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
|
Azad MG, Russell T, Gu X, Zhao X, Richardson V, Wijesinghe TP, Babu G, Guo X, Kaya B, Dharmasivam M, Deng Z, Richardson DR. NDRG1 and its Family Members: More than Just Metastasis Suppressor Proteins and Targets of Thiosemicarbazones. J Biol Chem 2025:110230. [PMID: 40378957 DOI: 10.1016/j.jbc.2025.110230] [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: 03/15/2025] [Revised: 04/19/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
N-Myc downstream regulated gene-1 (NDRG1) and the other three members of this family (NDRG2, 3, and 4) play various functional roles in the cellular stress response, differentiation, migration, and development. These proteins are involved in regulating key signaling proteins and pathways that are often dysregulated in cancer, such as EGFR, PI3K/AKT, c-Met, and the Wnt pathway. NDRG1 is the primary, well-examined member of the NDRG family, and is generally characterized as a metastasis suppressor that inhibits the first step in metastasis, the epithelial-mesenchymal transition. While NDRG1 is well-studied, emerging evidence suggests NDRG2, NDRG3, and NDRG4 also play significant roles in modulating oncogenic signaling and cellular homeostasis. NDRG family members are regulated by multiple mechanisms, including transcriptional control by hypoxia-inducible factors, p53, and Myc, as well as post-translational modifications such as phosphorylation, ubiquitination, and acetylation. Pharmacological targeting of the NDRG family is a therapeutic strategy against cancer. For instance, di-2-pyridylketone 4,4-dimethyl-3-thiosemicarbazone (Dp44mT) and di-2-pyridylketone-4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) have been extensively shown to up-regulate NDRG1 expression, leading to metastasis suppression and inhibition of tumor growth in multiple cancer models. Similarly, targeting NDRG2 demonstrates its pro-apoptotic and anti-proliferative effects, particularly in glioblastoma and colorectal cancer. This review provides a comprehensive analysis of the structural features, regulatory mechanisms, and biological functions of the NDRG family and their roles in cancer and neurodegenerative diseases. Additionally, NDRG1-4 are explored as therapeutic targets in oncology, focusing on recent advances in anti-cancer agents that induce the expression of these proteins. Implications for future research and clinical applications are also discussed.
Collapse
Affiliation(s)
- Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Tiffany Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xuanling Gu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xiao Zhao
- 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
| | - Tharushi P Wijesinghe
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Golap Babu
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Xinnong Guo
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Zhao Deng
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Nathan, Brisbane, 4111, Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and 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.
| |
Collapse
|
3
|
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
|
4
|
Prabhune NM, Ameen B, Prabhu S. Therapeutic potential of synthetic and natural iron chelators against ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3527-3555. [PMID: 39601820 DOI: 10.1007/s00210-024-03640-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024]
Abstract
Ferroptosis, a regulated form of cell death, is characterized by iron accumulation that results in the production of reactive oxygen species. This further causes lipid peroxidation and damage to the cellular components, eventually culminating into oxidative stress. Recent studies have highlighted the pivotal role of ferroptosis in the pathophysiological development and progression of various diseases such as β-thalassemia, hemochromatosis, and neurodegenerative disorders like AD and PD. Extensive efforts are in progress to understand the molecular mechanisms governing the role of ferroptosis in these conditions, and chelation therapy stands out as a potential approach to mitigate ferroptosis and its related implications in their development. There are currently both synthetic and natural iron chelators that are being researched for their potential as ferroptosis inhibitors. While synthetic chelators are relatively well-established and studied, their short plasma half-life and toxic side effects necessitate the exploration and identification of natural products that can act as efficient and safe iron chelators. In this review, we comprehensively discuss both synthetic and natural iron chelators as potential therapeutic strategies against ferroptosis-induced pathologies.
Collapse
Affiliation(s)
- Nupura Manish Prabhune
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bilal Ameen
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sudharshan Prabhu
- Department of Cellular and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
5
|
Kharga K, Jha S, Vishwakarma T, Kumar L. Current developments and prospects of the antibiotic delivery systems. Crit Rev Microbiol 2025; 51:44-83. [PMID: 38425122 DOI: 10.1080/1040841x.2024.2321480] [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: 07/26/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Antibiotics have remained the cornerstone for the treatment of bacterial infections ever since their discovery in the twentieth century. The uproar over antibiotic resistance among bacteria arising from genome plasticity and biofilm development has rendered current antibiotic therapies ineffective, urging the development of innovative therapeutic approaches. The development of antibiotic resistance among bacteria has further heightened the clinical failure of antibiotic therapy, which is often linked to its low bioavailability, side effects, and poor penetration and accumulation at the site of infection. In this review, we highlight the potential use of siderophores, antibodies, cell-penetrating peptides, antimicrobial peptides, bacteriophages, and nanoparticles to smuggle antibiotics across impermeable biological membranes to achieve therapeutically relevant concentrations of antibiotics and combat antimicrobial resistance (AMR). We will discuss the general mechanisms via which each delivery system functions and how it can be tailored to deliver antibiotics against the paradigm of mechanisms underlying antibiotic resistance.
Collapse
Affiliation(s)
- Kusum Kharga
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Shubhang Jha
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Tanvi Vishwakarma
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| | - Lokender Kumar
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Himachal Pradesh, India
| |
Collapse
|
6
|
Neeraj MA, Choi J. Absence of Cysteine and Iron Chelation Induces Ferroptosis in Triple-Negative Breast Cancer Cells. Breast Cancer (Auckl) 2025; 19:11782234241311012. [PMID: 39822769 PMCID: PMC11736731 DOI: 10.1177/11782234241311012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/11/2024] [Indexed: 01/19/2025] Open
Abstract
Background Ferroptosis is a recently studied form of programmed cell death characterized by lipid peroxides accumulation in the cells. This process occurs when a cell's antioxidant capacity is disturbed resulting in the inability of the cell to detoxify the toxic peroxides. Two major components that regulate ferroptosis are cysteine and iron. Objective This study aimed to determine the effect of cysteine deficiency and iron chelation on triple-negative breast cancer (TNBC) ferroptosis in a lipid-enriched microenvironment. Design The study has a laboratory-based experimental design. This study used the MDA-MB-231 cell line in various in vitro cell culture systems to investigate the research question. Methods For the first part of the study, we subjected MDA-MB-231 cells to grow in cysteine-absent adipocyte-conditioned media. In the second half, we treated MDA-MB-231 cells with iron chelator, deferoxamine. BODIPY imaging and western blot were carried out to observe ferroptosis in the cells under the 2 conditions. Results The results showed that cysteine absence in the conditioned media was able to reduce the formation of lipid droplets, which increased the greater access to free fatty acids to undergo oxidation, therefore inducing ferroptosis. On the contrary, cells when treated with deferoxamine along with erastin (ferroptosis-inducing drug), showed an increase in cell iron content was observed, later inducing ferroptosis. Conclusion Our results show an alternative function of cysteine and deferoxamine, one regulating lipid droplets and the other inducing ferroptosis, although an inhibitor of the same, respectively.
Collapse
Affiliation(s)
| | - JunJeong Choi
- Department of Pharmacy, Yonsei University, Incheon, South Korea
| |
Collapse
|
7
|
Streit WJ, Phan L, Bechmann I. Ferroptosis and pathogenesis of neuritic plaques in Alzheimer disease. Pharmacol Rev 2025; 77:100005. [PMID: 39952690 DOI: 10.1124/pharmrev.123.000823] [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: 12/14/2023] [Revised: 07/25/2024] [Accepted: 09/13/2024] [Indexed: 10/09/2024] Open
Abstract
Neuritic plaques are pathognomonic and terminal lesions of Alzheimer disease (AD). They embody AD pathogenesis because they harbor in one space critical pathologic features of the disease: amyloid deposits, neurofibrillary degeneration, neuroinflammation, and iron accumulation. Neuritic plaques are thought to arise from the conversion of diffuse extracellular deposits of amyloid-β protein (Aβ), and it is believed that during conversion, amyloid toxicity creates the dystrophic neurites of neuritic plaques, as well as neurofibrillary tangles However, recent evidence from human postmortem studies suggests a much different mechanism of neuritic plaque formation, where the first step in their creation is neuronal degeneration driven by iron overload and ferroptosis. Similarly, neurofibrillary tangles represent the corpses of iron-laden neurons that develop independently of Aβ deposits. In this review, we will focus on the role of free redox-active iron in the development of typical AD pathology, as determined largely by evidence obtained in the human temporal lobe during early, preclinical stages of AD. The findings have allowed the construction of a scheme of AD pathogenesis where brain iron is center stage and is involved in every step of the sequence of events that produce characteristic AD pathology. We will discuss how the study of preclinical AD has produced a fresh and revised assessment of AD pathogenesis that may be important for reconsidering current therapeutic efforts and guiding future ones. SIGNIFICANCE STATEMENT: This review offers a novel perspective on Alzheimer disease pathogenesis where elevated brain iron plays a central role and is involved throughout the development of lesions. Herein, we review arguments against the amyloid cascade theory and explain how recent findings in humans during early preclinical disease support iron-mediated cell death and endogenous iron containment mechanisms as critical components of neuritic plaque formation and ensuing dementia.
Collapse
Affiliation(s)
- Wolfgang J Streit
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida.
| | - Leah Phan
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| |
Collapse
|
8
|
He R, Liu Y, Fu W, He X, Liu S, Xiao D, Tao Y. Mechanisms and cross-talk of regulated cell death and their epigenetic modifications in tumor progression. Mol Cancer 2024; 23:267. [PMID: 39614268 PMCID: PMC11606237 DOI: 10.1186/s12943-024-02172-y] [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: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024] Open
Abstract
Cell death is a fundamental part of life for metazoans. To maintain the balance between cell proliferation and metabolism of human bodies, a certain number of cells need to be removed regularly. Hence, the mechanisms of cell death have been preserved during the evolution of multicellular organisms. Tumorigenesis is closely related with exceptional inhibition of cell death. Mutations or defects in cell death-related genes block the elimination of abnormal cells and enhance the resistance of malignant cells to chemotherapy. Therefore, the investigation of cell death mechanisms enables the development of drugs that directly induce tumor cell death. In the guidelines updated by the Cell Death Nomenclature Committee (NCCD) in 2018, cell death was classified into 12 types according to morphological, biochemical and functional classification, including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, PARP-1 parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence and mitotic catastrophe. The mechanistic relationships between epigenetic controls and cell death in cancer progression were previously unclear. In this review, we will summarize the mechanisms of cell death pathways and corresponding epigenetic regulations. Also, we will explore the extensive interactions between these pathways and discuss the mechanisms of cell death in epigenetics which bring benefits to tumor therapy.
Collapse
Affiliation(s)
- Ruimin He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yifan Liu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Weijie Fu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Xuan He
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China.
- Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China.
- Department of Pathology, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya Hospital, Central South University, Hunan, 410078, China.
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
- Furong Laboratory, Xiangya School of Medicine, Central South University, Hunan, 410078, China.
| |
Collapse
|
9
|
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
|
10
|
Lamačová LJ, Trnka J. Chelating mitochondrial iron and copper: Recipes, pitfalls and promise. Mitochondrion 2024; 78:101903. [PMID: 38777220 DOI: 10.1016/j.mito.2024.101903] [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: 10/20/2023] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Iron and copper chelation therapy plays a crucial role in treating conditions associated with metal overload, such as hemochromatosis or Wilson's disease. However, conventional chelators face challenges in reaching the core of iron and copper metabolism - the mitochondria. Mitochondria-targeted chelators can specifically target and remove metal ions from mitochondria, showing promise in treating diseases linked to mitochondrial dysfunction, including neurodegenerative diseases and cancer. Additionally, they serve as specific mitochondrial metal sensors. However, designing these new molecules presents its own set of challenges. Depending on the chelator's intended use to prevent or to promote redox cycling of the metals, the chelating moiety must possess different donor atoms and an optimal value of the electrode potential of the chelator-metal complex. Various targeting moieties can be employed for selective delivery into the mitochondria. This review also provides an overview of the current progress in the design of mitochondria-targeted chelators and their biological activity investigation.
Collapse
Affiliation(s)
- Lucie J Lamačová
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Praha, Czech Republic
| | - Jan Trnka
- Department of Biochemistry, Cell and Molecular Biology, Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Praha, Czech Republic.
| |
Collapse
|
11
|
Peña-Montes DJ, Huerta-Cervantes M, Riveros-Rosas H, Manzo-Avalos S, Aguilera-Méndez A, Huerta M, Trujillo X, Cortés-Rojo C, Montoya-Pérez R, Salgado-Garciglia R, Saavedra-Molina A. Iron chelation mitigates mitochondrial dysfunction and oxidative stress by enhancing nrf2-mediated antioxidant responses in the renal cortex of a murine model of type 2 diabetes. Mitochondrion 2024; 78:101937. [PMID: 39004262 DOI: 10.1016/j.mito.2024.101937] [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: 04/02/2024] [Revised: 06/20/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Renal iron overload is a common complication of diabetes that leads to oxidative stress and mitochondrial dysfunction in the kidneys. This study investigated the effects of iron chelation using deferiprone on mitochondrial dysfunction and oxidative stress in the renal cortex of a murine model of type 2 diabetes. Diabetic rats were treated with deferiprone (50 mg/kg BW) for 16 weeks. Our results show that iron chelation with deferiprone significantly increased the nuclear accumulation of Nrf2, a transcription factor that regulates the expression of antioxidant enzymes. This led to enhanced antioxidant capacity, reduced production of reactive oxygen species, and improved mitochondrial bioenergetic function in diabetic rats. However, chronic iron chelation led to altered mitochondrial respiration and increased oxidative stress in non-diabetic rats. In conclusion, our findings suggest that iron chelation with deferiprone protects mitochondrial bioenergetics and mitigates oxidative stress in the renal cortex, involving the NRF2 pathway in type 2 diabetes.
Collapse
Affiliation(s)
- Donovan J Peña-Montes
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Mexico
| | | | - Héctor Riveros-Rosas
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico
| | - Salvador Manzo-Avalos
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Mexico
| | - Asdrubal Aguilera-Méndez
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Mexico
| | - Miguel Huerta
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Mexico
| | - Xóchitl Trujillo
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Mexico
| | - Christian Cortés-Rojo
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Mexico
| | - Rocío Montoya-Pérez
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Mexico
| | - Rafael Salgado-Garciglia
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Mexico
| | - Alfredo Saavedra-Molina
- Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Mexico.
| |
Collapse
|
12
|
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
|
13
|
Gholam Azad M, Hussaini M, Russell TM, Richardson V, Kaya B, Dharmasivam M, Richardson DR. Multi-modal mechanisms of the metastasis suppressor, NDRG1: Inhibition of WNT/β-catenin signaling by stabilization of protein kinase Cα. J Biol Chem 2024; 300:107417. [PMID: 38815861 PMCID: PMC11261793 DOI: 10.1016/j.jbc.2024.107417] [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: 02/23/2024] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024] Open
Abstract
The metastasis suppressor, N-myc downstream regulated gene-1 (NDRG1), inhibits pro-oncogenic signaling in pancreatic cancer (PC). This investigation dissected a novel mechanism induced by NDRG1 on WNT/β-catenin signaling in multiple PC cell types. NDRG1 overexpression decreased β-catenin and downregulated glycogen synthase kinase-3β (GSK-3β) protein levels and its activation. However, β-catenin phosphorylation at Ser33, Ser37, and Thr41 are classically induced by GSK-3β was significantly increased after NDRG1 overexpression, suggesting a GSK-3β-independent mechanism. Intriguingly, NDRG1 overexpression upregulated protein kinase Cα (PKCα), with PKCα silencing preventing β-catenin phosphorylation at Ser33, Ser37, and Thr41, and decreasing β-catenin expression. Further, NDRG1 and PKCα were demonstrated to associate, with PKCα stabilization occurring after NDRG1 overexpression. PKCα half-life increased from 1.5 ± 0.8 h (3) in control cells to 11.0 ± 2.5 h (3) after NDRG1 overexpression. Thus, NDRG1 overexpression leads to the association of NDRG1 with PKCα and PKCα stabilization, resulting in β-catenin phosphorylation at Ser33, Ser37, and Thr41. The association between PKCα, NDRG1, and β-catenin was identified, with the formation of a potential metabolon that promotes the latter β-catenin phosphorylation. This anti-oncogenic activity of NDRG1 was multi-modal, with the above mechanism accompanied by the downregulation of the nucleo-cytoplasmic shuttling protein, p21-activated kinase 4 (PAK4), which is involved in β-catenin nuclear translocation, inhibition of AKT phosphorylation (Ser473), and decreased β-catenin phosphorylation at Ser552 that suppresses its transcriptional activity. These mechanisms of NDRG1 activity are important to dissect to understand the marked anti-cancer efficacy of NDRG1-inducing thiosemicarbazones that upregulate PKCα and inhibit WNT signaling.
Collapse
Affiliation(s)
- Mahan Gholam Azad
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Mohammed Hussaini
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Tiffany M Russell
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Vera Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Busra Kaya
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Mahendiran Dharmasivam
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Des R Richardson
- Centre for Cancer Cell Biology and Drug Discovery, Griffith University, Brisbane, Queensland, Australia; Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
14
|
Salimi Z, Afsharinasab M, Rostami M, Eshaghi Milasi Y, Mousavi Ezmareh SF, Sakhaei F, Mohammad-Sadeghipour M, Rasooli Manesh SM, Asemi Z. Iron chelators: as therapeutic agents in diseases. Ann Med Surg (Lond) 2024; 86:2759-2776. [PMID: 38694398 PMCID: PMC11060230 DOI: 10.1097/ms9.0000000000001717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/03/2024] [Indexed: 05/04/2024] Open
Abstract
The concentration of iron is tightly regulated, making it an essential element. Various cellular processes in the body rely on iron, such as oxygen sensing, oxygen transport, electron transfer, and DNA synthesis. Iron excess can be toxic because it participates in redox reactions that catalyze the production of reactive oxygen species and elevate oxidative stress. Iron chelators are chemically diverse; they can coordinate six ligands in an octagonal sequence. Because of the ability of chelators to trap essential metals, including iron, they may be involved in diseases caused by oxidative stress, such as infectious diseases, cardiovascular diseases, neurodegenerative diseases, and cancer. Iron-chelating agents, by tightly binding to iron, prohibit it from functioning as a catalyst in redox reactions and transfer iron and excrete it from the body. Thus, the use of iron chelators as therapeutic agents has received increasing attention. This review investigates the function of various iron chelators in treating iron overload in different clinical conditions.
Collapse
Affiliation(s)
- Zohreh Salimi
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan
| | - Mehdi Afsharinasab
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran
| | - Mehdi Rostami
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad
| | - Yaser Eshaghi Milasi
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan
| | - Seyedeh Fatemeh Mousavi Ezmareh
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan
| | - Fariba Sakhaei
- Department of Clinical Biochemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan
| | - Maryam Mohammad-Sadeghipour
- Department of Clinical Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman
| | | | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
| |
Collapse
|
15
|
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
|
16
|
Meng J, Xin L, Zou B, Wang L, Zhao X, Gao J, Zhang R. A manual controlled theranostic nanoplatform with real-time photoacoustic quantification of drug release for chemophotothermal therapy. J Colloid Interface Sci 2023; 651:1020-1027. [PMID: 37586151 DOI: 10.1016/j.jcis.2023.07.150] [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: 12/31/2022] [Revised: 07/15/2023] [Accepted: 07/24/2023] [Indexed: 08/18/2023]
Abstract
The development of intelligent nanodrug delivery systems that can visually guide the on-demand quantitative control of drug release has received extensive attention. Herein, two chemotherapeutic drugs, gallic acid and 5-fluorouracil, and Fe(III) were selected to prepare nanomedicine GF-Fe via polyphenol-metal self-assembly and infinite coordination of drug-metal. GF-Fe has good biocompatibility, photothermal properties and photoacoustic (PA) signals. When deferoxamine (DFO) was artificially applied and interacted with GF-Fe, GF-Fe began to disassemble, gallic acid and 5-fluorouracil were gradually released, while the PA signal of the nanomedicine decayed synchronously. Based on this, the relationship between the intensity of the PA signal and the drug release amount was established, so as to realize the precise quantitative control of the drug release in real-time under the guidance of PA imaging. Besides, the combined effect of the two therapeutic drugs in combination with photothermal therapy (PTT) can improve the therapeutic effect, resulting in significant superadditiveness. This nanoplatform constructed by facile synthesis provided good clinical translation potential for the implementation of precise multimodal combination therapy strategies for tumors.
Collapse
Affiliation(s)
- Jian Meng
- The Radiology Department of First Hospital of Shanxi Medical University, Taiyuan, China; Shanxi Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China; The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, China
| | - Lei Xin
- Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Bocheng Zou
- The Radiology Department of First Hospital of Shanxi Medical University, Taiyuan, China; Shanxi Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Lei Wang
- Shanxi Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China; The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, China
| | - Xuhui Zhao
- The Radiology Department of First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- The Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Taiyuan, China
| | - Ruiping Zhang
- The Radiology Department of First Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
17
|
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
|
18
|
Huang S, Zhang D, Yi X, Liu C, Jian C, Yu A. 3-AP inhibits the growth of human osteosarcoma by decreasing the activity of the iron-dependent pathway. Med Oncol 2023; 40:353. [PMID: 37952032 DOI: 10.1007/s12032-023-02215-2] [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: 09/11/2023] [Accepted: 10/08/2023] [Indexed: 11/14/2023]
Abstract
3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP) has broad-spectrum antitumor activity. However, its role in osteosarcoma (OS) remains unclear. Therefore, this study explored the effects of 3-AP on OS in vitro and in vivo using three human OS cell lines (MG-63, U2-OS, and 143B) and a nude mice model generated by transplanting 143B cells. The cells and mice were treated with DMSO (control) or gradient concentrations of 3-AP. Then, various assays (e.g., cell counting kit-8, flow cytometry, immunohistochemistry, and western blotting) were performed to assess cell viability and apoptosis levels, as well as γH2A.X (DNA damage correlation), ribonucleotide reductase catalytic subunit M1 and M2 (RRM1 and RRM2, respectively) protein levels (iron-dependent correlation). 3-AP time- and dose-dependably suppressed growth and induced apoptosis in all three OS cell lines, and ferric ammonium citrate (FAC) blocked these effects. Moreover, 3-AP decreased RRM2 and total ribonucleotide reductase (RRM1 plus RRM2) protein expression but significantly increased γH2A.X expression; treatment did not affect RRM1 expression. Again, FAC treatment attenuated these effects. In vivo, the number of apoptotic cells in the tumor slices increased in the 3-AP-treated mice compared to the control mice. 3-AP treatment also decreased Ki-67 and p21 expression, suggesting inhibited OS growth. Furthermore, the expression of RRM1, RRM2, and transferrin receptor protein 1 (i.e., Tfr1) indicated that 3-AP inhibited OS growth via an iron-dependent pathway. In conclusion, 3-AP exhibits anticancer activity in OS by decreasing the activity of iron-dependent pathways, which could be a promising therapeutic strategy for OS.
Collapse
Affiliation(s)
- Siyuan Huang
- Department of Orthopedics, Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China
| | - Dong Zhang
- Department of Orthopedics, Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China
| | - Xinzeyu Yi
- Department of Orthopedics, Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China
| | - Changjiang Liu
- Department of Orthopedics, Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China
| | - Chao Jian
- Department of Orthopedics, Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China.
- Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China.
| | - Aixi Yu
- Department of Orthopedics, Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, 430071, Hubei, People's Republic of China.
- Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, People's Republic of China.
| |
Collapse
|
19
|
Feng J, She Y, Li C, Shen L. Metal ion mediated aggregation of Alzheimer's disease peptides and proteins in solutions and at surfaces. Adv Colloid Interface Sci 2023; 320:103009. [PMID: 37776735 DOI: 10.1016/j.cis.2023.103009] [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: 06/20/2023] [Revised: 08/29/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
Although the pathogenesis of Alzheimer's disease (AD) is still unclear, abnormally high concentrations of metal ions, like copper, iron and zinc, were found in senile plaques of AD brain, which inspires extensive studies on the fundamental molecular interactions of metal ions with the pathogenic hallmarks, amyloid-β (Aβ) peptides and tau proteins, respectively forming senile plaques and neurofibrillary tangles (NFTs) in AD brains. Early works concern the concentration effect of the metal ions on Aβ and tau aggregation. Yet, it is obvious that the surrounding environment of the metal ions must also be considered, not just the metal ions as free accessible forms in the solution phase. The most important surrounding environment in vivo is a very large surface area from cell membranes and other macromolecular surfaces. These bio-interfaces make the kinetic pathways of metal ion mediated Aβ and tau aggregation radically different from those in the solution phase. To better understand the role of metal ions in AD peptide and protein aggregation, we summarize and discuss the recent achievements in the research of metal ion mediated Aβ and tau aggregation, particularly the corresponding mechanism differences between the solution phase and the surface environment. The metal ion chelation therapy for AD is also discussed from the point of the surface pool of metal ions.
Collapse
Affiliation(s)
- Jiahao Feng
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Yifei She
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Chongjia Li
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Lei Shen
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
20
|
Kinoshita N, Gessho M, Torii T, Ashida Y, Akamatsu M, Guo AK, Lee S, Katsuno T, Nakajima W, Budirahardja Y, Miyoshi D, Todokoro T, Ishida H, Nishikata T, Kawauchi K. The iron chelator deferriferrichrysin induces paraptosis via extracellular signal-related kinase activation in cancer cells. Genes Cells 2023; 28:653-662. [PMID: 37264202 DOI: 10.1111/gtc.13053] [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: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Cancer cells generally exhibit increased iron uptake, which contributes to their abnormal growth and metastatic ability. Iron chelators have thus recently attracted attention as potential anticancer agents. Here, we show that deferriferrichrysin (Dfcy), a natural product from Aspergillus oryzae acts as an iron chelator to induce paraptosis (a programmed cell death pathway characterized by ER dilation) in MCF-7 human breast cancer cells and H1299 human lung cancer cells. We first examined the anticancer efficacy of Dfcy in cancer cells and found that Dfcy induced ER dilation and reduced the number of viable cells. Extracellular signal-related kinase (ERK) was activated by Dfcy treatment, and the MEK inhibitor U0126, a small molecule commonly used to inhibit ERK activity, prevented the increase in ER dilation in Dfcy-treated cells. Concomitantly, the decrease in the number of viable cells upon treatment with Dfcy was attenuated by U0126. Taken together, these results demonstrate that the iron chelator Dfcy exhibits anticancer effects via induction of ERK-dependent paraptosis.
Collapse
Affiliation(s)
- Natsuki Kinoshita
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Masaya Gessho
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Takeru Torii
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Yukako Ashida
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Minori Akamatsu
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Alvin Kunyao Guo
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Sunmin Lee
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Tatsuya Katsuno
- Center of Anatomical, Pathological and Forensic Medical Researches, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Wataru Nakajima
- Department of Molecular Oncology, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo, Japan
| | - Yemima Budirahardja
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Daisuke Miyoshi
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | | | - Hiroki Ishida
- Research Institute, Gekkeikan Sake Co., Ltd, Kyoto, Japan
| | - Takahito Nishikata
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| | - Keiko Kawauchi
- Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Japan
| |
Collapse
|
21
|
Tian YM, Holdship P, To TQ, Ratcliffe PJ, Keeley TP. Comparative analysis of N-terminal cysteine dioxygenation and prolyl-hydroxylation as oxygen-sensing pathways in mammalian cells. J Biol Chem 2023; 299:105156. [PMID: 37572852 PMCID: PMC10506105 DOI: 10.1016/j.jbc.2023.105156] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/25/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023] Open
Abstract
In animals, adaptation to changes in cellular oxygen levels is coordinated largely by 2-oxoglutarate-dependent prolyl-hydroxylase domain (PHD) dioxygenase family members, which regulate the stability of their hypoxia-inducible factor (HIF) substrates to promote expression of genes that adapt cells to hypoxia. Recently, 2-aminoethanethiol dioxygenase (ADO) was identified as a novel O2-sensing enzyme in animals. Through N-terminal cysteine dioxygenation and the N-degron pathway, ADO regulates the stability of a set of non-transcription factor substrates; the regulators of G-protein signaling 4, 5. and 16 and interleukin-32. Here, we set out to compare and contrast the in cellulo characteristics of ADO and PHD enzymes in an attempt to better understand their co-evolution in animals. We find that ADO operates to regulate the stability of its substrates rapidly and with similar O2-sensitivity to the PHD/HIF pathway. ADO appeared less sensitive to iron chelating agents or transition metal exposure than the PHD enzymes, possibly due to tighter catalytic-site Fe2+ coordination. Unlike the PHD/HIF pathway, the ADO/N-degron pathway was not subject to feedback by hypoxic induction of ADO, and induction of ADO substrates was well sustained in response to prolonged hypoxia. The data also reveal strong interactions between proteolytic regulation of targets by ADO and transcriptional induction of those targets, that shape integrated cellular responses to hypoxia. Collectively, our comparative analysis provides further insight into ADO/N-degron-mediated oxygen sensing and its integration into established mechanisms of oxygen homeostasis.
Collapse
Affiliation(s)
- Ya-Min Tian
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip Holdship
- Department of Earth Sciences, University of Oxford, Oxford, UK
| | - Trang Quynh To
- Department of Earth Sciences, University of Oxford, Oxford, UK
| | - Peter J Ratcliffe
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK; The Francis Crick Institute, London, UK.
| | - Thomas P Keeley
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
22
|
Xu Z, Sung YS, Tomat E. Design of Tetrazolium Cations for the Release of Antiproliferative Formazan Chelators in Mammalian Cells. J Am Chem Soc 2023; 145:15197-15206. [PMID: 37410992 PMCID: PMC10521327 DOI: 10.1021/jacs.3c02033] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cancer cells generally present a higher demand for iron, which plays crucial roles in tumor progression and metastasis. This iron addiction provides opportunities to develop broad spectrum anticancer drugs that target iron metabolism. In this context, prochelation approaches are investigated to release metal-binding compounds under specific conditions, thereby limiting off-target toxicity. Here, we demonstrate a prochelation strategy inspired by the bioreduction of tetrazolium cations widely employed to assess the viability of mammalian cells. We designed a series of tetrazolium-based compounds for the intracellular release of metal-binding formazan ligands. The combination of reduction potentials appropriate for intracellular reduction and an N-pyridyl donor on the formazan scaffold led to two effective prochelators. The reduced formazans bind as tridentate ligands and stabilize low-spin Fe(II) centers in complexes of 2:1 ligand-to-metal stoichiometry. The tetrazolium salts are stable in blood serum for over 24 h, and antiproliferative activities at micromolar levels were recorded in a panel of cancer cell lines. Additional assays confirmed the intracellular activation of the prochelators and their ability to affect cell cycle progression, induce apoptotic death, and interfere with iron availability. Demonstrating the role of iron in their intracellular effects, the prochelators impacted the expression levels of key iron regulators (i.e., transferrin receptor 1 and ferritin), and iron supplementation mitigated their cytotoxicity. Overall, this work introduces the tetrazolium core as a platform to build prochelators that can be tuned for activation in the reducing environment of cancer cells and produce antiproliferative formazan chelators that interfere with cellular iron homeostasis.
Collapse
Affiliation(s)
- Zoufeng Xu
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson AZ 85721 (USA)
| | - Yu-Shien Sung
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson AZ 85721 (USA)
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson AZ 85721 (USA)
| |
Collapse
|
23
|
Ouyang R, Wang S, Feng K, Liu C, Silva DZ, Chen Y, Zhao Y, Liu B, Miao Y, Zhou S. Potent saccharinate-containing palladium(II) complexes for sensitization to cancer therapy. J Inorg Biochem 2023; 244:112205. [PMID: 37028114 DOI: 10.1016/j.jinorgbio.2023.112205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/07/2023]
Abstract
Palladium(II) (PdII) complexes are among the most promising anticancer compounds. Both 2`-benzoylpyridine thiosemicarbazone (BpT) and saccharinate (Sac) are efficient metal chelators with potent anticancer activity. To explore a more effective new anticancer drug, we synthesized a series of Sac and BpT-containing PdII complexes coordinated with thiosemicarbazone (TSC)-derived ligands, and characterized them through nuclear magnetic resonance (NMR), Fourier transformed infrared spectroscopy (FT-IR), elemental analysis, ultraviolet-visible spectroscopy (UV-Vis) and thermogravimetric analysis (TGA). Each target complex was composed of PdII, BpT, and one or two Sac molecules. Both the in vitro and in vivo anti-growth effects of those ligands and the obtained PdII complexes were investigated in the human lung adenocarcinoma cell lines A549 and Spc-A1. The coordination of PdII with the TSC-derivatives and Sac resulted in clearly greater anticancer activity than single ligands. These compounds were demonstrated to be safe for 293 T normal human kidney epithelial cells. The introduction of Sac into the TSC-derived PdII complex significantly enhanced anti-growth effects, and induced apoptosis of human lung cancer cells in vitro and in vivo in a dose dependent manner. Moreover, the PdII complex containing two Sac molecules showed the most promising therapeutic effects, thereby confirming that Sac increases the cancer therapeutic efficacy of PdII complexes and providing a new strategy for exploring anticancer drugs for potential clinical treatment.
Collapse
|
24
|
Tomat E. Targeting iron to contrast cancer progression. Curr Opin Chem Biol 2023; 74:102315. [PMID: 37187095 PMCID: PMC10225354 DOI: 10.1016/j.cbpa.2023.102315] [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: 12/30/2022] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
An altered metabolism of iron fuels cancer growth, invasion, metastasis, and recurrence. Ongoing research in cancer biology is delineating a complex iron-trafficking program involving both malignant cells and their support network of cancer stem cells, immune cells, and other stromal components in the tumor microenvironment. Iron-binding strategies in anticancer drug discovery are being pursued in clinical trials and in multiple programs at various levels of development. Polypharmacological mechanisms of action, combined with emerging iron-associated biomarkers and companion diagnostics, are poised to offer new therapeutic options. By targeting a fundamental player in cancer progression, iron-binding drug candidates (either alone or in combination therapy) have the potential to impact a broad range of cancer types and to address the major clinical problems of recurrence and resistance to therapy.
Collapse
Affiliation(s)
- Elisa Tomat
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA.
| |
Collapse
|
25
|
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
|
26
|
Papsdorf K, Miklas JW, Hosseini A, Cabruja M, Morrow CS, Savini M, Yu Y, Silva-García CG, Haseley NR, Murphy LM, Yao P, de Launoit E, Dixon SJ, Snyder MP, Wang MC, Mair WB, Brunet A. Lipid droplets and peroxisomes are co-regulated to drive lifespan extension in response to mono-unsaturated fatty acids. Nat Cell Biol 2023; 25:672-684. [PMID: 37127715 PMCID: PMC10185472 DOI: 10.1038/s41556-023-01136-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/23/2023] [Indexed: 05/03/2023]
Abstract
Dietary mono-unsaturated fatty acids (MUFAs) are linked to longevity in several species. But the mechanisms by which MUFAs extend lifespan remain unclear. Here we show that an organelle network involving lipid droplets and peroxisomes is critical for MUFA-induced longevity in Caenorhabditis elegans. MUFAs upregulate the number of lipid droplets in fat storage tissues. Increased lipid droplet number is necessary for MUFA-induced longevity and predicts remaining lifespan. Lipidomics datasets reveal that MUFAs also modify the ratio of membrane lipids and ether lipids-a signature associated with decreased lipid oxidation. In agreement with this, MUFAs decrease lipid oxidation in middle-aged individuals. Intriguingly, MUFAs upregulate not only lipid droplet number but also peroxisome number. A targeted screen identifies genes involved in the co-regulation of lipid droplets and peroxisomes, and reveals that induction of both organelles is optimal for longevity. Our study uncovers an organelle network involved in lipid homeostasis and lifespan regulation, opening new avenues for interventions to delay aging.
Collapse
Affiliation(s)
| | - Jason W Miklas
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Amir Hosseini
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Matias Cabruja
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Christopher S Morrow
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Marzia Savini
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
| | - Yong Yu
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Carlos G Silva-García
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Pallas Yao
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Meng C Wang
- Department of Molecular and Human Genetics, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - William B Mair
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, CA, USA.
- Wu Tsai Institute of Neurosciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Wu L, Xian X, Tan Z, Dong F, Xu G, Zhang M, Zhang F. The Role of Iron Metabolism, Lipid Metabolism, and Redox Homeostasis in Alzheimer's Disease: from the Perspective of Ferroptosis. Mol Neurobiol 2023; 60:2832-2850. [PMID: 36735178 DOI: 10.1007/s12035-023-03245-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
In the development of Alzheimer's disease (AD), cell death is common. Novel cell death form-ferroptosis is discovered in recent years. Ferroptosis is an iron-regulated programmed cell death mechanism and has been identified in AD clinical samples. Typical characteristics of ferroptosis involve the specific changes in cell morphology, iron-dependent aggregation of reactive oxygen species (ROS) and lipid peroxides, loss of glutathione (GSH), inactivation of glutathione peroxidase 4 (GPX4), and a unique group of regulatory genes. Increasing evidence demonstrates that ferroptosis may be associated with neurological dysfunction in AD. However, the underlying mechanisms have not been fully elucidated. This article reviews the potential role of ferroptosis in AD, the involvement of ferroptosis in the pathological progression of AD through the mechanisms of iron metabolism, lipid metabolism, and redox homeostasis, as well as a range of potential therapies targeting ferroptosis for AD. Intervention strategies based on ferroptosis are promising for Alzheimer's disease treatment at present, but further researches are still needed.
Collapse
Affiliation(s)
- Linyu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Xiaohui Xian
- Department of Pathophysiology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050051, Hebei, People's Republic of China
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China
| | - Zixuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, No. 361 East Zhongshan Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, 050051, Hebei, People's Republic of China.
- Hebei Key Laboratory of Critical Disease Mechanism and intervention, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
28
|
Botta M, Geraldes CFGC, Tei L. High spin Fe(III)-doped nanostructures as T 1 MR imaging probes. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1858. [PMID: 36251471 DOI: 10.1002/wnan.1858] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/05/2022]
Abstract
Magnetic Resonance Imaging (MRI) T1 contrast agents based on Fe(III) as an alternative to Gd-based compounds have been under intense scrutiny in the last 6-8 years and a number of nanostructures have been designed and proposed for in vivo diagnostic and theranostic applications. Excluding the large family of superparamagnetic iron oxides widely used as T2 -MR imaging agents that will not be covered by this review, a considerable number and type of nanoparticles (NPs) have been employed, ranging from amphiphilic polymer-based NPs, NPs containing polyphenolic binding units such as melanin-like or polycatechols, mixed metals such as Fe/Gd or Fe/Au NPs and perfluorocarbon nanoemulsions. Iron(III) exhibits several favorable magnetic properties, high biocompatibility and improved toxicity profile that place it as the paramagnetic ion of choice for the next generation of nanosized MRI and theranostic contrast agents. An analysis of the examples reported in the last decade will show the opportunities for relaxivity and MR-contrast enhancement optimization that could bring Fe(III)-doped NPs to really compete with Gd(III)-based nanosystems. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Diagnostic Tools > Diagnostic Nanodevices Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Mauro Botta
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Carlos F G C Geraldes
- Faculty of Science and Technology, Department of Life Sciences and Coimbra Chemistry Center, University of Coimbra, Coimbra, Portugal.,CIBIT-Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Coimbra, Portugal
| | - Lorenzo Tei
- Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| |
Collapse
|
29
|
Zhao X, Richardson DR. The role of the NDRG1 in the pathogenesis and treatment of breast cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:188871. [PMID: 36841367 DOI: 10.1016/j.bbcan.2023.188871] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/18/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
Breast cancer (BC) is the leading cause of cancer death in women. This disease is heterogeneous, with clinical subtypes being estrogen receptor-α (ER-α) positive, having human epidermal growth factor receptor 2 (HER2) overexpression, or being triple-negative for ER-α, progesterone receptor, and HER2 (TNBC). The ER-α positive and HER2 overexpressing tumors can be treated with agents targeting these proteins, including tamoxifen and pertuzumab, respectively. Despite these treatments, resistance and metastasis are problematic, while TNBC is challenging to treat due to the lack of suitable targets. Many studies examining BC and other tumors indicate a role for N-myc downstream-regulated gene-1 (NDRG1) as a metastasis suppressor. The ability of NDRG1 to inhibit metastasis is due, in part, to the inhibition of the initial step in metastasis, namely the epithelial-to-mesenchymal transition. Paradoxically, there are also reports of NDRG1 playing a pro-oncogenic role in BC pathogenesis. The oncogenic effects of NDRG1 in BC have been reported to relate to lipid metabolism or the mTOR signaling pathway. The molecular mechanism(s) of how NDRG1 regulates the activity of multiple signaling pathways remains unclear. Therapeutic strategies that up-regulate NDRG1 have been developed and include agents of the di-2-pyridylketone thiosemicarbazone class. These compounds target oncogenic drivers in BC cells, suppressing the expression of multiple key hormone receptors including ER-α, progesterone receptor, androgen receptor, and prolactin receptor, and can also overcome tamoxifen resistance. Considering the varying role of NDRG1 in BC pathogenesis, further studies are required to examine what subset of BC patients would benefit from pharmacopeia that up-regulate NDRG1.
Collapse
Affiliation(s)
- Xiao Zhao
- Centre for Cancer Cell Biology and Drug Discovery, Griffith Institute for Drug Discovery, Griffith University, Nathan, Brisbane, Queensland 4111, Australia
| | - Des R Richardson
- 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
|
30
|
Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators. Cancers (Basel) 2023; 15:cancers15020468. [PMID: 36672419 PMCID: PMC9856351 DOI: 10.3390/cancers15020468] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Excess iron is known to be a risk factor of carcinogenesis. Although iron chelators show anti-cancer effects, they have not been used successfully to treat cancer patients. Triple-negative breast cancer (TNBC) is a disease with poor prognosis without effective treatments. Thus, we aimed to evaluate a possibility of iron chelators as a therapy for TNBC. Deferasirox (DFX), an iron chelator, suppressed the growth of 4T1 murine TNBC cell line cells in vitro and in vivo. Lung metastasis was further significantly reduced, leading to the hypothesis that iron metabolism between metastatic and non-metastatic cells may be different. An analysis of existing database demonstrated that the expression of iron-uptake genes was significantly suppressed in TNBC cells that metastasized to lymph nodes or lungs compared to those in primary tumors. A highly metastatic clone of the murine 4T1 TNBC cells (4T1-HM) did not proliferate well under iron-rich or iron-depleted conditions by iron chelators compared to a low-metastatic clone (4T1-LM). Bulk RNA-seq analysis of RNA from 4T1-HM and 4T1-LM cells suggested that the PI3K-AKT pathway might be responsible for this difference. Indeed, DFX suppressed the proliferation via the AKT-mTOR pathway in 4T1-HM and the human MDA-MB-231 cells, a human mesenchymal-like TNBC cell line. DFX also suppressed the growth of 4T1-HM tumors in comparison to 4T1-LM tumors, and reduced lung metastases after surgical resection of primary 4T1 tumors. These results indicated, for the first time, that highly metastatic TNBC cells have limited iron metabolism, and they can be more effectively targeted by iron chelators.
Collapse
|
31
|
Sahoo K, Sharma A. Understanding the mechanistic roles of environmental heavy metal stressors in regulating ferroptosis: adding new paradigms to the links with diseases. Apoptosis 2023; 28:277-292. [PMID: 36611106 DOI: 10.1007/s10495-022-01806-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2022] [Indexed: 01/09/2023]
Abstract
Ferroptosis is a new type of iron-dependent cell death induced by a failure of the lipid repair protein GPX4 or the Xc- antiporter, which is essential for glutathione production. Some heavy metals such as arsenic (As), cobalt (Co), cadmium (Cd), iron (Fe), magnesium (Mg), manganese (Mn), nickel (Ni), mercury (Hg) as well as zinc (Zn) are shown to induce ferroptotic cell death involving the generation of oxidative stress, mitochondrial dysfunctioning, lipid peroxidation, and several other cellular etiologies. However, selenium (Se) treatment has been shown to enhance adaptive transcription responses to protect cells from ferroptosis. Heavy metals like Cadmium exposure activated ALK4/5 signaling via Smad3 and Akt signaling which leads to cell death mechanism. Continuous exposure to a small dose of mercury can damage tissues, and methylmercury bind to sulfhydryl proteins and GSH, this elevates oxidative stress, free radical accumulation, glutathione depletion, mitochondrial damage, and inhibited the nuclear factor-κB pathway which leads to ferroptotic cell death. Animals exposed to nickel and cobalt may have increased lipid peroxidation which can induce ferroptosis. Glutathione depletion is caused by Zn intoxication and exposure to manganese. These metals are systemic toxins that have been shown adverse effects on humans. Ferroptosis has recently been related to several pathological disorders, including, Alzheimer's disease, Parkinson's disease, Huntington's disease, as well as cardiovascular disease, and any type of cancer. For these disorders and some heavy metal toxicity, ferroptosis suppression needs to be looked upon as a promising therapeutic choice.
Collapse
Affiliation(s)
- Kumudini Sahoo
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow, 226002, India.,School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Ankita Sharma
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow, 226002, India.
| |
Collapse
|
32
|
Jones G, Zeng L, Kim J. Mechanism-Based Pharmacokinetic Modeling of Absorption and Disposition of a Deferoxamine-Based Nanochelator in Rats. Mol Pharm 2023; 20:481-490. [PMID: 36378830 DOI: 10.1021/acs.molpharmaceut.2c00737] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Deferoxamine (DFO) is an effective FDA-approved iron chelator. However, its use is considerably limited by off-target toxicities and an extremely cumbersome dose regimen with daily infusions. The recent development of a deferoxamine-based nanochelator (DFO-NP) with selective renal excretion has shown promise in ameliorating animal models of iron overload with a substantially improved safety profile. To further the preclinical development of this promising nanochelator and to inform on the feasibility of clinical development, it is necessary to fully characterize the dose and administration-route-dependent pharmacokinetics and to develop predictive pharmacokinetic (PK) models describing absorption and disposition. Herein, we have evaluated the absorption, distribution, and elimination of DFO-NPs after intravenous and subcutaneous (SC) injection at therapeutically relevant doses in Sprague Dawley rats. We also characterized compartment-based model structures and identified model-based parameters to quantitatively describe the PK of DFO-NPs. Our modeling efforts confirmed that disposition could be described using a three-compartment mamillary model with elimination and saturable reabsorption both occurring from the third compartment. We also determined that absorption was nonlinear and best described by parallel saturable and first-order processes. Finally, we characterized a novel pathway for saturable SC absorption of an ultrasmall organic nanoparticle directly into the systemic circulation, which offers a novel strategy for improving drug exposure for nanotherapeutics.
Collapse
Affiliation(s)
- Gregory Jones
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Lingxue Zeng
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| | - Jonghan Kim
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| |
Collapse
|
33
|
Fathy A, Ibrahim AB, Elkhalik SA, Villinger A, Abbas S. New iron(III) complexes with 2-formylpyridine thiosemicarbazones: Synthetic aspects, structural and spectral analyses and cytotoxicity screening against MCF-7 human cancer cells. Heliyon 2023; 9:e13008. [PMID: 36711299 PMCID: PMC9880397 DOI: 10.1016/j.heliyon.2023.e13008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
2-Formylpyridine thiosemicarbazone - iron (III) chelates [F e L 2 ] C l • 2 H 2 O {L = L1 (C1) [HL 1 = 4-(4-Nitrophenyl)-1-((pyridin-2-yl)methylene)thiosemicarbazide] and L = L2 (C2) [HL 2 = 4-(2,5-Dimethoxyphenyl)-1-((pyridin-2-yl)methylene)thiosemicarbazide]} were prepared. The two ligand anions in each complex resulted in saturation of the iron coordination number and consequently the existence of these complexes as 1:1 electrolytes. As well, the iron in these complexes exhibits low-spin electronic configuration. X-ray crystallography of complex C1 indicated its triclinic crystal system and P1 ‾ space group. In addition, it proved the ligation through a thiol sulfur atom and two nitrogen atoms of pyridine and azomethine groups. This is while the presence of two water molecules of crystallization in the complex structure was also indicated. The ligand HL 1 was selected for cytotoxicity screening against human MCF-7, A-549, HEPG-2 and HCT-116 cancer cells and the most enhanced activities were detected against the breast cells. Against these cells, the compounds HL 1 , HL 2 , C1 and C2 induced cytotoxicity, respectively, with IC50 values of 52.4, 145.4, 34.3 and 62.0 μM. However, against the healthy BHK cells, HL 1 and HL 2 caused cytotoxicity, respectively, with IC50 values of 54.8 and 110.6 μM and cytotoxicity with percent viabilities of 56.7 and 55.4% of the BHK cells by the complexes (137.4 μM of C1 and 131.9 μM of C2) was determined. These activities against MCF-7 cells are less significant compared with the measured value for doxorubicin. But this standard is more toxic to normal cells than the thiosemicarbazones (IC50 (doxorubicin) = 9.66 μM against MCF-7 cells and 36.42 μM against BHK cells).
Collapse
Affiliation(s)
- Amany Fathy
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Ahmed B.M. Ibrahim
- Department of Chemistry, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - S. Abd Elkhalik
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Alexander Villinger
- Institut für Chemie, Universität Rostock, Albert-Einstein-Str. 3a, 18059 Rostock, Germany
| | - S.M. Abbas
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt
| |
Collapse
|
34
|
Wu W, Sung YS, Tomat E. Thiol-Reactive Arylsulfonate Masks for Phenolate Donors in Antiproliferative Iron Prochelators. Inorg Chem 2022; 61:19974-19982. [PMID: 36455205 PMCID: PMC10188280 DOI: 10.1021/acs.inorgchem.2c03250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Tridentate thiosemicarbazones, among several families of iron chelators, have shown promising results in anticancer drug discovery because they target the increased need for iron that characterizes malignant cells. Prochelation strategies, in which the chelator is released under specific conditions, have the potential to avoid off-target metal binding (for instance, in the bloodstream) and minimize unwanted side effects. We report a prochelation approach that employs arylsulfonate esters to mask the phenolate donor of salicylaldehyde-based chelators. The new prochelators liberate a tridentate thiosemicarbazone intracellularly upon reaction with abundant nucleophile glutathione (GSH). A 5-bromo-substituted salicylaldehyde thiosemicarbazone (STC4) was selected for the chelator unit because of its antiproliferative activity at low micromolar levels in a panel of six cancer cell lines. The arylsulfonate prochelators were assessed in vitro with respect to their stability, ability to abolish metal binding, and reactivity in the presence of GSH. Cell-based assays indicated that the arylsulfonate-masked prochelators present higher antiproliferative activities relative to the parent compound after 24 h. The activation and release of the chelator intracellularly were corroborated by assays of cytosolic iron binding and iron supplementation effects as well as cell cycle analysis. This study introduces the 1,3,4-thiadiazole sulfonate moiety to mask the phenolate donor of an iron chelator and impart good solubility and stability to prochelator constructs. The reactivity of these systems can be tuned to release the chelator at high glutathione levels, as encountered in several cancer phenotypes.
Collapse
Affiliation(s)
- Wangbin Wu
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yu-Shien Sung
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
35
|
Khan T, Raza S, Lawrence AJ. Medicinal Utility of Thiosemicarbazones with Special Reference to Mixed Ligand and Mixed Metal Complexes: A Review. RUSS J COORD CHEM+ 2022. [DOI: 10.1134/s1070328422600280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
36
|
Amsri A, Srichairatanakool S, Teerawutgulrag A, Youngchim S, Pongpom M. Genetic Engineering of Talaromyces marneffei to Enhance Siderophore Production and Preliminary Testing for Medical Application Potential. J Fungi (Basel) 2022; 8:1183. [PMID: 36354950 PMCID: PMC9692399 DOI: 10.3390/jof8111183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 06/16/2025] Open
Abstract
Siderophores are compounds with low molecular weight with a high affinity and specificity for ferric iron, which is produced by bacteria and fungi. Fungal siderophores have been characterized and their feasibility for clinical applications has been investigated. Fungi may be limited in slow growth and low siderophore production; however, they have advantages of high diversity and affinity. Hence, the purpose of this study was to generate a genetically modified strain in Talaromyces marneffei that enhanced siderophore production and to identify the characteristics of siderophore to guide its medical application. SreA is a transcription factor that negatively controls iron acquisition mechanisms. Therefore, we deleted the sreA gene to enhance the siderophore production and found that the null mutant of sreA (ΔsreA) produced a high amount of extracellular siderophores. The produced siderophore was characterized using HPLC-MS, HPLC-DAD, FTIR, and 1H- and 13C-NMR techniques and identified as a coprogen B. The compound showed a powerful iron-binding activity and could reduce labile iron pool levels in iron-loaded hepatocellular carcinoma (Huh7) cells. In addition, the coprogen B showed no toxicity to the Huh7 cells, demonstrating its potential to serve as an ideal iron chelator. Moreover, it inhibits the growth of Candida albicans and Escherichia coli in a dose-dependent manner. Thus, we have generated the siderophore-enhancing strain of T. marneffei, and the coprogen B isolated from this strain could be useful in the development of a new iron-chelating agent or other medical applications.
Collapse
Affiliation(s)
- Artid Amsri
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Somdet Srichairatanakool
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Aphiwat Teerawutgulrag
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirida Youngchim
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Monsicha Pongpom
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
37
|
Feng W, Xiao Y, Zhao C, Zhang Z, Liu W, Ma J, Ganz T, Zhang J, Liu S. New Deferric Amine Compounds Efficiently Chelate Excess Iron to Treat Iron Overload Disorders and to Prevent Ferroptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202679. [PMID: 36031399 PMCID: PMC9561787 DOI: 10.1002/advs.202202679] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/04/2022] [Indexed: 05/09/2023]
Abstract
Excess iron accumulation occurs in organs of patients with certain genetic disorders or after repeated transfusions. No physiological mechanism is available to excrete excess iron and iron overload to promote lipid peroxidation to induce ferroptosis, thus iron chelation becomes critical for preventing ion toxicity in these patients. To date, several iron chelators have been approved for iron chelation therapy, such as deferiprone and deferoxamine, but the current iron chelators suffer from significant limitations. In this context, new agents are continuously sought. Here, a library of new deferric amine compounds (DFAs) with adjustable skeleton and flexibility is synthesized by adopting the beneficial properties of conventional chelators. After careful evaluations, compound DFA1 is found to have greater efficacy in binding iron through two molecular oxygens in the phenolic hydroxyl group and the nitrogen atom in the amine with a 2:1 stoichiometry. This compound remarkably ameliorates iron overload in diverse murine models through both oral and intravenous administration, including hemochromatosis, high iron diet-induced, and iron dextran-stimulated iron accumulation. Strikingly, this compound is found to suppress iron-induced ferroptosis by modulating the intracellular signaling that drives lipid peroxidation. This study opens a new approach for the development of iron chelators to treat iron overload.
Collapse
Affiliation(s)
- Wenya Feng
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Yuanjing Xiao
- School of Chemistry and Molecular EngineeringEast China Normal University500 Dongchuan RoadShanghai200241P. R. China
| | - Chuanfang Zhao
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Zhanming Zhang
- Department of ChemistryFudan University2005 Songhu RoadShanghai200438P. R. China
| | - Wei Liu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Tomas Ganz
- Department of MedicineDavid Geffen School of MedicineUniversity of CaliforniaLos AngelesCA90095USA
| | - Junliang Zhang
- Department of ChemistryFudan University2005 Songhu RoadShanghai200438P. R. China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and EcotoxicologyResearch Center for Eco‐Environmental SciencesChinese Academy of SciencesBeijing100085P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| |
Collapse
|
38
|
Mousavi Z, Ghasemi JB, Mohammadi Ziarani G, Saidi M, Badiei A. Dihydropyrano quinoline derivatives functionalized nanoporous silica as novel fluorescence sensor for Fe3+ in aqueous solutions(aq). J Mol Struct 2022; 1265:133408. [DOI: 10.1016/j.molstruc.2022.133408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Sung YS, Kerimoglu B, Ooi A, Tomat E. Aroylhydrazone Glycoconjugate Prochelators Exploit Glucose Transporter 1 (GLUT1) to Target Iron in Cancer Cells. ACS Med Chem Lett 2022; 13:1452-1458. [PMID: 36105345 PMCID: PMC9465708 DOI: 10.1021/acsmedchemlett.2c00250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Glycoconjugation strategies in anticancer drug discovery exploit the high expression of glucose transporters in malignant cells to achieve preferential uptake and hence attractive pharmacological characteristics of increased therapeutic windows and decreased unwanted toxicity. Here we present the design of glycoconjugated prochelators of aroylhydrazone AH1, an antiproliferative scavenger that targets the increased iron demand of rapidly proliferating malignant cells. The constructs feature a monosaccharide (d-glucose, d-glucosamine, or glycolytic inhibitor 2-deoxy-d-glucose) connected at the C2 or C6 position via a short linker, which masks the chelator through a disulfide bond susceptible to intracellular reduction. Cellular assays showed that the glycoconjugates rely on the GLUT1 transporter for uptake, lead to intracellular iron deprivation, and present antiproliferative activity. Ectopic overexpression of GLUT1 in malignant and normal cells increased the uptake and toxicity of the glycoconjugated prochelators, demonstrating that these compounds are well suited for targeting cells overexpressing glucose transporters and therefore for selective iron sequestration in malignant cells.
Collapse
Affiliation(s)
- Yu-Shien Sung
- Department
of Chemistry and Biochemistry, The University
of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721-0041, United States
| | - Baris Kerimoglu
- Department
of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E. Mabel St., Tucson, Arizona 85721, United
States
| | - Aikseng Ooi
- Department
of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, 1703 E. Mabel St., Tucson, Arizona 85721, United
States
| | - Elisa Tomat
- Department
of Chemistry and Biochemistry, The University
of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721-0041, United States
| |
Collapse
|
40
|
Pal A, Cerchiaro G, Rani I, Ventriglia M, Rongioletti M, Longobardi A, Squitti R. Iron in Alzheimer's Disease: From Physiology to Disease Disabilities. Biomolecules 2022; 12:1248. [PMID: 36139084 PMCID: PMC9496246 DOI: 10.3390/biom12091248] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022] Open
Abstract
Reactive oxygen species (ROS) play a key role in the neurodegeneration processes. Increased oxidative stress damages lipids, proteins, and nucleic acids in brain tissue, and it is tied to the loss of biometal homeostasis. For this reason, attention has been focused on transition metals involved in several biochemical reactions producing ROS. Even though a bulk of evidence has uncovered the role of metals in the generation of the toxic pathways at the base of Alzheimer's disease (AD), this matter has been sidelined by the advent of the Amyloid Cascade Hypothesis. However, the link between metals and AD has been investigated in the last two decades, focusing on their local accumulation in brain areas known to be critical for AD. Recent evidence revealed a relation between iron and AD, particularly in relation to its capacity to increase the risk of the disease through ferroptosis. In this review, we briefly summarize the major points characterizing the function of iron in our body and highlight why, even though it is essential for our life, we have to monitor its dysfunction, particularly if we want to control our risk of AD.
Collapse
Affiliation(s)
- Amit Pal
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Kalyani 741245, West Bengal, India
| | - Giselle Cerchiaro
- Center for Natural Sciences and Humanities, Federal University of ABC (UFABC), Avenida dos Estados, 5001, Bl.B, Santo André 09210-580, SP, Brazil
| | - Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar University (MMU), Mullana, Ambala 133203, Haryana, India
| | - Mariacarla Ventriglia
- Fatebenefratelli Foundation for Health Research and Education, AFaR Division, 00186 Rome, Italy
| | - Mauro Rongioletti
- Department of Laboratory Medicine, Research and Development Division, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy
| | - Antonio Longobardi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Rosanna Squitti
- Department of Laboratory Medicine, Research and Development Division, Fatebenefratelli Isola Tiberina, Gemelli Isola, 00186 Rome, Italy
| |
Collapse
|
41
|
Jones G, Zeng L, Stiles WR, Park SH, Kang H, Choi HS, Kim J. Pharmacokinetics and tissue distribution of deferoxamine-based nanochelator in rats. Nanomedicine (Lond) 2022; 17:1649-1662. [PMID: 36547231 PMCID: PMC9869290 DOI: 10.2217/nnm-2022-0159] [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: 06/25/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022] Open
Abstract
Aim: To characterize the pharmacokinetics of deferoxamine-conjugated nanoparticles (DFO-NPs), a novel nanochelator for removing excess iron. Materials & methods: The pharmacokinetics of DFO-NPs were evaluated in Sprague-Dawley rats at three doses (3.3, 10 and 30 μmol/kg) after intravenous and subcutaneous administration. Results: DFO-NPs exhibited a biphasic concentration-time profile after intravenous administration with a short terminal half-life (2.0-3.2 h), dose-dependent clearance (0.111-0.179 l/h/kg), minimal tissue distribution and exclusive renal excretion with a possible saturable reabsorption mechanism. DFO-NPs after subcutaneous administration exhibited absorption-rate-limited kinetics with a prolonged half-life (5.7-10.1 h) and favorable bioavailability (47-107%). Conclusion: DFO-NPs exhibit nonlinear pharmacokinetics with increasing dose, and subcutaneous administration substantially improves drug exposure, thereby making it a clinically viable administration route for iron chelation.
Collapse
Affiliation(s)
- Gregory Jones
- Department of Pharmaceutical Sciences, Bouve College of Health Sciences, Northeastern University, Boston, MA 02115, USA
| | - Lingxue Zeng
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Wesley R Stiles
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Seung Hun Park
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Homan Kang
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Hak Soo Choi
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02114, USA
| | - Jonghan Kim
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| |
Collapse
|
42
|
Ismail M, Yang W, Li Y, Wang Y, He W, Wang J, Muhammad P, Chaston TB, Rehman FU, Zheng M, Lovejoy DB, Shi B. Biomimetic Dp44mT-nanoparticles selectively induce apoptosis in Cu-loaded glioblastoma resulting in potent growth inhibition. Biomaterials 2022; 289:121760. [PMID: 36044788 DOI: 10.1016/j.biomaterials.2022.121760] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 08/11/2022] [Accepted: 08/20/2022] [Indexed: 12/25/2022]
Abstract
Selective targeting of elevated copper (Cu) in cancer cells by chelators to induce tumor-toxic reactive oxygen species (ROS) may be a promising approach in the treatment of glioblastoma multiforme (GBM). Previously, the Cu chelator di-2-pyridylketone-4,4-dimethyl-3-thiosemicarbazone (Dp44mT) attracted much interest due to its potent anti-tumor activity mediated by the formation of a highly redox-active Cu-Dp44mT complex. However, its translational potential was limited by the development of toxicity in murine models of cancer reflecting poor selectivity. Here, we overcame the limitations of Dp44mT by incorporating it in new biomimetic nanoparticles (NPs) optimized for GBM therapy. Biomimetic design elements enhancing selectivity included angiopeptide-2 functionalized red blood cell membrane (Ang-M) camouflaging of the NPs carrier. Co-loading Dp44mT with regadenoson (Reg), that transiently opens the blood-brain-barrier (BBB), yielded biomimetic Ang-MNPs@(Dp44mT/Reg) NPs that actively targeted and traversed the BBB delivering Dp44mT specifically to GBM cells. To further improve selectivity, we innovatively pre-loaded GBM tumors with Cu. Oral dosing of U87MG-Luc tumor bearing mice with diacetyl-bis(4-methylthiosemicarbazonato)-copperII (Cu(II)-ATSM), significantly enhanced Cu-level in GBM tumor. Subsequent treatment of mice bearing Cu-enriched orthotopic U87MG-Luc GBM with Ang-MNPs@(Dp44mT/Reg) substantially prevented orthotopic GBM growth and led to maximal increases in median survival time. These results highlighted the importance of both angiopeptide-2 functionalization and tumor Cu-loading required for greater selective cytotoxicity. Targeting Ang-MNPs@(Dp44mT/Reg) NPs also down-regulated antiapoptotic Bcl-2, but up-regulated pro-apoptotic Bax and cleaved-caspase-3, demonstrating the involvement of the apoptotic pathway in GBM suppression. Notably, Ang-MNPs@(Dp44mT/Reg) showed negligible systemic drug toxicity in mice, further indicating therapeutic potential that could be adapted for other central nervous system disorders.
Collapse
Affiliation(s)
- Muhammad Ismail
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Wen Yang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Yanfei Li
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Wenya He
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Jiefei Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Pir Muhammad
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Timothy B Chaston
- University Centre for Rural Health, School of Public Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Fawad Ur Rehman
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Centre for Regenerative Medicine and Stem Cells Research, The Aga Khan University, Stadium Road, Karachi, 78400, Pakistan
| | - Meng Zheng
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences & School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - David B Lovejoy
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, 2109, Australia.
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China; Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, 2109, Australia.
| |
Collapse
|
43
|
Roy C, Avril S, Legendre C, Lelièvre B, Vellenriter H, Boni S, Cayon J, Guillet C, Guilloux Y, Chérel M, Hindré F, Garcion E. A role for ceruloplasmin in the control of human glioblastoma cell responses to radiation. BMC Cancer 2022; 22:843. [PMID: 35918659 PMCID: PMC9347084 DOI: 10.1186/s12885-022-09808-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/16/2022] [Indexed: 11/08/2022] Open
Abstract
Background Glioblastoma (GB) is the most common and most aggressive malignant brain tumor. In understanding its resistance to conventional treatments, iron metabolism and related pathways may represent a novel avenue. As for many cancer cells, GB cell growth is dependent on iron, which is tightly involved in red-ox reactions related to radiotherapy effectiveness. From new observations indicating an impact of RX radiations on the expression of ceruloplasmin (CP), an important regulator of iron metabolism, the aim of the present work was to study the functional effects of constitutive expression of CP within GB lines in response to beam radiation depending on the oxygen status (21% O2 versus 3% O2). Methods and results After analysis of radiation responses (Hoechst staining, LDH release, Caspase 3 activation) in U251-MG and U87-MG human GB cell lines, described as radiosensitive and radioresistant respectively, the expression of 9 iron partners (TFR1, DMT1, FTH1, FTL, MFRN1, MFRN2, FXN, FPN1, CP) were tested by RTqPCR and western blots at 3 and 8 days following 4 Gy irradiation. Among those, only CP was significantly downregulated, both at transcript and protein levels in the two lines, with however, a weaker effect in the U87-MG, observable at 3% O2. To investigate specific role of CP in GB radioresistance, U251-MG and U87-MG cells were modified genetically to obtain CP depleted and overexpressing cells, respectively. Manipulation of CP expression in GB lines demonstrated impact both on cell survival and on activation of DNA repair/damage machinery (γH2AX); specifically high levels of CP led to increased production of reactive oxygen species, as shown by elevated levels of superoxide anion, SOD1 synthesis and cellular Fe2 + . Conclusions Taken together, these in vitro results indicate for the first time that CP plays a positive role in the efficiency of radiotherapy on GB cells. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09808-6.
Collapse
Affiliation(s)
- Charlotte Roy
- Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France
| | - Sylvie Avril
- Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France
| | - Claire Legendre
- Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France
| | - Bénédicte Lelièvre
- Centre Régional de Pharmacovigilance, Laboratoire de Pharmacologie-Toxicologie, CHU Angers, 4 rue Larrey, F-49100, Angers, France
| | - Honorine Vellenriter
- Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France
| | - Sébastien Boni
- Université d'Angers, SFR ICAT, Lentivec, F-49000, Angers, France
| | - Jérôme Cayon
- Université d'Angers, SFR ICAT, PACeM, F-49000, Angers, France
| | | | - Yannick Guilloux
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
| | - Michel Chérel
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, F-44000, Nantes, France
| | - François Hindré
- Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France.,Université d'Angers, SFR ICAT, PRIMEX, F-49000, Angers, France
| | - Emmanuel Garcion
- Université d'Angers, Inserm UMR 1307, CNRS UMR 6075, Nantes Université, CRCI2NA, F-49000, Angers, France. .,Université d'Angers, SFR ICAT, PACeM, F-49000, Angers, France. .,Université d'Angers, SFR ICAT, PRIMEX, F-49000, Angers, France. .,GLIAD - Design and Application of Innovative Local Treatments in Glioblastoma, CRCI2NA, Team 5, Inserm UMR 1307, CNRS UMR 6075, Institut de Biologie en Santé (IBS) - CHU, 4 rue Larrey, Angers, France.
| |
Collapse
|
44
|
Lanthanide coordination polymers functionalized by 5-nitroisophthalic acid: Synthesis, structure-DFT correlation and photoluminescent sensor of Cd2+ ion. J SOLID STATE CHEM 2022. [DOI: 10.1016/j.jssc.2022.123229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
45
|
Abedi M, Rahgozar S. Puzzling Out Iron Complications in Cancer Drug Resistance. Crit Rev Oncol Hematol 2022; 178:103772. [PMID: 35914667 DOI: 10.1016/j.critrevonc.2022.103772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/23/2022] [Accepted: 07/28/2022] [Indexed: 12/09/2022] Open
Abstract
Iron metabolism are frequently disrupted in cancer. Patients with cancer are prone to anemia and receive transfusions frequently; the condition which results in iron overload, contributing to serious therapeutic complications. Iron is introduced as a carcinogen that may increase tumor growth. However, investigations regarding its impact on response to chemotherapy, particularly the induction of drug resistance are still limited. Here, iron contribution to cell signaling and various molecular mechanisms underlying iron-mediated drug resistance are described. A dual role of this vital element in cancer treatment is also addressed. On one hand, the need to administer iron chelators to surmount iron overload and improve the sensitivity of tumor cells to chemotherapy is discussed. On the other hand, the necessary application of iron as a therapeutic option by iron-oxide nanoparticles or ferroptosis inducers is explained. Authors hope that this paper can help unravel the clinical complications related to iron in cancer therapy.
Collapse
Affiliation(s)
- Marjan Abedi
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Soheila Rahgozar
- Department of Cell and Molecular biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
46
|
KAYA B. An Iron(III)-S-methylthiosemicarbazone Complex: Synthesis, Spectral Characterization, and Antioxidant Potency Measured by CUPRAC and DPPH Methods. JOURNAL OF THE TURKISH CHEMICAL SOCIETY, SECTION A: CHEMISTRY 2022. [DOI: 10.18596/jotcsa.1058398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
An iron(III) complex, [Fe(L1)Cl].H2O, was synthesized by template condensation reaction of 1,1,1-Trifluoroacetylacetone-S-methylthiosemicarbazone hydrogen iodide (L) and 2,3-dihydroxybenzaldehyde in the presence of iron(III) ions. The complex was characterized by IR, ESI MS and X-ray diffraction techniques. Free radical scavenging (FRS) ability and antioxidant capacity of the S-methylthiosemicarbazone and the iron(III) complex were evaluated through DPPH and CUPRAC methods, respectively. The complex exerted better than the S-methylthiosemicarbazone in both TEAC and FRS% values. In addition, iron(III) complex was found to be 3.1 times more antioxidant than the reference ascorbic acid according to the CUPRAC method.
Collapse
Affiliation(s)
- Büşra KAYA
- İSTANBUL ÜNİVERSİTESİ-CERRAHPAŞA, MÜHENDİSLİK FAKÜLTESİ
| |
Collapse
|
47
|
Singh J, Sharma M, Jain N, Aftab I, Vikram N, Singh TP, Sharma P, Sharma S. Lactoferrin and its nano-formulations in rare eye diseases. Indian J Ophthalmol 2022; 70:2328-2334. [PMID: 35791114 PMCID: PMC9426081 DOI: 10.4103/ijo.ijo_303_22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/22/2022] [Accepted: 06/03/2022] [Indexed: 11/05/2022] Open
Abstract
Lactoferrin (LF) is an iron-binding glycoprotein released from mucous secreting cells and neutrophils. LF can be used in a broad range of eye diseases related to the retina, cornea, and optic nerve. The retina is particularly affected by oxidative stress inside the photoreceptor being constantly exposed to light which induces accumulation of reactive oxygen species (ROS) in the retinal pigmented epithelium (RPE) causing damage to photoreceptor recycling. Retinitis pigmentosa (RP) and macular degeneration are inherited retinopathies that consist of different disease-causing genes, that cause mutations with highly varied clinical consequences. Age-related macular degeneration is a chronic disease of the retina and one of the major causes of sight loss. This review provides an application of lactoferrin and LF-based nano-formulations or nanoparticles in the field of retinal diseases or corneal diseases such as retinitis pigmentosa, retinoblastoma, age-related macular degeneration (AMD), keratoconus and uveitis. Several studies have found that lactoferrin's antibacterial activity is not limited to its iron sequestration, but also its ability as a nanoparticle that acts as a carrier to deliver drugs by crossing the blood-retina barrier (BRB) and its involvement in cell cycle control, which is not possible by many transferrin proteins.
Collapse
Affiliation(s)
- Jiya Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Mohita Sharma
- Tirupati Eye Centre and Research Institute, Noida, Uttar Pradesh, India
| | - Neha Jain
- The Cornea Institute, KAR Campus, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Insha Aftab
- Tirupati Eye Centre and Research Institute, Noida, Uttar Pradesh, India
| | - Naval Vikram
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Tej P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
48
|
Yang J, Wei H, Liu M, Huang T, Fang X, Ren X, Yuan H, Zheng Y, Wang Y, Zhou Y. Prognostic biomarker HAMP and associates with immune infiltration in gastric cancer. Int Immunopharmacol 2022; 108:108839. [DOI: 10.1016/j.intimp.2022.108839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 04/20/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
49
|
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
|
50
|
Pita-Grisanti V, Chasser K, Sobol T, Cruz-Monserrate Z. Understanding the Potential and Risk of Bacterial Siderophores in Cancer. Front Oncol 2022; 12:867271. [PMID: 35785195 PMCID: PMC9248441 DOI: 10.3389/fonc.2022.867271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/06/2022] [Indexed: 01/19/2023] Open
Abstract
Siderophores are iron chelating molecules produced by nearly all organisms, most notably by bacteria, to efficiently sequester the limited iron that is available in the environment. Siderophores are an essential component of mammalian iron homeostasis and the ongoing interspecies competition for iron. Bacteria produce a broad repertoire of siderophores with a canonical role in iron chelation and the capacity to perform versatile functions such as interacting with other microbes and the host immune system. Siderophores are a vast area of untapped potential in the field of cancer research because cancer cells demand increased iron concentrations to sustain rapid proliferation. Studies investigating siderophores as therapeutics in cancer generally focused on the role of a few siderophores as iron chelators; however, these studies are limited and some show conflicting results. Moreover, siderophores are biologically conserved, structurally diverse molecules that perform additional functions related to iron chelation. Siderophores also have a role in inflammation due to their iron acquisition and chelation properties. These diverse functions may contribute to both risks and benefits as therapeutic agents in cancer. The potential of siderophore-mediated iron and bacterial modulation to be used in the treatment of cancer warrants further investigation. This review discusses the wide range of bacterial siderophore functions and their utilization in cancer treatment to further expand their functional relevance in cancer detection and treatment.
Collapse
Affiliation(s)
- Valentina Pita-Grisanti
- The Ohio State University Interdisciplinary Nutrition Program, The Ohio State University, Columbus, OH, United States
- Division of Gastroenterology, Hepatology, and Nutrition, Division of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, United States
| | - Kaylin Chasser
- Division of Gastroenterology, Hepatology, and Nutrition, Division of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, United States
| | - Trevor Sobol
- Division of Gastroenterology, Hepatology, and Nutrition, Division of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, United States
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, Division of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- The Comprehensive Cancer Center–Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, The Ohio State University, Columbus, OH, United States
- *Correspondence: Zobeida Cruz-Monserrate,
| |
Collapse
|