1
|
Yu Y, Liu S, Yang L, Song P, Liu Z, Liu X, Yan X, Dong Q. Roles of reactive oxygen species in inflammation and cancer. MedComm (Beijing) 2024; 5:e519. [PMID: 38576456 PMCID: PMC10993368 DOI: 10.1002/mco2.519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 01/21/2024] [Accepted: 02/23/2024] [Indexed: 04/06/2024] Open
Abstract
Reactive oxygen species (ROS) constitute a spectrum of oxygenic metabolites crucial in modulating pathological organism functions. Disruptions in ROS equilibrium span various diseases, and current insights suggest a dual role for ROS in tumorigenesis and the immune response within cancer. This review rigorously examines ROS production and its role in normal cells, elucidating the subsequent regulatory network in inflammation and cancer. Comprehensive synthesis details the documented impacts of ROS on diverse immune cells. Exploring the intricate relationship between ROS and cancer immunity, we highlight its influence on existing immunotherapies, including immune checkpoint blockade, chimeric antigen receptors, and cancer vaccines. Additionally, we underscore the promising prospects of utilizing ROS and targeting ROS modulators as novel immunotherapeutic interventions for cancer. This review discusses the complex interplay between ROS, inflammation, and tumorigenesis, emphasizing the multifaceted functions of ROS in both physiological and pathological conditions. It also underscores the potential implications of ROS in cancer immunotherapy and suggests future research directions, including the development of targeted therapies and precision oncology approaches. In summary, this review emphasizes the significance of understanding ROS-mediated mechanisms for advancing cancer therapy and developing personalized treatments.
Collapse
Affiliation(s)
- Yunfei Yu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Shengzhuo Liu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Luchen Yang
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Pan Song
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Zhenghuan Liu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Xiaoyang Liu
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Xin Yan
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| | - Qiang Dong
- Department of UrologyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
2
|
Chen L, Yu W, Tang H, Zhang S, Wang J, Ouyang Q, Guo M, Zhu X, Huang Z, Chen J. Cyclometalated ruthenium complexes overcome cisplatin resistance through PI3K/mTOR/Nrf2 signaling pathway. Metallomics 2024; 16:mfae002. [PMID: 38183290 DOI: 10.1093/mtomcs/mfae002] [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: 10/01/2023] [Accepted: 01/04/2024] [Indexed: 01/08/2024]
Abstract
Currently, cisplatin resistance remains a primary clinical obstacle in the successful treatment of non-small cell lung cancer. Here, we designed, synthesized, and characterized two novel cyclometalated Ru(II) complexes, [Ru(bpy)2(1-Ph-7-OCH3-IQ)] (PF6) (bpy = 2,2'-bipyridine, IQ = isoquinoline, RuIQ7)and [Ru(bpy)2(1-Ph-6,7-(OCH3)2-IQ)] (PF6) (RuIQ8). As experimental controls, we prepared complex [Ru(bpy)2(1-Ph-IQ)](PF6) (RuIQ6) lacking a methoxy group in the main ligand. Significantly, complexes RuIQ6-8 displayed higher in vitro cytotoxicity when compared to ligands, precursor cis-[Ru(bpy)2Cl2], and clinical cisplatin. Mechanistic investigations revealed that RuIQ6-8 could inhibit cell proliferation by downregulating the phosphorylation levels of Akt and mTOR proteins, consequently affecting the rapid growth of human lung adenocarcinoma cisplatin-resistant cells A549/DDP. Moreover, the results from qRT-PCR demonstrated that these complexes could directly suppress the transcription of the NF-E2-related factor 2 gene, leading to the inhibition of downstream multidrug resistance-associated protein 1 expression and effectively overcoming cisplatin resistance. Furthermore, the relationship between the chemical structures of these three complexes and their anticancer activity, ability to induce cell apoptosis, and their efficacy in overcoming cisplatin resistance has been thoroughly examined and discussed. Notably, the toxicity test conducted on zebrafish embryos indicated that the three Ru-IQ complexes displayed favorable safety profiles. Consequently, the potential of these developed compounds as innovative therapeutic agents for the efficient and low-toxic treatment of NSCLC appears highly promising.
Collapse
Affiliation(s)
- Lanmei Chen
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| | - Wenzhu Yu
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| | - Hong Tang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| | - Shenting Zhang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| | - Jie Wang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
| | - Qianqian Ouyang
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| | - Miao Guo
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
| | - Xufeng Zhu
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| | - Jincan Chen
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, Guangdong 523808, P. R. China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, P. R. China
| |
Collapse
|
3
|
Schoeller M, Piroš M, Litecká M, Koňariková K, Jozefíková F, Šagátová A, Zahradníková E, Valentová J, Moncol J. Bipyridine Ruthenium(II) Complexes with Halogen-Substituted Salicylates: Synthesis, Crystal Structure, and Biological Activity. Molecules 2023; 28:4609. [PMID: 37375164 DOI: 10.3390/molecules28124609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 05/28/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Ruthenium complexes currently represent a perspective subject of investigation in terms of potential anticancer therapeutics. Eight novel octahedral ruthenium(II) complexes are the subject of this article. Complexes contain 2,2'-bipyridine molecules and salicylates as ligands, differing in position and type of halogen substituent. The structure of the complexes was determined via X-ray structural analysis and NMR spectroscopy. All complexes were characterized by spectral methods-FTIR, UV-Vis, ESI-MS. Complexes show sufficient stability in solutions. Therefore, their biological properties were studied. Binding ability to BSA, interaction with DNA, as well as in vitro antiproliferative effects against MCF-7 and U-118MG cell lines were investigated. Several complexes showed anticancer effects against these cell lines.
Collapse
Affiliation(s)
- Martin Schoeller
- Department of Inorganic Chemistry, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Milan Piroš
- Department of Inorganic Chemistry, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Miroslava Litecká
- Department of Materials Chemistry, Institute of Inorganic Chemistry of the CAS, Husinec-Řež č.p. 1001, 250 68 Řež, Czech Republic
| | - Katarína Koňariková
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Sasinkova 2, 811 08 Bratislava, Slovakia
| | - Flóra Jozefíková
- Department of Inorganic Chemistry, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia
- Department of Chemical Theory of Drugs, Faculty of Pharmacy, Comenius University, Kalinčiakova 8, 832 32 Bratislava, Slovakia
| | - Alexandra Šagátová
- Department of Inorganic Chemistry, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Eva Zahradníková
- Department of Inorganic Chemistry, Faculty of Science, Palacký University, 17. Listopadu 12, 771 46 Olomouc, Czech Republic
| | - Jindra Valentová
- Department of Chemical Theory of Drugs, Faculty of Pharmacy, Comenius University, Kalinčiakova 8, 832 32 Bratislava, Slovakia
| | - Ján Moncol
- Department of Inorganic Chemistry, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia
| |
Collapse
|
4
|
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
|
5
|
Shahzad K, Asad M, Asiri AM, Irfan M, Iqbal MA. In-vitro anticancer profile of recent ruthenium complexes against liver cancer. REV INORG CHEM 2022. [DOI: 10.1515/revic-2021-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Ruthenium complexes are considered as the most favorable alternatives to traditional platinum-based cancer drugs owing to their acceptable toxicity level, selectivity, variant oxidation states and ability to treat platinum-resistant cancer cells. They have similar ligand exchange kinetics as platinum drugs but can be tailored according to our desire by ligands influence. In the current study, we illustrate the in-vitro anticancer profile of some ruthenium complexes (2016–2021) against human hepatocellular carcinoma (HepG2). The anticancer activity of ruthenium complexes is determined by comparing their IC50 values with one another and positive controls. Fortunately, some ruthenium complexes including 3, 4, 6, 14, 15, 20, 42, and 48 exhibit surpassed in-vitro anticancer profile than that of positive controls promising as potential candidates against liver cancer. We also explored the structure-activity relationship (SAR) which is a key factor in the rational designing and synthesis of new ruthenium drugs. It covers the factors affecting anticancer activity including lipophilicity, planarity, area and bulkiness, the steric influence of different ligands, and electronic effects induced by ligands, stability, aqueous solubility and bioavailability to the target sites. The data reported here will provide strong support in the plausible design and synthesis of ruthenium anticancer drugs in the upcoming days.
Collapse
Affiliation(s)
- Khurram Shahzad
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| | - Mohammad Asad
- Center of Excellence for Advanced Materials Research (CEAMR) , King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
- Chemistry Department , Faculty of Science, King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
| | - Abdullah M. Asiri
- Center of Excellence for Advanced Materials Research (CEAMR) , King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
- Chemistry Department , Faculty of Science, King Abdulaziz University , P.O. Box 80203 , Jeddah 21589 , Saudi Arabia
| | - Muhammad Irfan
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
| | - Muhammad Adnan Iqbal
- Department of Chemistry , University of Agriculture , Faisalabad , 38000 , Pakistan
- Organometallic and Coordination Chemistry Laboratory , University of Agriculture , Faisalabad , 38000 , Pakistan
| |
Collapse
|
6
|
Hildebrandt J, Häfner N, Kritsch D, Görls H, Dürst M, Runnebaum IB, Weigand W. Highly Cytotoxic Osmium(II) Compounds and Their Ruthenium(II) Analogues Targeting Ovarian Carcinoma Cell Lines and Evading Cisplatin Resistance Mechanisms. Int J Mol Sci 2022; 23:ijms23094976. [PMID: 35563367 PMCID: PMC9102668 DOI: 10.3390/ijms23094976] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 12/14/2022] Open
Abstract
(1) Background: Ruthenium and osmium complexes attract increasing interest as next generation anticancer drugs. Focusing on structure-activity-relationships of this class of compounds, we report on 17 different ruthenium(II) complexes and four promising osmium(II) analogues with cinnamic acid derivatives as O,S bidentate ligands. The aim of this study was to determine the anticancer activity and the ability to evade platin resistance mechanisms for these compounds. (2) Methods: Structural characterizations and stability determinations have been carried out with standard techniques, including NMR spectroscopy and X-ray crystallography. All complexes and single ligands have been tested for cytotoxic activity on two ovarian cancer cell lines (A2780, SKOV3) and their cisplatin-resistant isogenic cell cultures, a lung carcinoma cell line (A549) as well as selected compounds on three non-cancerous cell cultures in vitro. FACS analyses and histone γH2AX staining were carried out for cell cycle distribution and cell death or DNA damage analyses, respectively. (3) Results: IC50 values show promising results, specifically a high cancer selective cytotoxicity and evasion of resistance mechanisms for Ru(II) and Os(II) compounds. Histone γH2AX foci and FACS experiments validated the high cytotoxicity but revealed diminished DNA damage-inducing activity and an absence of cell cycle disturbance thus pointing to another mode of action. (4) Conclusion: Ru(II) and Os(II) compounds with O,S-bidentate ligands show high cytotoxicity without strong effects on DNA damage and cell cycle, and this seems to be the basis to circumvent resistance mechanisms and for the high cancer cell specificity.
Collapse
Affiliation(s)
- Jana Hildebrandt
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Norman Häfner
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Daniel Kritsch
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Helmar Görls
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
| | - Matthias Dürst
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
| | - Ingo B. Runnebaum
- Department of Gynecology, Jena University Hospital—Friedrich-Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (N.H.); (D.K.); (M.D.)
- Correspondence: (I.B.R.); (W.W.); Tel.: +49-3641-9329101 (I.B.R.); +49-3641-948160 (W.W.)
| | - Wolfgang Weigand
- Institut für Anorganische und Analytische Chemie Friedrich-Schiller Universität Jena, Humboldtstraße 8, 07743 Jena, Germany; (J.H.); (H.G.)
- Correspondence: (I.B.R.); (W.W.); Tel.: +49-3641-9329101 (I.B.R.); +49-3641-948160 (W.W.)
| |
Collapse
|
7
|
Nunes DCDOS, Costa MS, Bispo-da-Silva LB, Ferro EAV, Zóia MAP, Goulart LR, Rodrigues RS, Rodrigues VDM, Yoneyama KAG. Mitochondrial dysfunction on Leishmania (Leishmania) amazonensis induced by ketoconazole: insights into drug mode of action. Mem Inst Oswaldo Cruz 2022; 117:e210157. [PMID: 35508030 PMCID: PMC9060495 DOI: 10.1590/0074-02760210157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 02/10/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Leishmania parasites cause leishmaniasis that range from self-limiting cutaneous lesions to more serious forms of the disease. The search for potential drug targets focusing on biochemical and metabolic pathways revealed the sterol biosynthesis inhibitors (SBIs) as a promising approach. In this class of inhibitors is found ketoconazole, a classical inhibitor of 14α-methysterol 14-demethylase. OBJECTIVE The present study aimed to better understand the biological response of Leishmania (Leishmania) amazonensis promastigotes at the cellular level after ketoconazole treatment. METHODS Herein, techniques, such as fluorimetry, flow cytometry, fluorescence microscopy, electron and scanning microscopy were used to investigate the cellular structures and to identify organelles affected by ketoconazole treatment. FINDINGS The study demonstrated, for the first time, the effect of ketoconazole on mitochondrion functioning and its probable relationship to cell cycle and death on L. (L.) amazonensis promastigotes (IFLA/BR/67/PH8 strain). MAIN CONCLUSIONS Ketoconazole-induced mitochondrial damages led to hyperpolarisation of this single organelle and autophagic vacuoles formation, as a parasite survival strategy. These damages did not reflect directly on the parasite cell cycle, but drove the parasites to death, making them susceptible to ketoconazole treatment in in vitro models.
Collapse
|
8
|
Metal Complexes or Chelators with ROS Regulation Capacity: Promising Candidates for Cancer Treatment. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010148. [PMID: 35011380 PMCID: PMC8746559 DOI: 10.3390/molecules27010148] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 12/25/2021] [Accepted: 12/26/2021] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) are rapidly eliminated and reproduced in organisms, and they always play important roles in various biological functions and abnormal pathological processes. Evaluated ROS have frequently been observed in various cancers to activate multiple pro-tumorigenic signaling pathways and induce the survival and proliferation of cancer cells. Hydrogen peroxide (H2O2) and superoxide anion (O2•-) are the most important redox signaling agents in cancer cells, the homeostasis of which is maintained by dozens of growth factors, cytokines, and antioxidant enzymes. Therefore, antioxidant enzymes tend to have higher activity levels to maintain the homeostasis of ROS in cancer cells. Effective intervention in the ROS homeostasis of cancer cells by chelating agents or metal complexes has already developed into an important anti-cancer strategy. We can inhibit the activity of antioxidant enzymes using chelators or metal complexes; on the other hand, we can also use metal complexes to directly regulate the level of ROS in cancer cells via mitochondria. In this review, metal complexes or chelators with ROS regulation capacity and with anti-cancer applications are collectively and comprehensively analyzed, which is beneficial for the development of the next generation of inorganic anti-cancer drugs based on ROS regulation. We expect that this review will provide a new perspective to develop novel inorganic reagents for killing cancer cells and, further, as candidates or clinical drugs.
Collapse
|
9
|
Song A, Cho GW, Vijayakumar KA, Moon C, Ang MJ, Kim J, Park I, Jang CH. Neuroprotective Effect of Valproic Acid on Salicylate-Induced Tinnitus. Int J Mol Sci 2021; 23:ijms23010023. [PMID: 35008469 PMCID: PMC8744959 DOI: 10.3390/ijms23010023] [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: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
High-dose salicylate induces temporary moderate hearing loss and the perception of a high-pitched tinnitus in humans and animals. Previous studies demonstrated that high doses of salicylate increase N-methyl-d-aspartate (NMDA) receptor levels, resulting in a rise in Ca2+ influx and induction of excitotoxicity. Glutamate excitotoxicity is associated with failure in the maintenance of calcium homeostasis, mitochondrial dysfunction, and production of reactive oxygen species (ROS). Valproic acid (VPA) is widely used for the management of bipolar disorder, epilepsy, and migraine headaches, and is known to regulate NMDA receptor activity. In this study, we examined the beneficial effects of VPA in a salicylate-induced tinnitus model in vitro and in vivo. Cells were pretreated with VPA followed by salicylate treatment. The expression levels of NMDA receptor subunit NR2B, phosphorylated cAMP response element-binding protein—an apoptosis marker, and intracellular levels of ROS were measured using several biochemical techniques. We observed increased expression of NR2B and its related genes TNFα and ARC, increased intracellular ROS levels, and induced expression of cleaved caspase-3. These salicylate-induced changes were attenuated in the neuronal cell line SH-SY5Y and rat cortical neurons after VPA pretreatment. Together, these results provide evidence of the beneficial effects of VPA in a salicylate-induced temporary hearing loss and tinnitus model.
Collapse
Affiliation(s)
- Anji Song
- Department of Biology, College of Natural Science, Chosun University, Gwangju 61452, Korea; (A.S.); (G.-W.C.); (K.A.V.)
- BK21 FOUR Education Research Group for Age-Associated Disorder Control Technology, Department of Integrative Biological Science, Chosun University, Gwangju 61452, Korea
| | - Gwang-Won Cho
- Department of Biology, College of Natural Science, Chosun University, Gwangju 61452, Korea; (A.S.); (G.-W.C.); (K.A.V.)
- BK21 FOUR Education Research Group for Age-Associated Disorder Control Technology, Department of Integrative Biological Science, Chosun University, Gwangju 61452, Korea
| | - Karthikeyan A. Vijayakumar
- Department of Biology, College of Natural Science, Chosun University, Gwangju 61452, Korea; (A.S.); (G.-W.C.); (K.A.V.)
- BK21 FOUR Education Research Group for Age-Associated Disorder Control Technology, Department of Integrative Biological Science, Chosun University, Gwangju 61452, Korea
| | - Changjong Moon
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea;
- Correspondence: (C.M.); (C.H.J.); Tel.: +82-62-220-6774 (C.H.J.)
| | - Mary Jasmin Ang
- Department of Veterinary Anatomy, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju 61186, Korea;
| | - Jahae Kim
- Department of Nuclear Medicine, Chonnam National University Hospital, Gwangju 61469, Korea;
| | - Ilyong Park
- Department of Biomedical Engineering, School of Medicine, Dankook University, Cheonan 31116, Korea;
| | - Chul Ho Jang
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju 61469, Korea
- Correspondence: (C.M.); (C.H.J.); Tel.: +82-62-220-6774 (C.H.J.)
| |
Collapse
|
10
|
|
11
|
Chen L, Wang J, Cai X, Chen S, Zhang J, Li B, Chen W, Guo X, Luo H, Chen J. Cyclometalated Ru(II)-isoquinoline complexes overcome cisplatin resistance of A549/DDP cells by downregulation of Nrf2 via Akt/GSK-3β/Fyn pathway. Bioorg Chem 2021; 119:105516. [PMID: 34856444 DOI: 10.1016/j.bioorg.2021.105516] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 01/03/2023]
Abstract
Both ruthenium (Ru) and isoquinoline (IQ) compounds are regarded as potential anticancer drug candidates. Here, we report the synthesis and characterization of three novel cyclometalated Ru(II)-isoquinoline complexes: RuIQ-3, RuIQ-4, and RuIQ-5, and evaluation of their in vitro cytotoxicities against a panel of cell lines including A549/DDP, a cisplatin-resistant human lung cancer cell line. A549/DDP 3D multicellular tumor spheroids (MCTSs) were also used to detect the drug resistance reversal effect of Ru(II)-IQ complexes. Our results indicated that the cytotoxic activities against cancer cells of Ru(II)-IQ complexes, especially RuIQ-5, were superior compared with cisplatin. In addition, RuIQ-5 exhibited low toxicity towards both normal HBE cells in vitro and zebrafish embryos in vivo. Further investigation on cellular mechanism of action indicated that after absorption by A549/DDP cells, RuIQ-5 was mainly distributed in the nucleus, which is different from cisplatin. Besides, RuIQ-5 could induce apoptosis through mitochondrial dysfunction, reactive oxygen species (ROS) accumulation, ROS-mediated DNA damage, and cycle arrest at both S and G2/M phases. Moreover, RuIQ-5 could inhibit the overexpression of Nrf2 through regulation of Akt/GSK-3β/Fyn signaling pathway and hindering the nuclear translocation of Nrf2. Based on these findings, we firmly believe that the studied Ru(II)-IQ complexes hold great promise as anticancer therapeutics with high effectiveness and low toxicity.
Collapse
Affiliation(s)
- Lanmei Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong 524023, China
| | - Jie Wang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Xianhong Cai
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Suxiang Chen
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, Western Australia 6150, Australia
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Disease of Guangdong Medical University, Zhanjiang 524001, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, China
| | - Baojun Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Weigang Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Xinhua Guo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China
| | - Hui Luo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, China.
| | - Jincan Chen
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, School of Pharmacy, Guangdong Medical University, Zhanjiang 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
12
|
Swaminathan S, Haribabu J, Mohamed Subarkhan MK, Gayathri D, Balakrishnan N, Bhuvanesh N, Echeverria C, Karvembu R. Impact of aliphatic acyl and aromatic thioamide substituents on the anticancer activity of Ru(II)- p-cymene complexes with acylthiourea ligands- in vitro and in vivo studies. Dalton Trans 2021; 50:16311-16325. [PMID: 34730582 DOI: 10.1039/d1dt02611a] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Six different acylthiourea ligands (L1-L6) and their corresponding Ru(II)-p-cymene complexes (P1-P6) were designed to explore the structure-activity relationship of the complexes upon aliphatic chain and aromatic conjugation on the C- and N-terminals, respectively. The compounds were synthesized and adequately characterized using various analytical and spectroscopic techniques. The structures of P2-P6, solved using single crystal X-ray diffraction (XRD), confirmed the neutral monodentate coordination of the S atoms of the acylthiourea ligands to Ru(II) ions. In silico studies showed an increase of lipophilicity for the ligands with an increase in alkyl chain length or aromatic conjugation at the C- or N-terminal, respectively. Subsequently, mitogen-activated protein kinases (MAPK) were predicted as one of the primary targets for the complexes, which showed good binding affinity towards extracellular signal-regulated kinases (ERK1, ERK2 and ERK5), c-Jun N-terminal kinase (JNK) and p38 of the MAPK pathway. Henceforth, the complexes were tested for their anticancer activity in lung carcinoma (A549) and cisplatin-resistant lung carcinoma (cisA549R) cells and human umbilical vein epithelial normal cells (HUVEC). Interestingly, an increase in chain length or aromatic conjugation led to an increase in the activity of the complexes, with P5 (7.73 and 13.04 μM) and P6 (6.52 and 14.45 μM) showing the highest activity in A549 and cisA549R cells, which is better than the positive control, cisplatin (8.72 and 44.28 μM). Remarkably, we report the highest activity yet observed for complexes of the type [(η6-p-cymene)RuIICl2(S-acylthiourea)] in the tested cell lines. Aqueous solution studies showed that complexes P5 and P6 are rapidly hydrolyzed to produce solely aquated species that remained stable for 24 h. Staining assays and flow cytometric analyses of P5 and P6 in A549 cells revealed that the complexes induced apoptosis and arrested the cell cycle predominantly in the S phase. In vivo studies demonstrated the higher toxicity of cisplatin and a comparatively higher survival rate of mice injected with the most active complex P6. Histological analyses revealed that treatment with P6 at high doses of up to 8 mg kg-1 did not cause any palpable damage to the tested organs.
Collapse
Affiliation(s)
- Srividya Swaminathan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India.
| | - Jebiti Haribabu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India.
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502 Copiapo, Chile
| | - Mohamed Kasim Mohamed Subarkhan
- The First Affiliated Hospital, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, 310003, PR China
| | - Dasararaju Gayathri
- Centre of Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai 600025, India
| | - Nithya Balakrishnan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India.
| | - Nattamai Bhuvanesh
- Department of Chemistry, Texas A & M University, College Station, Texas 77842, USA
| | - Cesar Echeverria
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502 Copiapo, Chile
| | - Ramasamy Karvembu
- Department of Chemistry, National Institute of Technology, Tiruchirappalli 620015, Tamil Nadu, India.
| |
Collapse
|
13
|
Kladnik J, Coverdale JPC, Kljun J, Burmeister H, Lippman P, Ellis FG, Jones AM, Ott I, Romero-Canelón I, Turel I. Organoruthenium Complexes with Benzo-Fused Pyrithiones Overcome Platinum Resistance in Ovarian Cancer Cells. Cancers (Basel) 2021; 13:2493. [PMID: 34065335 PMCID: PMC8160969 DOI: 10.3390/cancers13102493] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/07/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023] Open
Abstract
Drug resistance to existing anticancer agents is a growing clinical concern, with many first line treatments showing poor efficacy in treatment plans of some cancers. Resistance to platinum agents, such as cisplatin, is particularly prevalent in the treatment of ovarian cancer, one of the most common cancers amongst women in the developing world. Therefore, there is an urgent need to develop next generation of anticancer agents which can overcome resistance to existing therapies. We report a new series of organoruthenium(II) complexes bearing structurally modified pyrithione ligands with extended aromatic scaffold, which overcome platinum and adriamycin resistance in human ovarian cancer cells. The mechanism of action of such complexes appears to be unique from that of cisplatin, involving G1 cell cycle arrest without generation of cellular ROS, as is typically associated with similar ruthenium complexes. The complexes inhibit the enzyme thioredoxin reductase (TrxR) in a model system and reduce cell motility towards wound healing. Importantly, this work highlights further development in our understanding of the multi-targeting mechanism of action exhibited by transition metal complexes.
Collapse
Affiliation(s)
- Jerneja Kladnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - James P. C. Coverdale
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK; (J.P.C.C.); (F.G.E.); (A.M.J.)
| | - Jakob Kljun
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| | - Hilke Burmeister
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, 38106 Braunschweig, Germany; (H.B.); (P.L.); (I.O.)
| | - Petra Lippman
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, 38106 Braunschweig, Germany; (H.B.); (P.L.); (I.O.)
| | - Francesca G. Ellis
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK; (J.P.C.C.); (F.G.E.); (A.M.J.)
| | - Alan M. Jones
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK; (J.P.C.C.); (F.G.E.); (A.M.J.)
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig, 38106 Braunschweig, Germany; (H.B.); (P.L.); (I.O.)
| | - Isolda Romero-Canelón
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK; (J.P.C.C.); (F.G.E.); (A.M.J.)
| | - Iztok Turel
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.K.); (J.K.)
| |
Collapse
|
14
|
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
|
15
|
Zhong Y, Liu J, Cheng X, Zhang H, Zhang C, Xia Z, Wu Z, Zhang L, Zheng Y, Gao Z, Jiang Z, Wang Z, Huang D, Lu Y, Jiang F. Design, synthesis and biological evaluations of diverse Michael acceptor-based phenazine hybrid molecules as TrxR1 inhibitors. Bioorg Chem 2021; 109:104736. [PMID: 33640630 DOI: 10.1016/j.bioorg.2021.104736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
A series of novel phenazine derivatives (1~27) containing the Michael acceptor scaffolds were designed and synthesized in this study. Some compounds exhibited selective cytotoxicity against Bel-7402 cancer cell line in vitro, in which compound 26 were found to have the best antiproliferative activity. Meanwhile, compound 26 showed no obvious cell toxicity against human normal liver epithelial L02 cells, which means this compound possessed a better safety potential. In the following research, compound 26 was verified to inhibit TrxR1 enzyme activity, ultimately resulting in cellular molecular mechanism events of apoptosis including growth of intracellular ROS level, depletion of reduced Trx1, liberation of ASK1 and up-regulation of p38, respectively. Together, all these evidences implicated that compound 26 acted as the TrxR1 inhibitor against Bel-7402 cells, and could activate apoptosis through the ROS-Trx-ASK1-p38 pathway.
Collapse
Affiliation(s)
- Yucheng Zhong
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Jing Liu
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Cheng
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Chunhua Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhuolu Xia
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongxi Wu
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Zhang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yuting Zheng
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhanyu Gao
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhidong Jiang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Zhixiang Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Dechun Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yuanyuan Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Feng Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
16
|
Lu J, Wei N, Cao J, Zhou Y, Gong H, Zhang H, Zhou J. Evaluation of enzymatic activity of Babesia microti thioredoxin reductase (Bmi TrxR)-mutants and screening of its potential inhibitors. Ticks Tick Borne Dis 2020; 12:101623. [PMID: 33418338 DOI: 10.1016/j.ttbdis.2020.101623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 01/25/2023]
Abstract
Babesia microti is a zoonotic pathogen that mainly parasitizes mammalian erythrocytes. Oxidative stress can induce gene mutation, protein denaturation and lipid peroxidation, such as reactive oxygen species (ROS) induced by hypoxic environment and the host immune system. An antioxidase, B. microti thioredoxin reductase (Bmi TrxR), has been identified in B. microti. We used a combination of homology modeling and domain prediction to explore the functional sites of Bmi TrxR and found that TrxR has three domains. Constructed a mutant pool which His-tag were at the N-terminus (TrxR-Nhis, C105-Nhis, C110-Nhis, C105110-Nhis, C547-Nhis, C552-Nhis, C547552-Nhis) and the His tag were at the N- and C-terminus (TrxR-NChis, C547-NChis, C552-NChis, C547552-NChis). The proteins were expressed as His-tagged fusion proteins in Escherichia coli. The His-tag of TrxR C-terminus were affected the reaction with Trx. The inhibitory efficiency of DNCB was decreased for mutant C547, compared with recombinant TrxR, indicating that the action site of DNCB might be cysteine at position 547. These results indicate that the N-terminal active site of Bmi TrxR plays an important role in accepting electrons and promotes electron transfer. The C-terminus His tag of Bmi TrxR affected the electron transfer and the reducing activity of Bmi TrxR. Reduce reactive oxygen produced in oxidative stress was reduced by Bmi TrxR, which is beneficial to Babesia survival. Therefore, reduction site of TrxR may become a potential target for Babesia microti treatment.
Collapse
Affiliation(s)
- Jinmiao Lu
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Nana Wei
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Jie Cao
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yongzhi Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Haiyan Gong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Houshuang Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Jinlin Zhou
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| |
Collapse
|
17
|
Habas K, Soldevila-Barreda JJ, Azmanova M, Rafols L, Pitto-Barry A, Anderson D, Barry NPE. Evaluation of the Toxicity of Two Electron-Deficient Half-Sandwich Complexes against Human Lymphocytes from Healthy Individuals. ChemMedChem 2020; 16:624-629. [PMID: 33119178 DOI: 10.1002/cmdc.202000672] [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] [Received: 08/28/2020] [Revised: 10/15/2020] [Indexed: 12/21/2022]
Abstract
Electron-deficient half-sandwich complexes are a class of under-studied organometallics with demonstrated potential as metallodrug candidates. This study investigates the effect of two 16-electron organoruthenium complexes ([(p-cym)Ru(benzene-1,2-dithiolato)] (1) and [(p-cym)Ru(maleonitriledithiolate)] (2)) on the cell viability of non-immortalised human lymphocytes from healthy individuals. The genotoxic effects of 1 and 2 in lymphocytes are also investigated by using the Comet and cytokinesis-block micronucleus assays. Gene expression studies were carried out on a panel of genes involved in apoptosis and the DNA damage-repair response. Results show that the two 16-electron complexes do not have significant effect on the cell viability of human lymphocytes from healthy individuals. However, an increase in DNA damage is induced by both compounds, presumably through oxidative stress production.
Collapse
Affiliation(s)
- Khaled Habas
- School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | | | - Maria Azmanova
- School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Laia Rafols
- School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Anaïs Pitto-Barry
- School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Diana Anderson
- School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| | - Nicolas P E Barry
- School of Chemistry and Biosciences, University of Bradford, Bradford, BD7 1DP, UK
| |
Collapse
|
18
|
Costa MS, Gonçalves YG, Borges BC, Silva MJB, Amstalden MK, Costa TR, Antunes LMG, Rodrigues RS, Rodrigues VDM, de Faria Franca E, Zoia MAP, de Araújo TG, Goulart LR, Von Poelhsitz G, Yoneyama KAG. Ruthenium (II) complex cis-[Ru II(ŋ 2-O 2CC 7H 7O 2)(dppm) 2]PF 6-hmxbato induces ROS-mediated apoptosis in lung tumor cells producing selective cytotoxicity. Sci Rep 2020; 10:15410. [PMID: 32958783 PMCID: PMC7506019 DOI: 10.1038/s41598-020-72420-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/31/2020] [Indexed: 12/24/2022] Open
Abstract
Ruthenium complexes have been extensively explored as potential molecules for cancer treatment. Considering our previous findings on the remarkable cytotoxic activity exhibited by the ruthenium (II) complex 3-hydroxy-4-methoxybenzoate (hmxbato)-cis-[RuII(ŋ2-O2CC7H7O2)(dppm)2]PF6 against Leishmania promastigotes and also the similar metabolic characteristics between trypanosomatids and tumor cells, the present study aimed to analyze the anticancer potential of hmxbato against lung tumor cells, as well as the partial death mechanisms involved. Hmxbato demonstrated selective cytotoxicity against A549 lung tumor cells. In addition, this complex at a concentration of 3.8 µM was able to expressively increase the generation of reactive oxygen species (ROS) in tumor cells, causing an oxidative stress that may culminate in: (1) reduction in cellular proliferation; (2) changes in cell morphology and organization patterns of the actin cytoskeleton; (3) cell arrest in the G2/M phase of the cell cycle; (4) apoptosis; (5) changes in the mitochondrial membrane potential and (6) initial DNA damage. Furthermore, we demonstrated that the induction of programmed cell death can occur by the intrinsic apoptotic pathway through the activation of caspases. It is also worth highlighting that hmxbato exhibited predominant actions on A549 tumor cells in comparison to BEAS-2B normal bronchial epithelium cells, which makes this complex an interesting candidate for the design of new drugs against lung cancer.
Collapse
Affiliation(s)
- Mônica Soares Costa
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil.
| | | | - Bruna Cristina Borges
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Marcelo José Barbosa Silva
- Laboratório de Osteoimunologia e Imunologia dos Tumores, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Martin Krähenbühl Amstalden
- Departamento de Análises Clínicas, Toxicologia e Ciências Alimentares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, CEP 14040-903, Brazil
| | - Tássia Rafaella Costa
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil
| | - Lusânia Maria Greggi Antunes
- Departamento de Análises Clínicas, Toxicologia e Ciências Alimentares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, CEP 14040-903, Brazil
| | - Renata Santos Rodrigues
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil
| | - Veridiana de Melo Rodrigues
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil
| | - Eduardo de Faria Franca
- Laboratório de Cristalografia e Química Computacional, Instituto de Química, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Mariana Alves Pereira Zoia
- Laboratório de Nanobiotecnologia, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Thaise Gonçalves de Araújo
- Laboratório de Nanobiotecnologia, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Luiz Ricardo Goulart
- Laboratório de Nanobiotecnologia, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Gustavo Von Poelhsitz
- Instituto de Química, Universidade Federal de Uberlândia, UFU, Uberlândia, MG, Brazil
| | - Kelly Aparecida Geraldo Yoneyama
- Laboratório de Bioquímica e Toxinas Animais, Instituto de Biotecnologia, Universidade Federal de Uberlândia, UFU, Pará avenue, 1720, Uberlândia, MG, CEP 38400-902, Brazil.
| |
Collapse
|
19
|
Novel ruthenium and palladium complexes as potential anticancer molecules on SCLC and NSCLC cell lines. CHEMICAL PAPERS 2020. [DOI: 10.1007/s11696-020-01129-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Ru(II) Complexes Bearing O, O-Chelated Ligands Induced Apoptosis in A549 Cells through the Mitochondrial Apoptotic Pathway. Bioinorg Chem Appl 2020; 2020:8890950. [PMID: 32879623 PMCID: PMC7448123 DOI: 10.1155/2020/8890950] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Two new Ru(II) complexes containing O, O-chelated ligands, Ru(dip)2(SA) (Ru-1) and Ru(dmp)2(SA) (Ru-2) (dip = 4,7-diphenyl-1,10-phenanthroline; dmp = 2,9-dimethyl-1,10-phenanthroline; SA = salicylate) were synthesized to evaluate their cytotoxicity in vitro. These complexes were found to exhibit moderate antitumor activity to different types of human cancers, including A549 (human lung carcinoma), MCF-7 (breast cancer), HeLa (human cervical cancer), and HepG2 (human hepatocellular carcinoma) cell lines, but displayed low toxicity to human normal cell lines BEAS-2B (immortalized human bronchial epithelial cells) when compared with that of cisplatin. Further studies revealed that these complexes could induce apoptosis in A549 cells, including activating caspase family proteins and poly (ADP-ribose) polymerase (PARP), reducing Bcl-2/Bax and Bcl-xl/Bad ratio, enhancing cellular reactive oxygen species (ROS) accumulation, triggering DNA damage, decreasing mitochondrial membrane potential (MMP), and leading cytochrome c release from mitochondria. Notably, complex Ru-1 showed low toxicity to developing zebrafish embryos. The obtained results suggest that these new synthetic complexes have the potential to be developed as low-toxicity agents for lung cancer treatment.
Collapse
|
21
|
Jiang GB, Zhang WY, He M, Gu YY, Bai L, Wang YJ, Yi QY, Du F. Development of four ruthenium polypyridyl complexes as antitumor agents: Design, biological evaluation and mechanism investigation. J Inorg Biochem 2020; 208:111104. [PMID: 32485635 DOI: 10.1016/j.jinorgbio.2020.111104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/21/2022]
Abstract
Ruthenium complexes are expected to be new opportunities for the development of antitumor agents. Herein, four ruthenium polypyridyl complexes ([Ru(bpy)2(CAPIP)](ClO4)2 (Ru(II)-1, bpy = 2,2'-bipyridine; CAPIP = (E)-2-(2-(furan-2-yl)vinyl)-1H-imidazo[4,5-f][1,10]phenanthroline), [Ru(phen)2(CA-PIP)](ClO4)2 (Ru(II)-2, phen = 1,10-phenanthroline), [Ru(dmb)2(CAPIP)](ClO4)2 (Ru(II)-3, dmb = 4,4'-dimethyl-2,2'-bipyridine), [Ru(dmb)2(ETPIP)](ClO4)2 (Ru(II)-4, ETPIP = 2-(4-(thiophen-2-ylethynyl)phenyl)-1H-imidazo[4,5-f][1,10]phen-anthroline)) have been investigated as mitochondria-targeted antitumor metallodrugs. DNA binding studies indicated that target Ru(II) complexes interacts with CT DNA (calf thymus DNA) by an intercalative mode. Cytotoxicity assay results demonstrate that Ru(II) complexes show high cytotoxicity against A549 cells with low IC50 value of 23.6 ± 2.3, 20.1 ± 1.9, 22.7 ± 1.8 and 18.4 ± 2.3 μM, respectively. Flow cytometry and morphological analysis revealed that these Ru(II) complexes can induce apoptosis in A549 cells. Intracellular reactive oxygen species (ROS) and mitochondrial membrane potential were also investigated by ImageXpress Micro XLS system. The experimental results indicate that the reactive oxygen species in A549 cells increased significantly and mitochondrial membrane potential decreased obviously. In addition, colocalization studies shown these complexes could get to the cytoplasm through the cell membrane and accumulate in the mitochondria. Furthermore, Ru(II) complexes can effectively induces cell cycle arrest at the S phase in A549 cells. Finally, cell invasion assay and quantitative studies were also performed to investigate the mechanism of this process. All in together, this study suggested that these Ru(II) complexes could induce apoptosis in A549 cells through cell cycle arrest and ROS-mediated mitochondrial dysfunction pathway.
Collapse
Affiliation(s)
- Guang-Bin Jiang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China.
| | - Wen-Yao Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Miao He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi-Ying Gu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lan Bai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yang-Jie Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qiao-Yan Yi
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Fan Du
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
22
|
Wang Y, Wang C, Bao S, Nie X. Responses of the Nrf2/Keap1 signaling pathway in Mugilogobius abei (M. abei) exposed to environmentally relevant concentration aspirin. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:15663-15673. [PMID: 32080815 DOI: 10.1007/s11356-020-07912-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/28/2020] [Indexed: 06/10/2023]
Abstract
Aspirin (ASA) is a widely used non-steroidal anti-inflammatory drug. Its high detection frequency in various waterborne and environmental residues has drawn wide attention. Limited information were provided for the effects of aspirin exposure on oxidative stress signaling pathway in fish, which is closely related to pathological and immunological process of fish. In this study, a small fish - Mugilogobius abei (M. abei) distributing widely in aquatic ecosystems in southern China, was employed as testing organism and the key genes of the detoxification metabolism were cloned for the first time. The responses of Nrf2/Keap1 signaling pathway were investigated under the environmentally relevant concentration aspirin exposure (0.5 μg L-1, 5 μg L-1, and 50 μg L-1) for 24 h, 72 h, and 168 h then. The transcriptional expression of the key genes (Nrf2, Keap1, GCLC, GPx, GST, SOD, CAT, Trx2, and TrxR) as well as the changes of the related enzymatic activities (GPx, GST, SOD, and CAT) and GSH and MDA content were also determined. Results showed that Nrf2 and Keap1 gene expression displayed a negative correlation to some extent under ASA exposure, the transcriptional expressions of the downstream related genes (GCLC, GST, SOD, CAT, Trx2, and TrxR) in Nrf2/Keap1 signaling pathway showed inhibition at 24 h but induction at 72 h and 168 h. At the protein level, ASA exposure can improve the antioxidant capacity by increasing GSH synthesis and enzymatic activity of GPx, GST, SOD, and CAT to reduce the degree of lipid peroxidation. We proposed that ASA exposure may interfere with the redox balance in M. abei at an early stage but sub-chronic ASA exposure can activate the Nrf2 signaling pathway to improve the antioxidant capacity of M. abei.
Collapse
Affiliation(s)
- Yimeng Wang
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Chao Wang
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Shuang Bao
- Department of Ecology, Jinan University, Guangzhou, 510632, China
| | - Xiangping Nie
- Department of Ecology, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Eutrophication and Red Tide Prevention of Guangdong Higher Education Institutes, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
23
|
Jiang GB, Zhang WY, He M, Gu YY, Bai L, Wang YJ, Yi QY, Du F. Design and synthesis of new ruthenium polypyridyl complexes with potent antitumor activity in vitro. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 220:117132. [PMID: 31146211 DOI: 10.1016/j.saa.2019.05.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/06/2019] [Accepted: 05/12/2019] [Indexed: 06/09/2023]
Abstract
We herein report the synthesis, characterization and anticancer activity of BTPIP (2-(4-(benzo[b]thiophen-2-yl)phenyl)-1H-imidazo[4,5-f][1,10]phenanthroline) and its four ruthenium(II) polypyridyl complexes [Ru(NN)2(BTPIP)](ClO4)2 (N-N = bpy = 2,2'-bipyridine, Ru(II)-1; phen = 1,10-phenanthroline, Ru(II)-2; dmb = 4,4'-dimethyl-2,2'-bipyridine, Ru(II)-3; dmp = 2,9-dimethyl-1,10-phenanthroline, Ru(II)-4). The DNA binding behaviors reveal that the complexes bind to calf thymus DNA by intercalation. Cytotoxicity of the complexes against A549, HepG-2, SGC-7901 and Hela cells were evaluated in vitro. Complexes Ru(II)-1, Ru(II)-2, Ru(II)-3, Ru(II)-4 show moderate activity on the cell proliferation in A549 cells with IC50 values of 9.3 ± 1.2, 12.1 ± 1.6, 10.3 ± 1.6, 8.9 ± 1.2 μM, respectively. Apoptosis assessment, intracellular mitochondrial membrane potential (MMP), location in mitochondria, reactive oxygen species (ROS), cell invasion assay and cell cycle arrest were also performed to explore the mechanism of this action. When the concentration of the ruthenium(II) complexes is increased, the amount of reactive oxygen species increases obviously and the mitochondrial membrane potential decreases dramatically in A549 cells. Most importantly, the ruthenium(II) polypyridyl complexes could arrive the cytoplasm through the cell membrane and accumulate in the mitochondria. These results showed that the ruthenium(II) complexes could induce apoptosis in A549 cells through an ROS-mediated mitochondrial dysfunction pathway.
Collapse
Affiliation(s)
- Guang-Bin Jiang
- Guangxi Key Laboratory of Electrochemical and Magneto-chemical Function Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin 541004, China.
| | - Wen-Yao Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Miao He
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yi-Ying Gu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lan Bai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yang-Jie Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Qiao-Yan Yi
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Fan Du
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| |
Collapse
|
24
|
Bian M, Fan R, Zhao S, Liu W. Targeting the Thioredoxin System as a Strategy for Cancer Therapy. J Med Chem 2019; 62:7309-7321. [PMID: 30963763 DOI: 10.1021/acs.jmedchem.8b01595] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Thioredoxin reductase (TrxR) participates in the regulation of redox reactions in organisms. It works mainly via its substrate molecule, thioredoxin, to maintain the redox balance and regulate signal transduction, which controls cell proliferation, differentiation, death, and other important physiological processes. In recent years, increasing evidence has shown that the overactivation of TrxR is related to the development of tumors. The exploration of TrxR-targeted antitumor drugs has attracted wide attention and is expected to provide new therapies for cancer treatment. In this perspective, we highlight the specific relationship between TrxR and apoptotic signaling pathways. The cytoplasm and mitochondria both contain TrxR, resulting in the activation of apoptosis. TrxR activity influences reactive oxygen species (ROS) and further regulates the inflammatory signaling pathway. In addition, we discuss representative TrxR inhibitors with anticancer activity and analyze the challenges in developing TrxR inhibitors as anticancer drugs.
Collapse
Affiliation(s)
- Mianli Bian
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China
| | - Rong Fan
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China
| | - Sai Zhao
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China.,Institute of New Medicine Research , Nanjing Hicin Pharmaceutical Co. Ltd. , Nanjing 210046 , P. R. China
| | - Wukun Liu
- Institute of Chinese Medicine, School of Pharmacy , Nanjing University of Chinese Medicine , Nanjing 210023 , P. R. China.,State Key Laboratory of Natural Medicines , China Pharmaceutical University , Nanjing 210009 , P. R. China
| |
Collapse
|
25
|
Zhang SL, Zhu HY, Zhou BY, Chu Y, Huo JR, Tan YY, Liu DL. Histone deacetylase 6 is overexpressed and promotes tumor growth of colon cancer through regulation of the MAPK/ERK signal pathway. Onco Targets Ther 2019; 12:2409-2419. [PMID: 31118659 PMCID: PMC6498393 DOI: 10.2147/ott.s194986] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/01/2019] [Indexed: 12/14/2022] Open
Abstract
Purpose: To investigate the expression of histone deacetylase 6 (HDAC6) in colon cancer and its role in colon cancer cell growth and migration. Materials and methods: We detected the expression of HDAC6 in a colon cancer tissue chip using immunochemical staining, and analyzed the difference in HDAC6 expression between cancer and adjacent noncancerous tissues. Then, we explored the relationship between HDAC6 expression and patients’ clinicopathological characteristics and prognoses. In adidition, the role of HDAC6 in colon cancer cell growth and migration, as well as its potential related signal pathway, through HDAC6 knockdown was explored. Results: The immunochemical score of HDAC6 expression was higher in cancer tissue than in the adjacent noncancerous tissue (4.54 vs 3.08, P<0.005); similarly, as well as the rate of high HDAC6 expression was higher in cancer tissue than in the adjacent noncancerous tissue (71.1% vs 40.9%, P<0.001). Patients showing high HDAC6 expression had a shorter overall survival time. Additionally, Cox regression analysis showed that high HDAC6 expression was an independent risk factor for poor prognosis. HDAC6 knockdown decreased cell viability, colony formation, and number of migrated colon cancer cells (HCT116 and HT29); the expression of p-MEK, p-ERK, and p-AKT was also decreased, but had no influence on MEK, ERK, and AKT expression. Conclusion: HDAC6 is highly expressed in colon cancer and associated with a poor prognosis. HDAC6 knockdown inhibits colon cancer cell growth and migration, partly through the MAPK/ERK pathway.
Collapse
Affiliation(s)
- Shi-Lan Zhang
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Hong-Yi Zhu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Bing-Yi Zhou
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Yi Chu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Ji-Rong Huo
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Yu-Yong Tan
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - De-Liang Liu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| |
Collapse
|
26
|
Soltan MY, Sumarni U, Assaf C, Langer P, Reidel U, Eberle J. Key Role of Reactive Oxygen Species (ROS) in Indirubin Derivative-Induced Cell Death in Cutaneous T-Cell Lymphoma Cells. Int J Mol Sci 2019; 20:ijms20051158. [PMID: 30866411 PMCID: PMC6429192 DOI: 10.3390/ijms20051158] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 01/05/2023] Open
Abstract
Cutaneous T-cell lymphoma (CTCL) may develop a highly malignant phenotype in its late phase, and patients may profit from innovative therapies. The plant extract indirubin and its chemical derivatives represent new and promising antitumor strategies. This first report on the effects of an indirubin derivative in CTCL cells shows a strong decrease of cell proliferation and cell viability as well as an induction of apoptosis, suggesting indirubin derivatives for therapy of CTCL. As concerning the mode of activity, the indirubin derivative DKP-071 activated the extrinsic apoptosis cascade via caspase-8 and caspase-3 through downregulation of the caspase antagonistic proteins c-FLIP and XIAP. Importantly, a strong increase of reactive oxygen species (ROS) was observed as an immediate early effect in response to DKP-071 treatment. The use of antioxidative pre-treatment proved the decisive role of ROS, which turned out upstream of all other proapoptotic effects monitored. Thus, reactive oxygen species appear as a highly active proapoptotic pathway in CTCL, which may be promising for therapeutic intervention. This pathway can be efficiently activated by an indirubin derivative.
Collapse
Affiliation(s)
- Marwa Y Soltan
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
- Department of Dermatology and Venereology, Faculty of Medicine, Ain Shams University, Cairo 11591, Egypt.
| | - Uly Sumarni
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Chalid Assaf
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
- Clinic for Dermatology and Venereology, Helios Klinikum Krefeld, Lutherplatz 40, 47805 Krefeld, Germany.
| | - Peter Langer
- Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 3a, 18059 Rostock, Germany.
- Leibniz Institute of Catalysis at the University of Rostock e.V., Albert-Einstein-Str. 29a, 18059 Rostock, Germany.
| | - Ulrich Reidel
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| | - Jürgen Eberle
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|