1
|
Koshman VE, Dmitriev AA, Timoshnikov VA, Arkhipova AS, Selyutina OY, Polyakov NE. Interaction of novel N-acridine thiosemicarbazones with lipid membrane: NMR and molecular dynamics simulations. Arch Biochem Biophys 2025; 768:110390. [PMID: 40090443 DOI: 10.1016/j.abb.2025.110390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
NMR and molecular dynamics simulations revealed differences in the localization of the novel thiosemicarbazones: 2-benzoyl ((E)-N-(acridin-9-yl)-2-(phenyl(pyridin-2-yl)methylene)hydrazine-1-carbothioamide (AOBP) and 2-dipyridyl ((E)-N-(acridin-9-yl)-2-(di(pyridin-2-yl)methylene)hydrazine-1-carbothioamide (AODP) within the lipid membrane. It turned out that both thiosemicarbazones can penetrate inside the membrane, but AOBP is able to pass into the center of the hydrophobic region of the lipid bilayer, while AODP is distributed closer to the surface and freely leaves the membrane into the aqueous environment. The presence of cholesterol was also found to prevent both thiosemicarbazones from penetrating the membrane. The mechanism of anti-proliferative activity of some TSCs is related to the penetration through the lysosomal membrane and formation of cytotoxic copper complexes, which generate ROS resulting in lysosomal membrane permeabilization and cell death. Hydrophobic drugs, including TSCs, could penetrate through lysosomal membrane via passive diffusion, thus the affinity of drug to the hydrophobic interior of the lipid membrane could be important for their activity. Since the mechanism of thiosemicarbazones anticancer activity is associated with their penetration into lysosomes, the results obtained are important for a better understanding of the mechanisms of activity of these compounds and the development of new drug agents.
Collapse
Affiliation(s)
- Vladimir E Koshman
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090, Novosibirsk, Russia
| | - Alexey A Dmitriev
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090, Novosibirsk, Russia
| | - Viktor A Timoshnikov
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090, Novosibirsk, Russia
| | - Alina S Arkhipova
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090, Novosibirsk, Russia
| | - Olga Yu Selyutina
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090, Novosibirsk, Russia; Institute of Solid State Chemistry and Mechanochemistry, Kutateladze St., 18, 630128, Novosibirsk, Russia.
| | - Nikolay E Polyakov
- Institute of Chemical Kinetics and Combustion, Institutskaya St., 3, 630090, Novosibirsk, Russia; Institute of Solid State Chemistry and Mechanochemistry, Kutateladze St., 18, 630128, Novosibirsk, Russia
| |
Collapse
|
2
|
Dharmasivam M, Zhang S, Zhao X, Richardson V, Wijesinghe TP, Suleymanoglu M, Gholam Azad M, Bernhardt PV, Kaya B, Richardson DR. Advantages of Novel Anti-cancer Selenosemicarbazones: Preferential Reactivity of Their Fe(III), Cu(II), and Zn(II) Complexes with Key Physiological Reductants/Ligands Versus Isosteric Thiosemicarbazones. J Med Chem 2025; 68:9594-9622. [PMID: 40265585 DOI: 10.1021/acs.jmedchem.5c00374] [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: 04/24/2025]
Abstract
Fe(III) complexes of clinically trialed thiosemicarbazones demonstrate deleterious oxy-myoglobin and oxy-hemoglobin oxidation. Therefore, the PPP4pSe selenosemicarbazone analogues were designed with several PPP4pSe Fe(III) complexes completely preventing deleterious oxy-myoglobin oxidation. This was ascribed to the decreased potentials of their Fe(III) complexes and steric hindrance effects. The Fe(III), Cu(II), and Zn(II) complexes of PPP4pSe demonstrated greater reactivity with physiological reductants/ligands (glutathione, l-cysteine, or l-ascorbate), than respective complexes of the isosteric thiosemicarbazone, PPP4pT. Considering this: (1) [Fe(PPP4pSe)2]+ demonstrated increased reduction relative to [Fe(PPP4pT)2]+ with glutathione and l-cysteine, while l-ascorbate led to comparable reduction; (2) glutathione led to complete dissociation of [Zn(PPP4pSe)2], while incomplete dissociation of [Zn(PPP4pT)2] occurred; and (3) [Cu(PPP4pSe)Cl] demonstrated complete coordinate sphere substitution with glutathione, l-cysteine, and l-ascorbate, whereas [Cu(PPP4pT)Cl] demonstrated partial substitution. The role of glutathione in all three latter reactions is significant, given the greater reactivity of the selenosemicarbazone, and glutathione's key role in selenosemicarbazone and thiosemicarbazone anticancer activity.
Collapse
Affiliation(s)
- Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Stanley Zhang
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Mediha Suleymanoglu
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34093, Turkey
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Institute for Biomedicine and Glycomics, Griffith University, Southport 4215 Queensland, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
3
|
Donohoe MN, Upadhyay A, Pratt DA. Ligand-Based Radical Reactivity of Metal Thiosemicarbazones Prompts the Identification of Platinum(II)-Based Cytoprotectants. J Am Chem Soc 2024; 146:31307-31320. [PMID: 39494512 DOI: 10.1021/jacs.4c12713] [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: 11/05/2024]
Abstract
CuATSM, a copper(II) complex of a bis(thiosemicarbazone) of diacetyl, prevents oxidative cell death and acts as a neuroprotectant in vivo, prompting its evaluation to treat amyotrophic lateral sclerosis and other neurodegenerative conditions in the clinic. We recently demonstrated that CuATSM functions as a potent radical-trapping antioxidant (RTA), inhibiting lipid peroxidation and associated ferroptotic cell death by a noncanonical mechanism based on radical addition to the ligand backbone. Herein we report our investigations of the generality of this reactivity, which include studies of corresponding complexes of various other metals, including Co, Ru, Ni, Pd, Pt, and Au. Inhibited autoxidations of styrene and dioxane reveal that most of these complexes exhibit RTA activity, consistent with ligand-based reactivity, but the identity of the metal atom nevertheless plays a role. In particular, analyses of the electronic structures of the complexes of metals within the same group (i.e., the group 10 metals Ni, Pd and Pt) highlight how the metal atom can modulate the ligand-based reactivity by enabling spin delocalization to the other thiosemicarbazone moiety. The RTA activity determined in organic solution largely translates to phospholipid bilayers and mammalian cells, where most complexes inhibited lipid peroxidation and associated ferroptotic cell death. A preliminary structure-activity study revealed Pt complexes with potencies eclipsing those of archetype ferroptosis inhibitors ferrostatin-1 and liproxstatin-1, suggesting that Pt (and to a lesser extent Ni) bis(thiosemicarbazone)s may be better suited to optimization for therapeutic development than those based on Cu.
Collapse
Affiliation(s)
- Michael N Donohoe
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Aditya Upadhyay
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa K1N 6N5, Ontario, Canada
| |
Collapse
|
4
|
Kaya B, Smith H, Chen Y, Azad MG, Russell TM, Richardson V, Dharmasivam M, Richardson DR. Innovative N-Acridine Thiosemicarbazones and Their Zn(II) Complexes Transmetallate with Cu(II): Redox Activity and Suppression of Detrimental Oxy-Myoglobin Oxidation. Inorg Chem 2024; 63:20840-20858. [PMID: 39404641 DOI: 10.1021/acs.inorgchem.4c03642] [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: 10/29/2024]
Abstract
The coordination chemistry and electrochemistry of novel N-acridine thiosemicarbazones (NATs) were investigated along with their redox activity, antiproliferative efficacy, transmetalation, and dissociation properties. The ability of NAT Fe(III) complexes to inhibit detrimental oxy-myoglobin (oxy-Mb) oxidation was also examined. The NATs act as tridentate ligands with a 2:1 L/Zn(II) complex crystal structure, revealing a distorted octahedral geometry, where both ligands bind Zn(II) in a meridional conformation. The NAT Fe(III) complexes exhibited fully reversible one-electron FeIII/II couples with more positive potentials than the Fe(III) complexes of a related clinically trialed thiosemicarbazone (e.g., [Fe(DpC)2]+) due to the electron-donating capacity of acridine. Surprisingly, the NAT-Zn(II) complexes showed generally greater or similar antiproliferative activity than their ligands, Cu(II), or Fe(III) complexes. This may be explained by (1) formation of a highly lipophilic Zn(II) complex that acts as a chaperone to promote cellular uptake and (2) the capacity of the Zn(II) complex to dissociate or undergo transmetalation to the redox-active Cu(II) complex. Of the NAT-Fe(III) complexes, [Fe(AOBP)2]+ demonstrated a significant (p < 0.0001) improvement in preventing oxy-Mb oxidation than the Fe(III) complex of the clinically trialed thiosemicarbazone, DpC. This article advances our understanding of NAT coordination chemistry, electrochemistry, and the intriguing biological activity of their complexes.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Henry Smith
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Yanbing Chen
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan 4111, Australia
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, New South Wales 2006, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
5
|
Kaya B, Smith H, Chen Y, Azad MG, M Russell T, Richardson V, Bernhardt PV, Dharmasivam M, Richardson DR. Targeting lysosomes by design: novel N-acridine thiosemicarbazones that enable direct detection of intracellular drug localization and overcome P-glycoprotein (Pgp)-mediated resistance. Chem Sci 2024:d4sc04339a. [PMID: 39165729 PMCID: PMC11331336 DOI: 10.1039/d4sc04339a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024] Open
Abstract
Innovative N-acridine thiosemicarbazones (NATs) were designed along with their iron(iii), copper(ii), and zinc(ii) complexes. Lysosomal targeting was promoted by specifically incorporating the lysosomotropic Pgp substrate, acridine, into the thiosemicarbazone scaffold to maintain the tridentate N, N, S-donor system. The acridine moiety enables a significant advance in thiosemicarbazone design, since: (1) it enables tracking of the drugs by confocal microscopy using its inherent fluorescence; (2) it is lysosomotropic enabling lysosomal targeting; and (3) as acridine is a P-glycoprotein (Pgp) substrate, it facilitates lysosomal targeting, resulting in the drug overcoming Pgp-mediated resistance. These new N-acridine analogues are novel, and this is the first time that acridine has been specifically added to the thiosemicarbazone framework to achieve the three important properties above. These new agents displayed markedly greater anti-proliferative activity against resistant Pgp-expressing cells than very low Pgp-expressing cells. The anti-proliferative activity of NATs against multiple Pgp-positive cancer cell-types (colon, lung, and cervical carcinoma) was abrogated by the third generation Pgp inhibitor, Elacridar, and also Pgp siRNA that down-regulated Pgp. Confocal microscopy demonstrated that low Pgp in KB31 (-Pgp) cells resulted in acridine's proclivity for DNA intercalation promoting NAT nuclear-targeting. In contrast, high Pgp in KBV1 (+Pgp) cells led to NAT lysosomal sequestration, preventing its nuclear localisation. High Pgp expression in KBV1 (+Pgp) cells resulted in co-localization of NATs with the lysosomal marker, LysoTracker™, that was significantly (p < 0.001) greater than the positive control, the di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) Zn(ii) complex, [Zn(DpC)2]. Incorporation of acridine into the thiosemicarbazone scaffold led to Pgp-mediated transport into lysosomes to overcome Pgp-resistance.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Henry Smith
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Yanbing Chen
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland Brisbane 4072 Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University Nathan Brisbane 4111 Queensland Australia
- Department of Pathology and Bosch Institute, Molecular Pharmacology and Pathology Program, University of Sydney Sydney New South Wales Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine Nagoya 466-8550 Japan
| |
Collapse
|
6
|
Kaya B, Gholam Azad M, Suleymanoglu M, Harmer JR, Wijesinghe TP, Richardson V, Zhao X, Bernhardt PV, Dharmasivam M, Richardson DR. Isosteric Replacement of Sulfur to Selenium in a Thiosemicarbazone: Promotion of Zn(II) Complex Dissociation and Transmetalation to Augment Anticancer Efficacy. J Med Chem 2024; 67:12155-12183. [PMID: 38967641 DOI: 10.1021/acs.jmedchem.4c00884] [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: 07/06/2024]
Abstract
We implemented isosteric replacement of sulfur to selenium in a novel thiosemicarbazone (PPTP4c4mT) to create a selenosemicarbazone (PPTP4c4mSe) that demonstrates potentiated anticancer efficacy and selectivity. Their design specifically incorporated cyclohexyl and styryl moieties to sterically inhibit the approach of their Fe(III) complexes to the oxy-myoglobin heme plane. Importantly, in contrast to the Fe(III) complexes of the clinically trialed thiosemicarbazones Triapine, COTI-2, and DpC, the Fe(III) complexes of PPTP4c4mT and PPTP4c4mSe did not induce detrimental oxy-myoglobin oxidation. Furthermore, PPTP4c4mSe demonstrated more potent antiproliferative activity than the homologous thiosemicarbazone, PPTP4c4mT, with their selectivity being superior or similar, respectively, to the clinically trialed thiosemicarbazone, COTI-2. An advantageous property of the selenosemicarbazone Zn(II) complexes relative to their thiosemicarbazone analogues was their greater transmetalation to Cu(II) complexes in lysosomes. This latter effect probably promoted their antiproliferative activity. Both ligands down-regulated multiple key receptors that display inter-receptor cooperation that leads to aggressive and resistant breast cancer.
Collapse
Affiliation(s)
- Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Mediha Suleymanoglu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Fatih, Istanbul 34093, Turkey
| | - Jeffrey R Harmer
- Centre for Advanced Imaging, University of Queensland, Brisbane 4072, Australia
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
7
|
Kumar A, Chaudhary A, Sonker H, Subhadarshini S, Jolly MK, Singh RG. Zinc(II) Complexes of SIRTi1/2 Analogues Transmetallating with Copper Ions and Inducing ROS Mediated Paraptosis. ACS ORGANIC & INORGANIC AU 2024; 4:319-328. [PMID: 38855338 PMCID: PMC11157505 DOI: 10.1021/acsorginorgau.3c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 06/11/2024]
Abstract
As the SIRTi analogue series (HL1-HL6) show potent antitumor activity in vitro, we synthesized their corresponding zinc(II) complexes (ZnL1-ZnL6) and investigated their potential as anticancer agents. The Zn(II) complexes showed substantially greater cytotoxicity than HL1-HL6 alone in several cancer cell-types. Notably, distinct structure-activity relationships confirmed the significance of tert-butyl (ZnL2) pharmacophore inclusion in their activity. ZnL2 complexes were found to transmetalate with copper ions inside cells, causing the formation of redox-active copper complexes that induced reactive oxygen species (ROS) production, mitochondrial membrane depolarization, ATP decay, and cell death. This is the first study to exhibit Zn(II) complexes that mediate their activity via transmetalation with copper ions to undergo paraptosis cell death pathway. To further confirm if the SIRT1/2 inhibitory property of SIRTi analogues is conserved, a docking simulation study is performed. The binding affinity and specific interactions of the Cu(II) complex obtained after transmetalation with ZnL2 were found to be higher for SIRT2 (K i = 0.06 μM) compared to SIRT1 (K i = 0.25 μM). Thus, the concurrent regulation of several biological targets using a single drug has been shown to have synergistic therapeutic effects, which are crucial for the effective treatment of cancer.
Collapse
Affiliation(s)
- Ashwini Kumar
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Ayushi Chaudhary
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | - Himanshu Sonker
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| | | | - Mohit K. Jolly
- Department
of Bioengineering, Indian Institute of Science, Bangalore 560012, India
| | - Ritika Gautam Singh
- Department
of Chemistry, Indian Institute of Technology
Kanpur, Kanpur 208016, India
| |
Collapse
|
8
|
Podolski-Renić A, Čipak Gašparović A, Valente A, López Ó, Bormio Nunes JH, Kowol CR, Heffeter P, Filipović NR. Schiff bases and their metal complexes to target and overcome (multidrug) resistance in cancer. Eur J Med Chem 2024; 270:116363. [PMID: 38593587 DOI: 10.1016/j.ejmech.2024.116363] [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: 02/27/2024] [Revised: 03/15/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024]
Abstract
Overcoming multidrug resistance (MDR) is one of the major challenges in cancer therapy. In this respect, Schiff base-related compounds (bearing a R1R2CNR3 bond) gained high interest during the past decades. Schiff bases are considered privileged ligands for various reasons, including the easiness of their preparation and the possibility to form complexes with almost all transition metal ions. Schiff bases and their metal complexes exhibit many types of biological activities and are used for the treatment and diagnosis of various diseases. Until now, 13 Schiff bases have been investigated in clinical trials for cancer treatment and hypoxia imaging. This review represents the first collection of Schiff bases and their complexes which demonstrated MDR-reversal activity. The areas of drug resistance covered in this article involve: 1) Modulation of ABC transporter function, 2) Targeting lysosomal ABCB1 overexpression, 3) Circumvention of ABC transporter-mediated drug efflux by alternative routes of drug uptake, 4) Selective activity against MDR cancer models (collateral sensitivity), 5) Targeting GSH-detoxifying systems, 6) Overcoming apoptosis resistance by inducing necrosis and paraptosis, 7) Reactivation of mutated p53, 8) Restoration of sensitivity to DNA-damaging anticancer therapy, and 9) Overcoming drug resistance through modulation of the immune system. Through this approach, we would like to draw attention to Schiff bases and their metal complexes representing highly interesting anticancer drug candidates with the ability to overcome MDR.
Collapse
Affiliation(s)
- Ana Podolski-Renić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Serbia
| | | | - Andreia Valente
- Centro de Química Estrutural and Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, Lisboa, Portugal
| | - Óscar López
- Departamento de Química Organica, Facultad de Química, Universidad de Sevilla, Sevilla, Spain
| | - Julia H Bormio Nunes
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria; Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| | - Nenad R Filipović
- Department of Chemistry and Biochemistry, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
9
|
Jiménez-Pérez A, Fernández-Fariña S, Pedrido R, García-Tojal J. Desulfurization of thiosemicarbazones: the role of metal ions and biological implications. J Biol Inorg Chem 2024; 29:3-31. [PMID: 38148423 DOI: 10.1007/s00775-023-02037-7] [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: 07/06/2023] [Accepted: 09/26/2023] [Indexed: 12/28/2023]
Abstract
Thiosemicarbazones are biologically active substances whose structural formula is formed by an azomethine, an hydrazine, and a thioamide fragments, to generate a R2C=N-NR-C(=S)-NR2 backbone. These compounds often act as ligands to generate highly stable metal-organic complexes. In certain experimental conditions, however, thiosemicarbazones undergo reactions leading to the cleavage of the chain. Sometimes, the breakage involves desulfurization processes. The present work summarizes the different chemical factors that influence the desulfurization reactions of thiosemicarbazones, such as pH, the presence of oxidant reactants or the establishment of redox processes as those electrochemically induced, the effects of the solvent, the temperature, and the electromagnetic radiation. Many of these reactions require coordination of thiosemicarbazones to metal ions, even those present in the intracellular environment. The nature of the products generated in these reactions, their detection in vivo and in vitro, together with the relevance for the biological activity of these compounds, mainly as antineoplastic agents, is discussed.
Collapse
Affiliation(s)
- Alondra Jiménez-Pérez
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001, Burgos, Spain
| | - Sandra Fernández-Fariña
- Departamento de Química Inorgánica, Facultade de Química, Campus Vida, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Rosa Pedrido
- Departamento de Química Inorgánica, Facultade de Química, Campus Vida, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| | - Javier García-Tojal
- Departamento de Química, Facultad de Ciencias, Universidad de Burgos, 09001, Burgos, Spain.
| |
Collapse
|
10
|
Kwan K, Castro-Sandoval O, Ma B, Martelino D, Saffari A, Liu XL, Orvain C, Mellitzer G, Gaiddon C, Storr T. Altering relative metal-binding affinities in multifunctional Metallochaperones for mutant p53 reactivation. J Inorg Biochem 2024; 251:112433. [PMID: 38043136 DOI: 10.1016/j.jinorgbio.2023.112433] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023]
Abstract
The p53 protein plays a major role in cancer prevention, and over 50% of cancer diagnoses can be attributed to p53 malfunction. p53 incorporates a structural Zn site that is required for proper protein folding and function, and in many cases point mutations can result in loss of the Zn2+ ion, destabilization of the tertiary structure, and eventual amyloid aggregation. Herein, we report a series of compounds designed to act as small molecule stabilizers of mutant p53, and feature Zn-binding fragments to chaperone Zn2+ to the metal depleted site and restore wild-type (WT) function. Many Zn metallochaperones (ZMCs) have been shown to generate intracellular reactive oxygen species (ROS), likely by chelating redox-active metals such as Fe2+/3+ and Cu+/2+ and undergoing associated Fenton chemistry. High levels of ROS can result in off-target effects and general toxicity, and thus, careful tuning of ligand Zn2+ affinity, in comparison to the affinity for other endogenous metals, is important for selective mutant p53 targeting. In this work we show that by using carboxylate donors in place of pyridine we can change the relative Zn2+/Cu2+ binding ability in a series of ligands, and we investigate the impact of donor group changes on metallochaperone activity and overall cytotoxicity in two mutant p53 cancer cell lines (NUGC3 and SKGT2).
Collapse
Affiliation(s)
- Kalvin Kwan
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Omar Castro-Sandoval
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Benjamin Ma
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Diego Martelino
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Ashkan Saffari
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Xi Lan Liu
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Christophe Orvain
- Inserm UMR_S 1113, Université de Strasbourg, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France
| | - Georg Mellitzer
- Inserm UMR_S 1113, Université de Strasbourg, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France
| | - Christian Gaiddon
- Inserm UMR_S 1113, Université de Strasbourg, Molecular Mechanisms of Stress Response and Pathologies, Strasbourg, France.
| | - Tim Storr
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada.
| |
Collapse
|
11
|
Dharmasivam M, Kaya B, Wijesinghe TP, Richardson V, Harmer JR, Gonzalvez MA, Lewis W, Azad MG, Bernhardt PV, Richardson DR. Differential transmetallation of complexes of the anti-cancer thiosemicarbazone, Dp4e4mT: effects on anti-proliferative efficacy, redox activity, oxy-myoglobin and oxy-hemoglobin oxidation. Chem Sci 2024; 15:974-990. [PMID: 38239703 PMCID: PMC10793205 DOI: 10.1039/d3sc05723b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
The di-2-pyridylthiosemicarbazone (DpT) analogs demonstrate potent and selective anti-proliferative activity against human tumors. The current investigation reports the synthesis and chemical and biological characterization of the Fe(iii), Co(iii), Ni(ii), Cu(ii), Zn(ii), Ga(iii), and Pd(ii) complexes of the promising second generation DpT analog, di-2-pyridylketone-4-ethyl-4-methyl-3-thiosemicarbazone (Dp4e4mT). These studies demonstrate that the Dp4e4mT Co(iii), Ni(ii), and Pd(ii) complexes display distinct biological activity versus those with Cu(ii), Zn(ii), and Ga(iii) regarding anti-proliferative efficacy against cancer cells and a detrimental off-target effect involving oxidation of oxy-myoglobin (oxy-Mb) and oxy-hemoglobin (oxy-Hb). With regards to anti-proliferative activity, the Zn(ii) and Ga(iii) Dp4e4mT complexes demonstrate facile transmetallation with Cu(ii), resulting in efficacy against tumor cells that is strikingly similar to the Dp4e4mT Cu(ii) complex (IC50: 0.003-0.006 μM and 72 h). Relative to the Zn(ii) and Ga(iii) Dp4e4mT complexes, the Dp4e4mT Ni(ii) complex demonstrates kinetically slow transmetallation with Cu(ii) and intermediate anti-proliferative effects (IC50: 0.018-0.076 μM after 72 h). In contrast, the Co(iii) and Pd(ii) complexes demonstrate poor anti-proliferative activity (IC50: 0.262-1.570 μM after 72 h), probably due to a lack of transmetallation with Cu(ii). The poor efficacy of the Dp4e4mT Co(iii), Ni(ii), and Pd(ii) complexes to transmetallate with Fe(iii) markedly suppresses the oxidation of oxy-Mb and oxy-Hb. In contrast, the 2 : 1 Dp4e4mT: Cu(ii), Zn(ii), and Ga(iii) complexes demonstrate facile reactions with Fe(iii), leading to the redox active Dp4e4mT Fe(iii) complex and oxy-Mb and oxy-Hb oxidation. This study demonstrates the key role of differential transmetallation of Dp4e4mT complexes that has therapeutic ramifications for their use as anti-cancer agents.
Collapse
Affiliation(s)
- Mahendiran Dharmasivam
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney Sydney New South Wales 2006 Australia
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University Nathan Brisbane Queensland 4111 Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University Nathan Brisbane Queensland 4111 Australia
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University Nathan Brisbane Queensland 4111 Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University Nathan Brisbane Queensland 4111 Australia
| | - Jeffrey R Harmer
- Centre for Advanced Imaging, University of Queensland Brisbane Queensland 4072 Australia
| | - Miguel A Gonzalvez
- School of Chemistry and Molecular Biosciences, University of Queensland Brisbane Queensland 4072 Australia
| | - William Lewis
- Department of Chemistry, University of Sydney New South Wales 2006 Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University Nathan Brisbane Queensland 4111 Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland Brisbane Queensland 4072 Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology and Bosch Institute, The University of Sydney Sydney New South Wales 2006 Australia
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University Nathan Brisbane Queensland 4111 Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine Nagoya 466-8550 Japan
| |
Collapse
|
12
|
Xu S, Luo W, Zhu M, Zhao L, Gao L, Liang H, Zhang Z, Yang F. Human Serum Albumin-Platinum(II) Agent Nanoparticles Inhibit Tumor Growth Through Multimodal Action Against the Tumor Microenvironment. Mol Pharm 2024; 21:346-357. [PMID: 38015620 DOI: 10.1021/acs.molpharmaceut.3c00881] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
To overcome the limitations of traditional platinum (Pt)-based drugs and further improve the targeting ability and therapeutic efficacy in vivo, we proposed to design a human serum albumin (HSA)-Pt agent complex nanoparticle (NP) for cancer treatment by multimodal action against the tumor microenvironment. We not only synthesized a series of Pt(II) di-2-pyridone thiosemicarbazone compounds and obtained a Pt(II) agent [Pt(Dp44mT)Cl] with significant anticancer activity but also successfully constructed a novel HSA-Pt(Dp44mT) complex nanoparticle delivery system. The structure of the HSA-Pt(Dp44mT) complex revealed that Pt(Dp44mT)Cl binds to the IIA subdomain of HSA and coordinates with His-242. The HSA-His242-Pt-Dp44mT NPs had an obvious effect on the inhibition of tumor growth, which was superior to that of Dp44mT and Pt(Dp44mT)Cl, and they had almost no toxicity. In addition, the HSA-His242-Pt-Dp44mT NPs were found to kill cancer cells by inducing apoptosis, autophagy, and inhibiting angiogenesis.
Collapse
Affiliation(s)
- Shihang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Weicong Luo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Lei Zhao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Lijuan Gao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| |
Collapse
|
13
|
Wang X, Zhu M, Li S, Xu G, Zhang Z, Yang F. Novel mono-, bi-, tri- and tetra-nuclear copper complexes that inhibit tumor growth through apoptosis and anti-angiogenesis. J Inorg Biochem 2024; 250:112403. [PMID: 37866112 DOI: 10.1016/j.jinorgbio.2023.112403] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
To develop the next-generation metal agents for efficiently inhibiting tumor growth, a series of novel mononuclear, binuclear and trinuclear copper (Cu) thiophene-2-formaldehyde thiosemicarbazone complexes and a tetranuclear Cu 1,2,4-triazole-derived complex have been synthesized and their structure-activity relationships have been studied. The trinucleated Cu complex showed the strongest inhibitory activity against T24 cells among all the Cu complexes. Its antitumor effect in vivo was superior to that of cisplatin, with reduced side effects. Further studies on the antitumor mechanism have showed that Cu complexes not only induced apoptosis of cancer cells but also inhibited tumor angiogenesis by inhibiting the migration and invasion of vascular endothelial cells, blocking the cell cycle in the G1 phase, and inducing autophagy.
Collapse
Affiliation(s)
- Xiaojun Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China.
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China.
| |
Collapse
|
14
|
Wijesinghe TP, Kaya B, Gonzálvez MA, Harmer JR, Gholam Azad M, Bernhardt PV, Dharmasivam M, Richardson DR. Steric Blockade of Oxy-Myoglobin Oxidation by Thiosemicarbazones: Structure-Activity Relationships of the Novel PPP4pT Series. J Med Chem 2023; 66:15453-15476. [PMID: 37922410 DOI: 10.1021/acs.jmedchem.3c01612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2023]
Abstract
The di-2-pyridylketone thiosemicarbazones demonstrated marked anticancer efficacy, prompting progression of DpC to clinical trials. However, DpC induced deleterious oxy-myoglobin oxidation, stifling development. To address this, novel substituted phenyl thiosemicarbazone (PPP4pT) analogues and their Fe(III), Cu(II), and Zn(II) complexes were prepared. The PPP4pT analogues demonstrated potent antiproliferative activity (IC50: 0.009-0.066 μM), with the 1:1 Cu:L complexes showing the greatest efficacy. Substitutions leading to decreased redox potential of the PPP4pT:Cu(II) complexes were associated with higher antiproliferative activity, while increasing potential correlated with increased redox activity. Surprisingly, there was no correlation between redox activity and antiproliferative efficacy. The PPP4pT:Fe(III) complexes attenuated oxy-myoglobin oxidation significantly more than the clinically trialed thiosemicarbazones, Triapine, COTI-2, and DpC, or earlier thiosemicarbazone series. Incorporation of phenyl- and styryl-substituents led to steric blockade, preventing approach of the PPP4pT:Fe(III) complexes to the heme plane and its oxidation. The 1:1 Cu(II):PPP4pT complexes were inert to transmetalation and did not induce oxy-myoglobin oxidation.
Collapse
Affiliation(s)
- Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Miguel A Gonzálvez
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Jeffrey R Harmer
- Centre for Advanced Imaging, University of Queensland, Brisbane 4072, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane 4072, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane 4111, Australia
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
15
|
Ciaffaglione V, Rizzarelli E. Carnosine, Zinc and Copper: A Menage a Trois in Bone and Cartilage Protection. Int J Mol Sci 2023; 24:16209. [PMID: 38003398 PMCID: PMC10671046 DOI: 10.3390/ijms242216209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Dysregulated metal homeostasis is associated with many pathological conditions, including arthritic diseases. Osteoarthritis and rheumatoid arthritis are the two most prevalent disorders that damage the joints and lead to cartilage and bone destruction. Recent studies show that the levels of zinc (Zn) and copper (Cu) are generally altered in the serum of arthritis patients. Therefore, metal dyshomeostasis may reflect the contribution of these trace elements to the disease's pathogenesis and manifestations, suggesting their potential for prognosis and treatment. Carnosine (Car) also emerged as a biomarker in arthritis and exerts protective and osteogenic effects in arthritic joints. Notably, its zinc(II) complex, polaprezinc, has been recently proposed as a drug-repurposing candidate for bone fracture healing. On these bases, this review article aims to provide an overview of the beneficial roles of Cu and Zn in bone and cartilage health and their potential application in tissue engineering. The effects of Car and polaprezinc in promoting cartilage and bone regeneration are also discussed. We hypothesize that polaprezinc could exchange Zn for Cu, present in the culture media, due to its higher sequestering ability towards Cu. However, future studies should unveil the potential contribution of Cu in the beneficial effects of polaprezinc.
Collapse
Affiliation(s)
- Valeria Ciaffaglione
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy
| | - Enrico Rizzarelli
- Institute of Crystallography, National Council of Research (CNR), P. Gaifami 18, 95126 Catania, Italy
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
16
|
Castillo CE, Gonzálvez MA, Algarra AG, Fernández-Trujillo MJ, Ferrer M, Martínez M, Basallote MG. Fe(II) complexes of pyridine-substituted thiosemicarbazone ligands as catalysts for oxidations with hydrogen peroxide. Dalton Trans 2023; 52:14606-14612. [PMID: 37786386 DOI: 10.1039/d3dt02442c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
The reaction of three [FeII(TSC)2] complexes, where TSC is a pyridine-substituted thiosemicarbazone of the HDpT or HBpT families, with H2O2 in acetonitrile solution does not result in the accumulation of the corresponding [FeIII(TSC)2]+ complexes. Instead, a mixture of diamagnetic low-spin FeII species is generated. According to the MS spectra, those species result from the sequential addition of up to five oxygen atoms to the complex. This capability for the addition of oxygen atoms suggested that oxygen atom transfer to external substrates may be possible, and these TSC complexes were tested in the oxidation of thioanisole and styrene with H2O2. As hypothesized, the complexes are active in both the oxidation of thioanisole to its sulfoxide and styrene to benzaldehyde, with time scales indicating the participation of the species containing added oxygen atoms. Interestingly, the free thiosemicarbazone ligands and the [Zn(Dp44mT)2] complex also catalyse the selective sulfoxidation of thioanisole, but they are ineffective in catalysing styrene oxidation to benzaldehyde. These findings open up new directions for the development of thiosemicarbazone-based metal catalysts for oxidation processes.
Collapse
Affiliation(s)
- Carmen E Castillo
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Instituto de Biomoléculas (INBIO), Universidad de Cádiz, Apartado 40, E-11510 Puerto Real, Cádiz, Spain.
| | - Miguel A Gonzálvez
- Departament de Química Inorgànica i Orgànica, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès 1-11, E-08028 Barcelona, Spain.
| | - Andrés G Algarra
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Instituto de Biomoléculas (INBIO), Universidad de Cádiz, Apartado 40, E-11510 Puerto Real, Cádiz, Spain.
| | - M Jesús Fernández-Trujillo
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Instituto de Biomoléculas (INBIO), Universidad de Cádiz, Apartado 40, E-11510 Puerto Real, Cádiz, Spain.
| | - Montserrat Ferrer
- Departament de Química Inorgànica i Orgànica, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès 1-11, E-08028 Barcelona, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Manuel Martínez
- Departament de Química Inorgànica i Orgànica, Secció de Química Inorgànica, Universitat de Barcelona, Martí i Franquès 1-11, E-08028 Barcelona, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, 08028 Barcelona, Spain
| | - Manuel G Basallote
- Departamento de Ciencia de los Materiales e Ingeniería Metalúrgica y Química Inorgánica, Instituto de Biomoléculas (INBIO), Universidad de Cádiz, Apartado 40, E-11510 Puerto Real, Cádiz, Spain.
| |
Collapse
|
17
|
Shehadeh-Tout F, Milioli HH, Roslan S, Jansson PJ, Dharmasivam M, Graham D, Anderson R, Wijesinghe T, Azad MG, Richardson DR, Kovacevic Z. Innovative Thiosemicarbazones that Induce Multi-Modal Mechanisms to Down-Regulate Estrogen-, Progesterone-, Androgen- and Prolactin-Receptors in Breast Cancer. Pharmacol Res 2023:106806. [PMID: 37244387 DOI: 10.1016/j.phrs.2023.106806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 05/29/2023]
Abstract
The estrogen receptor-α (ER-α) is a key driver of breast cancer (BC) and the ER-antagonist, tamoxifen, is a central pillar of BC treatment. However, cross-talk between ER-α, other hormone and growth factor receptors enables development of de novo resistance to tamoxifen. Herein, we mechanistically dissect the activity of a new class of anti-cancer agents that inhibit multiple growth factor receptors and down-stream signaling for the treatment of ER-positive BC. Using RNA sequencing and comprehensive protein expression analysis, we examined the activity of di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC), on the expression and activation of hormone and growth factor receptors, co-factors, and key resistance pathways in ER-α-positive BC. DpC differentially regulated 106 estrogen-response genes, and this was linked to decreased mRNA levels of 4 central hormone receptors involved in BC pathogenesis, namely ER, progesterone receptor (PR), androgen receptor (AR), and prolactin receptor (PRL-R). Mechanistic investigation demonstrated that due to DpC and Dp44mT binding metal ions, these agents caused a pronounced decrease in ER-α, AR, PR, and PRL-R protein expression. DpC and Dp44mT also inhibited activation and down-stream signaling of the epidermal growth factor (EGF) family receptors, and expression of co-factors that promote ER-α transcriptional activity, including SRC3, NF-κB p65, and SP1. In vivo, DpC was highly tolerable and effectively inhibited ER-α-positive BC growth. Through bespoke, non-hormonal, multi-modal mechanisms, Dp44mT and DpC decrease the expression of PR, AR, PRL-R, and tyrosine kinases that act with ER-α to promote BC, constituting an innovative therapeutic approach.
Collapse
Affiliation(s)
- Faten Shehadeh-Tout
- School of Medical Sciences, University of Sydney, NSW 2006, Australia; Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Heloisa H Milioli
- Connie Johnson Breast Cancer Research Laboratory, Garvan Institute of Medical Research, NSW 2010 Australia
| | - Suraya Roslan
- Metastasis Research Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg Vic 3084, Australia
| | - Patric J Jansson
- Cancer Drug Resistance and Stem Cell Program, School of Medical Sciences, University of Sydney, NSW 2006, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Dinny Graham
- Breast Cancer Group, The Westmead Institute for Medical Research and Westmead Clinical School, University of Sydney, NSW 2145 Australia
| | - Robin Anderson
- Metastasis Research Laboratory, Olivia Newton-John Cancer Research Institute, Heidelberg Vic 3084, Australia; School of Cancer Medicine, La Trobe University, Bundoora, 3086, Victoria, Australia
| | - Tharushi Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Zaklina Kovacevic
- School of Medical Sciences, University of Sydney, NSW 2006, Australia; Department of Physiology, School of Biomedical Sciences, University of NSW, NSW 2052 Australia.
| |
Collapse
|
18
|
Selyutina OY, Timoshnikov VA, Polyakov NE, Kontoghiorghes GJ. Metal Complexes of Omadine ( N-Hydroxypyridine-2-thione): Differences of Antioxidant and Pro-Oxidant Behavior in Light and Dark Conditions with Possible Toxicity Implications. Molecules 2023; 28:molecules28104210. [PMID: 37241949 DOI: 10.3390/molecules28104210] [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: 04/23/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Omadine or N-hydroxypyridine-2-thione and its metal complexes are widely used in medicine and show bactericidal, fungicidal, anticancer, and photochemical activity. The redox activity of omadine complexes with iron, copper, and zinc on lipid peroxidation under light and dark conditions has been investigated. The monitoring of the oxidation of linoleic acid micelles, resembling a model of lipid membrane, was carried out using nuclear magnetic resonance (1H-NMR). It has been shown that the omadine-zinc complex can induce the oxidation of linoleic acid under light irradiation, whereas the complexes with iron and copper are photochemically stable. All the chelating complexes of omadine appear to be redox-inactive in the presence of hydrogen peroxide under dark conditions. These findings suggest that omadine can demonstrate antioxidant behavior in processes involving reactive oxygen species generation induced by transition metals (Fenton and photo-Fenton reactions). However, the omadine complex with zinc, which is widely used in shampoos and ointments, is photochemically active and may cause oxidative cell membrane damage when exposed to light, with possible implications to health.
Collapse
Affiliation(s)
- Olga Yu Selyutina
- Institute of Chemical Kinetics & Combustion, 630090 Novosibirsk, Russia
- Institute of Solid Chemistry and Mechanochemistry, 630090 Novosibirsk, Russia
| | | | - Nikolay E Polyakov
- Institute of Chemical Kinetics & Combustion, 630090 Novosibirsk, Russia
- Institute of Solid Chemistry and Mechanochemistry, 630090 Novosibirsk, Russia
| | - George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, CY-3021 Limassol, Cyprus
| |
Collapse
|
19
|
Cortezon-Tamarit F, Song K, Kuganathan N, Arrowsmith RL, Mota Merelo de Aguiar SR, Waghorn PA, Brookfield A, Shanmugam M, Collison D, Ge H, Kociok-Köhn G, Pourzand C, Dilworth JR, Pascu SI. Structural and Functional Diversity in Rigid Thiosemicarbazones with Extended Aromatic Frameworks: Microwave-Assisted Synthesis and Structural Investigations. ACS OMEGA 2023; 8:16047-16079. [PMID: 37179648 PMCID: PMC10173449 DOI: 10.1021/acsomega.2c08157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/10/2023] [Indexed: 05/15/2023]
Abstract
The long-standing interest in thiosemicarbazones (TSCs) has been largely driven by their potential toward theranostic applications including cellular imaging assays and multimodality imaging. We focus herein on the results of our new investigations into: (a) the structural chemistry of a family of rigid mono(thiosemicarbazone) ligands characterized by extended and aromatic backbones and (b) the formation of their corresponding thiosemicarbazonato Zn(II) and Cu(II) metal complexes. The synthesis of new ligands and their Zn(II) complexes was performed using a rapid, efficient and straightforward microwave-assisted method which superseded their preparation by conventional heating. We describe hereby new microwave irradiation protocols that are suitable for both imine bond formation reactions in the thiosemicabazone ligand synthesis and for Zn(II) metalation reactions. The new thiosemicarbazone ligands, denoted HL, mono(4-R-3-thiosemicarbazone)quinone, and their corresponding Zn(II) complexes, denoted ZnL2, mono(4-R-3-thiosemicarbazone)quinone, where R = H, Me, Ethyl, Allyl, and Phenyl, quinone = acenapthnenequinone (AN), aceanthrenequinone (AA), phenanthrenequinone (PH), and pyrene-4,5-dione (PY) were isolated and fully characterized spectroscopically and by mass spectrometry. A plethora of single crystal X-ray diffraction structures were obtained and analyzed and the geometries were also validated by DFT calculations. The Zn(II) complexes presented either distorted octahedral geometry or tetrahedral arrangements of the O/N/S donors around the metal center. The modification of the thiosemicarbazide moiety at the exocyclic N atoms with a range of organic linkers was also explored, opening the way to bioconjugation protocols for these compounds. The radiolabeling of these thiosemicarbazones with 64Cu was achieved under mild conditions for the first time: this cyclotron-available radioisotope of copper (t1/2 = 12.7 h; β+ 17.8%; β- 38.4%) is well-known for its proficiency in positron emission tomography (PET) imaging and for its theranostic potential, on the basis of the preclinical and clinical cancer research of established bis(thiosemicarbazones), such as the hypoxia tracer 64Cu-labeled copper(diacetyl-bis(N4-methylthiosemicarbazone)], [64Cu]Cu(ATSM). Our labeling reactions proceeded in high radiochemical incorporation (>80% for the most sterically unencumbered ligands) showing promise of these species as building blocks for theranostics and synthetic scaffolds for multimodality imaging probes. The corresponding "cold" Cu(II) metalations were also performed under the mild conditions mimicking the radiolabeling protocols. Interestingly, room temperature or mild heating led to Cu(II) incorporation in the 1:1, as well as 1:2 metal: ligand ratios in the new complexes, as evident from extensive mass spectrometry investigations backed by EPR measurements, and the formation of Cu(L)2-type species prevails, especially for the AN-Ph thiosemicarbazone ligand (L-). The cytotoxicity levels of a selection of ligands and Zn(II) complexes in this class were further tested in commonly used human cancer cell lines (HeLa, human cervical cancer cells, and PC-3, human prostate cancer cells). Tests showed that their IC50 levels are comparable to that of the clinical drug cis-platin, evaluated under similar conditions. The cellular internalizations of the selected ZnL2-type compounds Zn(AN-Allyl)2, Zn(AA-Allyl)2, Zn(PH-Allyl)2, and Zn(PY-Allyl)2 were evaluated in living PC-3 cells using laser confocal fluorescent spectroscopy and these experiments showed exclusively cytoplasmic distributions.
Collapse
Affiliation(s)
| | - Kexin Song
- Department
of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United
Kingdom
| | - Navaratnarajah Kuganathan
- Department
of Materials, Imperial College London, Royal School of Mines, Exhibition
Road, London SW7 2AZ, U.K.
| | - Rory L. Arrowsmith
- Department
of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United
Kingdom
| | | | - Philip A. Waghorn
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Adam Brookfield
- Department
of Chemistry, and Photon Science Institute, The University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| | - Muralidharan Shanmugam
- Department
of Chemistry, and Photon Science Institute, The University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| | - David Collison
- Department
of Chemistry, and Photon Science Institute, The University of Manchester, Oxford Road, Manchester M13 9PL, United Kingdom
| | - Haobo Ge
- Department
of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United
Kingdom
- Department
of Life Sciences, University of Bath, Bath BA2 7AY, U.K.
| | - Gabriele Kociok-Köhn
- Department
of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United
Kingdom
| | - Charareh Pourzand
- Department
of Life Sciences, University of Bath, Bath BA2 7AY, U.K.
- Centre of
Therapeutic Innovation, University of Bath, Bath BA2 7AY, U.K.
| | - Jonathan Robin Dilworth
- Department
of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United
Kingdom
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Sofia Ioana Pascu
- Department
of Chemistry, University of Bath, Claverton Down, Bath, BA2 7AY, United
Kingdom
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, United Kingdom
- Centre of
Therapeutic Innovation, University of Bath, Bath BA2 7AY, U.K.
| |
Collapse
|
20
|
Banerjee A, Patra SA, Sahu G, Sciortino G, Pisanu F, Garribba E, Carvalho MFNN, Correia I, Pessoa JC, Reuter H, Dinda R. A Series of Non-Oxido V IV Complexes of Dibasic ONS Donor Ligands: Solution Stability, Chemical Transformations, Protein Interactions, and Antiproliferative Activity. Inorg Chem 2023; 62:7932-7953. [PMID: 37154533 PMCID: PMC10367067 DOI: 10.1021/acs.inorgchem.3c00753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
A series of mononuclear non-oxido vanadium(IV) complexes, [VIV(L1-4)2] (1-4), featuring tridentate bi-negative ONS chelating S-alkyl/aryl-substituted dithiocarbazate ligands H2L1-4, are reported. All the synthesized non-oxido VIV compounds are characterized by elemental analysis, spectroscopy (IR, UV-vis, and EPR), ESI-MS, as well as electrochemical techniques (cyclic voltammetry). Single-crystal X-ray diffraction studies of 1-3 reveal that the mononuclear non-oxido VIV complexes show distorted octahedral (1 and 2) or trigonal prismatic (3) arrangement around the non-oxido VIV center. EPR and DFT data indicate the coexistence of mer and fac isomers in solution, and ESI-MS results suggest a partial oxidation of [VIV(L1-4)2] to [VV(L1-4)2]+ and [VVO2(L1-4)]-; therefore, all these three complexes are plausible active species. Complexes 1-4 interact with bovine serum albumin (BSA) with a moderate binding affinity, and docking calculations reveal non-covalent interactions with different regions of BSA, particularly with Tyr, Lys, Arg, and Thr residues. In vitro cytotoxic activity of all complexes is assayed against the HT-29 (colon cancer) and HeLa (cervical cancer) cells and compared with the NIH-3T3 (mouse embryonic fibroblast) normal cell line by MTT assay and DAPI staining. The results suggest that complexes 1-4 are cytotoxic in nature and induce cell death in the cancer cell lines by apoptosis and that a mixture of VIV, VV, and VVO2 species could be responsible for the biological activity.
Collapse
Affiliation(s)
- Atanu Banerjee
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Sushree Aradhana Patra
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Gurunath Sahu
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Giuseppe Sciortino
- Institute of Chemical Research of Catalonia (ICIQ), The Barcelona Institute of Science and Technology, Tarragona 43007, Spain
| | - Federico Pisanu
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, Sassari I-07100, Italy
| | - Eugenio Garribba
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, Sassari I-07100, Italy
| | - M Fernanda N N Carvalho
- Centro de Química Estrutural and Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, Lisboa 1049-001, Portugal
| | - Isabel Correia
- Centro de Química Estrutural and Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, Lisboa 1049-001, Portugal
| | - João Costa Pessoa
- Centro de Química Estrutural and Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, Lisboa 1049-001, Portugal
| | - Hans Reuter
- Institute of Chemistry of New Materials, University of Osnabrück, Barbarastraße 6, Osnabruck 49069, Germany
| | - Rupam Dinda
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| |
Collapse
|
21
|
Miller JJ, Kwan K, Blanchet A, Orvain C, Mellitzer G, Smith J, Lento C, Nouchikian L, Omoregbee-Leichnitz S, Sabatou M, Wilson D, Gaiddon C, Storr T. Multifunctional metallochaperone modifications for targeting subsite cavities in mutant p53-Y220C. J Inorg Biochem 2023; 242:112164. [PMID: 36871418 DOI: 10.1016/j.jinorgbio.2023.112164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023]
Abstract
The p53 protein, known as the 'guardian of the genome', plays an important role in cancer prevention. Unfortunately, p53 mutations result in compromised activity with over 50% of cancers resulting from point mutations to p53. There is considerable interest in mutant p53 reactivation, with the development of small-molecule reactivators showing promise. We have focused our efforts on the common p53 mutation Y220C, which causes protein unfolding, aggregation, and can result in the loss of a structural Zn from the DNA-binding domain. In addition, the Y220C mutant creates a surface pocket that can be stabilized using small molecules. We previously reported the bifunctional ligand L5 as a Zn metallochaperone and reactivator of the p53-Y220C mutant. Herein we report two new ligands L5-P and L5-O that are designed to act as Zn metallochaperones and non-covalent binders in the Y220C mutant pocket. For L5-P the distance between the Zn-binding di-(2-picolyl)amine function and the pocket-binding diiodophenol was extended in comparison to L5, while for L5-O we extended the pocket-binding moiety via attachment of an alkyne function. While both new ligands displayed similar Zn-binding affinity to L5, neither acted as efficient Zn-metallochaperones. However, the new ligands exhibited significant cytotoxicity in the NCI-60 cell line screen as well as in the NUGC3 Y220C mutant cell line. We identified that the primary mode of cytotoxicity is likely reactive oxygen species (ROS) generation for L5-P and L5-O, in comparison to mutant p53 reactivation for L5, demonstrating that subtle changes to the ligand scaffold can change the toxicity pathway.
Collapse
Affiliation(s)
- Jessica J Miller
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Kalvin Kwan
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Anaïs Blanchet
- Laboratory Streinth, Université de Strasbourg; Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
| | - Christophe Orvain
- Laboratory Streinth, Université de Strasbourg; Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
| | - Georg Mellitzer
- Laboratory Streinth, Université de Strasbourg; Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France
| | - Jason Smith
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Cristina Lento
- York University, Chemistry Department, 6 Thompson Road, Toronto, Ontario, M3J 1L3, Canada
| | - Lucienne Nouchikian
- York University, Chemistry Department, 6 Thompson Road, Toronto, Ontario, M3J 1L3, Canada
| | | | - Marie Sabatou
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada
| | - Derek Wilson
- York University, Chemistry Department, 6 Thompson Road, Toronto, Ontario, M3J 1L3, Canada
| | - Christian Gaiddon
- Laboratory Streinth, Université de Strasbourg; Inserm, UMR_S 1113 IRFAC, 67200 Strasbourg, France.
| | - Tim Storr
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, Canada.
| |
Collapse
|
22
|
Zhang Z, Zhang J, Yang T, Li S, Xu G, Liang H, Yang F. Developing an Anticancer Platinum(II) Compound Based on the Uniqueness of Human Serum Albumin. J Med Chem 2023; 66:5669-5684. [PMID: 37071741 DOI: 10.1021/acs.jmedchem.3c00001] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
To develop the next-generation Pt drug with remarkable activity and low toxicity to maximally inhibit tumor growth, we optimized a Pt(II) thiosemicarbazone compound (C4) with remarkable cytotoxicity to SK-N-MC cells and then constructed a new human serum albumin-C4 (HSA-C4) complex delivery system. The in vivo results showed that C4 and the HSA-C4 complex have remarkable therapeutic efficiency and almost no toxicity; they induced apoptosis and inhibited tumor angiogenesis. This system showed potential as a practical Pt drug. This study could pave the way for developing next-generation dual-targeted Pt drugs and achieving their targeting therapy for cancer.
Collapse
Affiliation(s)
- Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Juzheng Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Tongfu Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| |
Collapse
|
23
|
Du LQ, Zhang TY, Huang XM, Xu Y, Tan MX, Huang Y, Chen Y, Qin QP. Synthesis and anticancer mechanisms of zinc(II)-8-hydroxyquinoline complexes with 1,10-phenanthroline ancillary ligands. Dalton Trans 2023; 52:4737-4751. [PMID: 36942929 DOI: 10.1039/d3dt00150d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Twenty new zinc(II) complexes with 8-hydroxyquinoline (H-Q1-H-Q6) in the presence of 1,10-phenanthroline derivatives (D1-D10) were synthesized and formulated as [Zn(Q1)2(D1)] (DQ1), [Zn(Q2)2(D2)]·CH3OH (DQ2), [Zn(Q1)2(D3)] (DQ3), [Zn(Q1)2(D4)] (DQ4), [Zn(Q3)2(D5)] (DQ5), [Zn(Q3)2(D4)] (DQ6), [Zn(Q4)2(D5)]·CH3OH (DQ7), [Zn(Q4)2(D6)] (DQ8), [Zn(Q4)2(D3)]·CH3OH (DQ9), [Zn(Q4)2(D1)]·H2O (DQ10), [Zn(Q5)2(D4)] (DQ11), [Zn(Q6)2(D6)]·CH3OH (DQ12), [Zn(Q5)2(D2)]·5CH3OH·H2O (DQ13), [Zn(Q5)2(D7)]·CH3OH (DQ14), [Zn(Q5)2(D8)]·CH2Cl2 (DQ15), [Zn(Q5)2(D9)] (DQ16), [Zn(Q5)2(D1)] (DQ17), [Zn(Q5)2(D5)] (DQ18), [Zn(Q5)2(D10)]·CH2Cl2 (DQ19) and [Zn(Q5)2(D3)] (DQ20). They were characterized using multiple techniques. The cytotoxicity of DQ1-DQ20 was screened using human cisplatin-resistant SK-OV-3/DDP ovarian cancer (SK-OV-3CR) cells and normal hepatocyte (HL-7702) cells. Complex DQ6 showed low IC50 values (2.25 ± 0.13 μM) on SK-OV-3CR cells, more than 3.0-8.0 times more cytotoxic than DQ1-DQ5 and DQ7-DQ20 (≥6.78 μM), and even 22.2 times more cytotoxic than the standard cisplatin, the corresponding free H-Q1-H-Q6 and D1-D10 alone (>50 μM). As a comparison, DQ1-DQ20 displayed nontoxic rates against healthy HL-7702 cells. Furthermore, DQ6 and DQ11 induced significant apoptosis via mitophagy pathways. DQ6 also significantly inhibited tumor growth in an in vivo SK-OV-3-xenograft model (ca. 49.7%). Thus, DQ6 may serve as a lead complex for the discovery of new antitumor agents.
Collapse
Affiliation(s)
- Ling-Qi Du
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Tian-Yu Zhang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Xiao-Mei Huang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Yue Xu
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Ming-Xiong Tan
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Yan Huang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Yuan Chen
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| |
Collapse
|
24
|
Stojičkov M, Zlatar M, Pio Mazzeo P, Bacchi A, Radovanović D, Stevanović N, Jevtović M, Novaković I, Anđelković K, Sladić D, Čobeljić B, Gruden M. The interplay between spin states, geometries and biological activity of Fe(III) and Mn(II) complexes with thiosemicarbazone. Polyhedron 2023. [DOI: 10.1016/j.poly.2023.116389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
25
|
Dharmasivam M, Kaya B, Wijesinghe T, Gholam Azad M, Gonzálvez MA, Hussaini M, Chekmarev J, Bernhardt PV, Richardson DR. Designing Tailored Thiosemicarbazones with Bespoke Properties: The Styrene Moiety Imparts Potent Activity, Inhibits Heme Center Oxidation, and Results in a Novel "Stealth Zinc(II) Complex". J Med Chem 2023; 66:1426-1453. [PMID: 36649565 DOI: 10.1021/acs.jmedchem.2c01600] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A novel, potent, and selective antitumor agent, namely (E)-3-phenyl-1-(2-pyridinyl)-2-propen-1-one 4,4-dimethyl-3-thiosemicarbazone (PPP44mT), and its analogues were synthesized and characterized and displayed strikingly distinctive properties. This activity was mediated by the inclusion of a styrene moiety, which through steric and electrochemical mechanisms prevented deleterious oxy-myoglobin or oxy-hemoglobin oxidation relative to other potent thiosemicarbazones, i.e., di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) or di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT). Structure-activity relationship analysis demonstrated specific tuning of PPP44mT electrochemistry further inhibited oxy-myoglobin or oxy-hemoglobin oxidation. Both PPP44mT and its Cu(II) complexes showed conspicuous almost immediate cytotoxicity against SK-N-MC tumor cells (within 3 h). In contrast, [Zn(PPP44mT)2] demonstrated a pronounced delay in activity, taking 48 h before marked antiproliferative efficacy was apparent. As such, [Zn(PPP44mT)2] was designated as a "stealth Zn(II) complex" that overcomes the near immediate cytotoxicity of PPP44mT or its copper complexes. Upon examination of the suppression of oncogenic signaling, [Zn(PPP44mT)2] was superior at inhibiting cyclin D1 expression compared to DpC or Dp44mT.
Collapse
Affiliation(s)
- Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan4111, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan4111, Australia.,Department of Chemistry, Istanbul University-Cerrahpasa, Avcilar, 34320Istanbul, Turkey
| | - Tharushi Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan4111, Australia
| | - Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan4111, Australia
| | - Miguel A Gonzálvez
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane4072, Australia
| | - Mohammad Hussaini
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan4111, Australia
| | - Jason Chekmarev
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan4111, Australia
| | - Paul V Bernhardt
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane4072, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan4111, Australia.,Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya466-8550, Japan
| |
Collapse
|
26
|
New Iron Metabolic Pathways and Chelation Targeting Strategies Affecting the Treatment of All Types and Stages of Cancer. Int J Mol Sci 2022; 23:ijms232213990. [PMID: 36430469 PMCID: PMC9696688 DOI: 10.3390/ijms232213990] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
There is new and increasing evidence from in vitro, in vivo and clinical studies implicating the pivotal role of iron and associated metabolic pathways in the initiation, progression and development of cancer and in cancer metastasis. New metabolic and toxicity mechanisms and pathways, as well as genomic, transcription and other factors, have been linked to cancer and many are related to iron. Accordingly, a number of new targets for iron chelators have been identified and characterized in new anticancer strategies, in addition to the classical restriction of/reduction in iron supply, the inhibition of transferrin iron delivery, the inhibition of ribonucleotide reductase in DNA synthesis and high antioxidant potential. The new targets include the removal of excess iron from iron-laden macrophages, which affects anticancer activity; the modulation of ferroptosis; ferritin iron removal and the control of hyperferritinemia; the inhibition of hypoxia related to the role of hypoxia-inducible factor (HIF); modulation of the function of new molecular species such as STEAP4 metalloreductase and the metastasis suppressor N-MYC downstream-regulated gene-1 (NDRG1); modulation of the metabolic pathways of oxidative stress damage affecting mitochondrial function, etc. Many of these new, but also previously known associated iron metabolic pathways appear to affect all stages of cancer, as well as metastasis and drug resistance. Iron-chelating drugs and especially deferiprone (L1), has been shown in many recent studies to fulfill the role of multi-target anticancer drug linked to the above and also other iron targets, and has been proposed for phase II trials in cancer patients. In contrast, lipophilic chelators and their iron complexes are proposed for the induction of ferroptosis in some refractory or recurring tumors in drug resistance and metastasis where effective treatments are absent. There is a need to readdress cancer therapy and include therapeutic strategies targeting multifactorial processes, including the application of multi-targeting drugs involving iron chelators and iron-chelator complexes. New therapeutic protocols including drug combinations with L1 and other chelating drugs could increase anticancer activity, decrease drug resistance and metastasis, improve treatments, reduce toxicity and increase overall survival in cancer patients.
Collapse
|
27
|
Zhang Q, Shao J, Wang J, Gong XJ, Liu WX, Wang S, Zhang Y, Yang S, Zhang QS, Wei JX, Tian JL. Antitumor effects of new glycoconjugated Pt II agents dual-targeting GLUT1 and Pgp proteins. Dalton Trans 2022; 51:16082-16092. [PMID: 36178270 DOI: 10.1039/d2dt02455a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel and highly efficient dual-targeting PtII system was designed to improve the drug delivery capacity and selectivity in cancer treatment. The dual-targeting monofunctional PtII complexes (1-8) having glycosylated pendants as tridentated ligand were achieved by introducing glycosylation modification in the thioaminocarbazone compounds with potential lysosomal targeting ability. The structures and stability of 1-8 were further established by various techniques. Molecular docking studies showed that 2 was efficiently docked into glucose transporters protein 1 (GLUT1) and P-glycoprotein (Pgp) proteins with the optimal CDocker-interaction-energy of -64.84 and -48.85 kcal mol-1. Complex 2 with higher protein binding capacity demonstrated significant and broad-spectrum antitumor efficacy in vitro, even exhibiting a half maximal inhibitory concentration (IC50) value (∼10 μM) than cisplatin (∼17 μM) against human lung adenocarcinoma cells (A549). The inhibitor experiment revealed GLUT-mediated uptake of 2, and the subcellular localization experiment in A549 also proved that 2 could be localized in the lysosome, thereby causing cell apoptosis. Moreover, cellular thermal shift assay (CETSA) confirmed the binding of 2 with the target proteins of GLUT1 and Pgp. The above results indicated that 2 represents a potential anticancer candidate with dual-targeting functions.
Collapse
Affiliation(s)
- Qiang Zhang
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Jia Shao
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin 300192, PR China. .,National Health Commission's Key Laboratory of Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Jin Wang
- Outpatient Office, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Xian-Jin Gong
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Wei-Xing Liu
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Shan Wang
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin 300192, PR China.
| | - Yi Zhang
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin 300192, PR China. .,National Health Commission's Key Laboratory of Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Shuang Yang
- Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Medical College of Nankai University, Tianjin 300071, PR China
| | - Quan-Sheng Zhang
- Tianjin Key Laboratory of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, PR China
| | - Jin-Xia Wei
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Jin-Lei Tian
- College of Chemistry, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
28
|
Zhou Z, Du LQ, Huang XM, Zhu LG, Wei QC, Qin QP, Bian H. Novel glycosylation zinc(II)-cryptolepine complexes perturb mitophagy pathways and trigger cancer cell apoptosis and autophagy in SK-OV-3/DDP cells. Eur J Med Chem 2022; 243:114743. [PMID: 36116236 DOI: 10.1016/j.ejmech.2022.114743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/04/2022]
Abstract
With the aim of shedding some light on the mechanism of action of zinc(II) complexes in antiproliferative processes and molecular signaling pathways, three novel glycosylated zinc(II)-cryptolepine complexes, i.e., [Zn(QA1)Cl2] (Zn(QA1)), [Zn(QA2)Cl2] (Zn(QA2)), and [Zn(QA3)Cl2] (Zn(QA3)), were prepared by conjugating a glucose moiety with cryptolepine, followed by complexation of the resulting glycosylated cryptolepine compounds N-((1-(2-morpholinoethyl)-1H-1,2,3-triazol-4-yl)methyl)-benzofuro[3,2-b]quinolin-11-amine (QA1), 2-(4-((benzofuro[3,2-b]quinolin-11-ylamino)methyl)-1H-1,2,3-triazol-1-yl)ethan-1-ol (QA2), and (2S,3S,4R,5R,6S)-2-(4-((benzofuro[3,2-b]quinolin-11-ylamino)-methyl)-1H-1,2,3-triazol-1-yl)-6-(hydroxymethyl)tetrahydro-2H-pyran-3,4,5-triol (QA3) with zinc(II), and their anticancer activity was evaluated. In MTT assays, Zn(QA1)-Zn(QA3) were more active against cisplatin-resistant ovarian SK-OV-3/DDP cancer cells (SK-OV-3cis) than ZnCl2 and the QA1-QA3 ligands, with IC50 values of 1.81 ± 0.50, 2.92 ± 0.32, and 1.01 ± 0.11 μM, respectively. Complexation of glycosylated cryptolepine QA3 with zinc(II) increased the antiproliferative activity of the ligand, suggesting that Zn(QA3) could act as a chaperone to deliver the active ligand intracellularly, in contrast with other cryptolepine metal complexes previously reported. In vivo and in vitro investigations suggested that Zn(QA3) exhibited enhanced anticancer activity with treatment effects comparable to those of the clinical drug cisplatin. Furthermore, Zn(QA1)-Zn(QA3) triggered SK-OV-3cis cell apoptosis through mitophagy pathways in the order Zn(QA1) > Zn(QA1) > Zn(QA2). These results demonstrate the potential of glycosylated zinc(II)-cryptolepine complexes for the development of chemotherapy drugs against cisplatin-resistant SK-OV-3cis cells.
Collapse
Affiliation(s)
- Zhen Zhou
- Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi University for Nationalities (Guangxi Minzu University), Nanning, 530006, China; Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin, 537000, PR China
| | - Ling-Qi Du
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin, 537000, PR China
| | - Xiao-Mei Huang
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin, 537000, PR China
| | - Li-Gang Zhu
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin, 537000, PR China.
| | - Qiao-Chang Wei
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin, 537000, PR China
| | - Qi-Pin Qin
- Guangxi Key Lab of Agricultural Resources Chemistry and Biotechnology, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin, 537000, PR China; State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, PR China.
| | - Hedong Bian
- Guangxi Key Laboratory of Chemistry and Engineering of Forest Products, Guangxi Collaborative Innovation Center for Chemistry and Engineering of Forest Products, School of Chemistry and Chemical Engineering, Guangxi University for Nationalities (Guangxi Minzu University), Nanning, 530006, China.
| |
Collapse
|
29
|
Zhang SH, Wang ZF, Tan H. Novel zinc(II)−curcumin molecular probes bearing berberine and jatrorrhizine derivatives as potential mitochondria-targeting anti-neoplastic drugs. Eur J Med Chem 2022; 243:114736. [DOI: 10.1016/j.ejmech.2022.114736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/04/2022]
|
30
|
Zheng Y, Li B, Ai Y, Chen M, Zheng X, Qi J. Synthesis, crystal structures and anti-cancer mechanism of Cu(II) complex derived from 2-acetylpyrazine thiosemicarbazone. J COORD CHEM 2022. [DOI: 10.1080/00958972.2022.2111660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Yunyun Zheng
- Medical School of Pingdingshan University, Pingdingshan, Henan, China
| | - Bin Li
- Medical School of Pingdingshan University, Pingdingshan, Henan, China
| | - Yu Ai
- Medical School of Pingdingshan University, Pingdingshan, Henan, China
| | - Mengyao Chen
- Medical School of Pingdingshan University, Pingdingshan, Henan, China
| | - Xinhua Zheng
- Medical School of Pingdingshan University, Pingdingshan, Henan, China
| | - Jinxu Qi
- Medical School of Pingdingshan University, Pingdingshan, Henan, China
| |
Collapse
|
31
|
Falcone E, Ritacca AG, Hager S, Schueffl H, Vileno B, El Khoury Y, Hellwig P, Kowol CR, Heffeter P, Sicilia E, Faller P. Copper-Catalyzed Glutathione Oxidation is Accelerated by the Anticancer Thiosemicarbazone Dp44mT and Further Boosted at Lower pH. J Am Chem Soc 2022; 144:14758-14768. [PMID: 35929814 PMCID: PMC9389589 DOI: 10.1021/jacs.2c05355] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
![]()
Glutathione (GSH) is the most abundant thiol in mammalian
cells
and plays a crucial role in maintaining redox cellular homeostasis.
The thiols of two GSH molecules can be oxidized to the disulfide GSSG.
The cytosolic GSH/GSSG ratio is very high (>100), and its reduction
can lead to apoptosis or necrosis, which are of interest in cancer
research. CuII ions are very efficient oxidants of thiols,
but with an excess of GSH, CuIn(GS)m clusters are formed, in which CuI is very slowly reoxidized by O2 at pH 7.4 and
even more slowly at lower pH. Here, the aerobic oxidation of GSH by
CuII was investigated at different pH values in the presence
of the anticancer thiosemicarbazone Dp44mT, which accumulates in lysosomes
and induces lysosomal membrane permeabilization in a Cu-dependent
manner. The results showed that CuII-Dp44mT catalyzes GSH
oxidation faster than CuII alone at pH 7.4 and hence accelerates
the production of very reactive hydroxyl radicals. Moreover, GSH oxidation
and hydroxyl radical production by CuII-Dp44mT were accelerated
at the acidic pH found in lysosomes. To decipher this unusually faster
thiol oxidation at lower pH, density functional theory (DFT) calculations,
electrochemical and spectroscopic studies were performed. The results
suggest that the acceleration is due to the protonation of CuII-Dp44mT on the hydrazinic nitrogen, which favors the rate-limiting
reduction step without subsequent dissociation of the CuI intermediate. Furthermore, preliminary biological studies in cell
culture using the proton pump inhibitor bafilomycin A1 indicated that
the lysosomal pH plays a role in the activity of CuII-Dp44mT.
Collapse
Affiliation(s)
- Enrico Falcone
- Institut de Chimie (UMR 7177), University of Strasbourg - CNRS, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Alessandra G Ritacca
- Department of Chemistry and Chemical Technologies, Università della Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende, (CS), Italy
| | - Sonja Hager
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Hemma Schueffl
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Bertrand Vileno
- Institut de Chimie (UMR 7177), University of Strasbourg - CNRS, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Youssef El Khoury
- Laboratoire de bioélectrochimie et spectroscopie, UMR 7140, CNRS, Université de Strasbourg, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Petra Hellwig
- Laboratoire de bioélectrochimie et spectroscopie, UMR 7140, CNRS, Université de Strasbourg, 4 Rue Blaise Pascal, 67081 Strasbourg, France
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 42, 1090 Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Emilia Sicilia
- Department of Chemistry and Chemical Technologies, Università della Calabria, Ponte P. Bucci, 87036 Arcavacata di Rende, (CS), Italy
| | - Peter Faller
- Institut de Chimie (UMR 7177), University of Strasbourg - CNRS, 4 Rue Blaise Pascal, 67081 Strasbourg, France.,Institut Universitaire de France (IUF), 1 rue Descartes, 75231 Paris, France
| |
Collapse
|
32
|
Thiosemicarbazones Can Act Synergistically with Anthracyclines to Downregulate CHEK1 Expression and Induce DNA Damage in Cell Lines Derived from Pediatric Solid Tumors. Int J Mol Sci 2022; 23:ijms23158549. [PMID: 35955683 PMCID: PMC9369312 DOI: 10.3390/ijms23158549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/10/2022] Open
Abstract
Anticancer therapy by anthracyclines often leads to the development of multidrug resistance (MDR), with subsequent treatment failure. Thiosemicarbazones have been previously suggested as suitable anthracycline partners due to their ability to overcome drug resistance through dual Pgp-dependent cytotoxicity-inducing effects. Here, we focused on combining anthracyclines (doxorubicin, daunorubicin, and mitoxantrone) and two thiosemicarbazones (DpC and Dp44mT) for treating cell types derived from the most frequent pediatric solid tumors. Our results showed synergistic effects for all combinations of treatments in all tested cell types. Nevertheless, further experiments revealed that this synergism was independent of Pgp expression but rather resulted from impaired DNA repair control leading to cell death via mitotic catastrophe. The downregulation of checkpoint kinase 1 (CHEK1) expression by thiosemicarbazones and the ability of both types of agents to induce double-strand breaks in DNA may explain the Pgp-independent synergism between anthracyclines and thiosemicarbazones. Moreover, the concomitant application of these agents was found to be the most efficient approach, achieving the strongest synergistic effect with lower concentrations of these drugs. Overall, our study identified a new mechanism that offers an avenue for combining thiosemicarbazones with anthracyclines to treat tumors regardless the Pgp status.
Collapse
|
33
|
Oliveri IP, Consiglio G, Munzi G, Failla S, Di Bella S. Deaggregation properties and transmetalation studies of a zinc(II) salen-type Schiff-base complex. Dalton Trans 2022; 51:11859-11867. [PMID: 35876090 DOI: 10.1039/d2dt01448c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This paper reports the synthesis and the deaggregation properties of a Lewis acidic Zn(II) salen-type Schiff-base complex derivative from diaminomaleonitrile and a systematic detailed study of its transmetalation with other metal ions in solution. In a solution of non-coordinating solvents, the complex is in a dimeric form, while in coordinating solvents or upon addition of a Lewis base it is stabilized as monomeric adducts. Experiments done in two solvents with different Lewis basicities indicate a major role of the stability of the starting adduct in transmetalation. Thus, using nitrate or perchlorate salts, acetonitrile solutions of the complex give an immediate and complete transmetalation with Cu2+, while with Co2+ and Ni2+ a much slower transmetalation rate is observed. Instead, using chloride salts a fast and complete transmetalation is observed for divalent ions of the first transition series (Mn2+, Fe2+, Co2+, Ni2+, Cu2+), indicating the role of the chloride in stabilizing the transition state of the transmetalation. On the other hand, DMF solutions of the complex are less prone to transmetalation, according with the greater basicity of the solvent and, hence, the greater stability of the related adducts with the complex. Therefore, the nature of the solvent and the counteranion allow controlling the transmetalation process of this Zn(II) Schiff-base complex.
Collapse
Affiliation(s)
- Ivan Pietro Oliveri
- Dipartimento di Scienze Chimiche, Università di Catania, I-95125 Catania, Italy.
| | - Giuseppe Consiglio
- Dipartimento di Scienze Chimiche, Università di Catania, I-95125 Catania, Italy.
| | - Gabriella Munzi
- Dipartimento di Scienze Chimiche, Università di Catania, I-95125 Catania, Italy.
| | - Salvatore Failla
- Dipartimento di Scienze Chimiche, Università di Catania, I-95125 Catania, Italy.
| | - Santo Di Bella
- Dipartimento di Scienze Chimiche, Università di Catania, I-95125 Catania, Italy.
| |
Collapse
|
34
|
Kim J, Park A, Hwang J, Zhao X, Kwak J, Kim HW, Ku M, Yang J, Kim TI, Jeong KS, Choi U, Lee H, Shin SJ. KS10076, a chelator for redox-active metal ions, induces ROS-mediated STAT3 degradation in autophagic cell death and eliminates ALDH1 + stem cells. Cell Rep 2022; 40:111077. [PMID: 35858554 DOI: 10.1016/j.celrep.2022.111077] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 03/17/2022] [Accepted: 06/20/2022] [Indexed: 12/21/2022] Open
Abstract
Redox-active metal ions are pivotal for rapid metabolism, proliferation, and aggression across cancer types, and this presents metal chelation as an attractive cancer cell-targeting strategy. Here, we identify a metal chelator, KS10076, as a potent anti-cancer drug candidate. A metal-bound KS10076 complex with redox potential for generating hydrogen peroxide and superoxide anions induces intracellular reactive oxygen species (ROS). The elevation of ROS by KS10076 promotes the destabilization of signal transducer and activator of transcription 3, removes aldehyde dehydrogenase isoform 1-positive cancer stem cells, and subsequently induces autophagic cell death. Bioinformatic analysis of KS10076 susceptibility in pan-cancer cells shows that KS10076 potentially targets cancer cells with increased mitochondrial function. Furthermore, patient-derived organoid models demonstrate that KS10076 efficiently represses cancer cells with active KRAS, and fluorouracil resistance, which suggests clinical advantages.
Collapse
Affiliation(s)
- Jaehee Kim
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Areum Park
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Jieon Hwang
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Xianghua Zhao
- Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jaesung Kwak
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Hyun Woo Kim
- Chemical Data-Driven Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Minhee Ku
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Severance of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jaemoon Yang
- Department of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Severance of Radiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Tae Il Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyu-Sung Jeong
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Uyeong Choi
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyuk Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea; Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Sang Joon Shin
- Department of Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Songdang Institute for Cancer Research, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
35
|
Qi J, Zheng Y, Li B, Ai Y, Chen M, Zheng X. Pyridoxal hydrochloride thiosemicarbazones with copper ions inhibit cell division via Topo-I and Topo-IIɑ. J Inorg Biochem 2022; 232:111816. [DOI: 10.1016/j.jinorgbio.2022.111816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 12/17/2022]
|
36
|
Richardson DR, Azad MG, Afroz R, Richardson V, Dharmasivam M. Thiosemicarbazones reprogram pancreatic cancer bidirectional oncogenic signaling between cancer cells and stellate cells to suppress desmoplasia. Future Med Chem 2022; 14:1005-1017. [PMID: 35670251 DOI: 10.4155/fmc-2022-0050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023] Open
Abstract
Standard treatments have shown dismal activity against pancreatic cancer (PC), due in part to the development of a dense stroma (desmoplasia). This perspective discusses the development of the di-2-pyridylketone thiosemicarbazones that overcomes bidirectional oncogenic signaling between PC cells and pancreatic stellate cells (PSCs), which is critical for desmoplasia development. This activity is induced by the up-regulation of the metastasis suppressor, N-myc downstream-regulated gene-1 (NDRG1), which inhibits oncogenic signaling via HGF, IGF-1 and Sonic Hedgehog pathway. More recent studies have deciphered additional pathways including those mediated by Wnt and tenascin C that are secreted by PSCs to activate β-catenin and YAP/TAZ signaling in PC cells. Suppression of bidirectional signaling between cell types presents a unique therapeutic opportunity.
Collapse
Affiliation(s)
- D R Richardson
- Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia
- Department of Pathology & Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - M Gholam Azad
- Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia
| | - R Afroz
- Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia
| | - V Richardson
- Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia
| | - M Dharmasivam
- Centre for Cancer Cell Biology & Drug Discovery, Griffith Institute of Drug Discovery, Griffith University & School of Environment & Science (N34), Nathan, Brisbane, Queensland, 4111, Australia
| |
Collapse
|
37
|
Miller JJ, Kwan K, Gaiddon C, Storr T. A role for bioinorganic chemistry in the reactivation of mutant p53 in cancer. J Biol Inorg Chem 2022; 27:393-403. [PMID: 35488931 DOI: 10.1007/s00775-022-01939-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/11/2022] [Indexed: 12/19/2022]
Abstract
Metal ion dysregulation has been implicated in a number of diseases from neurodegeneration to cancer. While defective metal ion transport mechanisms are known to cause specific diseases of genetic origin, the role of metal dysregulation in many diseases has yet to be elucidated due to the complicated function (both good and bad!) of metal ions in the body. A breakdown in metal ion speciation can manifest in several ways from increased reactive oxygen species (ROS) generation to an increase in protein misfolding and aggregation. In this review, we will discuss the role of Zn in the proper function of the p53 protein in cancer. The p53 protein plays a critical role in the prevention of genome mutations via initiation of apoptosis, DNA repair, cell cycle arrest, anti-angiogenesis, and senescence pathways to avoid propagation of damaged cells. p53 is the most frequently mutated protein in cancer and almost all cancers exhibit malfunction along the p53 pathway. Thus, there has been considerable effort dedicated to restoring normal p53 expression and activity to mutant p53. This includes understanding the relative populations of the Zn-bound and Zn-free p53 in wild-type and mutant forms, and the development of metallochaperones to re-populate the Zn binding site to restore mutant p53 activity. Parallels will be made to the development of multifunctional metal binding agents for modulating the aggregation of the amyloid-beta peptide in Alzheimer's Disease (AD).
Collapse
Affiliation(s)
- Jessica J Miller
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Kalvin Kwan
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Christian Gaiddon
- Inserm UMR_S1113, IRFAC, team Streinth, Strasbourg University, Strasbourg, France
| | - Tim Storr
- Department of Chemistry, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
38
|
Gou Y, Jia X, Hou LX, Deng JG, Huang GJ, Jiang HW, Yang F. Dithiocarbazate-Fe III, -Co III, -Ni II, and -Zn II Complexes: Design, Synthesis, Structure, and Anticancer Evaluation. J Med Chem 2022; 65:6677-6689. [PMID: 35446587 DOI: 10.1021/acs.jmedchem.1c02186] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Non-platinum-metal complexes show great potential as anticancer agents. Herein, a series of dithiocarbazate non-Pt-metal complexes, including [FeIII(L)2]·Cl·2H2O 1, [CoIII(L)2]·NO3·2.5H2O 2, [NiII(L)2] 3, and [ZnII(L)2] 4, have been designed and evaluated for their efficacy as antineoplastic agents. Among them, complex 2 exhibited higher anticancer efficacy than complexes 1, 3, 4, and cisplatin against several cancer cell lines. Hemolysis assays revealed that complex 2 showed comparable hemolysis with cisplatin. In vivo anticancer evaluations showed that complex 2 could retard tumor xenograft growth effectively with low systemic toxicity. Further studies revealed that complex 2 suppressed cancer cells by triggering multiple mechanisms involving the simultaneous inhibition of mitochondria and glycolytic bioenergetics. Overall, our study provides new insights into the anticancer mechanism of Co complexes, which can be used as a good strategy to overcome the flexibility of cancer cells to chemotherapy adaptation.
Collapse
Affiliation(s)
- Yi Gou
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Xiaoying Jia
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Norma University, Guilin 541004, Guangxi, China
| | - Li Xia Hou
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Jun Gang Deng
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Guo Jin Huang
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | - Hao Wen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
| | - Feng Yang
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China.,State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Norma University, Guilin 541004, Guangxi, China
| |
Collapse
|
39
|
Calatayud DG, Neophytou S, Nicodemou E, Giuffrida SG, Ge H, Pascu SI. Nano-Theranostics for the Sensing, Imaging and Therapy of Prostate Cancers. Front Chem 2022; 10:830133. [PMID: 35494646 PMCID: PMC9039169 DOI: 10.3389/fchem.2022.830133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/16/2022] [Indexed: 01/28/2023] Open
Abstract
We highlight hereby recent developments in the emerging field of theranostics, which encompasses the combination of therapeutics and diagnostics in a single entity aimed for an early-stage diagnosis, image-guided therapy as well as evaluation of therapeutic outcomes of relevance to prostate cancer (PCa). Prostate cancer is one of the most common malignancies in men and a frequent cause of male cancer death. As such, this overview is concerned with recent developments in imaging and sensing of relevance to prostate cancer diagnosis and therapeutic monitoring. A major advantage for the effective treatment of PCa is an early diagnosis that would provide information for an appropriate treatment. Several imaging techniques are being developed to diagnose and monitor different stages of cancer in general, and patient stratification is particularly relevant for PCa. Hybrid imaging techniques applicable for diagnosis combine complementary structural and morphological information to enhance resolution and sensitivity of imaging. The focus of this review is to sum up some of the most recent advances in the nanotechnological approaches to the sensing and treatment of prostate cancer (PCa). Targeted imaging using nanoparticles, radiotracers and biomarkers could result to a more specialised and personalised diagnosis and treatment of PCa. A myriad of reports has been published literature proposing methods to detect and treat PCa using nanoparticles but the number of techniques approved for clinical use is relatively small. Another facet of this report is on reviewing aspects of the role of functional nanoparticles in multimodality imaging therapy considering recent developments in simultaneous PET-MRI (Positron Emission Tomography-Magnetic Resonance Imaging) coupled with optical imaging in vitro and in vivo, whilst highlighting feasible case studies that hold promise for the next generation of dual modality medical imaging of PCa. It is envisaged that progress in the field of imaging and sensing domains, taken together, could benefit from the biomedical implementation of new synthetic platforms such as metal complexes and functional materials supported on organic molecular species, which can be conjugated to targeting biomolecules and encompass adaptable and versatile molecular architectures. Furthermore, we include hereby an overview of aspects of biosensing methods aimed to tackle PCa: prostate biomarkers such as Prostate Specific Antigen (PSA) have been incorporated into synthetic platforms and explored in the context of sensing and imaging applications in preclinical investigations for the early detection of PCa. Finally, some of the societal concerns around nanotechnology being used for the detection of PCa are considered and addressed together with the concerns about the toxicity of nanoparticles–these were aspects of recent lively debates that currently hamper the clinical advancements of nano-theranostics. The publications survey conducted for this review includes, to the best of our knowledge, some of the most recent relevant literature examples from the state-of-the-art. Highlighting these advances would be of interest to the biomedical research community aiming to advance the application of theranostics particularly in PCa diagnosis and treatment, but also to those interested in the development of new probes and methodologies for the simultaneous imaging and therapy monitoring employed for PCa targeting.
Collapse
Affiliation(s)
- David G. Calatayud
- Department of Chemistry, University of Bath, Bath, United Kingdom
- Department of Electroceramics, Instituto de Ceramica y Vidrio - CSIC, Madrid, Spain
- *Correspondence: Sofia I. Pascu, ; David G. Calatayud,
| | - Sotia Neophytou
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Eleni Nicodemou
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | | | - Haobo Ge
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Sofia I. Pascu
- Department of Chemistry, University of Bath, Bath, United Kingdom
- Centre of Therapeutic Innovations, University of Bath, Bath, United Kingdom
- *Correspondence: Sofia I. Pascu, ; David G. Calatayud,
| |
Collapse
|
40
|
Novel bifluorescent Zn(II)–cryptolepine–cyclen complexes trigger apoptosis induced by nuclear and mitochondrial DNA damage in cisplatin-resistant lung tumor cells. Eur J Med Chem 2022; 238:114418. [DOI: 10.1016/j.ejmech.2022.114418] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/20/2022] [Accepted: 04/26/2022] [Indexed: 12/15/2022]
|
41
|
Mechanism of vitamin B6 benzoyl hydrazone platinum(II) complexes overcomes multidrug resistance in lung cancer. Eur J Med Chem 2022; 237:114415. [DOI: 10.1016/j.ejmech.2022.114415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022]
|
42
|
Dharmasivam M, Azad MG, Afroz R, Richardson V, Jansson PJ, Richardson DR. The thiosemicarbazone, DpC, broadly synergizes with multiple anti-cancer therapeutics and demonstrates temperature- and energy-dependent uptake by tumor cells. Biochim Biophys Acta Gen Subj 2022; 1866:130152. [DOI: 10.1016/j.bbagen.2022.130152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/25/2022] [Accepted: 04/11/2022] [Indexed: 12/22/2022]
|
43
|
Wang ZF, Nong QX, Yu HL, Qin QP, Pan FH, Tan MX, Liang H, Zhang SH. Complexes of Zn(II) with a mixed tryptanthrin derivative and curcumin chelating ligands as new promising anticancer agents. Dalton Trans 2022; 51:5024-5033. [PMID: 35274641 DOI: 10.1039/d1dt04095b] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this study, two novel curcumin (H-Cur)-tryptanthrin metal compounds-[Zn(TA)Cl2], i.e., Zn(TA), and [Zn(TA)(Cur)]Cl, i.e., Zn(TAC)-were synthesized and investigated using 5-(bis-pyridin-2-ylmethyl-amino)-pentanoic acid (6,12-dioxo-6,12-dihydro-indolo[2,1-b]quinazolin-8-yl)-amide (TA) and H-Cur as the targeting and high-activity anticancer chemotherapeutic moieties, respectively. They were then compared with the di-(2-picolyl)amine (PA) Zn(II) complex [Zn(PA)Cl2], i.e., Zn(PA). When compared with Zn(PA) and cisplatin, the IC50 values of Zn(TA) and Zn(TAC) indicated that the compounds had high cytotoxicity against A549/DDP cancer cells, implying that the H-Cur-tryptanthrin Zn(II) compounds have the potential for use as anticancer drugs. We propose the use of synthesized theragnostic H-Cur-tryptanthrin Zn(II) complexes with nuclear-targeting and DNA-damaging capabilities as a simple therapeutic strategy against tumors. The Zn(TA) and Zn(TAC) complexes could be traced via red fluorescence and were found to accumulate in the cell nuclei and induce DNA damage, cell cycle arrest, mitochondrial dysfunction, and cell apoptosis both in vitro and in vivo. In addition, Zn(TAC) exhibited a higher antiproliferative effect on A549/DDP than Zn(TA) and Zn(PA), which was undoubtedly associated with the key roles of the novel tryptanthrin derivative TA and H-Cur in the Zn(TAC) complex.
Collapse
Affiliation(s)
- Zhen-Feng Wang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, P. R. China. .,College of Chemistry, Guangdong University of Petrochemical Technology, Maoming, Guangdong, 525000, P. R. China.
| | - Qun-Xue Nong
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Hua-Lian Yu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Qi-Pin Qin
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China. .,State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China
| | - Feng-Hua Pan
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Ming-Xiong Tan
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, College of Chemistry and Food Science, Yulin Normal University, 1303 Jiaoyudong Road, Yulin 537000, PR China.
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, 15 Yucai Road, Guilin 541004, PR China
| | - Shu-Hua Zhang
- Guangxi Key Laboratory of Electrochemical and Magnetochemical Functional Materials, College of Chemistry and Bioengineering, Guilin University of Technology, Guilin, 541004, P. R. China. .,College of Chemistry, Guangdong University of Petrochemical Technology, Maoming, Guangdong, 525000, P. R. China.
| |
Collapse
|
44
|
Geleta B, Tout FS, Lim SC, Sahni S, Jansson PJ, Apte MV, Richardson DR, Kovačević Ž. Targeting Wnt/tenascin C-mediated cross talk between pancreatic cancer cells and stellate cells via activation of the metastasis suppressor NDRG1. J Biol Chem 2022; 298:101608. [PMID: 35065073 PMCID: PMC8881656 DOI: 10.1016/j.jbc.2022.101608] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/02/2022] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
A major barrier to successful pancreatic cancer (PC) treatment is the surrounding stroma, which secretes growth factors/cytokines that promote PC progression. Wnt and tenascin C (TnC) are key ligands secreted by stromal pancreatic stellate cells (PSCs) that then act on PC cells in a paracrine manner to activate the oncogenic β-catenin and YAP/TAZ signaling pathways. Therefore, therapies targeting oncogenic Wnt/TnC cross talk between PC cells and PSCs constitute a promising new therapeutic approach for PC treatment. The metastasis suppressor N-myc downstream-regulated gene-1 (NDRG1) inhibits tumor progression and metastasis in numerous cancers, including PC. We demonstrate herein that targeting NDRG1 using the clinically trialed anticancer agent di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) inhibited Wnt/TnC-mediated interactions between PC cells and the surrounding PSCs. Mechanistically, NDRG1 and DpC markedly inhibit secretion of Wnt3a and TnC by PSCs, while also attenuating Wnt/β-catenin and YAP/TAZ activation and downstream signaling in PC cells. This antioncogenic activity was mediated by direct inhibition of β-catenin and YAP/TAZ nuclear localization and by increasing the Wnt inhibitor, DKK1. Expression of NDRG1 also inhibited transforming growth factor (TGF)-β secretion by PC cells, a key mechanism by which PC cells activate PSCs. Using an in vivo orthotopic PC mouse model, we show DpC downregulated β-catenin, TnC, and YAP/TAZ, while potently increasing NDRG1 expression in PC tumors. We conclude that NDRG1 and DpC inhibit Wnt/TnC-mediated interactions between PC cells and PSCs. These results further illuminate the antioncogenic mechanism of NDRG1 and the potential of targeting this metastasis suppressor to overcome the oncogenic effects of the PC-PSC interaction.
Collapse
Affiliation(s)
- Bekesho Geleta
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Faten S Tout
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Department of Medical Laboratory Science, Faculty of Allied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Syer Choon Lim
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia
| | - Sumit Sahni
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Patric J Jansson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia; Cancer Drug Resistance & Stem Cell Program, Faculty of Medicine and Health, School of Medical Science, University of Sydney, Sydney, New South Wales, Australia
| | - Minoti V Apte
- Pancreatic Research Group, South Western Sydney Clinical School, UNSW Sydney, Sydney, New South Wales, Australia; Pancreatic Research Group, Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Des R Richardson
- Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Žaklina Kovačević
- Cancer Metastasis and Tumor Microenvironment Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia; Molecular Pharmacology and Pathology Program, Department of Pathology, University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
45
|
Selyutina OY, Kononova PA, Koshman VE, Fedenok LG, Polyakov NE. The Interplay of Ascorbic Acid with Quinones-Chelators—Influence on Lipid Peroxidation: Insight into Anticancer Activity. Antioxidants (Basel) 2022; 11:antiox11020376. [PMID: 35204258 PMCID: PMC8869476 DOI: 10.3390/antiox11020376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Ascorbic acid is a multifaceted compound that can perform both antioxidant and pro-oxidant activities in the redox reactions induced by transition metal ions, so its role in nature and especially in the human body is still the subject of debate. In the present study, we have examined the influence of ascorbic acid on lipid peroxidation in a model system that mimics the cell membrane, namely micelles of linoleic acid (LA), induced by chelate complexes of iron and copper ions with quinone-chelator 2-phenyl-4-(butylamino)-naphtholquinoline-7,12-dione (Q1). This quinone effectively generates reactive oxygen species and semiquinone radicals inside cancer cells via a cycling redox reaction. Here it was demonstrated that in the absence of quinone-chelator ascorbic acid significantly accelerates the lipid peroxidation induced by both Fe(II) and Cu(II) ions. It has been shown also that Q1 chelate complexes with Fe(II) and Cu(II) ions are redox active in the LA micelles oxidation. No effect of ascorbate was detected on the reactivity of chelate complex with Fe(II) ions. On the other hand, ascorbate performs pro-oxidant activity in Q1-Cu(II) complex induced reaction. We can conclude that ascorbate-driven redox cycling of Q1 may promote its anti-tumor activity.
Collapse
|
46
|
Aguirre AR, Parrilha GL, Louro SR, Alves OC, Diniz R, Durval F, Rocha W, Beraldo H. Structural and theoretical studies on copper(II) and zinc(II) complexes with a 9-anthraldehyde-derived thiosemicarbazone. Polyhedron 2022. [DOI: 10.1016/j.poly.2022.115724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
47
|
Sohtun WP, Kathiravan A, Asha Jhonsi M, Aashique M, Bera S, Velusamy M. Synthesis, crystal structure, BSA binding and antibacterial studies of Ni(II) complexes derived from dithiocarbazate based ligands. Inorganica Chim Acta 2022. [DOI: 10.1016/j.ica.2022.120888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Mechanistic Insights of Chelator Complexes with Essential Transition Metals: Antioxidant/Pro-Oxidant Activity and Applications in Medicine. Int J Mol Sci 2022; 23:ijms23031247. [PMID: 35163169 PMCID: PMC8835618 DOI: 10.3390/ijms23031247] [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: 12/29/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
The antioxidant/pro-oxidant activity of drugs and dietary molecules and their role in the maintenance of redox homeostasis, as well as the implications in health and different diseases, have not yet been fully evaluated. In particular, the redox activity and other interactions of drugs with essential redox metal ions, such as iron and copper, need further investigation. These metal ions are ubiquitous in human nutrition but also widely found in dietary supplements and appear to exert major effects on redox homeostasis in health, but also on many diseases of free radical pathology. In this context, the redox mechanistic insights of mainly three prototype groups of drugs, namely alpha-ketohydroxypyridines (alpha-hydroxypyridones), e.g., deferiprone, anthraquinones, e.g., doxorubicin and thiosemicarbazones, e.g., triapine and their metal complexes were examined; details of the mechanisms of their redox activity were reviewed, with emphasis on the biological implications and potential clinical applications, including anticancer activity. Furthermore, the redox properties of these three classes of chelators were compared to those of the iron chelating drugs and also to vitamin C, with an emphasis on their potential clinical interactions and future clinical application prospects in cancer, neurodegenerative and other diseases.
Collapse
|
49
|
Scrivner O, Dao L, Newell-Rogers MK, Shahandeh B, Meyskens FL, Kozawa SK, Liu-Smith F, Plascencia-Villa G, José-Yacamán M, Jia S, Chang CJ, Farmer PJ. The ionophore thiomaltol induces rapid lysosomal accumulation of copper and apoptosis in melanoma. Metallomics 2022; 14:mfab074. [PMID: 34958363 PMCID: PMC8763036 DOI: 10.1093/mtomcs/mfab074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/05/2021] [Indexed: 12/30/2022]
Abstract
In this report, we investigate the toxicity of the ionophore thiomaltol (Htma) and Cu salts to melanoma. Divalent metal complexes of thiomaltol display toxicity against A375 melanoma cell culture resulting in a distinct apoptotic response at submicromolar concentrations, with toxicity of Cu(tma)2 > Zn(tma)2 >> Ni(tma)2. In metal-chelated media, Htma treatment shows little toxicity, but the combination with supplemental CuCl2, termed Cu/Htma treatment, results in toxicity that increases with suprastoichiometric concentrations of CuCl2 and correlates with the accumulation of intracellular copper. Electron microscopy and confocal laser scanning microscopy of Cu/Htma treated cells shows a rapid accumulation of copper within lysosomes over the course of hours, concurrent with the onset of apoptosis. A buildup of ubiquitinated proteins due to proteasome inhibition is seen on the same timescale and correlates with increases of copper without additional Htma.
Collapse
Affiliation(s)
- Ottis Scrivner
- Department of Chemistry & Biochemistry, Baylor University, Waco, TX 76706, USA
| | - Long Dao
- Department of Medical Physiology, College of Medicine, Texas A&M Health Sciences Center, Bryan, TX 77807, USA
| | - M Karen Newell-Rogers
- Department of Medical Physiology, College of Medicine, Texas A&M Health Sciences Center, Bryan, TX 77807, USA
| | | | | | - Susan Kurumi Kozawa
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Feng Liu-Smith
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Germán Plascencia-Villa
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Miguel José-Yacamán
- Applied Physics and Materials Science Department and MIRA Center, Northern Arizona University, Flagstaff, AZ 86011, USA
| | - Shang Jia
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Christopher J Chang
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Patrick J Farmer
- Department of Chemistry & Biochemistry, Baylor University, Waco, TX 76706, USA
| |
Collapse
|
50
|
New thiosemicarbazone-based Zinc(II) complexes. In vitro cytotoxicity competing with cisplatin on malignant melanoma A375 cells and its relation to neuraminidase inhibition. Chem Biol Interact 2022; 351:109757. [PMID: 34848165 DOI: 10.1016/j.cbi.2021.109757] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023]
Abstract
New thiosemicarbazone-based zinc(II) complexes were synthesized to study their cytotoxicity on A375 malignant melanoma cells. The complexes containing salicylidene (Zn1a), 3-methoxy-salicylidene (Zn1b) or 4-methoxy-salicylidene (Zn1c) moiety were characterized by analytical and spectroscopic methods. Anticancer potential of the complexes was determined by MTT test and HUVEC endothelial cells line was used to comprehend the effect on normal cells. Zn1b with an IC50 of 13 μM was found to be highly cytotoxic against A375 cancer cells, more effective than cisplatin (IC50: 37 μM). Zn1a and Zn1c did not have a negative effect on cell viability in the normal cells and gave the impression that they are more advantageous than cisplatin in this respect. Further, the ability of Zn1a-c to inhibit neuraminidase enzyme and its role in cytotoxicity was discussed. The test revealed that the Zn1b with 3-methoxy substituent exhibited higher inhibition activity against the neuraminidase than the Zn1a and Zn1c as analogical to the cytotoxicity results. In neuraminidase inhibition, IC50 values of Zn1b and Zn1c were 14 and 66 μM, respectively. These concentrations were very close to the cytotoxicity concentrations for Zn1b and Zn1c. The findings may indicate the role of neuraminidase enzyme inhibition in cell death for Zn1b and Zn1c.
Collapse
|