1
|
D’Amico M, De Amicis F. Challenges of Regulated Cell Death: Implications for Therapy Resistance in Cancer. Cells 2024; 13:1083. [PMID: 38994937 PMCID: PMC11240625 DOI: 10.3390/cells13131083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Regulated cell death, a regulatory form of cell demise, has been extensively studied in multicellular organisms. It plays a pivotal role in maintaining organismal homeostasis under normal and pathological conditions. Although alterations in various regulated cell death modes are hallmark features of tumorigenesis, they can have divergent effects on cancer cells. Consequently, there is a growing interest in targeting these mechanisms using small-molecule compounds for therapeutic purposes, with substantial progress observed across various human cancers. This review focuses on summarizing key signaling pathways associated with apoptotic and autophagy-dependent cell death. Additionally, it explores crucial pathways related to other regulated cell death modes in the context of cancer. The discussion delves into the current understanding of these processes and their implications in cancer treatment, aiming to illuminate novel strategies to combat therapy resistance and enhance overall cancer therapy.
Collapse
Affiliation(s)
- Maria D’Amico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
- Health Center, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
2
|
Wang Y, Chen Y, Zhang J, Yang Y, Fleishman JS, Wang Y, Wang J, Chen J, Li Y, Wang H. Cuproptosis: A novel therapeutic target for overcoming cancer drug resistance. Drug Resist Updat 2024; 72:101018. [PMID: 37979442 DOI: 10.1016/j.drup.2023.101018] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/20/2023]
Abstract
Cuproptosis is a newly identified form of cell death driven by copper. Recently, the role of copper and copper triggered cell death in the pathogenesis of cancers have attracted attentions. Cuproptosis has garnered enormous interest in cancer research communities because of its great potential for cancer therapy. Copper-based treatment exerts an inhibiting role in tumor growth and may open the door for the treatment of chemotherapy-insensitive tumors. In this review, we provide a critical analysis on copper homeostasis and the role of copper dysregulation in the development and progression of cancers. Then the core molecular mechanisms of cuproptosis and its role in cancer is discussed, followed by summarizing the current understanding of copper-based agents (copper chelators, copper ionophores, and copper complexes-based dynamic therapy) for cancer treatment. Additionally, we summarize the emerging data on copper complexes-based agents and copper ionophores to subdue tumor chemotherapy resistance in different types of cancers. We also review the small-molecule compounds and nanoparticles (NPs) that may kill cancer cells by inducing cuproptosis, which will shed new light on the development of anticancer drugs through inducing cuproptosis in the future. Finally, the important concepts and pressing questions of cuproptosis in future research that should be focused on were discussed. This review article suggests that targeting cuproptosis could be a novel antitumor therapy and treatment strategy to overcome cancer drug resistance.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China.
| | - Yongming Chen
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, PR China
| | - Junjing Zhang
- Department of Hepato-Biliary Surgery, Department of Surgery, Huhhot First Hospital, Huhhot 010030, PR China
| | - Yihui Yang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yan Wang
- Hunan Provincial Key Laboratory of Hepatobiliary Disease Research & Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, The Second Xiangya Hospital of Central South University, Changsha 410011, PR China
| | - Jinhua Wang
- Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, PR China
| | - Jichao Chen
- Department of Respiratory and Critical Care Medicine, Aerospace Center Hospital, Peking University Aerospace School of Clinical Medicine, Beijing 100049, PR China
| | - Yuanfang Li
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Guangzhou, PR China.
| | - Hongquan Wang
- Department of Neurology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, PR China.
| |
Collapse
|
3
|
Hassan I, Ebaid H, Alhazza IM, Al-Tamimi J, Rady AM. Disulfiram Enhances the Antineoplastic Activity and Sensitivity of Murine Hepatocellular Carcinoma to 5-FU via Redox Management. Pharmaceuticals (Basel) 2023; 16:169. [PMID: 37259318 PMCID: PMC9967644 DOI: 10.3390/ph16020169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 11/20/2023] Open
Abstract
The efficacy of anticancer drug 5-FU is suppressed due to various factors, including severe side effects and decreased insensitivity during prolonged chemotherapy. Elevated endogenous copper (Cu) levels are one of the prominent hallmark features of cancer cells. In the present investigation, this feature was targeted in diethyl nitrosamine-phenobarbital-induced hepatocellular carcinoma (HCC) in a rat model system by an established anticancer drug, 5-FU, co-administered with copper and its chelating agent, disulfiram. After treatment with the test chemicals in HCC-induced rats, blood and liver samples were subjected to biochemical, molecular, and histopathological analyses. The analysis revealed that reactive oxygen species-mediated oxidative stress is the crucial etiological reason for the pathogenesis of HCC in rats, as evidenced by the significantly compromised activity of major antioxidant enzymes and elevated levels of oxidative damaged products with major histological alterations compared to the control. However, the combination of 5-FU with DSF demonstrated a significant improvement in most of the parameters, followed by 5-FU-Cu in the combination-treated groups. The combination treatment improved the histological details and triggered apoptosis in the cancer cells to a remarkable extent, as the levels of cleaved PARP and caspase-3 were significantly higher than those in the HCC rats treated with the drug alone. The present study envisages that manipulating the Cu-level greatly enhances the antineoplastic activity of 5-FU and sensitizes cancer cells to the increased efficacy of the drug.
Collapse
Affiliation(s)
| | | | - Ibrahim M. Alhazza
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | | | | |
Collapse
|
4
|
Khan MS, Alomari A, Tabrez S, Hassan I, Wahab R, Bhat SA, Alafaleq NO, Altwaijry N, Shaik GM, Zaidi SK, Nouh W, Alokail MS, Ismael MA. Anticancer Potential of Biogenic Silver Nanoparticles: A Mechanistic Study. Pharmaceutics 2021; 13:pharmaceutics13050707. [PMID: 34066092 PMCID: PMC8151171 DOI: 10.3390/pharmaceutics13050707] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 01/18/2023] Open
Abstract
The continuous loss of human life due to the paucity of effective drugs against different forms of cancer demands a better/noble therapeutic approach. One possible way could be the use of nanostructures-based treatment methods. In the current piece of work, we have synthesized silver nanoparticles (AgNPs) using plant (Heliotropiumbacciferum) extract using AgNO3 as starting materials. The size, shape, and structure of synthesized AgNPs were confirmed by various spectroscopy and microscopic techniques. The average size of biosynthesized AgNPs was found to be in the range of 15 nm. The anticancer potential of these AgNPs was evaluated by a battery of tests such as MTT, scratch, and comet assays in breast (MCF-7) and colorectal (HCT-116) cancer models. The toxicity of AgNPs towards cancer cells was confirmed by the expression pattern of apoptotic (p53, Bax, caspase-3) and antiapoptotic (BCl-2) genes by RT-PCR. The cell viability assay showed an IC50 value of 5.44 and 9.54 µg/mL for AgNPs in MCF-7 and HCT-116 cell lines respectively. We also observed cell migration inhibiting potential of AgNPs in a concentration-dependent manner in MCF-7 cell lines. A tremendous rise (150–250%) in the production of ROS was observed as a result of AgNPs treatment compared with control. Moreover, the RT-PCR results indicated the difference in expression levels of pro/antiapoptotic proteins in both cancer cells. All these results indicate that cell death observed by us is mediated by ROS production, which might have altered the cellular redox status. Collectively, we report the antimetastasis potential of biogenic synthesized AgNPs against breast and colorectal cancers. The biogenic synthesis of AgNPs seems to be a promising anticancer therapy with greater efficacy against the studied cell lines.
Collapse
Affiliation(s)
- Mohd Shahnawaz Khan
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (N.O.A.); (N.A.); (G.M.S.); (M.S.A.); (M.A.I.)
- Correspondence:
| | - Alya Alomari
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (N.O.A.); (N.A.); (G.M.S.); (M.S.A.); (M.A.I.)
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Iftekhar Hassan
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (I.H.); (R.W.)
| | - Rizwan Wahab
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (I.H.); (R.W.)
| | - Sheraz Ahmad Bhat
- Department of Biochemistry, New Science Block, SP College, Cluster University, Srinagar, Jammu and Kashmir 190008, India;
| | - Nouf Omar Alafaleq
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (N.O.A.); (N.A.); (G.M.S.); (M.S.A.); (M.A.I.)
| | - Nojood Altwaijry
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (N.O.A.); (N.A.); (G.M.S.); (M.S.A.); (M.A.I.)
| | - Gouse M. Shaik
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (N.O.A.); (N.A.); (G.M.S.); (M.S.A.); (M.A.I.)
| | - Syed Kashif Zaidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Wessam Nouh
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Majed S. Alokail
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (N.O.A.); (N.A.); (G.M.S.); (M.S.A.); (M.A.I.)
| | - Mohamed A. Ismael
- Protein Research Chair, Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.A.); (N.O.A.); (N.A.); (G.M.S.); (M.S.A.); (M.A.I.)
| |
Collapse
|
5
|
Alhazza IM, Hassan I, Ebaid H, Al-Tamimi J, Alwasel SH. Chemopreventive effect of riboflavin on the potassium bromate-induced renal toxicity in vivo. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2355-2364. [PMID: 32666286 DOI: 10.1007/s00210-020-01938-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/02/2020] [Indexed: 02/06/2023]
Abstract
Potassium bromate (PB) is a general food additive, flavor enhancer, a by-product of water disinfection, and a class 2 carcinogen. It exerts various toxic effects in a dose- and time-dependent manner in vivo. This study is to explore the chemopreventive efficacy of vitamin B2 (riboflavin, RF) in PB-administered Swiss albino rats. The rats were distributed into five groups: control (group 1), PB alone (group 2, 150 mg/kg), RF alone (group 3, 2 mg/kg), PB + RF1 (group 4, 150 and 2 mg/kg), and PB + RF2 (group 5, 150 and 4 mg/kg). All the rodents were sacrificed after the completion of the treatment cycle. Then, blood and kidney samples were subjected to biochemical analysis. Group 2 demonstrated vivid signs of renal toxicities evidenced by altered renal function markers (urea, creatinine, albumin, glutathione-S-transferase) and redox status parameters (superoxide dismutase, catalase, glutathione reductase, reduced glutathione, lipid, and protein oxidation products). However, group 3 exhibited a slight alteration in many of the parameters while groups 4 and 5 demonstrated dose-dependent chemopreventive efficiency of RF against PB-induced alterations. Besides, RF seemed to facilitate apoptosis as well as inhibition of the necrosis in the PB-pre-challenged groups, as demonstrated by the cleaved PARP and lactate dehydrogenase activity. Also, the histopathological analysis and comet assay validate the biochemical results of the treatment groups significantly. All these results plead that RF has a significant chemopreventive property against PB-induced toxicity in vivo. Therefore, RF is a suitable agent in preventing the PB-induced toxicities at the clinical and industrial levels.
Collapse
Affiliation(s)
- Ibrahim M Alhazza
- Department of Zoology, College of Science, Building 05, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Iftekhar Hassan
- Department of Zoology, College of Science, Building 05, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Hossam Ebaid
- Department of Zoology, College of Science, Building 05, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Jameel Al-Tamimi
- Department of Zoology, College of Science, Building 05, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| | - Saleh H Alwasel
- Department of Zoology, College of Science, Building 05, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia
| |
Collapse
|
6
|
Moura GAD, Monteiro PB. Cytotoxic Activity of Antineoplastic Agents on Fertility: A Systematic Review. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2020; 42:759-768. [PMID: 33254272 PMCID: PMC10309244 DOI: 10.1055/s-0040-1713911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE To analyze the long-term effects of antineoplastic treatments on patient fertility. SELECTION OF STUDIES The studies were selected through the New PubMed, Scielo and Lilacs databases along with references used for the creation of the present work. For the selection of studies, articles published between the periods from January 1, 2015 to April 6, 2020 in the English, Portuguese and Spanish languages were used. As inclusion criteria: cohort studies and studies conducted in vitro. As exclusion criteria: review articles, reported cases, studies that do not address thematic reproduction, studies that do not address the cancer theme, articles that used animals, articles that address the preservation of fertility and articles in duplicate in the bases. DATA COLLECTION The collected data included: age of the patient at the beginning of treatment, type of neoplasm, type of antineoplastic treatment, chemotherapy used, radiotherapy dosage, radiotherapy site, effect of antineoplastic agents on fertility and number of patients in the study. DATA SYNTHESIS Thirty studies were evaluated, antineoplastic chemotherapy agents and radiotherapy modulate serum hormone levels, reduces germ cell quantities and correlated with an increase in sterility rates. The effects mentioned occur in patients in the prepubertal and postpubertal age. CONCLUSION Antineoplastic treatments have cytotoxic effects on the germ cells leading to hormonal modulation, and pubertal status does not interfere with the cytotoxic action of therapies.
Collapse
|
7
|
Rieber M. Cancer Pro-oxidant Therapy Through Copper Redox Cycling: Repurposing Disulfiram and Tetrathiomolybdate. Curr Pharm Des 2020; 26:4461-4466. [DOI: 10.2174/1381612826666200628022113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022]
Abstract
Background:
Copper (Cu) is a transition metal active in Fenton redox cycling from reduced Cu+ and
H2O2, to oxidized Cu2+ and the hydroxyl radical (·OH) highly reactive oxygen species (ROS). At homeostatic Cu
levels, ROS promote cell proliferation, migration, angiogenesis, and wound repair. To limit ROS toxicity, cells
use Cu-dependent chaperone proteins, Cu-binding ceruloplasmin, and Cu-modulated enzymes like superoxide
dismutases (SOD) like SOD1 and SOD3 to scavenge excess superoxide anions which favour Cu+ reduction, and
mitochondrial cytochrome c oxidase, important in aerobic energy production. Because Cu helps drive tumor cell
proliferation by promoting growth factor-independent receptor tyrosine kinase signaling, and Cu-dependent
MEK1 involved in oncogenic BRAF-V600E signaling, further augmenting bioavailable Cu may promote ROS overproduction,
cancer progression and eventually tumor cell death. For these reasons, the following clinically approved
copper chelators are being repurposed as anti-cancer agents: a) ammonium tetrathiomolybdate (TTM)
used to treat Wilson’s disease (copper overload) and Menkes disease (copper deficiency); b) Disulfiram (DSF),
used against alcoholism, since it inhibits Aldehyde Dehydrogenase (ALDH1) enzyme, important in ethanol detoxification,
and a key target against cancer stem cells. Moreover, TTM and DSF are also relevant in cancer clinical
trials, because they increase the uptake of both Cu and Platinum (Pt)-containing anti-cancer drugs, since Pt
and Cu share the same CTR1 copper transporter.
Purpose:
The majority of reports on Cu chelators dealt separately with either TTM, DSF or others. Here, we
compare in parallel, the anti-cancer efficacy of low doses of TTM and DSF, asking whether they can be synergistic
or antagonistic. The relevance of their unequal ROS inducing abilities and their different behavior as ionophores
is also addressed.
Significance:
The potential of Cu chelators as repurposed anti-cancer drugs, should be greater in patients with
higher endogenous Cu levels. Since platinum and Cu share uptake receptors, the synergism by drugs containing
these metals should not be under-estimated. The potential of disulfiram or its metabolically active Cu-containing
form, to inhibit ALDH1-positive tumor cells is therapeutically very important.
Collapse
Affiliation(s)
- Manuel Rieber
- IVIC, Cancer Cell Biology Laboratory, CMBC, Caracas 1020A, Venezuela
| |
Collapse
|
8
|
The Alleviative Effect of Vitamin B 2 on Potassium Bromate-Induced Hepatotoxicity in Male Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8274261. [PMID: 32802879 PMCID: PMC7415125 DOI: 10.1155/2020/8274261] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/24/2020] [Accepted: 07/16/2020] [Indexed: 12/30/2022]
Abstract
Potassium bromate (PB) is a food enhancer, water disinfection by-product, and a proven carcinogen. It elicits toxicities in the living organism due to exposure and in a dose-dependent manner. The present study discourses the ameliorative efficacy of riboflavin (RF) in PB-administered rodents. The animals were distributed into five treatment groups: control (group I), PB alone (group II, 150 mg/kg), RF alone (group III, 2 mg/kg), PB+RF1 (group IV, 150 mg/kg + 2 mg/kg), and PB+RF2 (group V, 150 mg/kg + 4 mg/kg). After the round of the treatment, the animals were sacrificed to collect their blood and liver samples for the detailed analysis. Group II depicted perturbed liver functions evidenced by altered serum and toxicity markers along with the disturbed redox balance. Also, these biochemical results were found harmonious with histopathological analysis and comet assay. However, group III showed no noticeable alteration in the same parameters, whereas the combination groups (IV and V) exhibited dose-dependent amelioration in the PB-induced toxicities. Interestingly, RF favored apoptosis concomitant with suppressing the necrosis in the PB-challenged groups, as shown by the activity of caspase-3 and lactate dehydrogenase. Histopathological analysis and comet assay further consolidate these results. Hence, RF has significant alleviative property against PB-induced hepatotoxicity in vivo that can be used in the consumer items containing the toxicant.
Collapse
|
9
|
Serra M, Columbano A, Ammarah U, Mazzone M, Menga A. Understanding Metal Dynamics Between Cancer Cells and Macrophages: Competition or Synergism? Front Oncol 2020; 10:646. [PMID: 32426284 PMCID: PMC7203474 DOI: 10.3389/fonc.2020.00646] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/07/2020] [Indexed: 12/13/2022] Open
Abstract
Metal ions, such as selenium, copper, zinc, and iron are naturally present in the environment (air, drinking water, and food) and are vital for cellular functions at chemical, molecular, and biological levels. These trace elements are involved in various biochemical reactions by acting as cofactors for many enzymes and control important biological processes by binding to the receptors and transcription factors. Moreover, they are essential for the stabilization of the cellular structures and for the maintenance of genome stability. A body of preclinical and clinical evidence indicates that dysregulation of metal homeostasis, both at intracellular and tissue level, contributes to the pathogenesis of many different types of cancer. These trace minerals play a crucial role in preventing or accelerating neoplastic cell transformation and in modulating the inflammatory and pro-tumorigenic response in immune cells, such as macrophages, by controlling a plethora of metabolic reactions. In this context, macrophages and cancer cells interact in different manners and some of these interactions are modulated by availability of metals. The current review discusses the new findings and focuses on the involvement of these micronutrients in metabolic and cellular signaling mechanisms that influence macrophage functions, onset of cancer and its progression. An improved understanding of "metallic" cross-talk between macrophages and cancer cells may pave the way for innovative pharmaceutical or dietary interventions in order to restore the balance of these trace elements and also strengthen the chemotherapeutic treatment.
Collapse
Affiliation(s)
- Marina Serra
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Amedeo Columbano
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Ummi Ammarah
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| | - Alessio Menga
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology (CCB), VIB, Leuven, Belgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center – MBC, University of Torino, Turin, Italy
| |
Collapse
|
10
|
Lu X, Lin B, Xu N, Huang H, Wang Y, Lin JM. Evaluation of the accumulation of disulfiram and its copper complex in A549 cells using mass spectrometry. Talanta 2020; 211:120732. [PMID: 32070566 DOI: 10.1016/j.talanta.2020.120732] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/04/2020] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
The famous alcohol-aversion drug disulfiram (DSF) is a promising candidate for repurposing in cancer therapy, as indicated by many ongoing and completed clinical trials. Existing researches focus on demonstrating that the anti-cancer activity of DSF is enhanced by copper ions, or solving the problem that DSF is easily decomposed in the body to lose its activity. However, the metabolic kinetics of its ultimate anti-cancer metabolite DDC-Cu (bis-diethyldithiocarbamate-copper) in cells and how it exerts anti-cancer mechanisms remain unclear. In this work, mass spectrometric evaluation of the intracellular and extracellular accumulation of DSF and its copper complex DDC-Cu was performed. Combined with cytotoxicity assay, staining analysis and flow cytometry, we found that DDC-Cu could easily pass through the cell membrane of A549 cells, and accumulate intracellularly for a long time. This process can lead to cellular morphological changes, an increase in ROS content, cell cycle arrest in the G0/G1 phase and apoptosis. Besides, molecular cancer-relevant targets of DDC-Cu in cancer cells were further discussed. This work investigated the cytotoxic mechanism of DDC-Cu, which has important clinical significance for its application in cancer therapy.
Collapse
Affiliation(s)
- Xinling Lu
- Department of Chemistry,School of Science,Tianjin University,Tianjin, 300075,China; Department of Chemistry,Beijing Key Laboratory of Microanalytical Methods and Instrumentation,MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University,Beijing, 100084,China
| | - Binxin Lin
- Department of Chemistry,Beijing Key Laboratory of Microanalytical Methods and Instrumentation,MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University,Beijing, 100084,China
| | - Ning Xu
- Department of Chemistry,Beijing Key Laboratory of Microanalytical Methods and Instrumentation,MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University,Beijing, 100084,China
| | - Hua Huang
- Department of Chemistry,School of Science,Tianjin University,Tianjin, 300075,China; Department of Chemistry,Beijing Key Laboratory of Microanalytical Methods and Instrumentation,MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University,Beijing, 100084,China
| | - Yong Wang
- Department of Chemistry,School of Science,Tianjin University,Tianjin, 300075,China.
| | - Jin-Ming Lin
- Department of Chemistry,Beijing Key Laboratory of Microanalytical Methods and Instrumentation,MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University,Beijing, 100084,China.
| |
Collapse
|
11
|
Alhazza IM, Ebaid H, Abdel-Salam B, Al-Tamimi JH, Hassan I, Rady AM, Mashaly AMA. Thymoquinone ameliorates Pachycondyla sennaarensis venom-induced acute toxic shock in male rats. BMC Pharmacol Toxicol 2019; 20:84. [PMID: 31847893 PMCID: PMC6918657 DOI: 10.1186/s40360-019-0375-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 11/26/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND For many decades, the sting of Samsun ant (Pachycondyla sennaarensis) has been a serious clinical challenge for the people living in some of the major Middle East and Asian countries. In the present study, the therapeutic potential of Nigella sativa derived plant extract component, thymoquinone (TQ) has been tested against the Samsun ant venom (SAV) at the toxic dose in the rats. METHODS The adult male rats were divided into four groups (n = 10): control, SAV treated, SAV + TQ treated and TQ alone treated. It was found that the sub-lethal dose of SAV alters not only many of the kidney and liver function markers but also induces oxidative stress in the animals. Moreover, the SAV also disturbs various immunological parameters including expression of PMNs, CD-80, CD-86, interleukins and other cytokines compromising the affected organism towards mild to severe allergic reactions including life-risking anaphylaxis. RESULTS The plant extract, TQ, effectively restores many of the biochemical and oxidative stress parameters comparable to the normal concomitant with improving the immunological aspects that might attributive in relieving from SAV-induced toxicity and allergic reactions in the affected organism to a greater extent. CONCLUSION Hence, TQ has an excellent antidote property against SAV-induced toxicities in vivo. Although the study is a vivid indication of the potential therapeutic potential of TQ against the SAV induced in vivo toxicity, yet the actual mechanism of interaction translating the toxicity amelioration warrants further investigations.
Collapse
Affiliation(s)
- Ibrahim M Alhazza
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Hossam Ebaid
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Bahaa Abdel-Salam
- Department of Biology, College of Science and Humanities in El-Quwiaya, 11961, Shaqra University, Shaqra, Saudi Arabia
| | - Jameel H Al-Tamimi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Iftekhar Hassan
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia.
| | - Ahmed M Rady
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Ashraf M A Mashaly
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
12
|
Li N, Zhou H, Wu H, Wu Q, Duan M, Deng W, Tang Q. STING-IRF3 contributes to lipopolysaccharide-induced cardiac dysfunction, inflammation, apoptosis and pyroptosis by activating NLRP3. Redox Biol 2019; 24:101215. [PMID: 31121492 PMCID: PMC6529775 DOI: 10.1016/j.redox.2019.101215] [Citation(s) in RCA: 398] [Impact Index Per Article: 66.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/27/2019] [Accepted: 05/06/2019] [Indexed: 12/18/2022] Open
Abstract
Mountainous evidence suggests that inflammation, cardiomyocyte apoptosis and pyroptosis are involved in the development of sepsis and sepsis-induced cardiomyopathy (SIC). Stimulator of interferon genes (STING) is an indispensable molecule that could regulate inflammation and immune response in multiple diseases. However, the role of STING in cardiovascular disease, especially SIC remains unclear. This study was designed to investigate the potential molecular mechanisms of STING in lipopolysaccharide (LPS)-induced cardiac injury using STING global knockout mice. In wild type mice and cardiomyocytes, LPS stimulation triggered the perinuclear translocation of STING, which further bound to Type-I interferons (IFN) regulatory factor 3 (IRF3) and phosphorylated IRF3. Phosphorylated (P-) IRF3 subsequently translocated into nucleus and increased the expression of NOD-like receptor protein 3 (NLRP3). Knockout of STING in mice significantly improved survival rate and cardiac function, apart from suppressing myocardial and serum inflammatory cytokines, apoptosis, as well as cardiomyocyte pyroptosis. In vitro experiments revealed that NLRP3 overexpression by adenovirus could offset protective effects of STING knockdown in LPS-induced cardiomyocytes. Additionally, LPS stimulation also promoted the production of intracellular reactive oxygen (ROS), which further induced the NLRP3 translocation to the cytoplasm from the nucleus. Dissociative TXNIP could directly interact with cytoplasmic NLRP3 and form inflammasome, eventually triggering cardiomyocyte injury. Collectively, our findings disclose that STING deficiency could alleviate LPS-induced SIC in mice. Hence, targeting STING in cardiomyocytes may be a promising therapeutic strategy for preventing SIC.
Collapse
Affiliation(s)
- Ning Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China.
| | - Haiming Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Qingqing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Mingxia Duan
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, PR China.
| |
Collapse
|
13
|
Hassan I, Husain FM, Khan RA, Ebaid H, Al-Tamimi J, Alhazza IM, Aman S, Ibrahim KE. Ameliorative effect of zinc oxide nanoparticles against potassium bromate-mediated toxicity in Swiss albino rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:9966-9980. [PMID: 30739294 DOI: 10.1007/s11356-019-04443-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/30/2019] [Indexed: 06/09/2023]
Abstract
Potassium bromate (PB) is a commonly used food additive, a prominent water disinfection by-product, and a class IIB carcinogen. It exerts a various degree of toxicity depending on its dose and exposure duration consumed with food and water in the living organisms. The present investigation aims to demonstrate the protective efficacy of zinc oxide nanoparticles (ZnO NPs) derived from Ochradenus arabicus (OA) leaf extract by green technology in PB-challenged Swiss albino rats. The rodents were randomly distributed, under the lab-standardized treatment strategy, into the following six treatment groups: control (group I), PB alone (group II), ZnO alone (group III), ZnO NP alone (group IV), PB + ZnO (group V), and PB + ZnO NPs (group VI). The rats were sacrificed after completion of the treatment, and their blood and liver samples were collected for further analysis. Group II showed extensive toxic effects with altered liver function markers (alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, lactate dehydrogenase, gamma-glutamyl transferase, glutathione-S-transferase, and thioredoxin reductase) and compromised redox status (SOD, CAT, GR, GPx, GSH, MDA, and total carbonyl content). The histopathological analysis and comet assay further supported the biochemical results of the same group. Besides, group III also showed moderate toxicity evidenced by an alteration in most of the studied parameters while group IV demonstrated mild toxicity after biochemical analysis indicating the excellent biocompatibility of the NPs. However, group VI exhibited attenuation of the PB-induced toxic insults to a significant level as compared to group II, whereas group V failed to show similar improvement in the studied parameters. All these findings entail that the ZnO NPs prepared by green synthesis have significant ameliorative property against PB-induced toxicity in vivo. Moreover, administration of the NPs improved the overall health of the treated animals profoundly. Hence, these NPs have significant therapeutic potential against the toxic effects of PB and similar compounds in vivo, and they are suitable to be used at the clinical and industrial levels.
Collapse
Affiliation(s)
- Iftekhar Hassan
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia.
| | - Fohad Mabood Husain
- Department of Food Science and Nutrition, College of Food and Agriculture, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Rais Ahmad Khan
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Hossam Ebaid
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| | - Jameel Al-Tamimi
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| | - Ibrahim M Alhazza
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| | - Shazia Aman
- Department of Biochemistry, J N Medical College and Hospital, Aligarh Muslim University, Aligarh, 202002, India
| | - Khalid Elfaki Ibrahim
- Department of Zoology, College of Science, King Saud University, Building 05, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
14
|
Copper Mediates Anti-Inflammatory and Antifibrotic Activity of Gleevec in Hepatocellular Carcinoma-Induced Male Rats. Can J Gastroenterol Hepatol 2019; 2019:9897315. [PMID: 30941331 PMCID: PMC6421053 DOI: 10.1155/2019/9897315] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/29/2019] [Accepted: 02/10/2019] [Indexed: 12/31/2022] Open
Abstract
The elevated level of copper is one of the hallmark features of cancer cells in most of the types of cancer. In the present study, this feature has been targeted to investigate if coadministration of exogenous copper (Cu+) and its chelating agent like disulfiram (DSF+) influence the antineoplastic activity of the anticancer drug, Gleevec (GLV+), in hepatocellular carcinoma (HCC)-induced rats via immunomodulation. After the treatment, the level of proinflammatory interleukins (IL-1, 2, 6, and 7), anti-inflammatory interleukin (IL-10) concomitant with transcription factors (NF-kB and TNF-a), and the apoptotic marker (cleaved PARP) was estimated. The cancer-induced group without treatment (CN+) demonstrated abnormally elevated level of all proinflammatory cytokines and transcription factors concomitant with a compromised level of cleaved PARP as compared to the control normal (CN-). The detailed histological analysis also supported the results exhibiting extensive inflammation and tissue fibrosis confirming the second stage of HCC. Cu+, DSF+, and GLV+ displayed mild improvement in most of the parameters, but the combination group GLV + Cu+ demonstrated remarkable recovery in histology and most of the parameters tended towards the CN- followed by GLV + DSF+. Therefore, the management of copper level is critical in realizing the antineoplastic activity of GLV up to its full potential in cancer treatment. These findings will help in improving chemoimmunotherapy and personalized cancer treatment.
Collapse
|
15
|
Calderon-Aparicio A, Cornejo A, Orue A, Rieber M. Anticancer response to disulfiram may be enhanced by co-treatment with MEK inhibitor or oxaliplatin: modulation by tetrathiomolybdate, KRAS/BRAF mutations and c-MYC/p53 status. Ecancermedicalscience 2019; 13:890. [PMID: 30792807 PMCID: PMC6369974 DOI: 10.3332/ecancer.2019.890] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Indexed: 12/22/2022] Open
Abstract
Ammonium tetrathiomolybdate (TTM) and disulfiram (DSF) are copper (Cu) chelators in cancer clinical trials partly because Cu chelation: a) restricts the activity of Cu-binding MEK1/2 enzymes which drive tumourigenesis by KRAS or BRAF oncogenic mutations and b) enhances uptake of oxaliplatin (OxPt), clinically used in advanced KRAS-mutant colorectal carcinomas (CRC). Whereas TTM decreases intracellular Cu trafficking, DSF can reach other Cu-dependent intracellular proteins. Since the use of individual or combined Cu chelation may help or interfere with anti-cancer therapy, this study investigated whether TTM modifies the response to DSF supplemented with: 1) UO126, a known MEK1/2 inhibitor; 2) other Cu chelators like neocuproine (NC) or 1, 10-o-phenanthroline (OPT) in wt p53 melanoma cells differing in BRAF or KRAS mutations; 3) OxPt in mutant p53 CRC cells devoid of KRAS and BRAF mutations or harbouring either KRAS or BRAF mutations. TTM was not toxic against V600E-mut-BRAF A375 and G12D-mut-KRAS/high c-myc C8161 melanoma cells. Moreover, TTM protected both melanoma types from toxicity induced by DSF, NC and co-treatment with sub-lethal levels of DSF and the MEK inhibitor, UO126. Toxicity by co-treatment with DSF+OPT was poorly reversed by TTM in C8161 melanoma cells. In contrast to the greater toxicity of 0.1 μM DSF against mutant p53 CRC cells irrespective of their KRAS mutation, TTM did not protect G12V-mut-KRAS/high c-myc SW620 CRC from DSF+OxPt compared to KRAS-WT/BRAF-WT Caco-2 CRC. Our results show that DSF co-treatment with: a) MEK inhibitors may enhance tumour suppression; b) OxPt in CRC may counteract impaired response to cetuximab by KRAS/BRAF mutations and c) as a single treatment, TTM may be less effective than DSF and decreases the efficacy of the latter.
Collapse
Affiliation(s)
| | | | - Andrea Orue
- Instituto Venezolano de Investigaciones Cientificas, Tumor Cell Biology Laboratory, Caracas 1020-A, Venezuela.,These authors contributed equally to this work
| | - Manuel Rieber
- Instituto Venezolano de Investigaciones Cientificas, Tumor Cell Biology Laboratory, Caracas 1020-A, Venezuela.,These authors contributed equally to this work
| |
Collapse
|