1
|
Balachandran C, Hirose M, Tanaka T, Zhu JJ, Yokoi K, Hisamatsu Y, Yamada Y, Aoki S. Design and Synthesis of Poly(2,2'-Bipyridyl) Ligands for Induction of Cell Death in Cancer Cells: Control of Anticancer Activity by Complexation/Decomplexation with Biorelevant Metal Cations. Inorg Chem 2023; 62:14615-14631. [PMID: 37642721 PMCID: PMC10498496 DOI: 10.1021/acs.inorgchem.3c01738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Indexed: 08/31/2023]
Abstract
Chelation therapy is a medical procedure for removing toxic metals from human organs and tissues and for the treatment of diseases by using metal-chelating agents. For example, iron chelation therapy is designed not only for the treatment of metal poisoning but also for some diseases that are induced by iron overload, cancer chemotherapy, and related diseases. However, the use of such metal chelators needs to be generally carried out very carefully, because of the side effects possibly due to the non-specific complexation with intracellular metal cations. Herein, we report on the preparation and characterization of some new poly(bpy) ligands (bpy: 2,2'-bipyridyl) that contain one-three bpy ligand moieties and their anticancer activity against Jurkat, MOLT-4, U937, HeLa S3, and A549 cell lines. The results of MTT assays revealed that the tris(bpy) and bis(bpy) ligands exhibit potent activity for inducing the cell death in cancer cells. Mechanistic studies suggest that the main pathway responsible for the cell death by these poly(bpy) ligands is apoptotic cell death. It was also found that the anticancer activity of the poly(bpy) ligands could be controlled by the complexation (anticancer activity is turned OFF) and decomplexation (anticancer activity is turned ON) with biorelevant metal cations. In this paper, these results will be described.
Collapse
Affiliation(s)
- Chandrasekar Balachandran
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
- Research
Institute for Biomedical Sciences, Tokyo
University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Masumi Hirose
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Tomohiro Tanaka
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Jun Jie Zhu
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Kenta Yokoi
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
| | - Yosuke Hisamatsu
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
- Graduate
School of Pharmaceutical Sciences, Nagoya
City University, 3-1
Tanabe-dori, Nagoya, Aichi 467-8603, Japan
| | - Yasuyuki Yamada
- Department
of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
- Research
Center for Materials Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
| | - Shin Aoki
- Faculty
of Pharmaceutical Sciences, Tokyo University
of Science, 2641 Yamazaki, Noda 278-8510, Japan
- Research
Institute for Biomedical Sciences, Tokyo
University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Research
Institute for Science and Technology, Tokyo
University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
2
|
Satoh H, Ochi S, Mizuno K, Saga Y, Ujita S, Toyoda M, Nishiyama Y, Tada K, Matsushita Y, Deguchi Y, Suzuki K, Tanaka Y, Ueda H, Inaba T, Hosoi Y, Morita A, Aoki S. Design, synthesis and biological evaluation of 2-pyrrolone derivatives as radioprotectors. Bioorg Med Chem 2022; 67:116764. [PMID: 35635928 DOI: 10.1016/j.bmc.2022.116764] [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: 03/08/2022] [Revised: 04/16/2022] [Accepted: 04/19/2022] [Indexed: 11/02/2022]
Abstract
It is known that p53 is an important transcription factor and plays a central role in ionizing radiation (IR)-induced DNA damage responses such as cell cycle arrest, DNA repair and apoptosis. We previously reported that regulating p53 protein is an effective strategy for modulating cell fate by reducing the acute side effects of radiation therapy. Herein, we report on the discovery of STK160830 as a new radioprotector from a chemical library at The University of Tokyo and the design, synthesis and biological evaluation of its derivatives. The radioprotective activity of STK160830 itself and its derivatives that were synthesized in this work was evaluated using a leukemia cell line, MOLT-4 cells as a model of normal cells that express the p53 protein in a structure-activity relationships (SAR) study. The experimental results suggest that a direct relationship exists between the inhibitory effect of these STK160830 derivatives on the expression level of p53 and their radioprotective activity and that the suppression of p53 by STK160830 derivatives contribute to protecting MOLT-4 cells from apoptosis that is induced by exposure to radiation.
Collapse
Affiliation(s)
- Hidetoshi Satoh
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Shintaro Ochi
- Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kosuke Mizuno
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yutaka Saga
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Shohei Ujita
- Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Miyu Toyoda
- Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuichi Nishiyama
- Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kasumi Tada
- Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yosuke Matsushita
- Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan; Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Yuichi Deguchi
- Center for Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Keiji Suzuki
- Department of Radiation Medical Sciences, Nagasaki University Atomic Bomb Disease Institute. 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Hiroshi Ueda
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Toshiya Inaba
- Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Yoshio Hosoi
- Department of Radiation Biology, Graduate School of Medicine, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Akinori Morita
- Tokushima University, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Research Institute for Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan; Research Institute for Biomedical Science, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
3
|
Ganesan S, Parvathi VD. Deconstructing the molecular genetics behind the PINK1/Parkin axis in Parkinson’s disease using Drosophila melanogaster as a model organism. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00208-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Abstract
Background
Parkinson’s disease (PD) is a multifactorial neurodegenerative disorder marked by the death of nigrostriatal dopaminergic neurons in response to the compounding effects of oxidative stress, mitochondrial dysfunction and protein aggregation. Transgenic Drosophila models have been used extensively to decipher the underlying genetic interactions that exacerbate neural health in PD. Autosomal recessive forms of the disease have been linked to mutations in the serine/threonine kinase PINK1(PTEN-Induced Putative Kinase 1) and E3 ligase Parkin, which function in an axis that is conserved in flies. This review aims to probe the current understanding of PD pathogenesis via the PINK1/Parkin axis while underscoring the importance of several molecular and pharmacologic rescues brought to light through studies in Drosophila.
Main body
Mutations in PINK1 and Parkin have been shown to affect the axonal transport of mitochondria within dopaminergic neurons and perturb the balance between mitochondrial fusion/fission resulting in abnormal mitochondrial morphology. As per studies in flies, ectopic expression of Fwd kinase and Atg-1 to promote fission and mitophagy while suppressing fusion via MUL1 E3 ligase may aid to halt mitochondrial aggregation and prolong the survival of dopaminergic neurons. Furthermore, upregulation of Hsp70/Hsp90 chaperone systems (Trap1, CHIP) to target misfolded mitochondrial respiratory complexes may help to preserve their bioenergetic capacity. Accumulation of reactive oxygen species as a consequence of respiratory complex dysfunction or antioxidant enzyme deficiency further escalates neural death by inducing apoptosis, lipid peroxidation and DNA damage. Fly studies have reported the induction of canonical Wnt signalling to enhance the activity of transcriptional co-activators (PGC1α, FOXO) which induce the expression of antioxidant enzymes. Enhancing the clearance of free radicals via uncoupling proteins (UCP4) has also been reported to ameliorate oxidative stress-induced cell death in PINK1/Parkin mutants.
Conclusion
While these novel mechanisms require validation through mammalian studies, they offer several explanations for the factors propagating dopaminergic death as well as promising insights into the therapeutic importance of transgenic fly models in PD.
Collapse
|
4
|
Morita A, Ochi S, Satoh H, Ujita S, Matsushita Y, Tada K, Toyoda M, Nishiyama Y, Mizuno K, Deguchi Y, Suzuki K, Tanaka Y, Ueda H, Inaba T, Hosoi Y, Aoki S. A Novel RNA Synthesis Inhibitor, STK160830, Has Negligible DNA-Intercalating Activity for Triggering A p53 Response, and Can Inhibit p53-Dependent Apoptosis. Life (Basel) 2021; 11:life11101087. [PMID: 34685458 PMCID: PMC8539076 DOI: 10.3390/life11101087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/04/2021] [Accepted: 10/10/2021] [Indexed: 11/16/2022] Open
Abstract
RNA synthesis inhibitors and protein synthesis inhibitors are useful for investigating whether biological events with unknown mechanisms require transcription or translation; however, the dependence of RNA synthesis has been difficult to verify because many RNA synthesis inhibitors cause adverse events that trigger a p53 response. In this study, we screened a library containing 9600 core compounds and obtained STK160830 that shows anti-apoptotic effects in irradiated wild-type-p53-bearing human T-cell leukemia MOLT-4 cells and murine thymocytes. In many of the p53-impaired cells and p53-knockdown cells tested, STK160830 did not show a remarkable anti-apoptotic effect, suggesting that the anti-apoptotic activity is p53-dependent. In the expression analysis of p53, p53-target gene products, and reference proteins by immunoblotting, STK160830 down-regulated the expression of many of the proteins examined, and the downregulation correlated strongly with its inhibitory effect on cell death. mRNA expression analyses by qPCR and nascent RNA capture kit revealed that STK160830 showed a decreased mRNA expression, which was similar to that induced by the RNA synthesis inhibitor actinomycin D but differed to some extent. Furthermore, unlike other RNA synthesis inhibitors such as actinomycin D, p53 accumulation by STK160830 alone was negligible, and a DNA melting-curve analysis showed very weak DNA-intercalating activity, indicating that STK160830 is a useful inhibitor for RNA synthesis without triggering p53-mediated damage responses.
Collapse
Affiliation(s)
- Akinori Morita
- Tokushima University, Tokushima 770-8503, Japan; (S.O.); (S.U.); (Y.M.); (K.T.); (M.T.); (Y.N.)
- Correspondence:
| | - Shintaro Ochi
- Tokushima University, Tokushima 770-8503, Japan; (S.O.); (S.U.); (Y.M.); (K.T.); (M.T.); (Y.N.)
| | - Hidetoshi Satoh
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (H.S.); (K.M.); (S.A.)
| | - Shohei Ujita
- Tokushima University, Tokushima 770-8503, Japan; (S.O.); (S.U.); (Y.M.); (K.T.); (M.T.); (Y.N.)
| | - Yosuke Matsushita
- Tokushima University, Tokushima 770-8503, Japan; (S.O.); (S.U.); (Y.M.); (K.T.); (M.T.); (Y.N.)
- Nagasaki University, Nagasaki 852-8521, Japan; (Y.D.); (K.S.); (Y.T.); (H.U.)
| | - Kasumi Tada
- Tokushima University, Tokushima 770-8503, Japan; (S.O.); (S.U.); (Y.M.); (K.T.); (M.T.); (Y.N.)
| | - Mihiro Toyoda
- Tokushima University, Tokushima 770-8503, Japan; (S.O.); (S.U.); (Y.M.); (K.T.); (M.T.); (Y.N.)
| | - Yuichi Nishiyama
- Tokushima University, Tokushima 770-8503, Japan; (S.O.); (S.U.); (Y.M.); (K.T.); (M.T.); (Y.N.)
| | - Kosuke Mizuno
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (H.S.); (K.M.); (S.A.)
| | - Yuichi Deguchi
- Nagasaki University, Nagasaki 852-8521, Japan; (Y.D.); (K.S.); (Y.T.); (H.U.)
| | - Keiji Suzuki
- Nagasaki University, Nagasaki 852-8521, Japan; (Y.D.); (K.S.); (Y.T.); (H.U.)
| | - Yoshimasa Tanaka
- Nagasaki University, Nagasaki 852-8521, Japan; (Y.D.); (K.S.); (Y.T.); (H.U.)
| | - Hiroshi Ueda
- Nagasaki University, Nagasaki 852-8521, Japan; (Y.D.); (K.S.); (Y.T.); (H.U.)
| | - Toshiya Inaba
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan;
| | - Yoshio Hosoi
- Department of Radiation Biology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan;
| | - Shin Aoki
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan; (H.S.); (K.M.); (S.A.)
| |
Collapse
|
5
|
Nishiyama Y, Morita A, Tatsuta S, Kanamaru M, Sakaue M, Ueda K, Shono M, Fujita R, Wang B, Hosoi Y, Aoki S, Sugai T. Isorhamnetin Promotes 53BP1 Recruitment through the Enhancement of ATM Phosphorylation and Protects Mice from Radiation Gastrointestinal Syndrome. Genes (Basel) 2021; 12:genes12101514. [PMID: 34680909 PMCID: PMC8535534 DOI: 10.3390/genes12101514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022] Open
Abstract
Flavonoids are a subclass of polyphenols which are attractive, due to possessing various physiological activities, including a radioprotective effect. Tumor suppressor p53 is a primary regulator in the radiation response and is involved in the pathogenesis of radiation injuries. In this study, we revealed that isorhamnetin inhibited radiation cell death, and investigated its action mechanism focusing on DNA damage response. Although isorhamnetin moderated p53 activity, it promoted phosphorylation of ataxia telangiectasia mutated (ATM) and enhanced 53BP1 recruitment in irradiated cells. The radioprotective effect of isorhamnetin was not observed in the presence of ATM inhibitor, indicating that its protective effect was dependent on ATM. Furthermore, isorhamnetin-treated mice survived gastrointestinal death caused by a lethal dose of abdominal irradiation. These findings suggested that isorhamnetin enhances the ATM-dependent DNA repair process, which is presumably associated with the suppressive effect against GI syndrome.
Collapse
Affiliation(s)
- Yuichi Nishiyama
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (Y.N.); (S.T.); (M.K.); (M.S.); (K.U.); (M.S.)
| | - Akinori Morita
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (Y.N.); (S.T.); (M.K.); (M.S.); (K.U.); (M.S.)
- Correspondence:
| | - Shogo Tatsuta
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (Y.N.); (S.T.); (M.K.); (M.S.); (K.U.); (M.S.)
| | - Misaki Kanamaru
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (Y.N.); (S.T.); (M.K.); (M.S.); (K.U.); (M.S.)
| | - Masahiro Sakaue
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (Y.N.); (S.T.); (M.K.); (M.S.); (K.U.); (M.S.)
| | - Kenta Ueda
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (Y.N.); (S.T.); (M.K.); (M.S.); (K.U.); (M.S.)
| | - Manami Shono
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; (Y.N.); (S.T.); (M.K.); (M.S.); (K.U.); (M.S.)
| | - Rie Fujita
- Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan; (R.F.); (T.S.)
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan;
| | - Yoshio Hosoi
- Department of Radiation Biology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan;
| | - Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-8510, Japan;
| | - Takeshi Sugai
- Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan; (R.F.); (T.S.)
| |
Collapse
|
6
|
La Piana L, Viaggi V, Principe L, Di Bella S, Luzzaro F, Viale M, Bertola N, Vecchio G. Polypyridine ligands as potential metallo-β-lactamase inhibitors. J Inorg Biochem 2020; 215:111315. [PMID: 33285370 DOI: 10.1016/j.jinorgbio.2020.111315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 11/19/2022]
Abstract
Bacteria have developed multiple resistance mechanisms against the most used antibiotics. In particular, zinc-dependent metallo-β-lactamase producing bacteria are a growing threat, and therapeutic options are limited. Zinc chelators have recently been investigated as metallo-β-lactamase inhibitors, as they are often able to restore carbapenem susceptibility. We synthesized polypyridyl ligands, N,N'-bis(2-pyridylmethyl)-ethylenediamine, N,N,N'-tris(2-pyridylmethyl)-ethylenediamine, N,N'-bis(2-pyridylmethyl)-ethylenediamine-N-acetic acid (N,N,N'-tris(2-pyridylmethyl)-ethylenediamine-N'-acetic acid, which can form zinc(II) complexes. We tested their ability to restore the antibiotic activity of meropenem against three clinical strains isolated from blood and metallo-β-lactamase producers (Klebsiella pneumoniae, Enterobacter cloacae, and Stenotrophomonas maltophilia). We functionalized N,N,N'-tris(2-pyridylmethyl)-ethylenediamine with D-alanyl-D-alanyl-D-alanine methyl ester with the aim to increase bacterial uptake. We observed synergistic activity of four polypyridyl ligands with meropenem against all tested isolates, while the combination N,N'-bis(2-pyridylmethyl)-ethylenediamine and meropenem was synergistic only against New Delhi and Verona integron-encoded metallo-β-lactamase-producing bacteria. All synergistic interactions restored the antimicrobial activity of meropenem, providing a significant decrease of minimal inhibitory concentration value (by 8- to 128-fold). We also studied toxicity of the ligands in two normal peripheral blood lymphocytes.
Collapse
Affiliation(s)
- Luana La Piana
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, V.le A. Doria 6, 95125 Catania, Italy
| | - Valentina Viaggi
- Clinical Microbiology and Virology Unit, A. Manzoni Hospital, Via dell'Eremo 9/11, 23900 Lecco, Italy
| | - Luigi Principe
- Clinical Pathology and Microbiology Unit, San Giovanni di Dio Hospital, Largo Bologna, 88900 Crotone, Italy
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, strada di Fiume 447, 34149 Trieste, Italy
| | - Francesco Luzzaro
- Clinical Microbiology and Virology Unit, A. Manzoni Hospital, Via dell'Eremo 9/11, 23900 Lecco, Italy
| | - Maurizio Viale
- IRCCS Ospedale Policlinico San Martino, U.O. Bioterapie, L.go R. Benzi 10, 16132 Genova, Italy
| | - Nadia Bertola
- IRCCS Ospedale Policlinico San Martino, U.O. Bioterapie, L.go R. Benzi 10, 16132 Genova, Italy
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, V.le A. Doria 6, 95125 Catania, Italy; Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB), Piazza Umberto I 1, 70121 Bari, Italy.
| |
Collapse
|
7
|
Khodamoradi E, Hoseini-Ghahfarokhi M, Amini P, Motevaseli E, Shabeeb D, Musa AE, Najafi M, Farhood B. Targets for protection and mitigation of radiation injury. Cell Mol Life Sci 2020; 77:3129-3159. [PMID: 32072238 PMCID: PMC11104832 DOI: 10.1007/s00018-020-03479-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Abstract
Protection of normal tissues against toxic effects of ionizing radiation is a critical issue in clinical and environmental radiobiology. Investigations in recent decades have suggested potential targets that are involved in the protection against radiation-induced damages to normal tissues and can be proposed for mitigation of radiation injury. Emerging evidences have been shown to be in contrast to an old dogma in radiation biology; a major amount of reactive oxygen species (ROS) production and cell toxicity occur during some hours to years after exposure to ionizing radiation. This can be attributed to upregulation of inflammatory and fibrosis mediators, epigenetic changes and disruption of the normal metabolism of oxygen. In the current review, we explain the cellular and molecular changes following exposure of normal tissues to ionizing radiation. Furthermore, we review potential targets that can be proposed for protection and mitigation of radiation toxicity.
Collapse
Affiliation(s)
- Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mojtaba Hoseini-Ghahfarokhi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
- Misan Radiotherapy Center, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
8
|
Haematoxylon campechianum Extract Ameliorates Neuropathic Pain via Inhibition of NF-κB/TNF-α/NOX/iNOS Signalling Pathway in a Rat Model of Chronic Constriction Injury. Biomolecules 2020; 10:biom10030386. [PMID: 32131490 PMCID: PMC7175380 DOI: 10.3390/biom10030386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/13/2020] [Accepted: 02/27/2020] [Indexed: 12/13/2022] Open
Abstract
: In this study, the phytochemical composition and the possible prophylactic effects of an aqueous ethanol extract of Haematoxylon campechianum flowers (HCF) on peripheral neuropathic pain in a chronic constriction injury (CCI) rat model are investigated. Rats with induced CCI were subjected to neuropathic pain behaviour tests and evaluated by chemical, thermal, and mechanical sensation tests and functional recovery of the brain stem and sciatic nerve at 7- and 14-day intervals. The effect of the extract on acute pain and inflammation is also investigated. The extract exerted both peripheral and central analgesic and anti-inflammatory properties in addition to antipyretic effects that are clear from targeting COX, LOX and PGE. It was found that CCI produced significant thermal and mechanical hyperalgesia, cold allodynia and deleterious structural changes in both sciatic nerve and brain stem. Treatments with HCF extract significantly improved cold and thermal withdrawal latency, mechanical sensibility and ameliorated deleterious changes of sciatic nerve and brain stem at different dose levels. The extract also ameliorated oxidative stress and inflammatory markers in brain stem and sciatic nerve. It suppressed the apoptotic marker, p53, and restored myelin sheath integrity. The effects of HCF extract were more potent than pregabalin. Fifteen secondary metabolites, mainly gallotannins and flavonoids, were characterized in the extract based on their retention times and MS/MS data. The identified phenolic constituents from the extract could be promising candidates to treat neuropathic pain due to their diverse biological activities, including antioxidant, anti-inflammatory and neuroprotective properties.
Collapse
|
9
|
Ochi S, Nishiyama Y, Morita A. Development of p53-targeting drugs that increase radioresistance in normal tissues. THE JOURNAL OF MEDICAL INVESTIGATION 2019; 66:219-223. [PMID: 31656277 DOI: 10.2152/jmi.66.219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Radiation damage to normal tissues is a serious concern in radiation therapy. Advances in radiotherapeutic technology have improved the dose distribution of the target volumes and risk organs, but damage to risk organs that are located within the irradiation field still limits the allowable prescription dose. To overcome this dose-limiting toxicity, and to further improve the efficacy of radiotherapy, the development of drugs that protect normal tissues but not cancer tissues from the effects of radiation are expected to be developed based on molecular target-based drugs. p53 is a well-known transcription factor that is closely associated with radiation-induced cell death. In radiation-injured tissues, p53 induces apoptosis in hematopoietic lineages, whereas it plays a radioprotective role in the gastrointestinal epithelium. These facts suggest that p53 inhibitor would be effective for radioprotection of the hematopoietic system, and that a drug that upregulates the radioprotective functions of p53 would enhance the radioresistance of gastrointestinal tissues. In this review, we summarize recent progress regarding the prevention of radiation injury by regulating p53 and provide new strategic insights into the development of radioprotectors in radiotherapy. J. Med. Invest. 66 : 219-223, August, 2019.
Collapse
Affiliation(s)
- Shintaro Ochi
- Department of Biomedical Science and Technology, Graduate School of Biomedical Sciences, Tokushima University
| | - Yuichi Nishiyama
- Department of Biomedical Science and Technology, Graduate School of Biomedical Sciences, Tokushima University
| | - Akinori Morita
- Department of Biomedical Science and Technology, Graduate School of Biomedical Sciences, Tokushima University
| |
Collapse
|
10
|
Sobeh M, Mahmoud MF, Rezq S, Alsemeh AE, Sabry OM, Mostafa I, Abdelfattah MAO, El-Allem KA, El-Shazly AM, Yasri A, Wink M. Salix tetrasperma Roxb. Extract Alleviates Neuropathic Pain in Rats via Modulation of the NF-κB/TNF-α/NOX/iNOS Pathway. Antioxidants (Basel) 2019; 8:antiox8100482. [PMID: 31614846 PMCID: PMC6826723 DOI: 10.3390/antiox8100482] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 11/16/2022] Open
Abstract
Patients with neuropathic pain experience chronic painful tingling, burning, and prickling sensations accompanied with hyperalgesia and/or allodynia. In this study, 38 secondary metabolites of a methanol extract from Salix tetrasperma flowers were identified by liquid chromatography-mass spectrometry (HPLC-MS/MS). The extract showed substantial anti-inflammatory, central and peripheral anti-nociceptive, antipyretic, and antioxidant activities in vitro and in different animal models. In the chronic constriction injury (CCI) rat model, the extract was able to attenuate and significantly relieve hyperalgesia and allodynia responses in a dose dependent manner and restore the myelin sheath integrity and Schwann cells average number in the sciatic nerve. The enzyme-linked immunosorbent assay (ELISA) showed that the extract significantly reduced the expression of various pro-inflammatory biomarkers including nuclear factor kabba B (NF-κB), tumor necrosis factor alpha (TNF-α), prostaglandin E2 (PGE2), 5-lipoxygenase (5-LOX), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and the oxidative stress biomarker NADPH oxidase 1 (NOX1), in brain stem and sciatic nerve tissues. These findings were supported by in vitro enzyme inhibition assays (COX-1, COX-2 and 5-LOX). Moreover, the extract significantly reduced p53 expression in the brain stem tissue. These findings support the use of S. tetrasperma in folk medicine to alleviate pain. It could be a promising natural product for further clinical investigations to treat inflammation, nociceptive pain and chronic neuropathic pain.
Collapse
Affiliation(s)
- Mansour Sobeh
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg 69120, Germany.
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660-Hay MoulayRachid, 43150 Ben-Guerir, Morocco.
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt.
| | - Samar Rezq
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt.
| | - Amira E Alsemeh
- Department of Anatomy and Embryology, Faculty of Medicine, Zagazig University, 44519 Zagazig, Egypt.
| | - Omar M Sabry
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Islam Mostafa
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt.
| | - Mohamed A O Abdelfattah
- College of Engineering and Technology, American University of the Middle East, Egaila 54200, Kuwait.
| | - Khadija Ait El-Allem
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660-Hay MoulayRachid, 43150 Ben-Guerir, Morocco.
| | - Assem M El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, 44519 Zagazig, Egypt.
| | - Aziz Yasri
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660-Hay MoulayRachid, 43150 Ben-Guerir, Morocco.
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg 69120, Germany.
| |
Collapse
|
11
|
Kocik J, Machula M, Wisniewska A, Surmiak E, Holak TA, Skalniak L. Helping the Released Guardian: Drug Combinations for Supporting the Anticancer Activity of HDM2 (MDM2) Antagonists. Cancers (Basel) 2019; 11:E1014. [PMID: 31331108 PMCID: PMC6678622 DOI: 10.3390/cancers11071014] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/13/2019] [Accepted: 07/16/2019] [Indexed: 01/22/2023] Open
Abstract
The protein p53, known as the "Guardian of the Genome", plays an important role in maintaining DNA integrity, providing protection against cancer-promoting mutations. Dysfunction of p53 is observed in almost every cancer, with 50% of cases bearing loss-of-function mutations/deletions in the TP53 gene. In the remaining 50% of cases the overexpression of HDM2 (mouse double minute 2, human homolog) protein, which is a natural inhibitor of p53, is the most common way of keeping p53 inactive. Disruption of HDM2-p53 interaction with the use of HDM2 antagonists leads to the release of p53 and expression of its target genes, engaged in the induction of cell cycle arrest, DNA repair, senescence, and apoptosis. The induction of apoptosis, however, is restricted to only a handful of p53wt cells, and, generally, cancer cells treated with HDM2 antagonists are not efficiently eliminated. For this reason, HDM2 antagonists were tested in combinations with multiple other therapeutics in a search for synergy that would enhance the cancer eradication. This manuscript aims at reviewing the recent progress in developing strategies of combined cancer treatment with the use of HDM2 antagonists.
Collapse
Affiliation(s)
- Justyna Kocik
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Monika Machula
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Aneta Wisniewska
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Ewa Surmiak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Tad A Holak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland
| | - Lukasz Skalniak
- Department of Organic Chemistry, Faculty of Chemistry, Jagiellonian University, ul. Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
12
|
Furusawa Y, Yamanouchi Y, Iizumi T, Zhao QL, Mitsuhashi Y, Morita A, Enomoto A, Tabuchi Y, Kondo T. Checkpoint kinase 2 is dispensable for regulation of the p53 response but is required for G 2/M arrest and cell survival in cells with p53 defects under heat stress. Apoptosis 2018; 22:1225-1234. [PMID: 28733865 DOI: 10.1007/s10495-017-1402-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hyperthermia induced by heat stress (HS) is known to inhibit proliferation and induce cell death in cancer. We previously demonstrated that checkpoint kinase 1 (Chk1) contributes to G2/M arrest and cell survival under HS; however, the role of Chk2, a functional analog of Chk1, in regulation of the cell cycle and cell death under HS is still unknown. Here, we addressed the role of Chk2 using Molt-4 cells with p53-targeted shRNA (Molt-4/shp53) and parental control cells (Molt-4/V). Chk2 inhibition suppressed C-terminal acetylation of p53 and delayed the induction of p53-target genes in Molt-4/V cells under HS; however, Chk2 inhibition failed to inhibit apoptosis induced by HS, indicating that Chk2 was dispensable for p53-dependent apoptosis under HS. In contrast, Chk2 inhibition abrogated G2/M arrest and promoted cell death induced by HS in HeLa cells and Molt-4/shp53 cells. Thus, we demonstrated for the first time that Chk2 was required for cell cycle arrest and cell survival, particularly in cells with p53 defects under HS. These findings indicated that Chk2 may be a selective target for p53-mutated or -deficient cancer treated with hyperthermia.
Collapse
Affiliation(s)
- Yukihiro Furusawa
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Kurokawa 5180, Toyama, 939-0398, Japan. .,Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.
| | - Yuka Yamanouchi
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Kurokawa 5180, Toyama, 939-0398, Japan
| | - Takashi Iizumi
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan.,Proton Beam Therapy Center, Tsukuba University Hospital Radiation Oncology, Tsukuba, Japan
| | - Qing-Li Zhao
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Yohei Mitsuhashi
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Akinori Morita
- Department of Radiological Science, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Atushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetic Research, Life Science Research Center, University of Toyama, Toyama, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| |
Collapse
|
13
|
Morita A, Takahashi I, Sasatani M, Aoki S, Wang B, Ariyasu S, Tanaka K, Yamaguchi T, Sawa A, Nishi Y, Teraoka T, Ujita S, Kawate Y, Yanagawa C, Tanimoto K, Enomoto A, Nenoi M, Kamiya K, Nagata Y, Hosoi Y, Inaba T. A Chemical Modulator of p53 Transactivation that Acts as a Radioprotective Agonist. Mol Cancer Ther 2017; 17:432-442. [PMID: 28939557 DOI: 10.1158/1535-7163.mct-16-0554] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 05/22/2017] [Accepted: 08/23/2017] [Indexed: 11/16/2022]
Abstract
Inhibiting p53-dependent apoptosis by inhibitors of p53 is an effective strategy for preventing radiation-induced damage in hematopoietic lineages, while p53 and p21 also play radioprotective roles in the gastrointestinal epithelium. We previously identified some zinc(II) chelators, including 8-quinolinol derivatives, that suppress apoptosis in attempts to discover compounds that target the zinc-binding site in p53. We found that 5-chloro-8-quinolinol (5CHQ) has a unique p53-modulating activity that shifts its transactivation from proapoptotic to protective responses, including enhancing p21 induction and suppressing PUMA induction. This p53-modulating activity also influenced p53 and p53-target gene expression in unirradiated cells without inducing DNA damage. The specificity of 5CHQ for p53 and p21 was demonstrated by silencing the expression of each protein. These effects seem to be attributable to the sequence-specific alteration of p53 DNA-binding, as evaluated by chromatin immunoprecipitation and electrophoretic mobility shift assays. In addition, 5-chloro-8-methoxyquinoline itself had no antiapoptotic activity, indicating that the hydroxyl group at the 8-position is required for its antiapoptotic activity. We applied this remarkable agonistic activity to protecting the hematopoietic and gastrointestinal system in mouse irradiation models. The dose reduction factors of 5CHQ in total-body and abdominally irradiated mice were about 1.2 and 1.3, respectively. 5CHQ effectively protected mouse epithelial stem cells from a lethal dose of abdominal irradiation. Furthermore, the specificity of 5CHQ for p53 in reducing the lethality induced by abdominal irradiation was revealed in Trp53-KO mice. These results indicate that the pharmacologic upregulation of radioprotective p53 target genes is an effective strategy for addressing the gastrointestinal syndrome. Mol Cancer Ther; 17(2); 432-42. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."
Collapse
Affiliation(s)
- Akinori Morita
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan. .,Department of Biomedical Science and Technology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Ippei Takahashi
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Department of Radiation Oncology, Hiroshima University, Hiroshima, Japan
| | - Megumi Sasatani
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shin Aoki
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan.,Center for Technologies against Cancer, Tokyo University of Science, Chiba, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Shinya Ariyasu
- Center for Technologies against Cancer, Tokyo University of Science, Chiba, Japan
| | - Kaoru Tanaka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Tetsuji Yamaguchi
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Akiko Sawa
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Yurie Nishi
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Tatsuro Teraoka
- Department of Medicinal and Life Science, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shohei Ujita
- Department of Biomedical Science and Technology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yosuke Kawate
- Department of Biomedical Science and Technology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Chihiro Yanagawa
- Department of Biomedical Science and Technology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Keiji Tanimoto
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Atsushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsuru Nenoi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kenji Kamiya
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Hiroshima University, Hiroshima, Japan
| | - Yoshio Hosoi
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.,Department of Radiation Biology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Toshiya Inaba
- Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
14
|
Wang P, Wang ZY. Metal ions influx is a double edged sword for the pathogenesis of Alzheimer's disease. Ageing Res Rev 2017; 35:265-290. [PMID: 27829171 DOI: 10.1016/j.arr.2016.10.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 09/08/2016] [Accepted: 10/17/2016] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is a common form of dementia in aged people, which is defined by two pathological characteristics: β-amyloid protein (Aβ) deposition and tau hyperphosphorylation. Although the mechanisms of AD development are still being debated, a series of evidence supports the idea that metals, such as copper, iron, zinc, magnesium and aluminium, are involved in the pathogenesis of the disease. In particular, the processes of Aβ deposition in senile plaques (SP) and the inclusion of phosphorylated tau in neurofibrillary tangles (NFTs) are markedly influenced by alterations in the homeostasis of the aforementioned metal ions. Moreover, the mechanisms of oxidative stress, synaptic plasticity, neurotoxicity, autophagy and apoptosis mediate the effects of metal ions-induced the aggregation state of Aβ and phosphorylated tau on AD development. More importantly, imbalance of these mechanisms finally caused cognitive decline in different experiment models. Collectively, reconstructing the signaling network that regulates AD progression by metal ions may provide novel insights for developing chelators specific for metal ions to combat AD.
Collapse
Affiliation(s)
- Pu Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, PR China.
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, No. 3-11, Wenhua Road, Shenyang, 110819, PR China.
| |
Collapse
|
15
|
Kamran MZ, Ranjan A, Kaur N, Sur S, Tandon V. Radioprotective Agents: Strategies and Translational Advances. Med Res Rev 2016; 36:461-93. [PMID: 26807693 DOI: 10.1002/med.21386] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/15/2015] [Accepted: 01/01/2016] [Indexed: 01/08/2023]
Abstract
Radioprotectors are agents required to protect biological system exposed to radiation, either naturally or through radiation leakage, and they protect normal cells from radiation injury in cancer patients undergoing radiotherapy. It is imperative to study radioprotectors and their mechanism of action comprehensively, looking at their potential therapeutic applications. This review intimately chronicles the rich intellectual, pharmacological story of natural and synthetic radioprotectors. A continuous effort is going on by researchers to develop clinically promising radioprotective agents. In this article, for the first time we have discussed the impact of radioprotectors on different signaling pathways in cells, which will create a basis for scientific community working in this area to develop novel molecules with better therapeutic efficacy. The bright future of exceptionally noncytotoxic derivatives of bisbenzimidazoles is also described as radiomodulators. Amifostine, an effective radioprotectant, has been approved by the FDA for limited clinical use. However, due to its adverse side effects, it is not routinely used clinically. Recently, CBLB502 and several analog of a peptide are under clinical trial and showed high success against radiotherapy in cancer. This article reviews the different types of radioprotective agents with emphasis on the strategies for the development of novel radioprotectors for drug development. In addition, direction for future strategies relevant to the development of radioprotectors is also addressed.
Collapse
Affiliation(s)
- Mohammad Zahid Kamran
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Atul Ranjan
- Kansas University of Medical Center, Kansas City, KS, 66160
| | - Navrinder Kaur
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, 110007, India
| | - Souvik Sur
- Department of Chemistry, University of Delhi, Delhi, 110007, India
| | - Vibha Tandon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India.,Department of Chemistry, University of Delhi, Delhi, 110007, India
| |
Collapse
|
16
|
Aoki S, Ariyasu S, Hanaya K, Hisamatsu Y, Sugai T. Chemical Reactions of 8-Quinolinol Derivatives and Their Applications to Biochemical Tools and Enzyme Inhibitors. J SYN ORG CHEM JPN 2016. [DOI: 10.5059/yukigoseikyokaishi.74.482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Shinya Ariyasu
- School of Physical & Mathematical Sciences, Nanyang Technological University
| | | | | | | |
Collapse
|
17
|
Amelio I, Antonov AA, Catani MV, Massoud R, Bernassola F, Knight RA, Melino G, Rufini A. TAp73 promotes anabolism. Oncotarget 2015; 5:12820-934. [PMID: 25514460 PMCID: PMC4350352 DOI: 10.18632/oncotarget.2667] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 12/18/2022] Open
Abstract
Metabolic adaptation has emerged as a hallmark of cancer and a promising therapeutic target, as rapidly proliferating cancer cells adapt their metabolism increasing nutrient uptake and reorganizing metabolic fluxes to support biosynthesis. The transcription factor p73 belongs to the p53-family and regulates tumorigenesis via its two N-terminal isoforms, with (TAp73) or without (ΔNp73) a transactivation domain. TAp73 acts as tumor suppressor, at least partially through induction of cell cycle arrest and apoptosis and through regulation of genomic stability. Here, we sought to investigate whether TAp73 also affects metabolic profiling of cancer cells. Using high throughput metabolomics, we unveil a thorough and unexpected role for TAp73 in promoting Warburg effect and cellular metabolism. TAp73-expressing cells show increased rate of glycolysis, higher amino acid uptake and increased levels and biosynthesis of acetyl-CoA. Moreover, we report an extensive TAp73-mediated upregulation of several anabolic pathways including polyamine and synthesis of membrane phospholipids. TAp73 expression also increases cellular methyl-donor S-adenosylmethionine (SAM), possibly influencing methylation and epigenetics, and promotes arginine metabolism, suggestive of a role in extracellular matrix (ECM) modeling. In summary, our data indicate that TAp73 regulates multiple metabolic pathways that impinge on numerous cellular functions, but that, overall, converge to sustain cell growth and proliferation.
Collapse
Affiliation(s)
- Ivano Amelio
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Alexey A Antonov
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Maria Valeria Catani
- Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Renato Massoud
- Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Francesca Bernassola
- Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy
| | - Richard A Knight
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK
| | - Gerry Melino
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK. Biochemistry Laboratory, IDI-IRCCS, University of Rome Tor Vergata, Rome 00133, Italy. Molecular Pharmacology Laboratory, Technological University, St-Petersburg, Russia
| | - Alessandro Rufini
- Medical Research Council, Toxicology Unit, Leicester University, Leicester LE1 9HN, UK. Department of Cancer Studies, Cancer Research UK, Leicester Centre, University of Leicester, Leicester, LE1 7RH, UK
| |
Collapse
|
18
|
Design and synthesis of 8-hydroxyquinoline-based radioprotective agents. Bioorg Med Chem 2014; 22:3891-905. [DOI: 10.1016/j.bmc.2014.06.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 11/22/2022]
|
19
|
AS-2, a novel inhibitor of p53-dependent apoptosis, prevents apoptotic mitochondrial dysfunction in a transcription-independent manner and protects mice from a lethal dose of ionizing radiation. Biochem Biophys Res Commun 2014; 450:1498-504. [PMID: 25026551 DOI: 10.1016/j.bbrc.2014.07.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 07/05/2014] [Indexed: 11/20/2022]
Abstract
In a previous study, we reported that some tetradentate zinc(II) chelators inhibit p53 through the denaturation of its zinc-requiring structure but a chelator, Bispicen, a potent inhibitor of in vitro apoptosis, failed to show any efficient radioprotective effect against irradiated mice because the toxicity of the chelator to mice. The unsuitability of using tetradentate chelators as radioprotectors prompted us to undertake a more extensive search for p53-inhibiting agents that are weaker zinc(II) chelators and therefore less toxic. Here, we show that an 8-hydroxyquinoline (8HQ) derivative, AS-2, suppresses p53-dependent apoptosis through a transcription-independent mechanism. A mechanistic study using cells with different p53 characteristics revealed that the suppressive effect of AS-2 on apoptosis is specifically mediated through p53. In addition, AS-2 was less effective in preventing p53-mediated transcription-dependent events than pifithrin-μ (PFTμ), an inhibitor of transcription-independent apoptosis by p53. Fluorescence visualization of the extranuclear distribution of AS-2 also supports that it is ineffective on the transcription-dependent pathway. Further investigations revealed that AS-2 suppressed mitochondrial apoptotic events, such as the mitochondrial release of intermembrane proteins and the loss of mitochondrial membrane potential, although AS-2 resulted in an increase in the mitochondrial translocation of p53 as opposed to the decrease of cytosolic p53, and did not affect the apoptotic interaction of p53 with Bcl-2. AS-2 also protected mice that had been exposed to a lethal dose of ionizing radiation. Our findings indicate that some types of bidentate 8HQ chelators could serve as radioprotectors with no substantial toxicity in vivo.
Collapse
|
20
|
Degradation of mutant p53H175 protein by Zn(II) through autophagy. Cell Death Dis 2014; 5:e1271. [PMID: 24874727 PMCID: PMC4044866 DOI: 10.1038/cddis.2014.217] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 04/12/2014] [Accepted: 04/15/2014] [Indexed: 01/01/2023]
Abstract
TP53, one of the most important oncosuppressors, is frequently mutated in cancer. Several p53 mutant proteins escape proteolytic degradation and are highly expressed in an aberrant conformation often acquiring pro-oncogenic activities that promote tumor progression and resistance to therapy. Therefore, it has been vastly proposed that reactivation of wild-type (wt) function(s) from mutant p53 (mutp53) may have therapeutic significance. We have previously reported that Zn(II) restores a folded conformation from mutp53 misfolding, rescuing wild-type (wt) p53/DNA-binding and transcription activities. However, whether Zn(II) affects mutp53 stability has never been investigated. Here we show that a novel Zn(II) compound induced mutp53 (R175H) protein degradation through autophagy, the proteolytic machinery specifically devoted to clearing misfolded proteins. Accordingly, pharmacological or genetic inhibition of autophagy prevented Zn(II)-mediated mutp53H175 degradation as well as the ability of the Zn(II) compound to restore wtp53 DNA-binding and transcription activity from this mutant. By contrast, inhibition of the proteasome failed to do so, suggesting that autophagy is the main route for p53H175 degradation. Mechanistically, Zn(II) restored the wtp53 ability to induce the expression of the p53 target gene DRAM (damage-regulated autophagy modulator), a key regulator of autophagy, leading to autophagic induction. Accordingly, inhibition of wtp53 transactivation by pifithrin-α (PFT-α) impaired both autophagy and mutp53H175 degradation induced by curcumin-based zinc compound (Zn(II)-curc). Viewed together, our results uncover a novel mechanism employed by Zn(II)-curc to reactivate mutp53H175, which involves, at least in part, induction of mutp53 degradation via wtp53-mediated autophagy.
Collapse
|
21
|
Singh CK, Pitschmann A, Ahmad N. Resveratrol-zinc combination for prostate cancer management. Cell Cycle 2014; 13:1867-74. [PMID: 24866157 DOI: 10.4161/cc.29334] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Zinc, an essential trace element, plays a critical role in cell signaling, and defect(s) in zinc homeostasis may contribute to adverse physiological and pathological conditions, including cancer. Zinc is present in healthy prostate at a very high concentration, where it is required for important prostatic functions. However, zinc levels are significantly diminished in cancerous tissue, and intracellular zinc level is inversely correlated with prostate cancer progression. During neoplastic transformation, zinc-accumulating, citrate-producing normal prostate cells are metabolically transformed to citrate oxidizing cells that lose the ability to accumulate zinc. Interestingly, zinc has been shown to function as chemopreventive agent against prostate cancer, albeit at high doses, which may lead to many adverse effects. Therefore, novel means to enhance bioaccumulation of sufficient zinc in prostate cells via increasing zinc transport could be useful against prostate cancer. On the basis of available evidence, we present a possibility that the grape antioxidant resveratrol, when given with zinc, may lead to retuning the zinc homeostasis in prostate, thereby abolishing or reversing malignancy. If experimentally verified in in vivo model(s) of prostate cancer, such as transgenic mouse models, this may lead to novel means toward management of prostate cancer and other conditions with compromised zinc homeostasis.
Collapse
Affiliation(s)
- Chandra K Singh
- Department of Dermatology; University of Wisconsin; Madison, WI USA
| | - Anna Pitschmann
- Department of Dermatology; University of Wisconsin; Madison, WI USA
| | - Nihal Ahmad
- Department of Dermatology; University of Wisconsin; Madison, WI USA
| |
Collapse
|