1
|
Martins C, Carvalho LM, Cabral IM, Saúde L, Dreij K, Costa PM. A mechanistic study on the interaction effects between legacy and pollutants of emerging concern: A case study with B[a]P and diclofenac. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 363:125189. [PMID: 39454814 DOI: 10.1016/j.envpol.2024.125189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/09/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
To study the intricate toxicological mechanisms triggered by exposure to mixed pollutants, we exposed zebrafish embryos to legacy and emerging pollutants through binary mixtures of benzo[a]pyrene (B[a]P) and diclofenac (DFC). The combination of next-generation transcriptomics and toxicopathology disclosed instances where exposure to mixtures did not attain the expected sum of acute effects of individual toxicants, indicating potential antagonism. Despite overall higher mortality in DFC treatments, the same antagonistic trend was noted in genotoxicity and molecular pathways related to RNA turnover, cell proliferation, apoptosis and cell-cycle control. The formation of oedemas in the heart cavity and yolk sac can be an adverse outcome (AO) resulting from exposure to DFC isolated or combined, whose potential key events (KEs) may involve cell cycle arrest and apoptosis via p53 and MAPK pathways. From the findings it can be hypothesised that, rather than genotoxicity, the molecular initiating event (MIE) maybe inflammation triggered by oxidative stress. Nonetheless, the exact role of ROS in the process needs further clarification. Impaired eye function by action of DFC and B[a]P combined may be another AO, in the case caused by ocular degeneration following the suppression of biologic processes and molecular functions involved in eye development and its functionalities, possibly linked to hindered regulation of the expression of hsf4 and cryaa. Altogether, toxicopathology suggests predominance of antagonistic effects, but its integration with mechanism suggests that interactions between DFC and B[a]P in environmentally-relevant concentrations that may lead to hindrance of key functions such as the control of inflammation and cell cycle. These outcomes suggest potentially severe implications for health and survival, in case of prolonged chronic exposure to combined toxicants.
Collapse
Affiliation(s)
- Carla Martins
- Associate Laboratory I4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal.
| | - Lara M Carvalho
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisboa, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-035 Lisboa, Portugal
| | - Inês Moutinho Cabral
- Associate Laboratory I4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal
| | - Leonor Saúde
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035 Lisboa, Portugal; Faculdade de Medicina, Universidade de Lisboa, 1649-035 Lisboa, Portugal
| | - Kristian Dreij
- Institute of Environmental Medicine, Karolinska Institutet, Box 210, SE-171 77 Stockholm, Sweden
| | - Pedro M Costa
- Associate Laboratory I4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, 2829-516, Caparica, Portugal.
| |
Collapse
|
2
|
Perontsis S, Hatzidimitriou AG, Psomas G. Coordination compounds of cobalt(II) with carboxylate non-steroidal anti-inflammatory drugs: structure and biological profile. Dalton Trans 2024; 53:15215-15235. [PMID: 39221624 DOI: 10.1039/d4dt01846j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Fourteen cobalt(II) complexes with the non-steroidal anti-inflammatory drugs sodium meclofenamate, tolfenamic acid, mefenamic acid, naproxen, sodium diclofenac, and diflunisal were prepared in the presence or absence of a series of nitrogen-donors (namely imidazole, pyridine, 3-aminopyridine, neocuproine, 2,2'-bipyridine, 1,10-phenanthroline and 2,2'-bipyridylamine) as co-ligands and were characterised by spectroscopic and physicochemical techniques. Single-crystal X-ray crystallography was employed to determine the crystal structure of eight complexes. The biological profile of the complexes was investigated regarding their interaction with serum albumins and DNA, and their antioxidant potency. The interaction of the compounds with calf-thymus DNA takes place via intercalation. The ability of the complexes to cleave pBR322 plasmid DNA at the concentration of 500 μM is rather low. The complexes demonstrated tight and reversible binding to human and bovine serum albumins and the binding site of bovine serum albumin was also examined. In order to assess the antioxidant activity of the compounds, the in vitro scavenging activity towards free radicals, namely 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid), and their ability to reduce H2O2 were studied.
Collapse
Affiliation(s)
- Spyros Perontsis
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece.
| | - Antonios G Hatzidimitriou
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece.
| | - George Psomas
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece.
| |
Collapse
|
3
|
Martins C, de Oliveira Galvão MF, Costa PM, Dreij K. Antagonistic effects of a COX1/2 inhibitor drug in human HepG2 cells exposed to an environmental carcinogen. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2024; 108:104453. [PMID: 38642625 DOI: 10.1016/j.etap.2024.104453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
Understanding interactions between legacy and emerging environmental contaminants has important implications for risk assessment, especially when mutagens and carcinogens are involved, whose critical effects are chronic and therefore difficult to predict. The current work aimed to investigate potential interactions between benzo[a]pyrene (B[a]P), a carcinogenic polycyclic aromatic hydrocarbon and legacy pollutant, and diclofenac (DFC), a non-steroidal anti-inflammatory drug and pollutant of emerging concern, and how DFC affects B[a]P toxicity. Exposure to binary mixtures of these chemicals resulted in substantially reduced cytotoxicity in human HepG2 cells compared to single-chemical exposures. Significant antagonistic effects were observed in response to high concentrations of B[a]P in combination with DFC at IC50 and ⅕ IC50. While additive effects were found for levels of intracellular reactive oxygen species, antagonistic mixture effects were observed for genotoxicity. B[a]P induced DNA strand breaks, γH2AX activation, and micronuclei formation at ½ IC50 concentrations or lower, whereas DFC induced only low levels of DNA strand breaks. Their mixture caused significantly lower levels of genotoxicity by all three endpoints compared to those expected based on concentration additivity. In addition, antagonistic mixture effects on CYP1 enzyme activity suggested that the observed reduced genotoxicity of B[a]P was due to its reduced metabolic activation as a result of enzymatic inhibition by DFC. Overall, the findings further support the growing concern that co-exposure to environmental toxicants and their non-additive interactions may be a confounding factor that should not be neglected in environmental and human health risk assessment.
Collapse
Affiliation(s)
- Carla Martins
- Associate Laboratory i4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, Caparica 2819 516, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, Caparica 2819 516, Portugal; Unit of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, Stockholm SE-171 77, Sweden.
| | - Marcos Felipe de Oliveira Galvão
- Unit of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, Stockholm SE-171 77, Sweden
| | - Pedro M Costa
- Associate Laboratory i4HB Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University of Lisbon, Caparica 2819 516, Portugal; UCIBIO Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University of Lisbon, Caparica 2819 516, Portugal
| | - Kristian Dreij
- Unit of Biochemical Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, Stockholm SE-171 77, Sweden.
| |
Collapse
|
4
|
Emre FB, Turhan DÖ, Güngördü A. Toxicity of commercial and pure forms of three nonsteroidal anti-inflammatory drugs in Xenopus laevis embryos before and after ozonation. Drug Chem Toxicol 2024:1-14. [PMID: 38465443 DOI: 10.1080/01480545.2024.2324325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/23/2024] [Indexed: 03/12/2024]
Abstract
In this study, the toxic and teratogenic effects of three commercial drugs and their active ingredients on Xenopus laevis embryos before and after ozonation were evaluated using the Frog Embryos Teratogenesis Assay-Xenopus (FETAX). First, the median lethal concentration (LC50) and, if data were available, the median effective concentration, teratogenic index and minimum growth inhibitory concentration were determined for each drug substance without ozonation. Then, the active substance amounts of three selected nominal concentrations (LC50/2, LC50, and LC50×2) of each test substance before ozonation were measured by HPLC analysis and the toxicity of these substances was evaluated after 2, 3, 4, and 5 h of ozonation. In addition, degradation products that may occur during ozonation were evaluated by LC-MS analysis. The 96-h LC50s of Dolphin-diflunisal, Dichloron-diclofenac sodium, and Apranax-naproxen drug-active substance pairs were determined to be 22.3 and 11.1, 25.7 and 18.7, and 47.8 mg active substance/L and 45.3 mg/L, respectively. According to the FETAX test results, the Dolphin-diflunisal drug-active ingredient pair did not cause growth retardation in exposed embryos. Dichloron-diclofenac sodium and Apranax-naproxen drug-active ingredient pairs were both teratogenic and growth inhibitory. In the second stage of the study, in which the effectiveness of ozonation in eliminating the toxic effects of drugs is evaluated, it is seen that ozonation is partially successful in eliminating the toxic effects of Dolphin-diflunisal and Dichloron-diclofenac sodium pairs, but insufficient for eliminating the effects of the Apranax-naproxen pair.
Collapse
Affiliation(s)
- Fatma Bilge Emre
- Department of Mathematics and Science Education, Faculty of Education, Inonu University, Malatya, Turkey
| | - Duygu Özhan Turhan
- Laboratory of Environmental Toxicology, Department of Biology, Faculty of Arts and Science, Inonu University, Malatya, Turkey
| | - Abbas Güngördü
- Laboratory of Environmental Toxicology, Department of Biology, Faculty of Arts and Science, Inonu University, Malatya, Turkey
| |
Collapse
|
5
|
Meligi NM, Dyab AKF. Natural sporopollenin microcapsules: biological evaluation and application in regulating hepatic toxicity of diclofenac sodium in vivo. Biomater Sci 2023; 11:6193-6209. [PMID: 37522344 DOI: 10.1039/d3bm00638g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Diclofenac sodium (DIC) is a pain reliever and anti-nociceptive medication. Significant limitations of DIC treatment stem from its adverse effects. This study investigates the feasibility of using natural Lycopodium clavatum sporopollenin (LCS) microcapsules loaded with DIC to mitigate the hepatotoxicity associated with DIC treatment. In addition, LCS microcapsules were tracked in the blood, stomach, small intestine, and feces of rats to demonstrate their morphological integrity and uptake behavior. Four groups (6 per group) of adult male albino rats were administered normal saline (control), empty LCS (30 mg kg-1), plain DIC (10 mg kg-1), and DIC-loaded LCS (40 mg kg-1) orally for seven consecutive days. The first comprehensive histological examination of the rat stomach demonstrated the robustness and bioadhesion ability of LCS under severe conditions. The findings suggested that these versatile microcapsules are unlikely to be digested in the gastrointestinal tract (GIT). The administration of DIC-loaded LCS was found to play a potential protective role in regulating DIC-induced substantially increased serum levels of transaminases, alkaline phosphatase, total bilirubin, and pro-inflammatory cytokines. In addition, DIC-loaded LCS restored the antioxidant enzymes, DNA damage, and liver histological architecture abnormalities caused by DIC. Microencapsulation of DIC into pollen-derived biomaterials could be employed as an efficient platform with enough safety coverage on rat liver, pending further clinical studies.
Collapse
Affiliation(s)
- Noha M Meligi
- Zoology Department, Faculty of Science, Minia University, Minia 61519, Egypt.
| | - Amro K F Dyab
- Department of Chemistry, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan.
| |
Collapse
|
6
|
Choi JY, Jeong M, Lee K, Kim JO, Lee WH, Park I, Kwon HC, Choi JH. Sedum middendorffianum Maxim Induces Apoptosis and Inhibits the Invasion of Human Ovarian Cancer Cells via Oxidative Stress Regulation. Antioxidants (Basel) 2023; 12:1386. [PMID: 37507925 PMCID: PMC10376315 DOI: 10.3390/antiox12071386] [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: 06/07/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Sedum middendorffianum Maxim (SMM) is a Korean endemic plant belonging to the Crassulaceae family. This study aimed to investigate the antitumor effects of the SMM extract on human ovarian cancer cells. Among five endemic plants grown in Korea, the SMM extract showed the most potent cytotoxicity in ovarian cancer cells and had little effect on normal ovarian surface epithelial cells. Furthermore, we revealed that the SMM extract dose-dependently induced apoptosis in human ovarian cancer A2780 and SKOV3 cells. The SMM extract markedly stimulated the activation of caspase-3/8, while the broad-spectrum caspase inhibitor and caspase-8 selective inhibitor significantly reversed SMM extract-induced apoptosis. In addition, the SMM extract significantly inhibited cell invasion and the expression levels of matrix metalloproteinase (MMP)-2 and MMP-9 in ovarian cancer cells. Notably, the SMM extract increased the generation of intracellular ROS, and pretreatment with antioxidant N-acetyl-L-cysteine (NAC) significantly suppressed SMM-induced cytotoxicity and anti-invasive activity. Moreover, NAC treatment reversed the SMM-induced inhibition of MMP-2/9 expression. Taken together, these data suggest that the SMM extract induces caspase-dependent apoptotic cell death and inhibits MMP-dependent invasion via ROS regulation.
Collapse
Affiliation(s)
- Ju-Yeon Choi
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Miran Jeong
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kijun Lee
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, Republic of Korea
- College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jin-Ok Kim
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wan Hee Lee
- Hantaek Botanical Garden, Yongin 17183, Republic of Korea
| | - InWha Park
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute, Gangneung 25451, Republic of Korea
| | - Hak Cheol Kwon
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST) Gangneung Institute, Gangneung 25451, Republic of Korea
| | - Jung-Hye Choi
- Department of Biomedical and Pharmaceutical Science, Kyung Hee University, Seoul 02447, Republic of Korea
- Hantaek Botanical Garden, Yongin 17183, Republic of Korea
| |
Collapse
|
7
|
Peng F, Xu Q, Jing X, Chi X, Zhang Z, Meng X, Liu X, Yan J, Liu X, Shao S. GPX2 promotes EMT and metastasis in non-small cell lung cancer by activating PI3K/AKT/mTOR/Snail signaling axis. FASEB Bioadv 2023; 5:233-250. [PMID: 37287867 PMCID: PMC10242197 DOI: 10.1096/fba.2022-00045] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/01/2023] [Accepted: 03/03/2023] [Indexed: 10/15/2023] Open
Abstract
Lung cancer, with non-small cell lung cancer (NSCLC) being the main subtype, is the leading cause of cancer death worldwide, which is mainly due to the cancer metastasis. Glutathione peroxidase 2 (GPX2), an antioxidant enzyme, is involved in tumor progression and metastasis. Nevertheless, the role of GPX2 in NSCLC metastasis has not been clarified. In this study, we found that GPX2 expression was elevated in NSCLC tissues and high GPX2 expression was correlated with poor prognosis in patients with NSCLC. In addtion, GPX2 expression was related to the patient's clinicopathological features, including lymph node metastasis, tumor size, and TNM stage. Overexpression of GPX2 promoted epithelial-mesenchymal transition (EMT), migration, and invasion of NSCLC cells in vitro. Knockdown of GPX2 showed the opposite effects in vitro and inhibited the metastasis of NSCLC cells in nude mice. Furthermore, GPX2 reduced reactive oxygen species (ROS) accumulation and activated the PI3K/AKT/mTOR/Snail signaling axis. Therefore, our results indicate that GPX2 promotes EMT and metastasis of NSCLC cells by activating the PI3K/AKT/mTOR/Snail signaling axis via the removal of ROS. GPX2 may be an effective diagnostic and prognostic biomarker for NSCLC.
Collapse
Affiliation(s)
- Fang Peng
- Liaoning Key Laboratory of ProteomicsDalian Medical UniversityDalianChina
- Department of PathologySecond Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Qiushi Xu
- Department of NeurosurgeryDalian Municipal Central HospitalDalianChina
| | - Xiaomeng Jing
- Department of PathologySecond Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xinming Chi
- Liaoning Key Laboratory of ProteomicsDalian Medical UniversityDalianChina
| | - Zheming Zhang
- Liaoning Key Laboratory of ProteomicsDalian Medical UniversityDalianChina
| | - Xiangpeng Meng
- Liaoning Key Laboratory of ProteomicsDalian Medical UniversityDalianChina
| | - Xinyuan Liu
- Liaoning Key Laboratory of ProteomicsDalian Medical UniversityDalianChina
| | - Jiao Yan
- Liaoning Key Laboratory of ProteomicsDalian Medical UniversityDalianChina
| | - Xuefeng Liu
- Institute of Cancer Stem CellDalian Medical UniversityDalianChina
| | - Shujuan Shao
- Liaoning Key Laboratory of ProteomicsDalian Medical UniversityDalianChina
| |
Collapse
|
8
|
Lazou M, Hatzidimitriou AG, Papadopoulos AN, Psomas G. Transition metal(II) complexes with the non–steroidal anti–inflammatory drug oxaprozin: Characterization and biological profile. J Inorg Biochem 2023; 243:112196. [PMID: 36966675 DOI: 10.1016/j.jinorgbio.2023.112196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/02/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
A series of copper(II), nickel(II) and cobalt(II) complexes with the non-steroidal anti-inflammatory drug oxaprozin (Hoxa) have been synthesized and characterized by diverse techniques. The crystal structures of two copper(II) complexes, namely the dinuclear complex [Cu2(oxa)4(DMF)2] (1) and the polymeric complex {[Cu2(oxa)4]·2MeOH·0.5MeOH}2 (12) were determined by single-crystal X-ray diffraction studies. In order to evaluate in vitro the antioxidant activity of the resultant complexes, their scavenging ability towards 1,1-diphenyl-picrylhydrazyl (DPPH), hydroxyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) radicals was investigated revealing their high effectiveness against these radicals. The binding of the complexes to bovine serum albumin and human serum albumin was examined and the corresponding determined albumin-binding constants showed a tight and reversible interaction. The interaction of the complexes with calf-thymus DNA was monitored by diverse techniques including UV-vis spectroscopy, cyclic voltammetry, DNA-viscosity measurements and competitive studies with ethidium bromide. Intercalation may be proposed as the most possible DNA-interaction mode of the complexes.
Collapse
|
9
|
Lae Lae Phoo N, Sukhamwang A, Dejkriengkraikul P, Yodkeeree S. Diclofenac Sensitizes Signet Ring Cell Gastric Carcinoma Cells to Cisplatin by Activating Autophagy and Inhibition of Survival Signal Pathways. Int J Mol Sci 2022; 23:ijms232012066. [PMID: 36292923 PMCID: PMC9602524 DOI: 10.3390/ijms232012066] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Gastric cancer has one of the highest incidence rates of cancer worldwide while also contributing to increased drug resistance among patients in clinical practice. Herein, we have investigated the role of diclofenac (DCF) on sensitizing cisplatin resistance in signet ring cell gastric carcinoma cells (SRCGC). Non-toxic concentrations of DCF significantly augmented cisplatin-induced cell death in cisplatin-resistant SRCGC cells (KATO/DDP) but not in cisplatin-sensitive SRCGC cells (KATOIII). Consistently, concomitant treatment of DCF and cisplatin significantly enhanced autophagic cell death due to overproduction of intracellular reactive oxygen species (ROS). At the molecular level, the induction of ROS has been associated with a reduction in antioxidant enzymes expression while inhibiting nuclear factor erythroid 2-related factor 2 (Nrf2) activity. Moreover, the combination of DCF and cisplatin also inhibited the expression of survival proteins including Bcl-2, Bcl-xL, cIAP1 and cyclin D1 in KATO/DDP cells when compared with cisplatin alone. This was due, at least in part, to reduce MAPKs, Akt, NF-κB, AP-1 and STAT-3 activation. Taken together, our results suggested that DCF potentiated the anticancer effect of cisplatin in SRCGC via the regeneration of intracellular ROS, which in turn promoted cell death as an autophagy mechanism and potentially modulated the cell survival signal transduction pathway.
Collapse
Affiliation(s)
- Nang Lae Lae Phoo
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Amonnat Sukhamwang
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornngarm Dejkriengkraikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Supachai Yodkeeree
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence:
| |
Collapse
|
10
|
Development and Challenges of Diclofenac-Based Novel Therapeutics: Targeting Cancer and Complex Diseases. Cancers (Basel) 2022; 14:cancers14184385. [PMID: 36139546 PMCID: PMC9496891 DOI: 10.3390/cancers14184385] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Diclofenac is a widely used drug for its anti-inflammatory and pain alleviating properties. This review summarizes the current understanding about the drug diclofenac. The potential applications of diclofenac beyond its well-known anti-inflammatory properties for other diseases such as cancer are discussed, along with existing limitations. Abstract Diclofenac is a highly prescribed non-steroidal anti-inflammatory drug (NSAID) that relieves inflammation, pain, fever, and aches, used at different doses depending on clinical conditions. This drug inhibits cyclooxygenase-1 and cyclooxygenase-2 enzymes, which are responsible for the generation of prostaglandin synthesis. To improve current diclofenac-based therapies, we require new molecular systematic therapeutic approaches to reduce complex multifactorial effects. However, the critical challenge that appears with diclofenac and other drugs of the same class is their side effects, such as signs of stomach injuries, kidney problems, cardiovascular issues, hepatic issues, and diarrhea. In this article, we discuss why defining diclofenac-based mechanisms, pharmacological features, and its medicinal properties are needed to direct future drug development against neurodegeneration and imperfect ageing and to improve cancer therapy. In addition, we describe various advance molecular mechanisms and fundamental aspects linked with diclofenac which can strengthen and enable the better designing of new derivatives of diclofenac to overcome critical challenges and improve their applications.
Collapse
|
11
|
Jozefíková F, Perontsis S, Koňáriková K, Švorc Ľ, Mazúr M, Psomas G, Moncol J. In vitro biological activity of copper(II) complexes with NSAIDs and nicotinamide: Characterization, DNA- and BSA-interaction study and anticancer activity. J Inorg Biochem 2021; 228:111696. [PMID: 35030390 DOI: 10.1016/j.jinorgbio.2021.111696] [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: 03/15/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 11/28/2022]
Abstract
Through the reaction of copper(II) acetate with nicotinamide (pyridine-3-carboxylic acid amide, niacinamide) and some derivatives of N-phenylanthranilic acid (fenamates), seven new mixed-ligand copper(II) compounds were isolated: [Cu(tolf-O)(tolf-O,O')nia-N)2(EtOH)] (1), [Cu(tolf-O)(tolf-O,O')(nia-N)2(MeOH)] (2), [Cu(meclf-O)(meclf-O,O')(nia-N)2(EtOH)] (3), [Cu(meclf-O)(meclf-O,O')(nia-N)2(MeOH)] (4), [Cu(meclf-O)(meclf-O,O')(nia-N)2(ACN)] (5), [Cu(mef-O)(mef-O,O')(nia-N)2(EtOH)] (6) and [Cu(mef-O)(mef-O,O')(nia-N)2(ACN)] (7) containing a molecule of relevant solvent as ligand in their primary crystal structure (tolf = tolfenamate, meclf = meclofenamate, mef = mefenamate, nia = nicotinamide, EtOH = ethanol, MeOH = methanol, ACN = acetonitrile). The structures of the complexes were determined by single-crystal X-ray analysis. The intermolecular interactions were studied by Hirshfeld surface analysis. The complexes were characterized by IR, UV-vis and EPR spectroscopy and their redox properties were determined by cyclic voltammetry. The interaction of the complexes with bovine serum albumin was studied by fluorescence emission spectroscopy and the albumin-binding constants of the compounds were calculated. The interaction of the complexes with calf-thymus DNA was monitored by diverse techniques (UV-vis spectroscopy, cyclic voltammetry, viscosity measurements) suggesting intercalation as the most possible mode of binding. DNA-competitive studies of the complexes with ethidium bromide were monitored by fluorescence emission spectroscopy. The cytotoxic effects of copper(II) complexes on lung carcinoma cells and healthy cells were determined by the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] colorimetric technique.
Collapse
Affiliation(s)
- Flóra Jozefíková
- Department of Inorganic Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia; Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Spyros Perontsis
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Katarína Koňáriková
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Sasinkova 2, 81372 Bratislava, Slovakia
| | - Ľubomír Švorc
- Institute of Analytical Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia
| | - Milan Mazúr
- Department of Physical Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia
| | - George Psomas
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece.
| | - Ján Moncol
- Department of Inorganic Chemistry, Faculty of Chemical and Food Technology, Slovak University of Technology, Radlinského 9, 812 37 Bratislava, Slovakia.
| |
Collapse
|
12
|
Busch M, Kämpfer AAM, Schins RPF. An inverted in vitro triple culture model of the healthy and inflamed intestine: Adverse effects of polyethylene particles. CHEMOSPHERE 2021; 284:131345. [PMID: 34216924 DOI: 10.1016/j.chemosphere.2021.131345] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/01/2021] [Accepted: 06/25/2021] [Indexed: 06/13/2023]
Abstract
As environmental pollution with plastic waste is increasing, numerous reports show the contamination of natural habitats, food and drinking water with plastic particles in the micro- and nanometer range. Since oral exposure to these particles is virtually unavoidable, health concerns towards the general population have been expressed and risk assessment regarding ingested plastic particles is of great interest. To study the intestinal effects of polymeric particles with a density of <1 g/cm³ in vitro, we spatially inverted a triple culture transwell model of the healthy and inflamed intestine (Caco-2/HT29-MTX-E12/THP-1), which allows contact between buoyant particles and cells. We validated the inverted model against the original model using the enterotoxic, non-steroidal anti-inflammatory drug diclofenac and subsequently assessed the cytotoxic and pro-inflammatory effects of polyethylene (PE) microparticles. The results show that the inverted model exhibits the same distinct features as the original model in terms of barrier development and inflammatory parameters. Treatment with 2 mM diclofenac causes severe cytotoxicity, DNA damage and complete barrier disruption in both models. PE particles induced cytotoxicity and pro-inflammatory effects in the inverted model, which would have remained undetected in conventional in vitro approaches, as no effect was observed in non-inverted control cultures.
Collapse
Affiliation(s)
- Mathias Busch
- IUF - Leibniz-Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz-Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Roel P F Schins
- IUF - Leibniz-Research Institute for Environmental Medicine, Duesseldorf, Germany.
| |
Collapse
|
13
|
Chitosan/Selenium Nanoparticles Attenuate Diclofenac Sodium-Induced Testicular Toxicity in Male Rats. CRYSTALS 2021. [DOI: 10.3390/cryst11121477] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The detrimental effect of diclofenac sodium (Diclo-Na) on male reproductive organs is reported upon in this paper. Chitosan is a polysaccharide composed of various amounts of glucosamine. Chitosan nanoparticles (CH-NPs) have attracted much attention owing to their biomedical activity. Selenium (Se) has a vital role in nutrition, plays an important role in enhancing male reproduction, and has a wide range of free radical scavenging activities. However, the study of the impact of chitosan nanoparticles in combination with Se (IV) (CH-NPs/Se) on male reproductive toxicity associated with Diclo-Na administration is lacking in recent literature. The current study assessed the ameliorative effects of complexes of CH-NPs/Se (IV) on Diclo-Na and the ways in which they alter reproductive toxicity in male rats. Male rats were treated for 30 days successively, either with Diclo-Na (10 mg/kg) or co-treated with a CH-NPs/Se complex (280 mg/kg). Sperm characteristics, marker enzymes of testicular function, LH, FSH, and testosterone were evaluated in addition to oxidative stress markers and histological alterations. CH-NPs/Se significantly alleviated Diclo-Na-induced decline in sperm count and motility, testicular function enzymes, and levels of LH and testosterone in serum. Additionally, CH-NPs/Se co-administration at 280 mg/Kg, inhibited the Diclo-Na-induced decline of antioxidant enzyme activities and elevated oxidative stress indices and reactive free radicals in testicular homogenates of male rats. CH-NPs/Se (280 mg/kg) alone improved Diclo-Na and ameliorated histological damages in exposed rats. In conclusion, chitosan improved testicular function in Diclo-Na-treated rats by enhancing the testosterone hormone levels, ameliorating testicular tissue, and inhibiting markers of oxidative stress in male rats.
Collapse
|
14
|
Nematollahzadeh A, Mirzaei-Kalar Z, Abolhasani H, Babapoor A. Synthesize and multi-spectroscopic studies of zinc-naproxen nanodrug as DNA intercalator agent. Anal Biochem 2021; 642:114454. [PMID: 34774837 DOI: 10.1016/j.ab.2021.114454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/07/2021] [Accepted: 11/08/2021] [Indexed: 11/27/2022]
Abstract
The zinc-naproxen complex as a nano-drug (NanoD) was synthesized successfully via fast and effective ultrasound-assisted processes. The chemicophysical properties of the NanoD were determined using FT-IR, XRD, SEM, and EDX mapping analyses. The results confirmed the formation of the 55 nm NanoD laminates. The interaction of the obtained NanoD with calf thymus deoxyribonucleic acid (CT-DNA) was studied as well. Structural and topography changes of DNA in interaction with the NanoD were investigated by atomic force microscopy (AFM). The results of electronic absorption spectroscopy, the DNA-viscosity studies, and competition fluorescence spectroscopy showed that CT-DNA binds to the NanoD through the intercalative binding mode. The data of AFM analysis indicated swollen CT-DNA upon interaction with the NanoD. The in vitro investigation of cytotoxicity of the NanoD on HT-29 and Hep G2 cancer cells demonstrated high cytotoxicity activity of the NanoD than that of cisplatin in HT-29 cell line, especially at lower concentrations.
Collapse
Affiliation(s)
- Ali Nematollahzadeh
- Department of Chemical Engineering, University of Mohaghegh Ardabili, P.O. Box 179, Ardabil, Iran.
| | - Zeinab Mirzaei-Kalar
- Department of Advanced Technologies, University of Mohaghegh Ardabili, Namin, Ardabil, Iran
| | - Hoda Abolhasani
- Cellular and Molecular Research Center and Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Aziz Babapoor
- Department of Chemical Engineering, University of Mohaghegh Ardabili, P.O. Box 179, Ardabil, Iran
| |
Collapse
|
15
|
Galisteo A, Jannus F, García-García A, Aheget H, Rojas S, Lupiañez JA, Rodríguez-Diéguez A, Reyes-Zurita FJ, Quílez del Moral JF. Diclofenac N-Derivatives as Therapeutic Agents with Anti-Inflammatory and Anti-Cancer Effect. Int J Mol Sci 2021; 22:ijms22105067. [PMID: 34064702 PMCID: PMC8151993 DOI: 10.3390/ijms22105067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/07/2021] [Accepted: 05/08/2021] [Indexed: 12/20/2022] Open
Abstract
A series of diclofenac N-derivatives (2, 4, 6, 8c, 9c, 10a-c) were synthesized in order to test their anti-cancer and anti-inflammatory effects. The anticarcinogen activity has been assayed against three cancer cell lines: HT29, human colon cancer cells; Hep-G2, human hepatic cells; and B16-F10, murine melanoma cells. First, we determined the cytotoxicity of the different compounds, finding that the most effective compound was compound 8c against all cell lines and both compounds 4 and 6 in human Hep-G2 and HT29 cell lines. Compounds 4 and 8c were selected for the percentage of apoptosis determination, cell cycle distribution, and mitochondrial membrane potential measure because these products presented the lowest IC50 values in two of the three cancer cell lines assayed (B16-F10 and HepG2), and were two of the three products with lowest IC50 in HT29 cell line. Moreover, the percentages of apoptosis induction were determined for compounds 4 and 8c, showing that the highest values were between 30 to 60%. Next, the effects of these two compounds were observed on the cellular cycle, resulting in an increase in the cell population in G2/M cell cycle phase after treatment with product 8c, whereas compound 4 increased the cells in phase G0/G1, by possible differentiation process induction. Finally, to determine the possible apoptosis mechanism triggered by these compounds, mitochondrial potential was evaluated, indicating the possible activation of extrinsic apoptotic mechanism. On the other hand, we studied the anti-inflammatory effects of these diclofenac (DCF) derivatives on lipopolysaccharide (LPS) activated RAW 264.7 macrophages-monocytes murine cells by inhibition of nitric oxide (NO) production. As a first step, we determined the cytotoxicity of the synthesized compounds, as well as DCF, against these cells. Then, sub-cytotoxic concentrations were used to determine NO release at different incubation times. The greatest anti-inflammatory effect was observed for products 2, 4, 8c, 10a, 10b, and 9c at 20 µg·mL-1 concentration after 48 h of treatment, with inhibition of produced NO between 60 to 75%, and a concentration that reduces to the 50% the production of NO (IC50 NO) between 2.5 to 25 times lower than that of DCF. In this work, we synthesized and determined for the first time the anti-cancer and anti-inflammatory potential of eight diclofenac N-derivatives. In agreement with the recent evidences suggesting that inflammation may contribute to all states of tumorigenesis, the development of these new derivatives capable of inducing apoptosis and anti-inflammatory effects at very low concentrations represent new effective therapeutic strategies against these diseases.
Collapse
Affiliation(s)
- Alberto Galisteo
- Department of Organic Chemistry, Institute of Biotechnology, University of Granada, 18071 Granada, Spain;
| | - Fatin Jannus
- Department of Biochemistry and Molecular Biology, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (F.J.); (J.A.L.)
| | - Amalia García-García
- Department of Inorganic Chemistry, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (A.G.-G.); (S.R.); (A.R.-D.)
| | - Houssam Aheget
- Centre for Genomics and Oncological Research, GENYO, C/Health Sciences Technology Park, Av. de la Illustration 114, 18016 Granada, Spain;
| | - Sara Rojas
- Department of Inorganic Chemistry, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (A.G.-G.); (S.R.); (A.R.-D.)
| | - José A. Lupiañez
- Department of Biochemistry and Molecular Biology, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (F.J.); (J.A.L.)
| | - Antonio Rodríguez-Diéguez
- Department of Inorganic Chemistry, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (A.G.-G.); (S.R.); (A.R.-D.)
| | - Fernando J. Reyes-Zurita
- Department of Biochemistry and Molecular Biology, University of Granada, C/Severo Ochoa s/n, 18071 Granada, Spain; (F.J.); (J.A.L.)
- Correspondence: (F.J.R.-Z.); (J.F.Q.d.M.); Tel.: +34-958243252 (F.J.R.-Z.); +34-958243185 (J.F.Q.d.M.)
| | - José F. Quílez del Moral
- Department of Organic Chemistry, Institute of Biotechnology, University of Granada, 18071 Granada, Spain;
- Correspondence: (F.J.R.-Z.); (J.F.Q.d.M.); Tel.: +34-958243252 (F.J.R.-Z.); +34-958243185 (J.F.Q.d.M.)
| |
Collapse
|
16
|
Queiroz HA, da Silva CR, de Andrade Neto JB, do Av Sá LG, do Nascimento FB, Moreno LS, Barroso FD, da Silva LJ, Cândido TM, de Oliveira LC, de Mesquita JR, de Moraes MO, Cavalcanti BC, Nobre Júnior HV. Synergistic activity of diclofenac sodium with oxacillin against planktonic cells and biofilm of methicillin-resistant Staphylococcus aureus strains. Future Microbiol 2021; 16:375-387. [PMID: 33870731 DOI: 10.2217/fmb-2020-0095] [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] [Indexed: 11/21/2022] Open
Abstract
Aim: To evaluate the activity of diclofenac sodium and synergism with oxacillin against clinical strains of SARM in plactonic cells, antibiofilm and biofilm. Materials & methods: Synergism activity was assessed using the fractional inhibitory concentration index and its possible mechanism of action by flow cytometry. Results: The synergistic activity of diclofenac sodium with oxacillin was observed against plactonic cells, antibiofilm and in biofilm formed from clinical methicillin-resistant Staphylococcus aureus strains. Conclusion: This combination caused damage to the integrity of the membrane and ruptures in the DNA of the cells, leading to apoptosis.
Collapse
Affiliation(s)
- Helaine A Queiroz
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Cecília R da Silva
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - João B de Andrade Neto
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,University Center Christus, Fortaleza, CE, Brazil
| | - Lívia G do Av Sá
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil.,University Center Christus, Fortaleza, CE, Brazil
| | - Francisca Bsa do Nascimento
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Leticia Ss Moreno
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Fátima Dd Barroso
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Lisandra J da Silva
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Thiago M Cândido
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Leilson C de Oliveira
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Manoel O de Moraes
- Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Bruno C Cavalcanti
- Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Hélio V Nobre Júnior
- Department of Clinical & Toxicological Analysis, School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Drug Research & Development Center, Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
17
|
Dimiza F, Hatzidimitriou AG, Sanakis Y, Papadopoulos AN, Psomas G. Trinuclear and tetranuclear iron(III) complexes with fenamates: Structure and biological profile. J Inorg Biochem 2021; 218:111410. [PMID: 33721718 DOI: 10.1016/j.jinorgbio.2021.111410] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/16/2023]
Abstract
The interaction of FeCl3 with the fenamate non-steroidal anti-inflammatory drugs has led to the formation and isolation of trinuclear iron(III) complexes, while in the presence of the nitrogen-donors 2,2'-bipyridine or pyridine tetranuclear iron(III) complexes were derived. The five resultant complexes were characterized by diverse techniques (including infrared, electronic and Mössbauer spectroscopy) and their crystal structures were determined by single-crystal X-ray crystallography. These complexes are the first structurally characterized Fe(III)-fenamato complexes. The complexes were evaluated for their ability to scavenge in vitro free radicals such as hydroxyl, 1,1-diphenyl-2-picrylhydrazyl and 2,2΄-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid). The in vitro binding affinity of the complexes to calf-thymus (CT) DNA was examined and their interaction with serum albumins was also investigated. In total, the complexes present promising activity against the radicals tested, and they may bind tightly to CT DNA possibly via intercalation and reversibly to serum albumins.
Collapse
Affiliation(s)
- Filitsa Dimiza
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Antonios G Hatzidimitriou
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Yiannis Sanakis
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Ag. Paraskevi, Attiki, Greece
| | - Athanasios N Papadopoulos
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Sindos, Thessaloniki, Greece
| | - George Psomas
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece.
| |
Collapse
|
18
|
Elshopakey GE, Elazab ST. Cinnamon Aqueous Extract Attenuates Diclofenac Sodium and Oxytetracycline Mediated Hepato-Renal Toxicity and Modulates Oxidative Stress, Cell Apoptosis, and Inflammation in Male Albino Rats. Vet Sci 2021; 8:vetsci8010009. [PMID: 33418920 PMCID: PMC7825122 DOI: 10.3390/vetsci8010009] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Among commonly consumed anti-inflammatory and antimicrobial drugs are diclofenac sodium (DFS) and oxytetracycline (OTC), especially in developing countries because they are highly effective and cheap. However, the concomitant administration of anti-inflammatory drugs with antibiotics may exaggerate massive toxic effects on many organs. Cinnamon (Cinnamomum zeylanicum, Cin) is considered one of the most broadly utilized plants with various antioxidant and anti-inflammatory actions. This study aimed to evaluate the possible protective effects of cinnamon aqueous extract (Cin) against DFS and OTC hepato-renal toxicity. Eight groups (8/group) of adult male albino rats were treated orally for 15 days with physiological saline (control), Cin aqueous extract (300 mg/kg b.w.), OTC (200 mg/kg b.w.), single dose of DFS at the 14th day (100 mg/kg b.w.), DFS + OTC, Cin + DFS, Cin + OTC, and Cin + DFS + OTC. The administration of DFS and/or OTC significantly increased (p < 0.05) the serum levels of alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, urea, creatinine, and uric acid. Serum levels of pro-inflammatory cytokines, as well as hepatic and renal malondialdehyde and nitric oxide metabolites, were also raised following DFS and OTC administration. Meanwhile, the activities of reduced glutathione, superoxide dismutase, and catalase in liver and kidney were significantly suppressed in DFS, OTC, and DFS + OTC treated rats. Moreover, hepatic and renal tissue sections from these rats exhibited overexpression of caspase-3 and cyclooxygenase-II on immunohistochemical investigation. The administration of Cin aqueous extract ameliorated the aforementioned deteriorations caused by DFS, OTC, and their combination. Conclusively, Cin is a promising protective plant extract capable of attenuating the oxidative damage, apoptosis, and inflammation induced by DFS and OTC either alone or combined, on hepatic and renal tissues.
Collapse
Affiliation(s)
- Gehad E. Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
- Correspondence: or ; Tel.: +20-102-392-3945
| | - Sara T. Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; or
| |
Collapse
|
19
|
Marinov L, Georgieva A, Voynikov Y, Toshkova R, Nikolova I, Malchev M. Cytotoxic and antiproliferative effects of the nonsteroidal anti-inflammatory drug diclofenac in human tumour cell lines. BIOTECHNOL BIOTEC EQ 2021. [DOI: 10.1080/13102818.2021.1953401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Affiliation(s)
- Lyubomir Marinov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Ani Georgieva
- Department Pathology, Institute of Experimental Morphology, Pathology, and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yulian Voynikov
- Department of Chemistry, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Reneta Toshkova
- Department Pathology, Institute of Experimental Morphology, Pathology, and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Irina Nikolova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Martin Malchev
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
20
|
Mousa AA, Elweza AE, Elbaz HT, Tahoun EAEA, Shoghy KM, Elsayed I, Hassan EB. Eucalyptus Globulus protects against diclofenac sodium induced hepatorenal and testicular toxicity in male rats. J Tradit Complement Med 2020; 10:521-528. [PMID: 33134128 PMCID: PMC7588335 DOI: 10.1016/j.jtcme.2019.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 10/01/2019] [Accepted: 11/23/2019] [Indexed: 12/11/2022] Open
Abstract
The current study was conducted to investigate the protective properties of Eucalyptus globulus leaves methanolic extract (EGLME) against diclofenac sodium (DS) induced hepatorenal and testicular toxicity in male rats. A total of 40 rats were equally divided into 4 groups, Control, Diclofenac sodium (DS), EGLME and DS + EGLME groups, respectively. DS and EGLME were administered orally at dose rate 2.5 and 100 mg/kg BW, 4 times/week for 8 weeks, respectively. Administration of DS distorted hepatorenal functions manifested by alteration of serum levels of ALT, AST, total protein and albumin, creatinine and urea with changes of histological architectures. DS caused reproductive toxicity represented by minimized sperm count, individual sperm motility and viability; depleted concentration of reduced glutathione (GSH) in testicular tissue; and decreased testosterone level with alteration in testicular histological features. In contrast, co-treatment of DS intoxicated rats with EGLME protected rats against the adverse effects of DS revealing enhancing properties of EGLME on rats' liver, kidney and testes. In conclusion, we demonstrated that EGLME had a potent protecting property against DS induced hepatic, renal and testicular toxicity in male rats, with special concern to testicular tissue via modulation of GSH as an oxidant marker. TAXONOMY (classification by EVISE): Diclofenac sodium toxicity (hepatorenal and testicular toxicity), co-treatment with natural herbal extract, blood biochemical assays, tissue anti-oxidants assay, histopathology and reproductive indices analyses.
Collapse
Affiliation(s)
- Ahmed Abdelmoniem Mousa
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Ahmed Essam Elweza
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Hamed Talaat Elbaz
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Enas Abd El-aziz Tahoun
- Department of Pathology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Khaled Mohamed Shoghy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, University of Sadat City, Sadat, City, 32897, Menoufia, Egypt
| | - Islam Elsayed
- Department of Sustainable Bioproducts, Mississippi State University, Box 9820, Mississippi State, MS, 39762, United States
| | - El Barbary Hassan
- Department of Sustainable Bioproducts, Mississippi State University, Box 9820, Mississippi State, MS, 39762, United States
| |
Collapse
|
21
|
Akinloye OA, Akinloye DI, Onigbinde SB, Metibemu DS. Phytosterols demonstrate selective inhibition of COX-2: In-vivo and in-silico studies of Nicotiana tabacum. Bioorg Chem 2020; 102:104037. [PMID: 32707278 DOI: 10.1016/j.bioorg.2020.104037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/26/2020] [Accepted: 06/18/2020] [Indexed: 12/28/2022]
Abstract
The inhibition of cyclooxygenase-1 (COX-1) enzyme by Nonsteroidal anti-inflammatory drugs (NSAIDs) exposes the gastrointestinal mucosa to peptic injuries. Selective inhibition of COX-2 generates surpassing anti-inflammatory drug candidates with reduced side effects over current NSAIDs. Phytosterols consumption is reported to decrease the risk of cardiovascular problems. Reports on the selective inhibition of COX-2 by phytosterols are scarce. The present study assesses the anti-inflammatory potentials of phytosterols from Nicotiana tabacum (of the family Solanaceae) through selective inhibition of COX-1 and/or COX-2. Virtual High Throughput Screening (vHTS) and Molecular Docking of phytochemicals from Nicotiana tabacum against the catalytic pockets of COX-1 and COX-2 were used to identify the lead bioactive(s) components of the plant. The hit phytosterols were isolated, histopathological examination of the stomach, in-vivo COX-1/COX-2 mRNAs expression patterns in the liver through reverse transcription-polymerase chain reactions, and enzymes activities of Nicotiana tabacum phytosterol isolates (NTPI) in HCl/ethanol-induced inflammation in Wistar rats were all investigated. Formation of hydrogen bonds favour selective inhibition of COX-2 while hydrophobic interactions favour selective inhibition of COX-1. NTPI demonstrates inhibition of COX-2 by down-regulate the expression of COX-2 mRNA and were ineffective against the expression COX-1 mRNA. NTPI demonstrates hepatoprotective abilities by improving the antioxidant defense system of the liver. Histopathological analyses show NTPI at 50 mg/kg bodyweight regenerates the parietal cells and maintain the gastrointestinal architecture. Drug likeness prediction and ADME toxicity screening show that phytosterols possess good oral bioavailability with no side effects. Phytosterols are selective inhibitors of COX-2, they are hepatoprotective, regenerate parietal cells, and non-toxic.
Collapse
Affiliation(s)
- O Adeboye Akinloye
- Department of Biochemistry, Phytomedicine, Phyto-chemistry and Bio-computing Research Laboratory, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - D Ibukun Akinloye
- Department of Biochemistry, Phytomedicine, Phyto-chemistry and Bio-computing Research Laboratory, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - S Bamidele Onigbinde
- Department of Biochemistry, Phytomedicine, Phyto-chemistry and Bio-computing Research Laboratory, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria
| | - D Samuel Metibemu
- Department of Biochemistry, Phytomedicine, Phyto-chemistry and Bio-computing Research Laboratory, College of Biosciences, Federal University of Agriculture, Abeokuta, Nigeria; Department of Biochemistry, Faculty of Science, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria.
| |
Collapse
|
22
|
Özbolat SN, Ayna A. Chrysin Suppresses HT-29 Cell Death Induced by Diclofenac through Apoptosis and Oxidative Damage. Nutr Cancer 2020; 73:1419-1428. [PMID: 32757685 DOI: 10.1080/01635581.2020.1801775] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Diclofenac (Dic) was shown to increase in reactive oxygen species (ROS) levels thereby resulting oxidative stress and apoptotic cell death in colon cancer. The antioxidants can prevent and repair oxidative damage caused by ROS. The aim of this study was to assess the effect of chrysin (Chr) on Dic-induced toxicity in HT-29 and molecular mechanisms underlying its effect. METHODS Cell proliferation and cytotoxicity assays were carried out by WST-1 and LDH leakage assay, apoptotic index was calculated by TUNEL Assay, antioxidant parameters were studied by measurement of ROS, LPO and TAS levels and catalase activity, expression of caspase-3 protein levels were analyzed by immunohistochemical staining, mRNA levels of apoptotic and anti-apoptotic genes were studied by qRT-PCR. RESULTS The cellular processes of Dic-triggered cell death was associated with increase in ROS, malondialdehyde levels and lactate dehydrogenase release, decrease in total antioxidant and catalase activity while pretreatment with Chr reversed these effects. The expression level of p53, cas-3, cas-8, Bax and cytochrome c increased in Dic-exposed group while they were reduced by Chr. CONCLUSION The use of antioxidant nutritional supplements, and in particular of Chr, may reduce the efficacy of Dic in inducing apoptosis of colon cancer cells.
Collapse
Affiliation(s)
- Seda Nur Özbolat
- Department of Chemistry, Faculty of Arts and Sciences, Bingol University, Bingol, Turkey
| | - Adnan Ayna
- Department of Chemistry, Faculty of Arts and Sciences, Bingol University, Bingol, Turkey
| |
Collapse
|
23
|
Owumi SE, Aliyu-Banjo NO, Odunola OA. Selenium attenuates diclofenac-induced testicular and epididymal toxicity in rats. Andrologia 2020; 52:e13669. [PMID: 32510627 DOI: 10.1111/and.13669] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
The adverse effect of diclofenac administration on the male reproductive organ in both humans and rats has been reported. Selenium, a trace element vital in nutrition, plays a significant part in cellular redox homeostasis, including male reproduction. However, the impact of selenium on male reproductive toxicity associated with diclofenac administration is lacking in the literature. The current investigation assessed the modulatory effects of selenium on diclofenac-mediated reproductive toxicity in rats. Rats were treated for fourteen consecutive days, either with diclofenac (10 mg/kg) or co-treated with selenium (0.125 and 0.25 mg/kg) body weight. Sperm parameters, enzymes of testicular function, luteinizing, follicle-stimulating hormone and testosterone were assessed in addition to oxidative stress indices and histopathological changes. Selenium significantly alleviated diclofenac-induced decreases in sperm count and motility, testicular function enzymes and levels of luteinizing hormone and testosterone in serum. Moreover, selenium co-administration at 0.125 and 0.25 mg/kg inhibited the diclofenac-induced decrease of antioxidant enzyme activities and increased oxidative stress parameters-lipid peroxidation, reactive nitrogen and oxygen species-in epididymis and testes of rats. Selenium (0.25 mg/kg) alone ameliorated diclofenac-mediated histological injuries in exposed rats. Collectively, selenium enhanced testicular and epididymal function in diclofenac-treated rats by suppressing nitrosative and oxidative stress in rats.
Collapse
Affiliation(s)
- Solomon E Owumi
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Nazirat O Aliyu-Banjo
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Oyeronke A Odunola
- Cancer Research and Molecular Biology Laboratories, Department of Biochemistry, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
24
|
Psomas G. Copper(II) and zinc(II) coordination compounds of non-steroidal anti-inflammatory drugs: Structural features and antioxidant activity. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213259] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
25
|
Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, Khattar E, Garg M. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol 2020; 68:258-278. [PMID: 32380233 DOI: 10.1016/j.semcancer.2020.04.006] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/20/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
Human malignancies are one of the major health-related issues though out the world and anticipated to rise in the future. The development of novel drugs/agents requires a huge amount of cost and time that represents a major challenge for drug discovery. In the last three decades, the number of FDA approved drugs has dropped down and this led to increasing interest in drug reposition or repurposing. The present review focuses on recent concepts and therapeutic opportunities for the utilization of antidiabetics, antibiotics, antifungal, anti-inflammatory, antipsychotic, PDE inhibitors and estrogen receptor antagonist, Antabuse, antiparasitic and cardiovascular agents/drugs as an alternative approach against human malignancies. The repurposing of approved non-cancerous drugs is an effective strategy to develop new therapeutic options for the treatment of cancer patients at an affordable cost in clinics. In the current scenario, most of the countries throughout the globe are unable to meet the medical needs of cancer patients because of the high cost of the available cancerous drugs. Some of these drugs displayed potential anti-cancer activity in preclinic and clinical studies by regulating several key molecular mechanisms and oncogenic pathways in human malignancies. The emerging pieces of evidence indicate that repurposing of drugs is crucial to the faster and cheaper discovery of anti-cancerous drugs.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India; Equal contribution
| | - Kavita Gala
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India; Equal contribution
| | - Stina George Fernandes
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India; Equal contribution
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India; Equal contribution
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Haryana, 122413, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India.
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India.
| |
Collapse
|
26
|
Dimiza F, Lazou M, Papadopoulos AN, Hatzidimitriou AG, Psomas G. Manganese(II) coordination compounds of carboxylate non-steroidal anti-inflammatory drugs. J Inorg Biochem 2020; 203:110906. [DOI: 10.1016/j.jinorgbio.2019.110906] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/30/2019] [Accepted: 10/24/2019] [Indexed: 02/06/2023]
|
27
|
Altindağ F, Rağbetli MÇ. The effect of maternal treatment with diclofenac sodium and thymoquinone on testicular parameters in rat offspring. Rev Int Androl 2019; 19:34-40. [PMID: 31628026 DOI: 10.1016/j.androl.2019.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 05/29/2019] [Accepted: 07/03/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION AND OBJECTIVE Diclofenac sodium (DS) can have toxic effects on various tissues and organs, as well as causing foetal and new-born malformations. Thymoquinone (TQ), the basic bioactive compound of black seed oil, is an antioxidant and antineoplastic substance. The aim of our study was to explore the effects of DS and TQ exposure during gestation on offspring rat testicular histology. MATERIALS AND METHODS Mother pregnant rats were divided into five groups: control, saline, DS, TQ and DS plus TQ (DS+TQ) four animals for each group. They were then treated as follows between day of 5 and 15 of gestation: the control group received no treatment. The saline group received physiological saline (1mg/kg/d) via the intraperitoneal (IP) route; the DS group received an intramuscular (IM) injection of DS (6.1mg/kg/d); the TQ group received TQ (5mg/kg/d) dissolved in drinking water; and the DS+TQ group received DS (6.1mg/kg/d) and TQ (5mg/kg/d) dissolved in water. After birth, the male rats were fed for four weeks, and at the end of this period offspring were sacrificed. Stereological methods, physical disector and Cavalieri principle were used for particle counting and volume estimation respectively. RESULTS The results revealed a significant decrease in the total number of Sertoli and Leydig cells in 4-week-old rats in the DS group (p<0.05), and TQ not have provide protection against this adverse effect of DS. CONCLUSIONS In this study, DS at a dose of 6.1mg/kg, equivalent to a dose of 1mg/kg in humans, decreased the number of Sertoli and Leydig cells, and TQ did not have a protective effect against the adverse effect of DS during the gestation period. These results show that new dose depend studies on TQ and DS interaction are requested to see protective effect of TQ.
Collapse
Affiliation(s)
- Fikret Altindağ
- Department of Histology and Embryology, Medical School, Van Yüzüncü Yıl University, Van, Turkey.
| | - Murat Çetin Rağbetli
- Department of Histology and Embryology, Medical School, Van Yüzüncü Yıl University, Van, Turkey
| |
Collapse
|
28
|
Batool SS, Gilani SR, Zainab SS, Tahir MN, Harrison WTA, Syed Q, Mazhar S. Synthesis and Structural Characterization of a Monomeric Mixed Ligand Copper(II) Complex Involving N,N,N′,N′-Tetramethylethylenediamine and Mefenamate. J STRUCT CHEM+ 2019. [DOI: 10.1134/s0022476619070187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
29
|
Slator C, Molphy Z, McKee V, Long C, Brown T, Kellett A. Di-copper metallodrugs promote NCI-60 chemotherapy via singlet oxygen and superoxide production with tandem TA/TA and AT/AT oligonucleotide discrimination. Nucleic Acids Res 2019; 46:2733-2750. [PMID: 29474633 PMCID: PMC5888725 DOI: 10.1093/nar/gky105] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 02/08/2018] [Indexed: 12/29/2022] Open
Abstract
In order to expand the current repertoire of cancer treatments and to help circumvent limitations associated with resistance, the identification of new metallodrugs with high potency and novel mechanisms of action is of significant importance. Here we present a class of di-copper(II) complex based on the synthetic chemical nuclease [Cu(Phen)2]+ (where Phen = 1,10-phenanthroline) that is selective against solid epithelial cancer cells from the National Cancer Institute's 60 human cell line panel (NCI-60). Two metallodrug leads are studied and in each case two [Cu(Phen)2]+ units are bridged by a dicarboxylate linker but the length and rigidity of the linkers differ distinctly. Both agents catalyze intracellular superoxide (O2•-) and singlet oxygen (1O2) formation with radical species mediating oxidative damage within nuclear DNA in the form of double strand breaks and to the mitochondria in terms of membrane depolarization. The complexes are effective DNA binders and can discriminate AT/AT from TA/TA steps of duplex DNA through induction of distinctive Z-like DNA or by intercalative interactions.
Collapse
Affiliation(s)
- Creina Slator
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Zara Molphy
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Vickie McKee
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Conor Long
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Tom Brown
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford OX1 3TA, UK
| | - Andrew Kellett
- School of Chemical Sciences and National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|
30
|
Adeyemi WJ, Omoniyi JA, Olayiwola A, Ibrahim M, Ogunyemi O, Olayaki LA. Elevated reproductive toxicity effects of diclofenac after withdrawal: Investigation of the therapeutic role of melatonin. Toxicol Rep 2019; 6:571-577. [PMID: 31293902 PMCID: PMC6595233 DOI: 10.1016/j.toxrep.2019.06.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 01/15/2023] Open
Abstract
Although there are several reports on the toxic actions of sodium diclofenac (DF), there is dearth information on its effect on the male reproductive system. Therefore, the study investigated the effects of DF and melatonin in male rats. Twenty rats were used in this study, which lasted for 6 weeks. The control group (vehicle treated) received normal saline (0.1 ml/day, p.o.). In the experimental groups, DF was administered during the first (group 2) and last (group 3) three weeks of the study. However, in group 4, melatonin was administered for 3 weeks, after 3 weeks of treatment with DF. DF and melatonin were administered at 1 and 10 mg/kg b.w./day (p.o.) respectively. The results showed that unlike melatonin, DF had no effect on gonadotrophins; however, it caused significant decreases in GNRH and testosterone, but a significant increase in prolactin. Melatonin attenuated the pro-antioxidant and pro-inflammatory effects of DF, which caused significant decreases in SOD, TAC, CAT, but significant elevations in LDH, MDA, uric acid and CRP. Moreover, the hormone reversed the adverse effect of DF on sperm count, sperm motility and sperm morphology. There were slight evidence of the precipitation of imbalance in lipid metabolism by DF and the antidyslipidaemic action of melatonin. Compared to DF, DF recovery showed more adverse effects on prolactin, testosterone, LDH, MDA, UA, CRP, semen parameters (except sperm motility), TC, LDL-c, HDL-c and phospholipid. The histological results agreed with the biochemical assays. In conclusion, the reproductive toxicity effects of DF seem to escalate after withdrawal; however, these effects could be attenuated by treatment with melatonin.
Collapse
Affiliation(s)
- Wale J Adeyemi
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Julius A Omoniyi
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Aluko Olayiwola
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Mariam Ibrahim
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olatinbo Ogunyemi
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Luqman A Olayaki
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
31
|
Manganese coordination compounds of mefenamic acid: In vitro screening and in silico prediction of biological activity. J Inorg Biochem 2019; 190:1-14. [DOI: 10.1016/j.jinorgbio.2018.09.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
|
32
|
Kumar S, Sharma RP, Venugopalan P, Ferretti V, Perontsis S, Psomas G. Copper(II) diclofenac complexes: Synthesis, structural studies and interaction with albumins and calf-thymus DNA. J Inorg Biochem 2018; 187:97-108. [DOI: 10.1016/j.jinorgbio.2018.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 06/30/2018] [Accepted: 07/19/2018] [Indexed: 12/24/2022]
|
33
|
Geromichalos GD, Tarushi A, Lafazanis K, Pantazaki AA, Kessissoglou DP, Psomas G. In vitro and in silico study of the biological activity of manganese(III) inverse-[9-MC-3]-metallacrowns and manganese(II) complexes with the anti-inflammatory drugs diclofenac or indomethacin. J Inorg Biochem 2018; 187:41-55. [PMID: 30055395 DOI: 10.1016/j.jinorgbio.2018.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 05/02/2018] [Accepted: 07/18/2018] [Indexed: 12/23/2022]
Abstract
In the present contribution, the biological properties of four manganese complexes with the non-steroidal anti-inflammatory drugs sodium diclofenac (Nadicl) or indomethacin (Hindo) in the presence or absence of salicylaldoxime (Η2sao), i.e. [Μn6(O)2(dicl)2(sao)6(CH3OH)6] 1, [Μn6(O)2(indo)2(sao)6(H2O)4], 2, [Μn(dicl)2(CH3OH)4], 3, and [Μn(indo)2(CH3OH)4], 4 are presented. More specifically, the in vitro cytotoxic effects of the complexes were evaluated against three cancer cell lines (HeLa, MCF-7 and A549 cells) as well as their combinatory activity with the well-known chemotherapeutic drugs irinotecan, cisplatin, paclitaxel and 5-fluorouracil. The biological activity of the complexes was investigated in vitro by studying their affinity to calf-thymus DNA and their binding towards bovine or human serum albumin (HSA). Molecular docking simulations on the crystal structure of HSA and human estrogen receptor alpha (hERa) were employed in order to study in silico the ability of the studied complexes to bind to these proteins.
Collapse
Affiliation(s)
- George D Geromichalos
- Laboratory of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece; Cell Culture, Molecular Modeling and Drug Design Lab, Symeonidion Research Center, Theagenion Cancer Hospital, Thessaloniki GR-54007, Greece
| | - Alketa Tarushi
- Laboratory of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Konstantinos Lafazanis
- Cell Culture, Molecular Modeling and Drug Design Lab, Symeonidion Research Center, Theagenion Cancer Hospital, Thessaloniki GR-54007, Greece; Laboratory of Biochemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Anastasia A Pantazaki
- Laboratory of Biochemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Dimitris P Kessissoglou
- Laboratory of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - George Psomas
- Laboratory of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece.
| |
Collapse
|
34
|
Ramachandran A, Visschers RGJ, Duan L, Akakpo JY, Jaeschke H. Mitochondrial dysfunction as a mechanism of drug-induced hepatotoxicity: current understanding and future perspectives. J Clin Transl Res 2018. [PMID: 30873497 PMCID: PMC6261533 DOI: 10.18053/jctres.04.201801.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are critical cellular organelles for energy generation and are now also recognized as playing important roles in cellular signaling. Their central role in energy metabolism, as well as their high abundance in hepatocytes, make them important targets for drug-induced hepatotoxicity. This review summarizes the current mechanistic understanding of the role of mitochondria in drug-induced hepatotoxicity caused by acetaminophen, diclofenac, anti-tuberculosis drugs such as rifampin and isoniazid, anti-epileptic drugs such as valproic acid and constituents of herbal supplements such as pyrrolizidine alkaloids. The utilization of circulating mitochondrial-specific biomarkers in understanding mechanisms of toxicity in humans will also be examined. In summary, it is well-established that mitochondria are central to acetaminophen-induced cell death. However, the most promising areas for clinically useful therapeutic interventions after acetaminophen toxicity may involve the promotion of adaptive responses and repair processes including mitophagy and mitochondrial biogenesis, In contrast, the limited understanding of the role of mitochondria in various aspects of hepatotoxicity by most other drugs and herbs requires more detailed mechanistic investigations in both animals and humans. Development of clinically relevant animal models and more translational studies using mechanistic biomarkers are critical for progress in this area. Relevance for patients:This review focuses on the role of mitochondrial dysfunction in liver injury mechanisms of clinically important drugs like acetaminophen, diclofenac, rifampicin, isoniazid, amiodarone and others. A better understanding ofthe mechanisms in animal models and their translation to patients will be critical for the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ruben G J Visschers
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Luqi Duan
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jephte Y Akakpo
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
35
|
Tarushi A, Geromichalos GD, Lafazanis K, Raptopoulou CP, Psycharis V, Lalioti N, Pantazaki AA, Kessissoglou DP, Tangoulis V, Psomas G. A step-ladder manganese(iii) metallacrown hosting mefenamic acid and a manganese(ii)–mefanamato complex: synthesis, characterization and cytotoxic activity. NEW J CHEM 2018. [DOI: 10.1039/c8nj01182f] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Manganese complexes of mefenamic acid in the presence or absence of salicylaldoxime have been evaluated for their cytotoxic activity.
Collapse
Affiliation(s)
- Alketa Tarushi
- Laboratory of Inorganic Chemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| | - George D. Geromichalos
- Laboratory of Inorganic Chemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| | - Kostas Lafazanis
- Cell Culture
- Molecular Modeling and Drug Design Lab
- Symeonidion Research Center
- Theagenion Cancer Hospital
- Thessaloniki GR-54007
| | - Catherine P. Raptopoulou
- Institute of Nanoscience and Nanotechnology
- NCSR “Demokritos”
- GR-15310 Aghia Paraskevi Attikis
- Greece
| | - Vassilis Psycharis
- Institute of Nanoscience and Nanotechnology
- NCSR “Demokritos”
- GR-15310 Aghia Paraskevi Attikis
- Greece
| | - Nikolia Lalioti
- Department of Chemistry
- University of Patras
- GR-26504 Patras
- Greece
| | - Anastasia A. Pantazaki
- Laboratory of Biochemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| | - Dimitris P. Kessissoglou
- Laboratory of Inorganic Chemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| | | | - George Psomas
- Laboratory of Inorganic Chemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| |
Collapse
|
36
|
Dimiza F, Raptopoulou CP, Psycharis V, Papadopoulos AN, Psomas G. Manganese(ii) complexes with the non-steroidal anti-inflammatory drugs naproxen and mefenamic acid: synthesis, structure, antioxidant capacity, and interaction with albumins and DNA. NEW J CHEM 2018. [DOI: 10.1039/c8nj03226b] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The structure, antioxidant activity and interaction with DNA and albumins of a series of manganese(ii) complexes of mefenamic acid or naproxen are presented herein.
Collapse
Affiliation(s)
- Filitsa Dimiza
- Department of General and Inorganic Chemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| | - Catherine P. Raptopoulou
- Institute of Nanoscience and Nanotechnology
- NCSR “Demokritos”
- GR-15310 Aghia Paraskevi Attikis
- Greece
| | - Vassilis Psycharis
- Institute of Nanoscience and Nanotechnology
- NCSR “Demokritos”
- GR-15310 Aghia Paraskevi Attikis
- Greece
| | - Athanasios N. Papadopoulos
- Department of Nutrition and Dietetics
- Faculty of Food Technology and Nutrition
- Alexandrion Technological Educational Institution
- Sindos
- Thessaloniki
| | - George Psomas
- Department of General and Inorganic Chemistry
- Faculty of Chemistry
- Aristotle University of Thessaloniki
- GR-54124 Thessaloniki
- Greece
| |
Collapse
|
37
|
Nishi K, Iwaihara Y, Tsunoda T, Doi K, Sakata T, Shirasawa S, Ishikura S. ROS-induced cleavage of NHLRC2 by caspase-8 leads to apoptotic cell death in the HCT116 human colon cancer cell line. Cell Death Dis 2017; 8:3218. [PMID: 29242562 PMCID: PMC5870588 DOI: 10.1038/s41419-017-0006-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 12/14/2022]
Abstract
Excess production of reactive oxygen species (ROS) is known to cause apoptotic cell death. However, the molecular mechanisms whereby ROS induce apoptosis remain elusive. Here we show that the NHL-repeat-containing protein 2 (NHLRC2) thioredoxin-like domain protein is cleaved by caspase-8 in ROS-induced apoptosis in the HCT116 human colon cancer cell line. Treatment of HCT116 cells with the oxidant tert-butyl hydroperoxide (tBHP) induced apoptosis and reduced NHLRC2 protein levels, whereas pretreatment with the antioxidant N-acetyl-l-cysteine prevented apoptosis and the decrease in NHLRC2 protein levels seen in tBHP-treated cells. Furthermore, the ROS-induced decrease in NHLRC2 protein levels was relieved by the caspase inhibitor z-VAD-fmk. We found that the thioredoxin-like domain of NHLRC2 interacted with a proenzyme form of caspase-8, and that caspase-8 cleaved NHLRC2 protein at Asp580 in vitro. Furthermore, siRNA-mediated knockdown of caspase-8 blocked the ROS-induced decrease in NHLRC2 protein levels. Both shRNA and CRISPR-Cas9-mediated loss of NHLRC2 resulted in an increased susceptibility of HCT116 cells to ROS-induced apoptosis. These results suggest that excess ROS production causes a caspase-8-mediated decrease in NHLRC2 protein levels, leading to apoptotic cell death in colon cancer cells, and indicate an important role of NHLRC2 in the regulation of ROS-induced apoptosis.
Collapse
Affiliation(s)
- Kensuke Nishi
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan.,Department of Otorhinolaryngology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Yuri Iwaihara
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Toshiyuki Tsunoda
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan.,Center for Advanced Molecular Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Keiko Doi
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan.,Center for Advanced Molecular Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Toshifumi Sakata
- Department of Otorhinolaryngology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Senji Shirasawa
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan.,Center for Advanced Molecular Medicine, Fukuoka University, Fukuoka, 814-0180, Japan
| | - Shuhei Ishikura
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180, Japan. .,Center for Advanced Molecular Medicine, Fukuoka University, Fukuoka, 814-0180, Japan.
| |
Collapse
|
38
|
Hu WL, Dong HY, Li Y, Ojcius DM, Li SJ, Yan J. Bid-Induced Release of AIF/EndoG from Mitochondria Causes Apoptosis of Macrophages during Infection with Leptospira interrogans. Front Cell Infect Microbiol 2017; 7:471. [PMID: 29184851 PMCID: PMC5694448 DOI: 10.3389/fcimb.2017.00471] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/27/2017] [Indexed: 12/20/2022] Open
Abstract
Leptospirosis is a global zoonotic infectious disease caused by pathogenic Leptospira species. Leptospire-induced macrophage apoptosis through the Fas/FasL-caspase-8/3 pathway plays an important role in the survival and proliferation of the pathogen in hosts. Although, the release of mitochondrial apoptosis-inducing factor (AIF) and endonuclease G (EndoG) in leptospire-infected macrophages has been described, the mechanisms linking caspase and mitochondrion-related host-cell apoptosis has not been determined. Here, we demonstrated that leptospire-infection induced apoptosis through mitochondrial damages in macrophages. Apoptosis was caused by the mitochondrial release and nuclear translocation of AIF and/or EndoG, leading to nuclear DNA fragmentation. However, the mitochondrion-related CytC-caspase-9/3 pathway was not activated. Next, we found that the release and translocation of AIF and/or EndoG was preceded by the activation of the BH3-interacting domain death agonist (Bid). Furthermore, our data demonstrated that caspase-8 was activated during the infection and caused the activation of Bid. Meanwhile, high reactive oxygen species (ROS) trigged by the infection caused the dephosphorylation of Akt, which also activated Bid. In conclusion, Bid-mediated mitochondrial release of AIF and/or EndoG followed by nuclear translocation is a major mechanism of leptospire- induced apoptosis in macrophages, and this process is modulated by both caspase-8 and ROS-Akt signal pathways.
Collapse
Affiliation(s)
- Wei-Lin Hu
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hai-Yan Dong
- Department of Medical Microbiology and Immunology, Wenzhou Medical University, Wenzhou, China
| | - Yang Li
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, CA, United States
| | - Shi-Jun Li
- Institute of Communicable Disease Control and Prevention, Guizhou Provincial Centre for Disease Control and Prevention, Guiyang, China
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China.,Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
39
|
Tarushi A, Raptopoulou CP, Psycharis V, Kessissoglou DP, Papadopoulos AN, Psomas G. Interaction of zinc(II) with the non-steroidal anti-inflammatory drug niflumic acid. J Inorg Biochem 2017; 176:100-112. [PMID: 28886446 DOI: 10.1016/j.jinorgbio.2017.08.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/16/2017] [Accepted: 08/28/2017] [Indexed: 12/20/2022]
Abstract
The reaction of ZnCl2 with the non-steroidal anti-inflammatory drug niflumic acid (Hnif) resulted in the formation of complex [Zn(nif-O)2(MeOH)4], 1. When this reaction was performed in the presence of a N,N'-donor heterocyclic ligand such as 2,2'-bipyridine (bipy), 2,2'-bipyridylamine (bipyam), 1,10-phenanthroline (phen) and 2,2'-dipyridylketone oxime (Hpko), the complexes [Zn(nif-O,O')(bipy)Cl], 2, [Zn(nif-O)(nif-O,O')2(bipyam)], 3, [Zn(nif-O,O')2(phen)], 4 and [Zn(nif-O)2(Hpko-N,N')2], 5 were formed, respectively. The complexes were characterized by physicochemical and spectroscopic techniques and X-ray crystallography (for complexes 1-3). The complexes can scavenge 1,1-diphenyl-picrylhydrazyl, 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) and hydroxyl radicals, may inhibit soybean lipoxygenase and are more active compounds than free Hnif. The interaction of the complexes with serum albumins was monitored by fluorescence emission spectroscopy and the corresponding binding constants were calculated. The affinity of the complexes with calf-thymus DNA was investigated by UV-vis spectroscopy, viscosity measurements and fluorescence emission spectroscopy for the competitive studies of the complexes with ethidium bromide revealing their interaction probably via intercalation.
Collapse
Affiliation(s)
- Alketa Tarushi
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, P.O. Box 135, GR-54124 Thessaloniki, Greece
| | - Catherine P Raptopoulou
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", GR-15310, Aghia Paraskevi, Attikis, Greece
| | - Vassilis Psycharis
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", GR-15310, Aghia Paraskevi, Attikis, Greece
| | - Dimitris P Kessissoglou
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, P.O. Box 135, GR-54124 Thessaloniki, Greece
| | - Athanasios N Papadopoulos
- Department of Nutrition and Dietetics, Faculty of Food Technology and Nutrition, Alexandrion Technological Educational Institution, Sindos, Thessaloniki, Greece
| | - George Psomas
- Department of General and Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, P.O. Box 135, GR-54124 Thessaloniki, Greece.
| |
Collapse
|
40
|
Tarushi A, Hatzidimitriou AG, Estrader M, Kessissoglou DP, Tangoulis V, Psomas G. Toward Multifunctional Materials Incorporating Stepladder Manganese(III) Inverse-[9-MC-3]-Metallacrowns and Anti-Inflammatory Drugs. Inorg Chem 2017; 56:7048-7057. [DOI: 10.1021/acs.inorgchem.7b00655] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Alketa Tarushi
- Laboratory
of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Antonios G. Hatzidimitriou
- Laboratory
of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | - Marta Estrader
- Departament
de Química Inorgànica, Universitat de Barcelona, Diagonal
645, 08028 Barcelona, Spain
| | - Dimitris P. Kessissoglou
- Laboratory
of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| | | | - George Psomas
- Laboratory
of Inorganic Chemistry, Faculty of Chemistry, Aristotle University of Thessaloniki, GR-54124 Thessaloniki, Greece
| |
Collapse
|
41
|
Al-Attrache H, Chamieh H, Hamzé M, Morel I, Taha S, Abdel-Razzak Z. N-acetylcysteine potentiates diclofenac toxicity in Saccharomyces cerevisiae: stronger potentiation in ABC transporter mutant strains. Drug Chem Toxicol 2017; 41:89-94. [DOI: 10.1080/01480545.2017.1320404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Houssein Al-Attrache
- Laboratory of Applied Biotechnology: Biomolecules, LBA3B – AZM Center, Tripoli, Lebanon
- Faculty of Pharmacy, Inserm U991, Rennes, France
- Rennes 1 University, Faculty of Pharmacy, Rennes, France
| | - Hala Chamieh
- Laboratory of Applied Biotechnology: Biomolecules, LBA3B – AZM Center, Tripoli, Lebanon
| | - Monzer Hamzé
- Laboratory of Medical Microbiology, AZM Center, Tripoli, Lebanon
| | - Isabelle Morel
- Faculty of Pharmacy, Inserm U991, Rennes, France
- Rennes 1 University, Faculty of Pharmacy, Rennes, France
- Laboratory of Emergency and Intensive Care, Pontchaillou Hospital, Rennes, France
| | - Samir Taha
- Laboratory of Applied Biotechnology: Biomolecules, LBA3B – AZM Center, Tripoli, Lebanon
| | - Ziad Abdel-Razzak
- Laboratory of Applied Biotechnology: Biomolecules, LBA3B – AZM Center, Tripoli, Lebanon
| |
Collapse
|
42
|
Zinc complexes of diflunisal: Synthesis, characterization, structure, antioxidant activity, and in vitro and in silico study of the interaction with DNA and albumins. J Inorg Biochem 2017; 170:85-97. [DOI: 10.1016/j.jinorgbio.2017.02.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 01/13/2017] [Accepted: 02/10/2017] [Indexed: 12/12/2022]
|
43
|
Amanullah A, Upadhyay A, Chhangani D, Joshi V, Mishra R, Yamanaka K, Mishra A. Proteasomal Dysfunction Induced By Diclofenac Engenders Apoptosis Through Mitochondrial Pathway. J Cell Biochem 2017; 118:1014-1027. [DOI: 10.1002/jcb.25666] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 08/01/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Arun Upadhyay
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Deepak Chhangani
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Ribhav Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology Research Institute of Environmental Medicine; Nagoya University Furo-cho; Chikusa-ku Nagoya 464-8601 Japan
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit; Indian Institute of Technology Jodhpur; Rajasthan 342011 India
| |
Collapse
|
44
|
Ghosh R, Hwang SM, Cui Z, Gilda JE, Gomes AV. Different effects of the nonsteroidal anti-inflammatory drugs meclofenamate sodium and naproxen sodium on proteasome activity in cardiac cells. J Mol Cell Cardiol 2016; 94:131-144. [PMID: 27049794 DOI: 10.1016/j.yjmcc.2016.03.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/10/2016] [Accepted: 03/28/2016] [Indexed: 02/06/2023]
Abstract
The use of nonsteroidal anti-inflammatory drugs (NSAIDs) like meclofenamate sodium (MS), used to reduce pain, has been associated with an increased risk of cardiovascular disease (CVD). Naproxen (NAP), another NSAID, is not associated with increased risk of CVD. The molecular mechanism(s) by which NSAIDs induce CVD is unknown. We investigated the effects of MS and NAP on protein homeostasis and cardiotoxicity in rat cardiac H9c2 cells and murine neonatal cardiomyocytes. MS, but not NAP, significantly inhibited proteasome activity and reduced cardiac cell viability at pharmacological levels found in humans. Although proteasome subunit gene and protein expression were unaffected by NSAIDs, MS treated cell lysates showed higher 20S proteasome content, while purified proteasomes from MS treated cells had lower proteasome activity and higher levels of oxidized subunits than proteasomes from control cells. Addition of exogenous proteasome to MS treated cells improved cell viability. Both MS and NAP increased ROS production, but the rate of ROS production was greater in MS than in NAP treated cells. The ROS production is likely from mitochondria, as MS inhibited mitochondrial Complexes I and III, major sources of ROS, while NAP inhibited Complex I. MS also impaired mitochondrial membrane potential while NAP did not. Antioxidants were able to prevent the reduced cell viability caused by MS treatment. These results suggest that NSAIDs induce cardiotoxicity by a ROS dependent mechanism involving mitochondrial and proteasome dysfunction and may explain why some NSAIDs should not be given to patients for long periods.
Collapse
Affiliation(s)
- Rajeshwary Ghosh
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, United States
| | - Soyun M Hwang
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, United States
| | - Ziyou Cui
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, United States
| | - Jennifer E Gilda
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, United States
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, United States; Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, United States.
| |
Collapse
|
45
|
Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-diclofenac as an anti-cancer agent. Ecancermedicalscience 2016; 10:610. [PMID: 26823679 PMCID: PMC4720497 DOI: 10.3332/ecancer.2016.610] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Diclofenac (DCF) is a well-known and widely used non-steroidal anti-inflammatory drug (NSAID), with a range of actions which are of interest in an oncological context. While there has long been an interest in the use of NSAIDs in chemoprevention, there is now emerging evidence that such drugs may have activity in a treatment setting. DCF, which is a potent inhibitor of COX-2 and prostaglandin E2 synthesis, displays a range of effects on the immune system, the angiogenic cascade, chemo- and radio-sensitivity and tumour metabolism. Both pre-clinical and clinical evidence of these effects, in multiple cancer types, is assessed and summarised and relevant mechanisms of action outlined. Based on this evidence the case is made for further clinical investigation of the anticancer effects of DCF, particularly in combination with other agents - with a range of possible multi-drug and multi-modality combinations outlined in the supplementary materials accompanying the main paper.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | | | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc; Newton MA 02459, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
46
|
Ghosh R, Alajbegovic A, Gomes AV. NSAIDs and Cardiovascular Diseases: Role of Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:536962. [PMID: 26457127 PMCID: PMC4592725 DOI: 10.1155/2015/536962] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/24/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most commonly used drugs worldwide. NSAIDs are used for a variety of conditions including pain, rheumatoid arthritis, and musculoskeletal disorders. The beneficial effects of NSAIDs in reducing or relieving pain are well established, and other benefits such as reducing inflammation and anticancer effects are also documented. The undesirable side effects of NSAIDs include ulcers, internal bleeding, kidney failure, and increased risk of heart attack and stroke. Some of these side effects may be due to the oxidative stress induced by NSAIDs in different tissues. NSAIDs have been shown to induce reactive oxygen species (ROS) in different cell types including cardiac and cardiovascular related cells. Increases in ROS result in increased levels of oxidized proteins which alters key intracellular signaling pathways. One of these key pathways is apoptosis which causes cell death when significantly activated. This review discusses the relationship between NSAIDs and cardiovascular diseases (CVD) and the role of NSAID-induced ROS in CVD.
Collapse
Affiliation(s)
- Rajeshwary Ghosh
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Azra Alajbegovic
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Aldrin V. Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA
| |
Collapse
|
47
|
A novel bioactive Cd(II) polymeric complex with mefenamic acid: Synthesis, crystal structure and biological evaluations. Inorganica Chim Acta 2015. [DOI: 10.1016/j.ica.2015.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
48
|
Behera B, Mishra D, Roy B, Devi KSP, Narayan R, Das J, Ghosh SK, Maiti TK. Abrus precatorius agglutinin-derived peptides induce ROS-dependent mitochondrial apoptosis through JNK and Akt/P38/P53 pathways in HeLa cells. Chem Biol Interact 2014; 222:97-105. [DOI: 10.1016/j.cbi.2014.08.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 07/17/2014] [Accepted: 08/28/2014] [Indexed: 01/14/2023]
|
49
|
Schwake M, Günes D, Köchling M, Brentrup A, Schroeteler J, Hotfilder M, Fruehwald MC, Stummer W, Ewelt C. Kinetics of porphyrin fluorescence accumulation in pediatric brain tumor cells incubated in 5-aminolevulinic acid. Acta Neurochir (Wien) 2014; 156:1077-84. [PMID: 24777761 DOI: 10.1007/s00701-014-2096-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 04/10/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Fluorescence-guided surgery with 5-aminolevulinic acid (5-ALA) enables more complete resections of tumors in adults. 5-ALA elicits accumulation of fluorescent porphyrins in various cancerous tissues, which can be visualized using a modified neurosurgical microscope with blue light. Although this technique is well established in adults, it has not been investigated systematically in pediatric brain tumors. Specifically, it is unknown how quickly, how long, and to what extent various pediatric tumors accumulate fluorescence. The purpose of this study was to determine utility and time course of 5-ALA-induced fluorescence in typical pediatric brain tumors in vitro. METHODS Cell cultures of medulloblastoma [DAOY and UW228], cPNET [PFSK] atypical teratoid rhabdoid tumor [BT16] and ependymoma [RES196] were incubated with 5-ALA for either 60 minutes or continuously. Porphyrin fluorescence intensities were determined using a fluorescence-activated cell sorter (FACS) after 1, 3, 6, 9, 12 and 24 hours. C6 and U87 cells served as controls. RESULTS All pediatric brain tumor cell lines displayed fluorescence compared to their respective controls without 5-ALA (p < 0.05). Sixty minutes of incubation resulted in peaks between 3 and 6 hours, whereas continuous incubation resulted in peaks at 12 hours or beyond. 60 minute incubation peak levels were between 52 and 91 % of maxima achieved with continuous incubation. Accumulation and clearance varied between cell types. CONCLUSIONS We demonstrate that 5-ALA exposure of cell lines derived from typical pediatric central nervous system (CNS) tumors induces accumulation of fluorescent porphyrins. Differences in uptake and clearance indicate that different application modes may be necessary for fluorescence-guided resection, depending on tumor type.
Collapse
Affiliation(s)
- Michael Schwake
- Department of Neurosurgery, University Hospital, Albert-Schweitzer-Campus 1, Gebäude A1, D-48149, Münster, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chakraborty S, Bose M, Sarkar M. Spectroscopic studies of the binding of Cu(II) complexes of oxicam NSAIDs to alternating G-C and homopolymeric G-C sequences. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2014; 122:690-697. [PMID: 24345609 DOI: 10.1016/j.saa.2013.11.078] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 11/12/2013] [Accepted: 11/13/2013] [Indexed: 06/03/2023]
Abstract
Drugs belonging to the Non-steroidal anti-inflammatory (NSAID) group are not only used as anti-inflammatory, analgesic and anti-pyretic agents, but also show anti-cancer effects. Complexing them with a bioactive metal like copper, show an enhancement in their anti-cancer effects compared to the bare drugs, whose exact mechanism of action is not yet fully understood. For the first time, it was shown by our group that Cu(II)-NSAIDs can directly bind to the DNA backbone. The ability of the copper complexes of NSAIDs namely meloxicam and piroxicam to bind to the DNA backbone could be a possible molecular mechanism behind their enhanced anticancer effects. Elucidating base sequence specific interaction of Cu(II)-NSAIDs to the DNA will provide information on their possible binding sites in the genome sequence. In this work, we present how these complexes respond to differences in structure and hydration pattern of GC rich sequences. For this, binding studies of Cu(II) complexes of piroxicam [Cu(II)-(Px)2 (L)2] and meloxicam [Cu(II)-(Mx)2 (L)] with alternating GC (polydG-dC) and homopolymeric GC (polydG-polydC) sequences were carried out using a combination of spectroscopic techniques that include UV-Vis absorption, fluorescence and circular dichroism (CD) spectroscopy. The Cu(II)-NSAIDs show strong binding affinity to both polydG-dC and polydG-polydC. The role reversal of Cu(II)-meloxicam from a strong binder of polydG-dC (Kb=11.5×10(3) M(-1)) to a weak binder of polydG-polydC (Kb=5.02×10(3) M(-1)), while Cu(II)-piroxicam changes from a strong binder of polydG-polydC (Kb=8.18×10(3) M(-1)) to a weak one of polydG-dC (Kb=2.18×10(3) M(-1)), point to the sensitivity of these complexes to changes in the backbone structures/hydration. Changes in the profiles of UV absorption band and CD difference spectra, upon complex binding to polynucleotides and the results of competitive binding assay using ethidium bromide (EtBr) fluorescence indicate different binding modes in each case.
Collapse
Affiliation(s)
- Sreeja Chakraborty
- Chemical Sciences Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | - Madhuparna Bose
- Department of Biochemistry, Ballygunge Science College, University of Calcutta, Kolkata, India
| | - Munna Sarkar
- Chemical Sciences Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India.
| |
Collapse
|