reductase and thioredoxin systems as candidate denitrosylases, years of rampant speculation centered around the identification of specific substrates and other candidate denitrosylases, subcellular localization of both substrates and denitrosylases, the position of susceptible thiols, mechanisms of S-denitrosylation under basal and stimulus-dependent conditions, impact on protein conformation and function, and extrapolating these findings towards the understanding of diseases, aging and the development of novel therapeutic strategies. However, newer insights in the ever-expanding field of redox biology reveal distinct gaps in exploring the crucial crosstalk between the redoxins/major denitrosylase systems. Clarifying the importance of the functional overlap of the glutaredoxin, glutathione, and thioredoxin systems and examining their complementary functions as denitrosylases and antioxidant enzymatic defense systems are essential prerequisites for devising a rationale that could aid in predicting the extent of cell survival under high oxidative/nitrosative stress while taking into account the existence of the alternative and compensatory regulatory mechanisms. This review thus attempts to highlight major gaps in our understanding of the robust cellular redox regulation system, which is upheld by the concerted efforts of various denitrosylases and antioxidants.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Esha Sircar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, Uttarakhand, India
| | - Camelia Bhattacharyya
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Akansha Mishra
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Sreejita Dutta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Sneha Bhatta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| | - Kumar Sachin
- Department of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata 700135, West Bengal, India
| |
Collapse
|
14
|
Gencheva R, Cheng Q, Arnér ESJ. Thioredoxin reductase selenoproteins from different organisms as potential drug targets for treatment of human diseases. Free Radic Biol Med 2022; 190:320-338. [PMID: 35987423 DOI: 10.1016/j.freeradbiomed.2022.07.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/25/2022] [Accepted: 07/26/2022] [Indexed: 11/15/2022]
Abstract
Human thioredoxin reductase (TrxR) is a selenoprotein with a central role in cellular redox homeostasis, utilizing a highly reactive and solvent-exposed selenocysteine (Sec) residue in its active site. Pharmacological modulation of TrxR can be obtained with several classes of small compounds showing different mechanisms of action, but most often dependent upon interactions with its Sec residue. The clinical implications of TrxR modulation as mediated by small compounds have been studied in diverse diseases, from rheumatoid arthritis and ischemia to cancer and parasitic infections. The possible involvement of TrxR in these diseases was in some cases serendipitously discovered, by finding that existing clinically used drugs are also TrxR inhibitors. Inhibiting isoforms of human TrxR is, however, not the only strategy for human disease treatment, as some pathogenic parasites also depend upon Sec-containing TrxR variants, including S. mansoni, B. malayi or O. volvulus. Inhibiting parasite TrxR has been shown to selectively kill parasites and can thus become a promising treatment strategy, especially in the context of quickly emerging resistance towards other drugs. Here we have summarized the basis for the targeting of selenoprotein TrxR variants with small molecules for therapeutic purposes in different human disease contexts. We discuss how Sec engagement appears to be an indispensable part of treatment efficacy and how some therapeutically promising compounds have been evaluated in preclinical or clinical studies. Several research questions remain before a wider application of selenoprotein TrxR inhibition as a first-line treatment strategy might be developed. These include further mechanistic studies of downstream effects that may mediate treatment efficacy, identification of isoform-specific enzyme inhibition patterns for some given therapeutic compounds, and the further elucidation of cell-specific effects in disease contexts such as in the tumor microenvironment or in host-parasite interactions, and which of these effects may be dependent upon the specific targeting of Sec in distinct TrxR isoforms.
Collapse
Affiliation(s)
- Radosveta Gencheva
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Qing Cheng
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; Department of Selenoprotein Research, National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary.
| |
Collapse
|
15
|
Li H, Jiang R, Lou L, Jia C, Zou L, Chen M. Formononetin Improves the Survival of Random Skin Flaps Through PI3K/Akt-Mediated Nrf2 Antioxidant Defense System. Front Pharmacol 2022; 13:901498. [PMID: 35662691 PMCID: PMC9160463 DOI: 10.3389/fphar.2022.901498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Random-pattern skin flap is widely used in plastic and reconstructive surgery. However, its clinical effect is limited by ischemia necrosis occurs at the distal part of flap. Previous studies have proved that the protective effect of formononetin was associated with its antioxidant, anti-inflammatory ability. However, further research is still needed on the effect of formononetin on flap viability. The purpose of our study was to investigate the effect of formononetin on flap survival and the underlying mechanisms. Two doses (25 mg/kg, 50 mg/kg)of formononetin were administered for seven consecutive days on flap model. Flap tissues were collected on postoperative day 7. Our results revealed that formononetin promoted skin flap viability in a dose-dependent manner. Using immunohistochemical staining and western blot, we found that formononetin significantly reduced oxidative stress and inflammation. Hematoxylin and eosin (H and E) staining, laser Doppler images and immunofluorescence staining showed the enhancement of angiogenesis after formononetin treatment. Mechanistically, we demonstrated that the antioxidation of formononetin was mediated by activation and nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2), while down-regulating cytoplasmic Kelch-like ECH-associated protein 1 (Keap1) expression. Co-treatment with formononetin and LY294002 (15 mg/kg), a potent Phosphatidylinositol-3-kinase (PI3K) inhibitor, which aborted nuclear Nrf2 expression and phosphorylated Akt, indicating that formononetin-mediated Nrf2 activation was related to PI3K/Akt pathway. Overall, our findings revealed that formononetin increased angiogenesis, reduced oxidative stress and inflammation, thus promoting flap survival. We highlighted the antioxidant effects of formononetin since the Nrf2 system was activated. Therefore, formononetin might be a promising candidate drug that can enhance survival of skin flaps.
Collapse
Affiliation(s)
- Haoliang Li
- Department of Orthopaedics, The Second Affiliated Hospital, Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Renhao Jiang
- Department of Orthopaedics, The Second Affiliated Hospital, Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Lejing Lou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chao Jia
- Department of Orthopaedics, The Second Affiliated Hospital, Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Linfang Zou
- Department of Orthopaedics, The Second Affiliated Hospital, Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
| | - Mochuan Chen
- Department of Orthopaedics, The Second Affiliated Hospital, Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Mochuan Chen,
| |
Collapse
|
16
|
Mecheliolide elicits ROS-mediated ERS driven immunogenic cell death in hepatocellular carcinoma. Redox Biol 2022; 54:102351. [PMID: 35671636 PMCID: PMC9168183 DOI: 10.1016/j.redox.2022.102351] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/13/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022] Open
Abstract
The nonnegligible reason for the poor prognosis of hepatocellular carcinoma (HCC) is resistance to conventional chemotherapy. Immunogenic cell death (ICD) is a rare immunostimulatory form of cell death that can reengage the tumor-specific immune system. ICD can improve the clinical outcomes of chemotherapeutics by promoting a long-term cancer immunity. The discovery of potential ICD inducers is emerging as a promising direction. In the present study, micheliolide (MCL), a natural guaianolide sesquiterpene lactone, was screened out by the virtual screening strategies, identified as an inhibitor of thioredoxin reductase (TrxR) and was evaluated to have high potential to induce ICD. Here, we showed that MCL induced ICD-associated DAMPs (damage-associated molecular patterns, such as CRT exposure, ATP secretion and HMGB1 release). MCL significantly triggered the regression of established tumors in an immunocompetent mouse vaccine model, and induced ICD (DCs maturation, the stimulation of CD4+, and CD8+ T-cells responses) in vivo. Mechanistically, we found that the magnitude of ICD-associated effects induced upon exposure of HCC cells to MCL was dependent on the generation of reactive oxygen species (ROS)-mediated endoplasmic reticulum stress (ERS). In addition, the suppression of ROS normalized MCL-induced ERS, in contrast, the downregulation of TrxR synergized with the ERS driven by MCL. We also systematically detected the H2O2 generation using Hyper7 sensors in HCC cells exposed to MCL. Notably, MCL inhibited the development of HCC organoids. Collectively, our results reveal a potential association between the TrxR inhibitors and ICD, presenting valuable insights into the MCL-activated ICD in HCC cells.
Collapse
|
17
|
Natural compounds as inhibitors of thioredoxin reductase (TrxR1). Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3431-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
18
|
Felber JG, Poczka L, Scholzen KC, Zeisel L, Maier MS, Busker S, Theisen U, Brandstädter C, Becker K, Arnér ESJ, Thorn-Seshold J, Thorn-Seshold O. Cyclic 5-membered disulfides are not selective substrates of thioredoxin reductase, but are opened nonspecifically. Nat Commun 2022; 13:1754. [PMID: 35365603 PMCID: PMC8975869 DOI: 10.1038/s41467-022-29136-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/01/2022] [Indexed: 11/08/2022] Open
Abstract
The cyclic five-membered disulfide 1,2-dithiolane has been widely used in chemical biology and in redox probes. Contradictory reports have described it either as nonspecifically reduced in cells, or else as a highly specific substrate for thioredoxin reductase (TrxR). Here we show that 1,2-dithiolane probes, such as "TRFS" probes, are nonspecifically reduced by thiol reductants and redox-active proteins, and their cellular performance is barely affected by TrxR inhibition or knockout. Therefore, results of cellular imaging or inhibitor screening using 1,2-dithiolanes should not be interpreted as reflecting TrxR activity, and previous studies may need re-evaluation. To understand 1,2-dithiolanes' complex behaviour, probe localisation, environment-dependent fluorescence, reduction-independent ring-opening polymerisation, and thiol-dependent cellular uptake must all be considered; particular caution is needed when co-applying thiophilic inhibitors. We present a general approach controlling against assay misinterpretation with reducible probes, to ensure future TrxR-targeted designs are robustly evaluated for selectivity, and to better orient future research.
Collapse
Affiliation(s)
- Jan G Felber
- Department of Pharmacy, Ludwig-Maximilians University Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Lena Poczka
- Department of Pharmacy, Ludwig-Maximilians University Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Karoline C Scholzen
- Department of Medical Biochemistry, Karolinska Institutet, Solnavägen 9, 171 77, Stockholm, Sweden
| | - Lukas Zeisel
- Department of Pharmacy, Ludwig-Maximilians University Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Martin S Maier
- Department of Pharmacy, Ludwig-Maximilians University Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Sander Busker
- Department of Medical Biochemistry, Karolinska Institutet, Solnavägen 9, 171 77, Stockholm, Sweden
- Pelago Bioscience AB, 171 48, Solna, Sweden
| | - Ulrike Theisen
- Zoological Institute, Cellular and Molecular Neurobiology, TU Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Christina Brandstädter
- Interdisciplinary Research Centre (IFZ), Justus-Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Katja Becker
- Interdisciplinary Research Centre (IFZ), Justus-Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Elias S J Arnér
- Department of Medical Biochemistry, Karolinska Institutet, Solnavägen 9, 171 77, Stockholm, Sweden
- Department of Selenoprotein Research, National Institute of Oncology, 1122, Budapest, Hungary
| | - Julia Thorn-Seshold
- Department of Pharmacy, Ludwig-Maximilians University Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Oliver Thorn-Seshold
- Department of Pharmacy, Ludwig-Maximilians University Munich, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
19
|
Patwardhan RS, Sharma D, Sandur SK. Thioredoxin reductase: An emerging pharmacologic target for radiosensitization of cancer. Transl Oncol 2022; 17:101341. [PMID: 35078017 PMCID: PMC8790659 DOI: 10.1016/j.tranon.2022.101341] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Novel agents are required to increase the radiosensitivity of cancer and improve the outcome of radiotherapy. Thioredoxin (Trx) and thioredoxin reductase (TrxR) reduce the oxidized cysteine thiols in several proteins, which regulate cellular redox, survival, proliferation, DNA synthesis, transcription factor activity and apoptosis. TrxR is essential for maintaining a conducive redox state for tumor growth, survival and resistance to therapy. Therefore, it is an appealing pharmacological target for the radiosensitization of tumors. Ionizing radiation (IR) is known to cause cytotoxicity through ROS, oxidative stress and DNA damage. Inhibition of thioredoxin system augments IR induced oxidative stress and potentiates cytotoxic effects. However, TrxR also regulates several critical cellular processes in normal cells. Here, we highlight the pre-clinical research and pharmacological studies to surmise possible utility of different TrxR inhibitors for radiosensitization. This review provides a succinct perspective on the role of TrxR inhibitors during the radiotherapy of cancer.
Collapse
Affiliation(s)
- Raghavendra S Patwardhan
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
20
|
Tolbatov I, Marrone A. Selenocysteine of thioredoxin reductase as the primary target for the antitumor metallodrugs: A computational point of view. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2022.122330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Xu Q, Zhang J. Novel strategies for targeting the thioredoxin system for cancer therapy. Expert Opin Drug Discov 2022; 17:437-442. [PMID: 35193453 DOI: 10.1080/17460441.2022.2045270] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION : The thioredoxin system is increasingly recognized as an important executor for maintaining cell redox homeostasis and regulating multiple cell signaling pathways. Targeting this system for cancer treatment has therefore attracted much attention. AREAS COVERED : The authors focus on providing coverage and emphasizing the strategy of targeting the thioredoxin system to develop anticancer therapeutics in the past five years, especially from the perspective of discovering novel protein functions or new downstream regulatory pathways, and designing new therapeutic reagents. The authors also provide the readers with their expert perspectives for future development. EXPERT OPINION : The limited pharmacophore of inhibitors and the slow progress of clinical research partially restrict the development of anticancer drugs targeting the thioredoxin system, necessitating thus novel strategies to accelerate the system for treating cancer. Nevertheless, the synergistic targeting of thioredoxin system for cancer therapy is a promising strategy, particularly with regards to chemotherapy resistance and/or sensitization immunotherapy.
Collapse
Affiliation(s)
- Qianhe Xu
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
22
|
Zhang J, Xu Q, Ma D. Inhibition of thioredoxin reductase by natural anticancer candidate β-lapachone accounts for triggering redox activation-mediated HL-60 cell apoptosis. Free Radic Biol Med 2022; 180:244-252. [PMID: 35091063 DOI: 10.1016/j.freeradbiomed.2022.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/11/2022] [Accepted: 01/22/2022] [Indexed: 02/06/2023]
Abstract
β-Lapachone as a natural novel anticancer candidate is under clinical trials. Previous studies suggested that β-lapachone works by redox activation to ablate cancer cells. However, it is still unclear whether thioredoxin reductase (TrxR), one of the key redox catalytic enzymes in cells, plays a role in the pharmacological effects of β-lapachone. Herein, we present that β-lapachone kills human promyelocytic leukemia HL-60 cells with preference over other cancer cells and normal cells. The follow-up studies demonstrate that β-lapachone induces the HL-60 cell apoptosis through inhibition of TrxR and further elevation of oxidative stress. Overexpression of the TrxR alleviates the efficiency of β-lapachone while knockdown of the enzyme increases the β-lapachone cytotoxicity, scientifically underpinning the correlation of the observed biological behaviors of β-lapachone to TrxR inhibition. The disclosure of the novel action mechanism of β-lapachone sheds light on understanding its capacity in interfering with cellular redox signaling and supports β-lapachone as an anticancer drug candidate.
Collapse
Affiliation(s)
- Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau (SAR), China.
| | - Qianhe Xu
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Di Ma
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
23
|
Xia D, Liu H, Cheng X, Maraswami M, Chen Y, Lv X. Recent Developments of Coumarin-based Hybrids in Drug Discovery. Curr Top Med Chem 2022; 22:269-283. [PMID: 34986774 DOI: 10.2174/1568026622666220105105450] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/23/2021] [Accepted: 12/05/2021] [Indexed: 11/22/2022]
Abstract
Coumarin scaffold is a highly significant O-heterocycle, namely benzopyran-2-ones, form an elite class of naturally occurring compounds that possess promising therapeutic perspectives. Based on its broad spectrum of biological activities, the privileged coumarin scaffold is applied to medicinal and pharmacological treatments by several rational design strategies and approaches. Structure-activity relationships of the coumarin-based hybrids with various bioactivity fragments revealed significant information toward the further development of highly potent and selective disorder therapeutic agents. The molecular docking studies between coumarins and critical therapeutic enzymes demonstrated mode of action by forming noncovalent interactions with more than one receptor, further rationally confirm information about structure-activity relationships. This review summarizes recent developments relating to coumarin-based hybrids with other pharmacophores aiming to numerous feasible therapeutic enzymatic targets to combat various therapeutic fields, including anticancer, antimicrobic, anti-Alzheimer, anti-inflammatory activities.
Collapse
Affiliation(s)
- Dongguo Xia
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| | - Hao Liu
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| | - Xiang Cheng
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| | - Manikantha Maraswami
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 637371, Singapore
| | - Yiting Chen
- Fujian Provincial University Engineering Research Center of Green Materials and Chemical Engineering, Minjiang University, 350108, Fuzhou, China
| | - Xianhai Lv
- School of Science, Anhui Agricultural University, 230036, Hefei, China
| |
Collapse
|
24
|
Yan J, Xu Y, Jin X, Zhang Q, Ouyang F, Han L, Zhan M, Li X, Liang B, Huang X. Structure modification and biological evaluation of indole-chalcone derivatives as anti-tumor agents through dual targeting tubulin and TrxR. Eur J Med Chem 2022; 227:113897. [PMID: 34649064 DOI: 10.1016/j.ejmech.2021.113897] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022]
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,β-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.
Collapse
Affiliation(s)
- Jun Yan
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510720, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Yuzhu Xu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510720, China
| | - Xing Jin
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510720, China
| | - Qiaoxuan Zhang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510720, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Feng Ouyang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Liqiao Han
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Min Zhan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Baoxia Liang
- The School of Food and Biological Engineering, Guangdong Polytechnic of Science and Trade, Guangzhou, 510430, China.
| | - Xianzhang Huang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510720, China; Department of Laboratory Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
25
|
The Toxic Mechanism of Gliotoxins and Biosynthetic Strategies for Toxicity Prevention. Int J Mol Sci 2021; 22:ijms222413510. [PMID: 34948306 PMCID: PMC8705807 DOI: 10.3390/ijms222413510] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 12/13/2022] Open
Abstract
Gliotoxin is a kind of epipolythiodioxopiperazine derived from different fungi that is characterized by a disulfide bridge. Gliotoxins can be biosynthesized by a gli gene cluster and regulated by a positive GliZ regulator. Gliotoxins show cytotoxic effects via the suppression the function of macrophage immune function, inflammation, antiangiogenesis, DNA damage by ROS production, peroxide damage by the inhibition of various enzymes, and apoptosis through different signal pathways. In the other hand, gliotoxins can also be beneficial with different doses. Low doses of gliotoxin can be used as an antioxidant, in the diagnosis and treatment of HIV, and as an anti-tumor agent in the future. Gliotoxins have also been used in the control of plant pathogens, including Pythium ultimum and Sclerotinia sclerotiorum. Thus, it is important to elucidate the toxic mechanism of gliotoxins. The toxic mechanism of gliotoxins and biosynthetic strategies to reduce the toxicity of gliotoxins and their producing strains are summarized in this review.
Collapse
|
26
|
Shutkov IA, Okulova YN, Tyurin VY, Sokolova EV, Babkov DA, Spasov AA, Gracheva YA, Schmidt C, Kirsanov KI, Shtil AA, Redkozubova OM, Shevtsova EF, Milaeva ER, Ott I, Nazarov AA. Ru(III) Complexes with Lonidamine-Modified Ligands. Int J Mol Sci 2021; 22:ijms222413468. [PMID: 34948263 PMCID: PMC8707700 DOI: 10.3390/ijms222413468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
A series of bifunctional Ru(III) complexes with lonidamine-modified ligands (lonidamine is a selective inhibitor of aerobic glycolysis in cancer cells) was described. Redox properties of Ru(III) complexes were characterized by cyclic voltammetry. An easy reduction suggested a perspective for these agents as their whole mechanism of action seems to be based on activation by metal atom reduction. New compounds demonstrated a more pronounced antiproliferative potency than the parental drug; individual new agents were more cytotoxic than cisplatin. Stability studies showed an increase in the stability of complexes along with the linker length. A similar trend was noted for antiproliferative activity, cellular uptake, apoptosis induction, and thioredoxin reductase inhibition. Finally, at concentrations that did not alter water solubility, the selected new complex evoked no acute toxicity in Balb/c mice.
Collapse
Affiliation(s)
- Ilya A. Shutkov
- Department of Medicinal Chemistry & Fine Organic Synthesis, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991 Moscow, Russia; (I.A.S.); (Y.N.O.); (V.Y.T.); (Y.A.G.); (E.R.M.)
| | - Yulia N. Okulova
- Department of Medicinal Chemistry & Fine Organic Synthesis, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991 Moscow, Russia; (I.A.S.); (Y.N.O.); (V.Y.T.); (Y.A.G.); (E.R.M.)
| | - Vladimir Yu. Tyurin
- Department of Medicinal Chemistry & Fine Organic Synthesis, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991 Moscow, Russia; (I.A.S.); (Y.N.O.); (V.Y.T.); (Y.A.G.); (E.R.M.)
| | - Elena V. Sokolova
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39 Novorossiyskaya Street, 400087 Volgograd, Russia; (E.V.S.); (D.A.B.); (A.A.S.)
| | - Denis A. Babkov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39 Novorossiyskaya Street, 400087 Volgograd, Russia; (E.V.S.); (D.A.B.); (A.A.S.)
| | - Alexander A. Spasov
- Scientific Center for Innovative Drugs, Volgograd State Medical University, 39 Novorossiyskaya Street, 400087 Volgograd, Russia; (E.V.S.); (D.A.B.); (A.A.S.)
| | - Yulia A. Gracheva
- Department of Medicinal Chemistry & Fine Organic Synthesis, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991 Moscow, Russia; (I.A.S.); (Y.N.O.); (V.Y.T.); (Y.A.G.); (E.R.M.)
| | - Claudia Schmidt
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, 55 Beethovenstrasse, 38106 Braunschweig, Germany; (C.S.); (I.O.)
| | - Kirill I. Kirsanov
- Blokhin Cancer Research Center, 24 Kashirskoye Shosse, 115478 Moscow, Russia; (K.I.K.); (A.A.S.)
- Institute of Medicine, RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russia
| | - Alexander A. Shtil
- Blokhin Cancer Research Center, 24 Kashirskoye Shosse, 115478 Moscow, Russia; (K.I.K.); (A.A.S.)
| | | | - Elena F. Shevtsova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 1 Severniy Proezd, 142432 Chernogolovka, Russia;
| | - Elena R. Milaeva
- Department of Medicinal Chemistry & Fine Organic Synthesis, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991 Moscow, Russia; (I.A.S.); (Y.N.O.); (V.Y.T.); (Y.A.G.); (E.R.M.)
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, 55 Beethovenstrasse, 38106 Braunschweig, Germany; (C.S.); (I.O.)
| | - Alexey A. Nazarov
- Department of Medicinal Chemistry & Fine Organic Synthesis, Lomonosov Moscow State University, 1/3 Leninskie Gory, 119991 Moscow, Russia; (I.A.S.); (Y.N.O.); (V.Y.T.); (Y.A.G.); (E.R.M.)
- Correspondence:
| |
Collapse
|
27
|
Li M, Cheng W, Zhang L. Maternal selenium deficiency suppresses proliferation, induces autophagy dysfunction and apoptosis in the placenta of mice. Metallomics 2021; 13:6406492. [PMID: 34669944 DOI: 10.1093/mtomcs/mfab058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/10/2021] [Indexed: 12/16/2022]
Abstract
Selenium deficiency is thought to be associated with the occurrence of gestational complications. However, the underlying mechanism of selenium deficiency impairs placental function remains unclear. In this study, female mice were separately supplemented with a Se-deficient (0.02 mg/kg Se) or control diet (0.2 mg/kg Se) for 12 weeks before mating and throughout gestation. Maternal liver and placentas were collected at embryonic day 15.5 and analyzed for Se content. Oxidative stress status, proliferation capability, autophagy, and apoptosis of the placenta were determined. We found that maternal selenium deficiency decreased placental Se concentration and some antioxidant selenoproteins expressions. The concentrations of catalase and glutathione in selenium-deficient placentas were reduced, along with an increase in hydrogen peroxide (H2O2) content. Selenium deficiency inhibited the expression of proliferating cell nuclear antigen. Autophagosomes, autophagolysosomes, and upregulation of autophagy-related protein microtubule-associated protein 1 light chain 3 alpha II (LC3B), Beclin1, PTEN-induced putative kinase 1 (PINK1), and Parkin were found in the selenium-deficient trophoblasts. Autophagic substrate p62/sequestosome 1 was surprisingly increased, indicating autophagy flux dysfunction. Selenium deficiency increased expressions of B cell leukemia/lymphoma 2 associated X protein (Bax), cleaved caspase-9/-3, and decreased the B cell leukemia/lymphoma 2 (Bcl2) level. Moreover, typical apoptotic ultrastructure and apoptosis-positive cells were observed in the selenium-deficient placenta. Our results suggested that maternal selenium deficiency impaired placental proliferation, induced autophagy dysfunction and apoptosis via increasing oxidative stress, and the Akt/mechanistic target of rapamycin (mTOR) pathway involved in this process. This study revealed a novel mechanism by which maternal selenium deficiency caused impairment of the placenta.
Collapse
Affiliation(s)
- Mengdi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lantian Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
28
|
Song ZL, Zhang J, Xu Q, Shi D, Yao X, Fang J. Structural Modification of Aminophenylarsenoxides Generates Candidates for Leukemia Treatment via Thioredoxin Reductase Inhibition. J Med Chem 2021; 64:16132-16146. [PMID: 34704769 DOI: 10.1021/acs.jmedchem.1c01441] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
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.
Collapse
Affiliation(s)
- Zi-Long Song
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,Botanical Agrochemicals Research & Development Center, Lanzhou Jiaotong University, Lanzhou 730070, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qianhe Xu
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Danfeng Shi
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.,State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, School of Pharmacy, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
29
|
Zhang J, Xu Q, Yang HY, Yang M, Fang J, Gao K. Inhibition of Thioredoxin Reductase by Santamarine Conferring Anticancer Effect in HeLa Cells. Front Mol Biosci 2021; 8:710676. [PMID: 34485384 PMCID: PMC8416462 DOI: 10.3389/fmolb.2021.710676] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/06/2021] [Indexed: 01/03/2023] Open
Abstract
Natural products frequently have unique physiological activities and new action mechanisms due to their structural diversity and novelty, and are an important source for innovative drugs and lead compounds. We present herein that natural product santamarine targeted thioredoxin reductase (TrxR) to weaken its antioxidative function in cells, accompanied by accumulation of high levels of reactive oxygen species (ROS), and finally induced a new mechanism of tumor cell oxidative stress-mediated apoptosis. TrxR knockdown or overexpression cell lines were employed to further evaluate the cytotoxicity of santamarine regulated by TrxR, demonstrated that TrxR played a key role in the physiological effect of santamarine on cells. Santamarine targeting TrxR reveals its previously unrecognized mechanism of antitumor and provides a basis for the further development of santamarine as a potential cancer therapeutic agent.
Collapse
Affiliation(s)
- Junmin Zhang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Qianhe Xu
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Hong-Ying Yang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Minghao Yang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Jianguo Fang
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Kun Gao
- School of Pharmacy, State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| |
Collapse
|
30
|
Thioredoxin reductase as a pharmacological target. Pharmacol Res 2021; 174:105854. [PMID: 34455077 DOI: 10.1016/j.phrs.2021.105854] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/15/2021] [Accepted: 08/23/2021] [Indexed: 02/07/2023]
Abstract
Thioredoxin reductases (TrxRs) belong to the pyridine nucleotide disulfide oxidoreductase family enzymes that reduce thioredoxin (Trx). The couple TrxR and Trx is one of the major antioxidant systems that control the redox homeostasis in cells. The thioredoxin system, comprised of TrxR, Trx and NADPH, exerts its activities via a disulfide-dithiol exchange reaction. Inhibition of TrxR is an important clinical goal in all conditions in which the redox state is perturbed. The present review focuses on the most critical aspects of the cellular functions of TrxRs and their inhibition mechanisms by metal ions or chemicals, through direct targeting of TrxRs or their substrates or protein interactors. To update the involvement of overactivation/dysfunction of TrxRs in various pathological conditions, human diseases associated with TrxRs genes were critically summarized by publicly available genome-wide association study (GWAS) catalogs and literature. The pieces of evidence presented here justify why TrxR is recognized as one of the most critical clinical targets and the growing current interest in developing molecules capable of interfering with the functions of TrxR enzymes.
Collapse
|
31
|
Abstract
The cytosolic selenoprotein thioredoxin reductase 1 (TrxR1, TXNRD1), and to some extent mitochondrial TrxR2 (TXNRD2), can be inhibited by a wide range of electrophilic compounds. Many such compounds also yield cytotoxicity toward cancer cells in culture or in mouse models, and most compounds are likely to irreversibly modify the easily accessible selenocysteine residue in TrxR1, thereby inhibiting its normal activity to reduce cytosolic thioredoxin (Trx1, TXN) and other substrates of the enzyme. This leads to an oxidative challenge. In some cases, the inhibited forms of TrxR1 are not catalytically inert and are instead converted to prooxidant NADPH oxidases, named SecTRAPs, thus further aggravating the oxidative stress, particularly in cells expressing higher levels of the enzyme. In this review, the possible molecular and cellular consequences of these effects are discussed in relation to cancer therapy, with a focus on outstanding questions that should be addressed if targeted TrxR1 inhibition is to be further developed for therapeutic use. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Radosveta Gencheva
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden; .,Department of Selenoprotein Research, National Institute of Oncology, Budapest 1122, Hungary
| |
Collapse
|
32
|
Karunanithi S, Liu R, Hou Y, Gonzalez G, Oldford N, Roe AJ, Idipilly N, Gupta K, Amara CS, Putluri S, Lee GK, Valentin-Goyco J, Stetson L, Moreton SA, Putluri V, Kavuri SM, Saunthararajah Y, de Lima M, Tochtrop GP, Putluri N, Wald DN. Thioredoxin reductase is a major regulator of metabolism in leukemia cells. Oncogene 2021; 40:5236-5246. [PMID: 34239044 PMCID: PMC8380733 DOI: 10.1038/s41388-021-01924-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 05/20/2021] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
Despite the fact that AML is the most common acute leukemia in adults, patient outcomes are poor necessitating the development of novel therapies. We identified that inhibition of Thioredoxin Reductase (TrxR) is a promising strategy for AML and report a highly potent and specific inhibitor of TrxR, S-250. Both pharmacologic and genetic inhibition of TrxR impairs the growth of human AML in mouse models. We found that TrxR inhibition leads to a rapid and marked impairment of metabolism in leukemic cells subsequently leading to cell death. TrxR was found to be a major and direct regulator of metabolism in AML cells through impacts on both glycolysis and the TCA cycle. Studies revealed that TrxR directly regulates GAPDH leading to a disruption of glycolysis and an increase in flux through the pentose phosphate pathway (PPP). The combined inhibition of TrxR and the PPP led to enhanced leukemia growth inhibition. Overall, TrxR abrogation, particularly with S-250, was identified as a promising strategy to disrupt AML metabolism.
Collapse
Affiliation(s)
- Sheelarani Karunanithi
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- CuronBiotech Inc, Cleveland, OH, USA
| | - Ruifu Liu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Giancarlo Gonzalez
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Natasha Oldford
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Anne Jessica Roe
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- CuronBiotech Inc, Cleveland, OH, USA
| | - Nethrie Idipilly
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
- CuronBiotech Inc, Cleveland, OH, USA
| | - Kalpana Gupta
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Chandra Sekhar Amara
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Satwikreddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Grace Kyueun Lee
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Juan Valentin-Goyco
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Lindsay Stetson
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | | | - Vasanta Putluri
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Shyam M Kavuri
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Marcos de Lima
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, OH, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - David N Wald
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.
- CuronBiotech Inc, Cleveland, OH, USA.
- Department of Pathology, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
33
|
Zaher DM, Ramadan WS, El-Awady R, Omar HA, Hersi F, Srinivasulu V, Hachim IY, Al-Marzooq FI, Vazhappilly CG, Merali S, Merali C, Soares NC, Schilf P, Ibrahim SM, Al-Tel TH. A Novel Benzopyrane Derivative Targeting Cancer Cell Metabolic and Survival Pathways. Cancers (Basel) 2021; 13:cancers13112840. [PMID: 34200264 PMCID: PMC8201054 DOI: 10.3390/cancers13112840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 02/03/2023] Open
Abstract
(1) Background: Today, the discovery of novel anticancer agents with multitarget effects and high safety margins represents a high challenge. Drug discovery efforts indicated that benzopyrane scaffolds possess a wide range of pharmacological activities. This spurs on building a skeletally diverse library of benzopyranes to identify an anticancer lead drug candidate. Here, we aim to characterize the anticancer effect of a novel benzopyrane derivative, aiming to develop a promising clinical anticancer candidate. (2) Methods: The anticancer effect of SIMR1281 against a panel of cancer cell lines was tested. In vitro assays were performed to determine the effect of SIMR1281 on GSHR, TrxR, mitochondrial metabolism, DNA damage, cell cycle progression, and the induction of apoptosis. Additionally, SIMR1281 was evaluated in vivo for its safety and in a xenograft mice model. (3) Results: SIMR1281 strongly inhibits GSHR while it moderately inhibits TrxR and modulates the mitochondrial metabolism. SIMR1281 inhibits the cell proliferation of various cancers. The antiproliferative activity of SIMR1281 was mediated through the induction of DNA damage, perturbations in the cell cycle, and the inactivation of Ras/ERK and PI3K/Akt pathways. Furthermore, SIMR1281 induced apoptosis and attenuated cell survival machinery. In addition, SIMR1281 reduced the tumor volume in a xenograft model while maintaining a high in vivo safety profile at a high dose. (4) Conclusions: Our findings demonstrate the anticancer multitarget effect of SIMR1281, including the dual inhibition of glutathione and thioredoxin reductases. These findings support the development of SIMR1281 in preclinical and clinical settings, as it represents a potential lead compound for the treatment of cancer.
Collapse
Affiliation(s)
- Dana M. Zaher
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Wafaa S. Ramadan
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Raafat El-Awady
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hany A. Omar
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Fatema Hersi
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Vunnam Srinivasulu
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
| | - Ibrahim Y. Hachim
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Farah I. Al-Marzooq
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Cijo G. Vazhappilly
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- School of Arts and Sciences, American University of Ras Al Khaimah, P.O. Box 10021, Ras Al Khaimah 10021, United Arab Emirates
| | - Salim Merali
- School of Pharmacy, Temple University, 3307 N Broad Street, Room 552, Philadelphia, PA 19140, USA; (S.M.); (C.M.)
| | - Carmen Merali
- School of Pharmacy, Temple University, 3307 N Broad Street, Room 552, Philadelphia, PA 19140, USA; (S.M.); (C.M.)
| | - Nelson C. Soares
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Paul Schilf
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany;
| | - Saleh M. Ibrahim
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany;
| | - Taleb H. Al-Tel
- Sharjah Institute for Medical Researches, University of Sharjah, Sharjah 27272, United Arab Emirates; (D.M.Z.); (W.S.R.); (R.E.-A.); (H.A.O.); (F.H.); (V.S.); (I.Y.H.); (F.I.A.-M.); (C.G.V.); (N.C.S.); (S.M.I.)
- College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: ; Tel.: +971-6505-7417
| |
Collapse
|
34
|
Zhang J, Duan D, Osama A, Fang J. Natural Molecules Targeting Thioredoxin System and Their Therapeutic Potential. Antioxid Redox Signal 2021; 34:1083-1107. [PMID: 33115246 DOI: 10.1089/ars.2020.8213] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Thioredoxin (Trx) and thioredoxin reductase are two core members of the Trx system. The system bridges the gap between the universal reducing equivalent NADPH and various biological molecules and plays an essential role in maintaining cellular redox homeostasis and regulating multiple cellular redox signaling pathways. Recent Advance: In recent years, the Trx system has been well documented as an important regulator of many diseases, especially tumorigenesis. Thus, the development of potential therapeutic molecules targeting the system is of great significance for disease treatment. Critical Issues: We herein first discuss the physiological functions of the Trx system and the role that the Trx system plays in various diseases. Then, we focus on the introduction of natural small molecules with potential therapeutic applications, especially the anticancer activity, and review their mechanisms of pharmacological actions via interfering with the Trx system. Finally, we further discuss several natural molecules that harbor therapeutic potential and have entered different clinical trials. Future Directions: Further studies on the functions of the Trx system in multiple diseases will not only improve our understanding of the pathogenesis of many human disorders but also help develop novel therapeutic strategies against these diseases. Antioxid. Redox Signal. 34, 1083-1107.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
- Shaanxi Key Laboratory of Phytochemistry, Baoji University of Arts and Sciences, Baoji, China
| | - Dongzhu Duan
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
- Shaanxi Key Laboratory of Phytochemistry, Baoji University of Arts and Sciences, Baoji, China
| | - Alsiddig Osama
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
- Shaanxi Key Laboratory of Phytochemistry, Baoji University of Arts and Sciences, Baoji, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, and School of Pharmacy, Lanzhou University, Lanzhou, China
- Shaanxi Key Laboratory of Phytochemistry, Baoji University of Arts and Sciences, Baoji, China
| |
Collapse
|
35
|
Dar'in D, Kantin G, Chupakhin E, Sharoyko V, Krasavin M. Natural-Like Spirocyclic Δ α,β -Butenolides Obtained from Diazo Homophthalimides. Chemistry 2021; 27:8221-8227. [PMID: 33848018 DOI: 10.1002/chem.202100880] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 01/07/2023]
Abstract
α-Diazo homophotalimides were reacted with various propiolic acids on Rh2 (esp)2 catalysis. The resulting propiolate esters were transformed into novel, heterocyclic Δα,β -spirobutenolides in good to excellent product yields. The approach represents a fundamentally novel entry into natural-like Δα,β -spirobutenolides present in many biologically active natural products as well as fully synthetic compounds endowed with diverse biological activities. The Δα,β -spirobutenolides thus obtained were shown to inhibit thioredoxin reductase, a selenocysteine enzyme target for cancer. Moreover, for the best compound in the series (TrxR IC50 1.49±0.08 μM), by using MALDI-TOF mass-spectrometry it was shown that it selectively binds selenocysteine in the presence of a 10-fold excess of cysteine. This validates the new compound as a promising lead for anticancer therapy development.
Collapse
Affiliation(s)
- Dmitry Dar'in
- Chair of Natural Products Chemistry, Saint Petersburg State University, Saint-Petersburg, 199034, Russian Federation
| | - Grigory Kantin
- Chair of Natural Products Chemistry, Saint Petersburg State University, Saint-Petersburg, 199034, Russian Federation
| | - Evgeny Chupakhin
- Chair of Natural Products Chemistry, Saint Petersburg State University, Saint-Petersburg, 199034, Russian Federation.,Immanuel Kant Baltic Federal University, Kaliningrad, 236041, Russian Federation
| | - Vladimir Sharoyko
- Chair of Natural Products Chemistry, Saint Petersburg State University, Saint-Petersburg, 199034, Russian Federation.,Laboratory for Cell Biotechnology, Saint Petersburg State Institute of Technology (University), Saint Petersburg, 190013, Russian Federation
| | - Mikhail Krasavin
- Chair of Natural Products Chemistry, Saint Petersburg State University, Saint-Petersburg, 199034, Russian Federation.,Immanuel Kant Baltic Federal University, Kaliningrad, 236041, Russian Federation
| |
Collapse
|
36
|
Liu Y, Yu Y, Meng Q, Jia X, Zhu J, Tang C, Zhao Q, Feng X, Zhang J. A Fluorescent Probe for the Specific Staining of Cysteine Containing Proteins and Thioredoxin Reductase in SDS-PAGE. BIOSENSORS 2021; 11:bios11050132. [PMID: 33922498 PMCID: PMC8146322 DOI: 10.3390/bios11050132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 06/12/2023]
Abstract
A naphthalimide-based fluorescent probe, Nap-I, with iodoacetamide as the alkylating group, has been synthesized, and its specific fluorescent staining of proteins containing cysteine (Cys) and selenocysteine (Sec) residues in sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) has been evaluated. This molecule shows good fluorescence properties in the labeling of protein Cys/Sec residues, while reducing steric hindrance and minimizing changes in the water solubility of proteins. Reaction parameters, such as labeling time and pH, have been investigated, and the optimal labeling conditions for Cys-containing proteins have been determined. Thioredoxin reductase (TXNRD) is best stained at low pH. The probe Nap-I has been successfully used for the quantification of serum proteins and hemoglobin in Tan sheep serum, and TXNRD in Tan sheep liver and muscle has been labeled at low pH. Based on the probe Nap-I, we have also distinguished TXNRD1 and TXNRD2 by SDS-PAGE. The results showed that, compared with the normal microenvironment in which the protein resides, the lower the pH value, the greater the TXNRD activity.
Collapse
Affiliation(s)
- Yuning Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanan Yu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qingshi Meng
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xueting Jia
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Jiawei Zhu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Chaohua Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qingyu Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaohui Feng
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junmin Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.L.); (Y.Y.); (Q.M.); (X.J.); (J.Z.); (C.T.); (Q.Z.)
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
37
|
Chupakhin E, Krasavin M. Thioredoxin reductase inhibitors: updated patent review (2017-present). Expert Opin Ther Pat 2021; 31:745-758. [PMID: 33666133 DOI: 10.1080/13543776.2021.1899160] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Thioredoxin reductase (TrxR) is a selenocysteine-containing enzyme which is responsible - as a part of the thioredoxin system - for maintaining redox homeostasis in cells. It is upregulated in cancerous state as a defense against oxidative stress. TrxR has been mostly considered an anticancer drug target although it has applications in other therapeutic areas such as neurodegeneration, inflammation, microbial infections, and neonatal hyperoxic lung injury.Areas covered: The present review covers the patent literature that appeared in the period 2017-2020, i.e. since the publication of the previous expert opinion patent review on TrxR inhibitors. The recent additions to the following traditional classes of inhibitors are discussed: metal complexes, Michael acceptors as well as arsenic and selenium compounds. At the same time, a novel group of nitro (hetero)aromatic compounds have emerged which likely acts via covalent inhibition mechanism. Several miscellaneous chemotypes are grouped under Miscellaneous subsection.Expert opinion: While specificity over glutathione reductase is achieved easily, TrxR is still moving toward the later stages of development at a very slow rate. Michael acceptors, particularly based on TRXR substrate-mimicking scaffolds, are gaining impetus and so are dual and hybrid compounds. The development prospects of the emerging nitro (hetero)aromatic chemotypes remain uncertain.
Collapse
Affiliation(s)
- Evgeny Chupakhin
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg Russian Federation.,Institute for Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad Russian Federation
| | - Mikhail Krasavin
- Institute of Chemistry, Saint Petersburg State University, Saint Petersburg Russian Federation.,Institute for Living Systems, Immanuel Kant Baltic Federal University, Kaliningrad Russian Federation
| |
Collapse
|
38
|
Sohrabi M, Saeedi M, Larijani B, Mahdavi M. Recent advances in biological activities of rhodium complexes: Their applications in drug discovery research. Eur J Med Chem 2021; 216:113308. [PMID: 33713976 DOI: 10.1016/j.ejmech.2021.113308] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/09/2021] [Accepted: 02/17/2021] [Indexed: 01/01/2023]
Abstract
Unique structure, characteristic reactivity, and facile synthesis of metal complexes have made them efficient ligands in drug development research. Among them, rhodium complexes have a limited history and there are a few discussions about their biological activities documented in the literature. However, investigation of kinetically inert rhodium complexes has recently attracted lots of attention and especially there are various evidences on their anti-cancer activity. It seems that they can be investigated as a versatile surrogates or candidates for the existing drugs which do not affect selectively or suffer from various side effects. In recent years, there has been an increasing interest in the use of mononuclear rhodium (III) organometallo drugs due to its versatile structurally important aspects to inhibit various enzymes. It has been demonstrated that organometallic Rh complexes profiting from both organic and inorganic aspects have shown more potent biological activities than classical inorganic compartments. In this respect, smart design, use of the appropriate organic ligands, and efficient and user-friendly synthesis of organometallic Rh complexes have played crucial roles in the inducing desirable biological activities. In this review, we focused on the recent advances published on the bioactivity of Rh (III/II/I) complexes especially inhibitory activity, from 2013 till now. Accordingly, considering the structure-activity relationship (SAR), the effect of oxidation state (+1, +2, and +3) and geometry (dimer or monomer complexes with coordination number of 4 and 6) of Rh complexes as well as various ligands on in vitro and in vivo studies was comprehensively discussed.
Collapse
Affiliation(s)
- Marzieh Sohrabi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Saeedi
- Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Bu L, Tian Y, Wen H, Jia W, Yang S. miR-195-5p exerts tumor-suppressive functions in human lung cancer cells through targeting TrxR2. Acta Biochim Biophys Sin (Shanghai) 2021; 53:189-200. [PMID: 33332541 DOI: 10.1093/abbs/gmaa159] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Indexed: 12/12/2022] Open
Abstract
miR-195-5p has been widely explored in various cancers and is considered as a tumor-suppressive microRNA. However, its roles in human lung cancer pathogenesis are not fully elucidated. In this study, we aimed to explore how miR-195-5p is involved in malignant behaviors of lung adenocarcinoma (LUAD) cells. miR-195-5p expression was examined in the tumor tissues of patients with LUAD and human LUAD cell lines including A549 and PC-9. Thioredoxin reductase 2 (TrxR2) was predicted to be an mRNA target of miR-195-5p using online tools and validated by the Dual-Luciferase Reporter Assay. Lentivirus infection was used for gene overexpression, while gene knockdown was achieved by RNA interference. Cell proliferation was determined by Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine methods, and cell migration and invasion were assayed with transwell experiments. Cell apoptosis was determined by annexin V staining-based flow cytometry. The antitumor effects of miR-195-5p were also evaluated in nude mice xenografted with A549 cells. We found that miR-195-5p was lowly expressed in human LUAD cells, and its overexpression markedly suppressed cell proliferation, migration, and invasion and increased the apoptosis of LUAD cells in vitro. TrxR2 knockdown phenocopied the tumor-suppressive effects of miR-195-5p overexpression, while simultaneous TrxR2 overexpression remarkably reversed the effects of miR-195-5p overexpression on malignant behaviors of A549 and PC-9 cells. Additionally, miR-195-5p overexpression inhibited the growth of xenografted A549 tumor in nude mice. Our work verified that miR-195-5p exerts tumor-suppressive functions in LUAD cells through targeting TrxR2 and suggested that the miR-195-5p/TrxR2 axis is a potential biomarker for LUAD therapy.
Collapse
Affiliation(s)
- Lina Bu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Northwest University, Xi’an No.3 Hospital, Xi’an 710018, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Yingxuan Tian
- Department of Geriatric Respiratory, Shaanxi Provincial People’s Hospital, Xi’an 710068, China
| | - Hongqing Wen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Northwest University, Xi’an No.3 Hospital, Xi’an 710018, China
| | - Weihong Jia
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Northwest University, Xi’an No.3 Hospital, Xi’an 710018, China
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| |
Collapse
|
40
|
Seki Kioshima E, de Souza Bonfim de Mendonça P, de Melo Teixeira M, Grenier Capoci IR, Amaral A, Vilugron Rodrigues-Vendramini FA, Lauton Simões B, Rodrigues Abadio AK, Fernandes Matos L, Soares Felipe MS. One Century of Study: What We Learned about Paracoccidioides and How This Pathogen Contributed to Advances in Antifungal Therapy. J Fungi (Basel) 2021; 7:106. [PMID: 33540749 PMCID: PMC7913102 DOI: 10.3390/jof7020106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/19/2021] [Accepted: 01/26/2021] [Indexed: 02/08/2023] Open
Abstract
Paracoccidioidomycosis (PCM) is a notable fungal infection restricted to Latin America. Since the first description of the disease by Lutz up to the present day, Brazilian researchers have contributed to the understanding of the life cycle of this pathogen and provided the possibility of new targets for antifungal therapy based on the structural and functional genomics of Paracoccidioides. In this context, in silico approaches have selected molecules that act on specific targets, such as the thioredoxin system, with promising antifungal activity against Paracoccidioides. Some of these are already in advanced development stages. In addition, the application of nanostructured systems has addressed issues related to the high toxicity of conventional PCM therapy. Thus, the contribution of molecular biology and biotechnology to the advances achieved is unquestionable. However, it is still necessary to transcend the boundaries of synthetic chemistry, pharmaco-technics, and pharmacodynamics, aiming to turn promising molecules into newly available drugs for the treatment of fungal diseases.
Collapse
Affiliation(s)
- Erika Seki Kioshima
- Program in Biosciences and Pathophysiology, Department of Clinical Analysis and Biomedicine, State University of Maringa (UEM), Maringa, Parana 87020-900, Brazil; (P.d.S.B.d.M.); (I.R.G.C.); (F.A.V.R.-V.); (B.L.S.)
| | - Patrícia de Souza Bonfim de Mendonça
- Program in Biosciences and Pathophysiology, Department of Clinical Analysis and Biomedicine, State University of Maringa (UEM), Maringa, Parana 87020-900, Brazil; (P.d.S.B.d.M.); (I.R.G.C.); (F.A.V.R.-V.); (B.L.S.)
| | - Marcus de Melo Teixeira
- Faculty of Medicine, University of Brasília (UnB), Brasilia, Distrito Federal 70910-900, Brazil;
| | - Isis Regina Grenier Capoci
- Program in Biosciences and Pathophysiology, Department of Clinical Analysis and Biomedicine, State University of Maringa (UEM), Maringa, Parana 87020-900, Brazil; (P.d.S.B.d.M.); (I.R.G.C.); (F.A.V.R.-V.); (B.L.S.)
| | - André Amaral
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia 74690-900, Brazil;
| | - Franciele Abigail Vilugron Rodrigues-Vendramini
- Program in Biosciences and Pathophysiology, Department of Clinical Analysis and Biomedicine, State University of Maringa (UEM), Maringa, Parana 87020-900, Brazil; (P.d.S.B.d.M.); (I.R.G.C.); (F.A.V.R.-V.); (B.L.S.)
| | - Bruna Lauton Simões
- Program in Biosciences and Pathophysiology, Department of Clinical Analysis and Biomedicine, State University of Maringa (UEM), Maringa, Parana 87020-900, Brazil; (P.d.S.B.d.M.); (I.R.G.C.); (F.A.V.R.-V.); (B.L.S.)
| | - Ana Karina Rodrigues Abadio
- Faculty of Agricultural Social Sciences, Mato Grosso State University, Nova Mutum, Mato Grosso 78450-000, Brazil;
| | - Larissa Fernandes Matos
- Faculty of Ceilandia, University of Brasília (UnB), Brasília, Distrito Federal 72220-275, Brazil;
- Program in Microbial Biology, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil
| | - Maria Sueli Soares Felipe
- Program of Genomic Sciences and Biotechnology, Catholic University of Brasilia, Brasília 70790-160, Brazil;
| |
Collapse
|
41
|
Liu Y, Yu Y, Zhao Q, Tang C, Zhang H, Qin Y, Feng X, Zhang J. Fluorescent probes based on nucleophilic aromatic substitution reactions for reactive sulfur and selenium species: Recent progress, applications, and design strategies. Coord Chem Rev 2021; 427:213601. [PMID: 33024340 PMCID: PMC7529596 DOI: 10.1016/j.ccr.2020.213601] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
Abstract
Reactive sulfur species (RSS) and reactive selenium species (RSeS) are important substances for the maintenance of physiological balance. Imbalance of RSS and RSeS is closely related to a series of human diseases, so it is considered to be an important biomarker in early diagnosis, treatment, and stage monitoring. Fast and accurate quantitative analysis of different RSS and RSeS in complex biological systems may promote the development of personalized diagnosis and treatment in the future. One way to explore the physiological function of various types of RSS and RSeS in vivo is to detect them at the molecular level, and one of the most effective methods for this is to use fluorescent probes. Nucleophilic aromatic substitution (SNAr) reactions are commonly exploited as a detection mechanism for RSS and RSeS in fluorescent probes. In this review, we cover recent progress in fluorescent probes for RSS and RSeS based on SNAr reactions, and discuss their response mechanisms, properties, and applications. Benzenesulfonate, phenyl-O ether, phenyl-S ether, phenyl-Se ether, 7-nitro-2,1,3-benzoxadiazole (NBD), benzoate, and selenium-nitrogen bonds are all good detection groups. Moreover, based on an integration of different reports, we propose the design and synthesis of RSS- and RSeS-selective probes based on SNAr reactions, current challenges, and future research directions, considering the selection of active sites, the effect of substituents on the benzene ring, and the introduction of other functional groups.
Collapse
Affiliation(s)
- Yuning Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yanan Yu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Qingyu Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Chaohua Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Huiyan Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yuchang Qin
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Xiaohui Feng
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Junmin Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Scientific Observing and Experiment Station of Animal Genetic Resources and Nutrition in North China of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
42
|
Zhang W, Zhu Y, Yu H, Liu X, Jiao B, Lu X. Libertellenone H, a Natural Pimarane Diterpenoid, Inhibits Thioredoxin System and Induces ROS-Mediated Apoptosis in Human Pancreatic Cancer Cells. Molecules 2021; 26:molecules26020315. [PMID: 33435380 PMCID: PMC7827531 DOI: 10.3390/molecules26020315] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 01/22/2023] Open
Abstract
Libertellenone H (LH), a marine-derived pimarane diterpenoid isolated from arctic fungus Eutypella sp. D-1, has shown effective cytotoxicity on a range of cancer cells. The present study is to explore the anticancer effect of LH on human pancreatic cancer cells and to investigate the intracellular molecular target and underlying mechanism. As shown, LH exhibited anticancer activity in human pancreatic cancer cells by promoting cell apoptosis. Mechanistic studies suggested that LH-induced reactive oxygen species (ROS) accumulation was responsible for apoptosis as antioxidant N-acetylcysteine (NAC) and antioxidant enzyme superoxide dismutase (SOD) antagonized the inhibitory effect of LH. Zymologic testing demonstrated that LH inhibited Trx system but had little effect on the glutathione reductase and glutaredoxin. Mass spectrometry (MS) analysis revealed that the mechanism of action was based on the direct conjugation of LH to the Cys32/Cys35 residue of Trx1 and Sec498 of TrxR, leading to a decrease in the cellular level of glutathione (GSH) and activation of downstream ASK1/JNK signaling pathway. Taken together, our findings revealed LH was a marine derived inhibitor of Trx system and an anticancer candidate.
Collapse
Affiliation(s)
- Weirui Zhang
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (W.Z.); (H.Y.); (X.L.)
| | - Yuping Zhu
- College of Basic Medical Sciences, Experimental Teacher Center, Naval Medical University, Shanghai 200433, China;
| | - Haobing Yu
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (W.Z.); (H.Y.); (X.L.)
| | - Xiaoyu Liu
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (W.Z.); (H.Y.); (X.L.)
| | - Binghua Jiao
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (W.Z.); (H.Y.); (X.L.)
- Correspondence: (B.J.); (X.L.); Tel.: +86-21-81870970 (ext. 8001) (B.J.); +86-21-81870970 (ext. 8004) (X.L.)
| | - Xiaoling Lu
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Shanghai 200433, China; (W.Z.); (H.Y.); (X.L.)
- Correspondence: (B.J.); (X.L.); Tel.: +86-21-81870970 (ext. 8001) (B.J.); +86-21-81870970 (ext. 8004) (X.L.)
| |
Collapse
|
43
|
Abstract
One of the systems responsible for maintaining cellular redox homeostasis is the thioredoxin-dependent system. An equally important function of this system is the regulation of the expression of many proteins by the transcription factor NF-κB or the apoptosis regulating kinase (ASK-1). Since it has been shown that the Trx-dependent system can contribute to both the enhancement of tumour angiogenesis and growth as well as apoptosis of neoplastic cells, the search for compounds that inhibit the level/activity of Trx and/or TrxR and thus modulate the course of the neoplastic process is ongoing. It has been shown that many naturally occurring polyphenolic compounds inactivate elements of the thioredoxin system. In addition, the effectiveness of Trx is inhibited by imidazole derivatives, while the activity of TrxR is reduced by transition metal ions complexes, dinitrohalobenzene derivatives, Michael acceptors, nitrosourea and ebselen. In addition, research is ongoing to identify new selective Trx/TrxR inhibitors.
Collapse
Affiliation(s)
- Anna Jastrząb
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Inorganic and Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
44
|
Xu J, Fang J. How can we improve the design of small molecules to target thioredoxin reductase for treating cancer? Expert Opin Drug Discov 2020; 16:331-333. [PMID: 33307863 DOI: 10.1080/17460441.2021.1854220] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jianqiang Xu
- School of Life and Pharmaceutical Sciences (LPS) and Panjin Institute of Industrial Technology (PIIT), Dalian University of Technology, Panjin, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry & College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| |
Collapse
|
45
|
Krasavin M, Sharonova T, Sharoyko V, Zhukovsky D, Kalinin S, Žalubovskis R, Tennikova T, Supuran CT. Combining carbonic anhydrase and thioredoxin reductase inhibitory motifs within a single molecule dramatically increases its cytotoxicity. J Enzyme Inhib Med Chem 2020; 35:665-671. [PMID: 32131646 PMCID: PMC7067156 DOI: 10.1080/14756366.2020.1734800] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A hypothesis that simultaneous targeting cancer-related carbonic anhydrase hCA IX and hCA XII isoforms (whose overexpression is a cancer cell’s defence mechanism against hypoxia) along with thioredoxin reductase (overexpressed in cancers as a defence against oxidative stress) may lead to synergistic antiproliferative effects was confirmed by testing combinations of the two inhibitor classes against pancreatic cancer cells (PANC-1). Combining both pharmacophoric motifs within one molecule led to a sharp increase of cytotoxicity. This preliminary observation sets the ground for a fundamentally new approach to anticancer agent design.
Collapse
Affiliation(s)
- Mikhail Krasavin
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Tatiana Sharonova
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Vladimir Sharoyko
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Daniil Zhukovsky
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Stanislav Kalinin
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Raivis Žalubovskis
- Latvian Institute of Organic Synthesis, Riga, Latvia.,Faculty of Materials Science and Applied Chemistry, Institute of Technology of Organic Chemistry, Riga Technical University, Riga, Latvia
| | - Tatiana Tennikova
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Claudiu T Supuran
- Neurofarba Department, Universita degli Studi di Firenze, Florence, Italy
| |
Collapse
|
46
|
Busker S, Page B, Arnér ESJ. To inhibit TrxR1 is to inactivate STAT3-Inhibition of TrxR1 enzymatic function by STAT3 small molecule inhibitors. Redox Biol 2020; 36:101646. [PMID: 32863208 PMCID: PMC7378686 DOI: 10.1016/j.redox.2020.101646] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 07/11/2020] [Indexed: 01/05/2023] Open
Abstract
The transcription factor STAT3 plays a key role in cancer and immunity, being widely explored as a potential drug target for the development of novel immunomodulatory or anticancer therapeutics. The mechanisms of small molecule-derived inhibition of STAT3 appear, however, to be more complex than initially perceived. Our recent discovery, that some novel STAT3 inhibitors were bona fide inhibitors of the cytosolic selenoprotein oxidoreductase TrxR1 (TXNRD1), led us to explore the effects of a wide array of previously described STAT3 inhibitors on TrxR1 function. We found that 17 out of 23 tested STAT3 small molecule inhibitors indeed inhibited purified TrxR1 at the reported concentrations yielding STAT3 inhibition. All tested compounds were electrophilic as shown by direct reactivities with GSH, and several were found to also be redox cycling substrates of TrxR1. Ten compounds previously shown to inhibit STAT3 were here found to irreversibly inhibit cellular TrxR1 activity (Auranofin, Stattic, 5,15-DPP, Galiellalactone, LLL12, Napabucasin, BP1-102, STA-21, S3I-201 and Degrasyn (WP1130)). Our findings suggest that targeting of TrxR1 may be a common feature for many small molecules that inhibit cellular STAT3 function. It is possible that prevention of STAT3 activation in cells by several small molecules classified as STAT3 inhibitors can be a downstream event following TrxR1 inhibition. Therefore, the relationship between TrxR1 and STAT3 should be considered when studying inhibition of either of these promising drug targets.
Collapse
Affiliation(s)
- Sander Busker
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Brent Page
- Department of Oncology-Pathology, Science for Life Laboratories, Karolinska Institutet, Stockholm, Sweden; Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden; Department of Selenoprotein Research, National Institute of Oncology, Budapest, Hungary.
| |
Collapse
|
47
|
6-Dithio-2'-deoxyguanosine analogs induce reactive oxygen species-mediated tumor cell apoptosis via bi-targeting thioredoxin 1 and telomerase. Toxicol Appl Pharmacol 2020; 401:115079. [PMID: 32497534 DOI: 10.1016/j.taap.2020.115079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/16/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023]
Abstract
Thioredoxin 1 (Trx1) and telomerase play key roles in the development and progression process of most tumors, and they both are promising drug therapy targets. We have, for the first time, discovered that Trx1 and telomerase had a dual-target synergistic effect. Based on that results, we designed a series of 6-dithio-2'-deoxyguanosine analogs (named as YLS00X) and verified whether they can inhibit Trx1 and telomerase simultaneously. TrxR1/Trx1 system activity and telomerase expression were significantly inhibited by 6-dithio-2'-deoxyguanosine analogs, especially YLS004. YLS004 can also cause ROS accumulation, and induce tumor cell apoptosis. The vitro antitumor activity of 6-dithio-2'-deoxyguanosine analogs using MTT assay on 11 different human cancer cells and found that human colon cancer cells(HCT116) and melanoma cells (A375) were the most sensitive cells to 6-dithio-2'-deoxyguanosine analogs treatment and vivo xenografts models also confirmed that. The serum biochemical parameters and multiple organs HE staining results of subacute experiments indicated that YLS004 might be mildly toxic to immune organs, including the thymus, spleen, and hematopoietic system. Besides, YLS004 was rapidly metabolized in the rats' blood. Our study revealed that YLS004, a Trx1 and telomerase inhibitor, has strong anti-tumor effects to colon cancer and melanoma cells and is a promising new candidate drug.
Collapse
|
48
|
Yao J, Duan D, Song ZL, Zhang J, Fang J. Sanguinarine as a new chemical entity of thioredoxin reductase inhibitor to elicit oxidative stress and promote tumor cell apoptosis. Free Radic Biol Med 2020; 152:659-667. [PMID: 31931095 DOI: 10.1016/j.freeradbiomed.2020.01.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/17/2022]
Abstract
The alteration of redox homeostasis is a hallmark of cancer cells. As a critical player in regulating cellular redox signaling, thioredoxin reductase (TrxR) enzymes are increasingly recognized as attractive targets for anticancer drug development. We reported herein the natural product sanguinarine (SAN) as a potent inhibitor of TrxR with a new chemical scaffold. Inhibition of TrxR leads to accumulation of the oxidized thioredoxin, elicits oxidative stress, and finally promotes apoptosis of cancer cells. Further synthesis of different model compounds of SAN demonstrated that the phenanthridinium unit is responsible for the TrxR inhibition. The core structure of SAN, e.g., the phenanthridinium moiety, is different from those of known TrxR inhibitors, and thus SAN is a new chemical entity of TrxR inhibitors and may serve a lead for further development. In addition, as the phenanthridinium scaffold is widely present in natural products, the disclosure of TrxR inhibition by such unit sheds light in understanding the pharmacological actions of these molecules.
Collapse
Affiliation(s)
- Juan Yao
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China; School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China
| | - Dongzhu Duan
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China; Shaanxi Key Laboratory of Phytochemistry, Baoji University of Arts and Sciences, Baoji, 721013, China
| | - Zi-Long Song
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
49
|
Waris S, Patel A, Ali A, Mahmood R. Acetaldehyde-induced oxidative modifications and morphological changes in isolated human erythrocytes: an in vitro study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:16268-16281. [PMID: 32124282 DOI: 10.1007/s11356-020-08044-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 02/10/2020] [Indexed: 06/10/2023]
Abstract
Acetaldehyde is a toxic, mutagenic and carcinogenic metabolite of alcohol which can bind to proteins, DNA and several other cellular macromolecules. Chronic alcohol consumption increases intracellular acetaldehyde levels which enhances the generation of reactive oxygen and nitrogen species (ROS and RNS). In this study, we have examined the effect of acetaldehyde on human erythrocytes under in vitro conditions. Treatment of human erythrocytes with different concentrations of acetaldehyde (0.05-2 mM) for 24 h at 37 °C increased intracellular generation of ROS and RNS. It also increased oxidation of proteins and lipids but decreased glutathione, total sulphhydryl and free amino group content. Methemoglobin level was increased accompanied by a decrease in methemoglobin reductase activity. Acetaldehyde impaired the antioxidant defence system and lowered the total antioxidant capacity of the cell. It decreased the activity of metabolic and membrane-bound enzymes and altered erythrocyte morphology. Our results show that acetaldehyde enhances the generation of ROS and RNS that results in oxidative modification of cellular components. This will lower the oxygen transporting ability of blood and shorten erythrocyte lifespan (red cell senescence).
Collapse
Affiliation(s)
- Sana Waris
- Department of Biochemistry, Faculty of Medicine, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, India
| | - Ayyub Patel
- Department of Clinical Biochemistry, King Khalid University, Abha, Saudi Arabia
| | - Asif Ali
- Department of Biochemistry, Faculty of Medicine, J. N. Medical College, Aligarh Muslim University, Aligarh, UP, India
| | - Riaz Mahmood
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, UP, India.
| |
Collapse
|
50
|
Dunigan-Russell K, Lin V, Silverberg M, Wall SB, Li R, Gotham J, Nicola T, Sridharan A, Snowball J, Delaney C, Li Q, Tipple TE. Aurothioglucose enhances proangiogenic pathway activation in lungs from room air and hyperoxia-exposed newborn mice. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1165-L1171. [PMID: 32292070 DOI: 10.1152/ajplung.00086.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD), a long-term respiratory morbidity of prematurity, is characterized by attenuated alveolar and vascular development. Supplemental oxygen and immature antioxidant defenses contribute to BPD development. Our group identified thioredoxin reductase-1 (TXNRD1) as a therapeutic target to prevent BPD. The present studies evaluated the impact of the TXNRD1 inhibitor aurothioglucose (ATG) on pulmonary responses and gene expression in newborn C57BL/6 pups treated with saline or ATG (25 mg/kg ip) within 12 h of birth and exposed to room air (21% O2) or hyperoxia (>95% O2) for 72 h. Purified RNA from lung tissues was sequenced, and differential expression was evaluated. Hyperoxic exposure altered ~2,000 genes, including pathways involved in glutathione metabolism, intrinsic apoptosis signaling, and cell cycle regulation. The isolated effect of ATG treatment was limited primarily to genes that regulate angiogenesis and vascularization. In separate studies, pups were treated as described above and returned to room air until 14 days. Vascular density analyses were performed, and ANOVA indicated an independent effect of hyperoxia on vascular density and alveolar architecture at 14 days. Consistent with RNA-seq analyses, ATG significantly increased vascular density in room air, but not in hyperoxia-exposed pups. These findings provide insights into the mechanisms by which TXNRD1 inhibitors may enhance lung development.
Collapse
Affiliation(s)
- Katelyn Dunigan-Russell
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Vivian Lin
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mary Silverberg
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephanie B Wall
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rui Li
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - John Gotham
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Teodora Nicola
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anusha Sridharan
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - John Snowball
- Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Cassidy Delaney
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Qian Li
- Division of Neonatology, Neonatal Redox Biology Laboratory, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama.,Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Trent E Tipple
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma, Oklahoma City, Oklahoma
| |
Collapse
|