1
|
Fu Y, Zeng S, Wang Z, Huang H, Zhao X, Li M. Mechanisms of Copper-Induced Autophagy and Links with Human Diseases. Pharmaceuticals (Basel) 2025; 18:99. [PMID: 39861161 PMCID: PMC11768742 DOI: 10.3390/ph18010099] [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: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
As a structural and catalytic cofactor, copper is involved in many biological pathways and is required for the biochemistry of all living organisms. However, excess intracellular copper can induce cell death due to its potential to catalyze the generation of reactive oxygen species, thus copper homeostasis is strictly regulated. And the deficiency or accumulation of intracellular copper is connected with various pathological conditions. Since the success of platinum-based compounds in the clinical treatment of various types of neoplasias, metal-based drugs have shown encouraging perspectives for drug development. Compared to platinum, copper is an essential intracellular trace element that may have better prospects for drug development than platinum. Recently, the potential therapeutic role of copper-induced autophagy in chronic diseases such as Parkinson's, Wilson's, and cardiovascular disease has already been demonstrated. In brief, copper ions, numerous copper complexes, and copper-based nano-preparations could induce autophagy, a lysosome-dependent process that plays an important role in various human diseases. In this review, we not only focus on the current advances in elucidating the mechanisms of copper or copper-based compounds/preparations on the regulation of autophagy but also outline the association between copper-induced autophagy and human diseases.
Collapse
Affiliation(s)
- Yuanyuan Fu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shuyan Zeng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhenlin Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huiting Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin Zhao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Min Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
2
|
Upadhyay TK, Trivedi R, Khan F, Al-Keridis LA, Pandey P, Sharangi AB, Alshammari N, Abdullah NM, Yadav DK, Saeed M. In vitro elucidation of antioxidant, antiproliferative, and apoptotic potential of yeast-derived β-1,3-glucan particles against cervical cancer cells. Front Oncol 2022; 12:942075. [PMID: 36059639 PMCID: PMC9436396 DOI: 10.3389/fonc.2022.942075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/12/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is the leading cause of mortality worldwide and in particular is the fourth most common cause of mortality in women every year. Conventional treatments for cancer are chemotherapy and radiation therapy, which have various kinds of side effects. Hence, there is a high need to develop alternative, efficient, and safer therapies for cancer treatment. β-Glucan, a novel polysaccharide isolated from baker’s yeast Saccharomyces cerevisiae, shows noteworthy cytotoxicity toward a variety of cancer cell lines in vitro. In this research, we characterized the β-glucan with high-performance thin-layer chromatography (HPTLC) analysis and found that d-glucose units with β-1,3 links are the major component of the extracted β-glucan particles. Fourier transform IR (FTIR) analysis confirmed a β-(1→3)-linked glucan structure. In vitro cell cytotoxicity was evaluated by MTT with IC50 136 μg/ml, and therapeutic potential was assessed by various assays using values below and above the IC50. A significant reactive oxygen species (ROS) generation at 50–150 μg/ml of concentrations indicated the apoptosis of cervical cancer cells. Along with ROS generation, these concentrations were also found to induce morphological changes such as fragmentation in DNA upon staining HeLa cells with DAPI. Mitochondrial membrane potential was significantly reduced after increasing the dose of treatment, assessed with the help of MitoTracker dye. Hence, by all these experimental supports, we observed that β-glucan has the potential to slow down the growth of cervical cancer cells, and it can be further investigated for unfolding its complete anticancer potential.
Collapse
Affiliation(s)
- Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, India
- *Correspondence: Mohd Saeed, ; Dharmendra Kumar Yadav, ; Tarun Kumar Upadhyay, ; Lamya Ahmed Al-Keridis,
| | - Rashmi Trivedi
- Department of Biotechnology, Parul Institute of Applied Sciences and Centre of Research for Development, Parul University, Vadodara, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, India
| | - Lamya Ahmed Al-Keridis
- Biology Department, Faculty of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
- *Correspondence: Mohd Saeed, ; Dharmendra Kumar Yadav, ; Tarun Kumar Upadhyay, ; Lamya Ahmed Al-Keridis,
| | - Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, India
| | - Amit Baran Sharangi
- Department of Plantation, Spices, Medicinal and Aromatic Crops, Bidhan Chandra Krishi Viswavidyalaya (BCKV)-Agricultural University, Mohanpur, India
| | - Nawaf Alshammari
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
| | - Nadiya M. Abdullah
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Dharmendra Kumar Yadav
- Department of Pharmacy and Gachon Institute of Pharmaceutical Science, College of Pharmacy, Gachon University, Yeonsu-gu, Incheon, South Korea
- *Correspondence: Mohd Saeed, ; Dharmendra Kumar Yadav, ; Tarun Kumar Upadhyay, ; Lamya Ahmed Al-Keridis,
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, Hail, Saudi Arabia
- *Correspondence: Mohd Saeed, ; Dharmendra Kumar Yadav, ; Tarun Kumar Upadhyay, ; Lamya Ahmed Al-Keridis,
| |
Collapse
|
3
|
Ferritinophagy-Mediated ROS Production Contributed to Proliferation Inhibition, Apoptosis, and Ferroptosis Induction in Action of Mechanism of 2-Pyridylhydrazone Dithiocarbamate Acetate. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5594059. [PMID: 34691357 PMCID: PMC8531783 DOI: 10.1155/2021/5594059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 08/28/2021] [Accepted: 09/03/2021] [Indexed: 01/17/2023]
Abstract
Reactive oxygen species (ROS) production is involved in the mechanism of action of a number of drugs, but the biological effects of ROS remain to be clarified. Furthermore, ferroptosis involves iron-dependent ROS production that may be derived from ferritinophagy; however, the association between ferroptosis and ferritinophagy has not been fully established. The present study demonstrated that dithiocarbamate derivatives (iron chelators) exhibited antineoplastic properties involving ferritinophagy induction, but whether the underlying mechanisms involved ferroptosis was unknown. To gain insight into the underlying mechanism, a dithiocarbamate derivative, 2-pyridylhydrazone dithiocarbamate s-acetic acid (PdtaA), was prepared. An MTT assay demonstrated that PdtaA inhibited proliferation involving ROS production (IC50 = 23.0 ± 1.5 μM for HepG2 cells). A preliminary mechanistic study revealed that PdtaA induced both apoptosis and cell cycle arrest. Notably, PdtaA also induced ferroptosis via downregulation of GPx4 and xCT, which was first reported for a dithiocarbamate derivative. Moreover, these cellular events were associated with ROS production. To explore the origin of ROS, expression of the ferritinophagy-related genes, ferritin, and nuclear receptor coactivator (NCOA4) were measured. Immunofluorescence and western blotting analysis indicated that PdtaA-induced ferritinophagy may contribute to ROS production. To investigate the role of ferritinophagy, autophagy inhibitor 3-methyladenin or genetic knockdown of NCOA4 was employed to inhibit ferritinophagy, which significantly neutralized the action of PdtaA in both apoptosis and ferroptosis. Taken together, PdtaA-induced cell cycle arrest, apoptosis, and ferroptosis were associated with ferritinophagy.
Collapse
|
4
|
Luo Y, Fu Y, Huang Z, Li M. Transition metals and metal complexes in autophagy and diseases. J Cell Physiol 2021; 236:7144-7158. [PMID: 33694161 DOI: 10.1002/jcp.30359] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/19/2021] [Accepted: 02/27/2021] [Indexed: 12/19/2022]
Abstract
Transition metals refer to the elements in the d and ds blocks of the periodic table. Since the success of cisplatin and auranofin, transition metal-based compounds have become a prospective source for drug development, particularly in cancer treatment. In recent years, extensive studies have shown that numerous transition metal-based compounds could modulate autophagy, promising a new therapeutic strategy for metal-related diseases and the design of metal-based agents. Copper, zinc, and manganese, which are common components in physiological pathways, play important roles in the progression of cancer, neurodegenerative diseases, and cardiovascular diseases. Furthermore, enrichment of copper, zinc, or manganese can regulate autophagy. Thus, we summarized the current advances in elucidating the mechanisms of some metals/metal-based compounds and their functions in autophagy regulation, which is conducive to explore the intricate roles of autophagy and exploit novel therapeutic drugs for human diseases.
Collapse
Affiliation(s)
- Yuping Luo
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Fu
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhiying Huang
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Min Li
- Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Molinaro C, Martoriati A, Pelinski L, Cailliau K. Copper Complexes as Anticancer Agents Targeting Topoisomerases I and II. Cancers (Basel) 2020; 12:E2863. [PMID: 33027952 PMCID: PMC7601307 DOI: 10.3390/cancers12102863] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
Organometallics, such as copper compounds, are cancer chemotherapeutics used alone or in combination with other drugs. One small group of copper complexes exerts an effective inhibitory action on topoisomerases, which participate in the regulation of DNA topology. Copper complexes inhibitors of topoisomerases 1 and 2 work by different molecular mechanisms, analyzed herein. They allow genesis of DNA breaks after the formation of a ternary complex, or act in a catalytic mode, often display DNA intercalative properties and ROS production, and sometimes display dual effects. These amplified actions have repercussions on the cell cycle checkpoints and death effectors. Copper complexes of topoisomerase inhibitors are analyzed in a broader synthetic view and in the context of cancer cell mutations. Finally, new emerging treatment aspects are depicted to encourage the expansion of this family of highly active anticancer drugs and to expend their use in clinical trials and future cancer therapy.
Collapse
Affiliation(s)
- Caroline Molinaro
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (C.M.); (A.M.)
| | - Alain Martoriati
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (C.M.); (A.M.)
| | - Lydie Pelinski
- Univ. Lille, CNRS, Centrale Lille, Univ. Artois, UMR 8181-UCCS-Unité de Catalyse et Chimie du Solide, F-59000 Lille, France;
| | - Katia Cailliau
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (C.M.); (A.M.)
| |
Collapse
|
6
|
Kim H, Jeon BT, Kim IM, Bennett SJ, Lorch CM, Viana MP, Myers JF, Trupp CJ, Whipps ZT, Kundu M, Chung S, Sun X, Khalimonchuk O, Lee J, Ro SH. Sestrin2 Phosphorylation by ULK1 Induces Autophagic Degradation of Mitochondria Damaged by Copper-Induced Oxidative Stress. Int J Mol Sci 2020; 21:ijms21176130. [PMID: 32854424 PMCID: PMC7504119 DOI: 10.3390/ijms21176130] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/05/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
Selective autolysosomal degradation of damaged mitochondria, also called mitophagy, is an indispensable process for maintaining integrity and homeostasis of mitochondria. One well-established mechanism mediating selective removal of mitochondria under relatively mild mitochondria-depolarizing stress is PINK1-Parkin-mediated or ubiquitin-dependent mitophagy. However, additional mechanisms such as LC3-mediated or ubiquitin-independent mitophagy induction by heavy environmental stress exist and remain poorly understood. The present study unravels a novel role of stress-inducible protein Sestrin2 in degradation of mitochondria damaged by transition metal stress. By utilizing proteomic methods and studies in cell culture and rodent models, we identify autophagy kinase ULK1-mediated phosphorylation sites of Sestrin2 and demonstrate Sestrin2 association with mitochondria adaptor proteins in HEK293 cells. We show that Ser-73 and Ser-254 residues of Sestrin2 are phosphorylated by ULK1, and a pool of Sestrin2 is strongly associated with mitochondrial ATP5A in response to Cu-induced oxidative stress. Subsequently, this interaction promotes association with LC3-coated autolysosomes to induce degradation of mitochondria damaged by Cu-induced ROS. Treatment of cells with antioxidants or a Cu chelator significantly reduces Sestrin2 association with mitochondria. These results highlight the ULK1-Sestrin2 pathway as a novel stress-sensing mechanism that can rapidly induce autophagic degradation of mitochondria under severe heavy metal stress.
Collapse
Affiliation(s)
- Heejeong Kim
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Byeong Tak Jeon
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Isaac M. Kim
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Sydney J. Bennett
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Carolyn M. Lorch
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
- Department of Biology, Grinnell College, Grinnell, IA 50112, USA
- Driskill Graduate Program in Life Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Martonio Ponte Viana
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Jacob F. Myers
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
- Department of Chemistry, The University of Scranton, Scranton, PA 18510, USA
| | - Caroline J. Trupp
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Zachary T. Whipps
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Mondira Kundu
- Departments of Pathology and Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Soonkyu Chung
- Department of Nutrition, University of Massachusetts, Amherst, MA 01003, USA;
| | - Xinghui Sun
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Oleh Khalimonchuk
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Jaekwon Lee
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
| | - Seung-Hyun Ro
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA; (H.K.); (B.T.J.); (I.M.K.); (S.J.B.); (C.M.L.); (M.P.V.); (J.F.M.); (C.J.T.); (Z.T.W.); (X.S.); (O.K.); (J.L.)
- Correspondence: ; Tel.: +1-402-472-5424
| |
Collapse
|
7
|
Redox cycling of copper by coumarin-di(2-picolyl)amine hybrid molecule leads to ROS-mediated modulation of redox scavengers, DNA damage and cell death in diethylnitrosamine induced hepatocellular carcinoma. Bioorg Chem 2020; 99:103818. [PMID: 32276135 DOI: 10.1016/j.bioorg.2020.103818] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022]
Abstract
Targeted therapy is a new strategy for cancer treatment that targets chemical entities specific to cancer cells than normal ones. One of the features associated with malignancy is the elevated copper which plays an integral role in angiogenesis. Work is in progress in our lab to identify new copper chelators to target elevated copper under targeted therapy for the killing of cancer cells. Recently, a coumarin-based copper chelator, di(2-picolyl)amine-3(bromoacetyl)coumarin hybrid molecule (ligand-L) has been synthesized by us, and also studied its copper-dependent macromolecular damage response in copper overloaded lymphocytes. The present study investigates the anticancer activity of ligand-L and its mode of action in rat model of diethylnitrosamine (DEN) induced hepatocellular carcinoma. It has been found that liver tissue has a marked increase in copper levels in DEN induced hepatocellular carcinoma. Ex vivo results showed that ligand-L inhibited cell viability, induced reactive oxygen species (ROS) generation, DNA damage, loss of mitochondrial membrane potential and caspase-3 activation in isolated hepatocellular carcinoma cells (HCC). All these effects induced by ligand-L were abrogated by neocuproine and N-acetylcysteine (ROS scavenger). Further, ligand-L treatment of animals bearing hepatocellular carcinoma results in an increment in the cellular redox scavengers, lipid peroxidation and DNA breakage in malignant hepatocytes. In vivo studies using ligand-L also showed that ligand-L possesses anticancer properties as evidenced by improvement in liver marker enzymes and liver surface morphology, and reduced alpha-fetoprotein in the treated group compared to untreated cancer-induced group. Overall, this study suggests that copper-ligand-L interaction leads to ROS generation which caused DNA damage and apoptosis in malignant cells. This study provides enough support to establish ligand-L as a clinically relevant lead molecule for the treatment of different malignancies.
Collapse
|
8
|
Karmakar S, Maji M, Mukherjee A. Modulation of the reactivity of nitrogen mustards by metal complexation: approaches to modify their therapeutic properties. Dalton Trans 2019; 48:1144-1160. [DOI: 10.1039/c8dt04503h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Metal complexation of nitrogen mustards shows promise with an ability to control the mustards’ reactivity, perform selective hypoxia activation, overcome resistance, and control GSH deactivation.
Collapse
Affiliation(s)
- Subhendu Karmakar
- Department of Chemical Sciences
- Indian Institute of Science Education and Research Kolkata
- Mohanpur-741246
- India
| | - Moumita Maji
- Department of Chemical Sciences
- Indian Institute of Science Education and Research Kolkata
- Mohanpur-741246
- India
| | - Arindam Mukherjee
- Department of Chemical Sciences
- Indian Institute of Science Education and Research Kolkata
- Mohanpur-741246
- India
| |
Collapse
|
9
|
Li C, Liu Y, Fu Y, Huang T, Kang L, Li C. The antiproliferative activity of di-2-pyridylketone dithiocarbamate is partly attributed to catalase inhibition: detailing the interaction by spectroscopic methods. MOLECULAR BIOSYSTEMS 2018; 13:1817-1826. [PMID: 28714505 DOI: 10.1039/c7mb00032d] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The bioactivity of drugs is attributed to their interaction with biological molecules, embodied in either their direct or indirect influence on enzyme activity and conformation. Di-2-pyridylketone hydrazine dithiocarbamate (DpdtC) exhibits significant antitumor activity in our preliminary study. We speculated that its activity may partly stem from enzyme inhibition due to strong metal chelating ability. To this end, we assessed its effect on catalase from erythrocytes and found evidence of inhibition, which was further confirmed by ROS determination in vivo. Thus, detailing the interaction between the agent and catalase via spectroscopic methods and molecular docking was required to obtain information on both the dynamics and thermodynamic parameters. The Lineweaver-Burk plot implied an uncompetitive pattern between DpdtC and catalase from beef liver, and IC50 = ∼7 μM. The thermodynamic parameters from fluorescence quenching measurements indicated that DpdtC could bind to catalase with moderate affinity (Ka = approximately 104 M-1). CD spectra revealed that DpdtC could significantly disrupt the secondary structure of catalase. Docking studies indicated that DpdtC bound to a flexible region of catalase, involving hydrogen bonds and salt bond; this was consistent with thermodynamic results from spectral investigations. Our data clearly showed that catalase inhibition of DpdtC was not due to direct chelation of iron from heme (killing), but through an allosteric effect. Thus, it can be concluded that the antiproliferative activity of DpdtC is partially attributed to its catalase inhibition.
Collapse
Affiliation(s)
- Cuiping Li
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P. R. China.
| | | | | | | | | | | |
Collapse
|
10
|
Wang T, Liu Y, Fu Y, Huang T, Yang Y, Li S, Li C. Antiproliferative activity of di-2-pyridylhydrazone dithiocarbamate acetate partly involved in p53 mediated apoptosis and autophagy. Int J Oncol 2017; 51:1909-1919. [PMID: 29039462 DOI: 10.3892/ijo.2017.4149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/04/2017] [Indexed: 11/05/2022] Open
Abstract
Cancer cells have higher demand of iron and copper ions for growth, disturbing the metal's homeostasis can inhibit proliferation of cancer cell. Dithiocarbamates possessing excellent metal chelating ability and antitumor activity are considered as candidates in chelation therapy, however, their antitumor molecular mechanisms remain to be elucidated. In the present study, a dithiocarbamate derivative, di-2-pyridylhydrazone dithiocarbamate s-acetic acid (DpdtaA) was prepared to address the issue whether the molecular mechanism behind biological behavior showed by dithiocarbamate was p53 mediated. The proliferation inhibition assay showed that DpdtaA exhibited excellent antiproliferative effect for hepatocellular carcinoma (IC50= 3.0±0.4 µM for HepG2, 6.1±0.6 µM for Bel-7402 cell). However, in the presence of copper ion, the antiproliferative activity of DpdtaA significantly attenuated (~3-fold for HepG2) due to formation of copper chelate. The ROS assay revealed that the antiproliferative activity of DpdtaA correlated with ROS generation. Western blotting demonstrated that DpdtaA could upregulate p53 via down-regulating the Mdm2, accordingly leading to changes of bcl family proteins, indicating that a p53-dependent intrinsic apoptosis was partly involved. Simulation from molecular docking hinted that DpdtaA could disrupt interaction between p53 and Mdm2, indicating the disruption might also contribute to the upregulation of p53. The alternations in lysosome membrane permeability and acidic vacuoles as well as LC3-II upregulation indicated that autophagy was involved. The copper addition led to significantly attenuate biological activity of DpdtaA, with few dithiocarbamates, but the mechanism in apoptosis induction was not altered except for weaker ability.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Youxun Liu
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yun Fu
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Tengfei Huang
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yun Yang
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Shaoshan Li
- Department of Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Changzheng Li
- Department of Molecular Biology and Biochemistry, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
11
|
Grasso G, Santoro AM, Lanza V, Sbardella D, Tundo GR, Ciaccio C, Marini S, Coletta M, Milardi D. The double faced role of copper in Aβ homeostasis: A survey on the interrelationship between metal dyshomeostasis, UPS functioning and autophagy in neurodegeneration. Coord Chem Rev 2017. [DOI: 10.1016/j.ccr.2017.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
12
|
Sahni S, Bae DH, Jansson PJ, Richardson DR. The mechanistic role of chemically diverse metal ions in the induction of autophagy. Pharmacol Res 2017; 119:118-127. [DOI: 10.1016/j.phrs.2017.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/12/2022]
|
13
|
Wang T, Fu Y, Huang T, Liu Y, Wu M, Yuan Y, Li S, Li C. Copper Ion Attenuated the Antiproliferative Activity of Di-2-pyridylhydrazone Dithiocarbamate Derivative; However, There Was a Lack of Correlation between ROS Generation and Antiproliferative Activity. Molecules 2016; 21:molecules21081088. [PMID: 27556432 PMCID: PMC6273760 DOI: 10.3390/molecules21081088] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/08/2016] [Accepted: 08/16/2016] [Indexed: 12/16/2022] Open
Abstract
The use of chelators for cancer treatment has been an alternative option. Dithiocarbamates have recently attracted considerable attention owning to their diverse biological activities; thus, the preparation of new dithiocarbamate derivatives with improved antitumor activity and selectivity as well as probing the underlying molecular mechanism are required. In this study, di-2-pyridylhydrazone dithiocarbamate S-propionic acid (DpdtpA) and its copper complex were prepared and characterized, and its antiproliferative activity was evaluated. The proliferation inhibition assay showed that DpdtpA exhibited excellent antiproliferative effect in hepatocellular carcinoma (IC50 = 1.3 ± 0.3 μM for HepG2, and 2.5 ± 0.6 μM for Bel-7402). However, in the presence of copper ion, the antiproliferative activity of DpdtpA was dramatically attenuated (20–30 fold) owing to the formation of copper chelate. A preliminarily mechanistic study revealed that reactive oxygen species (ROS) generation mediated the antiproliferative activity of DpdtpA, and accordingly induced apoptosis, DNA cleavage, and autophagy. Surprisingly, the cytotoxicity of DpdtpA copper complex (DpdtpA–Cu) was also involved in ROS generation; however, a paradoxical relation between cellular ROS level and cytotoxicity was observed. Further investigation indicated that DpdtpA could induce cell cycle arrest at the S phase; however, DpdtpA–Cu lacked this effect, which explained the difference in their antiproliferative activity.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
- Henan Collaborative Innovation Center of Molecular Diagnostics and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Yun Fu
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Tengfei Huang
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Youxun Liu
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Meihao Wu
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Yanbin Yuan
- Department of Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Shaoshan Li
- Department of Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, Henan, China.
| | - Changzheng Li
- Department of Molecular Biology & Biochemistry, Xinxiang Medical University, Xinxiang 453003, Henan, China.
- Henan Collaborative Innovation Center of Molecular Diagnostics and Laboratory Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China.
| |
Collapse
|