1
|
Fuochi V, Furnari S, Floresta G, Patamia V, Zagni C, Drago F, Rescifina A, Furneri PM. Antiviral efficacy of heparan sulfate and enoxaparin sodium against SARS-CoV-2. Arch Pharm (Weinheim) 2024:e2400545. [PMID: 39520338 DOI: 10.1002/ardp.202400545] [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: 07/05/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
As the world transitions from the acute phase of the COVID-19 pandemic caused by SARS-CoV-2, the scientific community continues to explore various therapeutic avenues to control its spread and mitigate its ongoing effects. Among the promising candidates are heparan sulfate (HS) and enoxaparin (EX), which have emerged as potential virus inhibitors. HS, a type of glycosaminoglycan, plays a prominent role in the attachment of the virus to host cells. At the same time, EX, a low-molecular-weight heparin, is being investigated for its ability to disrupt the interaction between the spike protein of SARS-CoV-2 and the ACE2 receptor in human cells. Understanding the mechanisms through which these substances operate could lay the foundation for new strategies in the ongoing management of COVID-19. This study aimed to examine the details of SARS-CoV-2's entry mechanisms and the role of HS in this process. Furthermore, it examines EX's mechanism of action, highlighting how it potentially inhibits SARS-CoV-2. The interactions between HS and the virus, alongside in-vitro and in-silico inhibition studies with HS and EX, are critically analyzed to assess their antiviral efficacy. Additionally, the antiviral activity of sulfated polysaccharides and the potential therapeutic applications of these findings are discussed.
Collapse
Affiliation(s)
- Virginia Fuochi
- Department of Biomedical and Biotechnological Sciences (Biometec), University of Catania, Catania, Italy
| | - Salvatore Furnari
- Department of Biomedical and Biotechnological Sciences (Biometec), University of Catania, Catania, Italy
| | - Giuseppe Floresta
- Department of Drug and Health Sciences (DSFS), University of Catania, Catania, Italy
| | - Vincenzo Patamia
- Department of Drug and Health Sciences (DSFS), University of Catania, Catania, Italy
| | - Chiara Zagni
- Department of Drug and Health Sciences (DSFS), University of Catania, Catania, Italy
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences (Biometec), University of Catania, Catania, Italy
| | - Antonio Rescifina
- Department of Drug and Health Sciences (DSFS), University of Catania, Catania, Italy
| | - Pio Maria Furneri
- Department of Biomedical and Biotechnological Sciences (Biometec), University of Catania, Catania, Italy
| |
Collapse
|
2
|
Salerno L, Sorrenti V, Pittalà V, Consoli V, Modica MN, Romeo G, Marrazzo A, Giuliano M, Zajdel P, Vanella L, Intagliata S. Discovery of SI 1/20 and SI 1/22 as Mutual Prodrugs of 5-Fluorouracil and Imidazole-Based Heme Oxygenase 1 Inhibitor with Improved Cytotoxicity in DU145 Prostate Cancer Cells. ChemMedChem 2023; 18:e202300047. [PMID: 36756924 DOI: 10.1002/cmdc.202300047] [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: 01/28/2023] [Accepted: 02/08/2023] [Indexed: 02/10/2023]
Abstract
In this work, we extend the concept of 5-fluorouracil/heme oxygenase 1 (5-FU/HO-1) inhibitor hybrid as an effective strategy for enhancing 5-FU-based anticancer therapies. For this purpose, we designed and synthesized new mutual prodrugs, named SI 1/20 and SI 1/22, in which the two active parent drugs (i. e., 5-FU and an imidazole-based HO-1 inhibitor) were connected through an easily cleavable succinic linker. Experimental hydrolysis rate, and in silico ADMET predictions were indicative of good drug-likeness and pharmacokinetic properties. Novel hybrids significantly reduced the viability of prostate DU145 cancer cells compared to the parent compounds 5-FU and HO-1 inhibitor administered alone or in combination. Interestingly, both compounds showed statistically significant lower toxicity, than 5-FU at the same dose, against non-tumorigenic human benign prostatic hyperplasia (BPH-1) cell line. Moreover, the newly synthesized mutual prodrugs inhibited the HO-1 activity both in a cell-free model and in vitro, as well as downregulated the HO-1 expression and increased the reactive oxygen species (ROS) levels.
Collapse
Affiliation(s)
- Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
- Department of Molecular Medicine, College of Medicine and Medical Sciences, Princess Al Jawhara Centre for Molecular Medicine, Arabian Gulf University, Manama, 329, Bahrain
| | - Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Maria N Modica
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Michela Giuliano
- Department of Biological, Chemical and Pharmaceutical Sciences, and Technologies (STEBICEF), University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Paweł Zajdel
- Department of Organic Chemistry, Jagiellonian University Medical College, 9 Medyczna Str., 30-688, Kraków, Poland
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
3
|
Xu J, Zhu K, Wang Y, Chen J. The dual role and mutual dependence of heme/HO-1/Bach1 axis in the carcinogenic and anti-carcinogenic intersection. J Cancer Res Clin Oncol 2023; 149:483-501. [PMID: 36310300 DOI: 10.1007/s00432-022-04447-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION In physiological concentrations, heme is nontoxic to the cell and is essential for cell survival and proliferation. Increasing intracellular heme concentrations beyond normal levels, however, will lead to carcinogenesis and facilitate the survival of tumor cells. Simultaneously, heme in an abnormally high quantity is also a potent inducer of tumor cell death, contributing to its ability to generate oxidative stress on the cells by boosting oxidative phosphorylation and suppressing tumors through ferroptosis. During tumorigenesis and progression, therefore, heme works as a double-edged sword. Heme oxygenase 1 (HO-1) is the rate-limiting enzyme in heme catabolism, which converts heme into physiologically active catabolites of carbon monoxide (CO), biliverdin, and ferrous iron (Fe2+). HO-1 maintains redox equilibrium in healthy cells and functions as a carcinogenesis inhibitor. It is widely recognized that HO-1 is involved in the adaptive response to cellular stress and the anti-inflammation effect. Notably, its expression level in cancer cells corresponds with tumor growth, aggressiveness, metastasis, and angiogenesis. Besides, heme-binding transcription factor BTB and CNC homology 1 (Bach1) play a critical regulatory role in heme homeostasis, oxidative stress and senescence, cell cycle, angiogenesis, immune cell differentiation, and autoimmune disorders. Moreover, it was found that Bach1 influences cancer cells' metabolism and metastatic capacity. Bach1 controls heme level by adjusting HO-1 expression, establishing a negative feedback loop. MATERIALS AND METHODS Herein, the authors review recent studies on heme, HO-1, and Bach1 in cancer. Specifically, they cover the following areas: (1) the carcinogenic and anticarcinogenic aspects of heme; (2) the carcinogenic and anticarcinogenic aspects of HO-1; (3) the carcinogenic and anticarcinogenic aspects of Bach1; (4) the interactions of the heme/HO-1/Bach1 axis involved in tumor progression. CONCLUSION This review summarized the literature about the dual role of the heme/HO-1/Bach1 axis and their mutual dependence in the carcinogenesis and anti-carcinogenesis intersection.
Collapse
Affiliation(s)
- Jinjing Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China
| | | | - Yali Wang
- Jiangsu Huai'an Maternity and Children Hospital, Huai'an, 223001, China
| | - Jing Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, 225009, China. .,College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
4
|
Consoli V, Sorrenti V, Pittalà V, Greish K, D’Amico AG, Romeo G, Intagliata S, Salerno L, Vanella L. Heme Oxygenase Modulation Drives Ferroptosis in TNBC Cells. Int J Mol Sci 2022; 23:ijms23105709. [PMID: 35628518 PMCID: PMC9143660 DOI: 10.3390/ijms23105709] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
The term ferroptosis refers to a peculiar type of programmed cell death (PCD) mainly characterized by extensive iron-dependent lipid peroxidation. Recently, ferroptosis has been suggested as a potential new strategy for the treatment of several cancers, including breast cancer (BC). In particular, among the BC subtypes, triple negative breast cancer (TNBC) is considered the most aggressive, and conventional drugs fail to provide long-term efficacy. In this context, our study's purpose was to investigate the mechanism of ferroptosis in breast cancer cell lines and reveal the significance of heme oxygenase (HO) modulation in the process, providing new biochemical approaches. HO's effect on BC was evaluated by MTT tests, gene silencing, Western blot analysis, and measurement of reactive oxygen species (ROS), glutathione (GSH) and lipid hydroperoxide (LOOH) levels. In order to assess HO's implication, different approaches were exploited, using two distinct HO-1 inducers (hemin and curcumin), a well-known HO inhibitor (SnMP) and a selective HO-2 inhibitor. The data obtained showed HO's contribution to the onset of ferroptosis; in particular, HO-1 induction seemed to accelerate the process. Moreover, our results suggest a potential role of HO-2 in erastin-induced ferroptosis. In view of the above, HO modulation in ferroptosis can offer a novel approach for breast cancer treatment.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
| | - Valeria Sorrenti
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
- CERNUT-Research Centre on Nutraceuticals and Health Products, University of Catania, 95125 Catania, Italy
- Correspondence:
| | - Valeria Pittalà
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
- CERNUT-Research Centre on Nutraceuticals and Health Products, University of Catania, 95125 Catania, Italy
| | - Khaled Greish
- Princess Al-Jawhara Centre for Molecular Medicine, Department of Molecular Medicine, School of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain;
| | - Agata Grazia D’Amico
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
| | - Giuseppe Romeo
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
- CERNUT-Research Centre on Nutraceuticals and Health Products, University of Catania, 95125 Catania, Italy
| | - Sebastiano Intagliata
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
- CERNUT-Research Centre on Nutraceuticals and Health Products, University of Catania, 95125 Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
- CERNUT-Research Centre on Nutraceuticals and Health Products, University of Catania, 95125 Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Science, University of Catania, 95125 Catania, Italy; (V.C.); (V.P.); (A.G.D.); (G.R.); (S.I.); (L.S.); (L.V.)
- CERNUT-Research Centre on Nutraceuticals and Health Products, University of Catania, 95125 Catania, Italy
| |
Collapse
|
5
|
Novel Tyrosine Kinase Inhibitors to Target Chronic Myeloid Leukemia. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103220. [PMID: 35630697 PMCID: PMC9143943 DOI: 10.3390/molecules27103220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 11/19/2022]
Abstract
This paper reports on a novel series of tyrosine kinase inhibitors (TKIs) potentially useful for the treatment of chronic myeloid leukemia (CML). The newly designed and synthesized compounds are structurally related to nilotinib (NIL), a second-generation oral TKI, and to a series of imatinib (IM)-based TKIs, previously reported by our research group, these latter characterized by a hybrid structure between TKIs and heme oxygenase-1 (HO-1) inhibitors. The enzyme HO-1 was selected as an additional target since it is overexpressed in many cases of drug resistance, including CML. The new derivatives 1a–j correctly tackle the chimeric protein BCR-ABL. Therefore, the inhibition of TK was comparable to or higher than NIL and IM for many novel compounds, while most of the new analogs showed only moderate potency against HO-1. Molecular docking studies revealed insights into the binding mode with BCR-ABL and HO-1, providing a structural explanation for the differential activity. Cytotoxicity on K562 CML cells, both NIL-sensitive and -resistant, was evaluated. Notably, some new compounds strongly reduced the viability of K562 sensitive cells.
Collapse
|
6
|
Feng YD, Ye W, Tian W, Meng JR, Zhang M, Sun Y, Zhang HN, Wang SJ, Wu KH, Liu CX, Liu SY, Cao W, Li XQ. Old targets, new strategy: Apigenin-7-O-β-d-(-6″-p-coumaroyl)-glucopyranoside prevents endothelial ferroptosis and alleviates intestinal ischemia-reperfusion injury through HO-1 and MAO-B inhibition. Free Radic Biol Med 2022; 184:74-88. [PMID: 35398494 DOI: 10.1016/j.freeradbiomed.2022.03.033] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022]
Abstract
With the increasing morbidity and mortality, intestinal ischemia/reperfusion injury (IIRI) has attracted more and more attention, but there is no efficient therapeutics at present. Apigenin-7-O-β-D-(-6″-p-coumaroyl)-glucopyranoside (APG) is a new flavonoid glycoside isolated from Clematis tangutica that has strong antioxidant abilities in previous studies. However, the pharmacodynamic function and mechanism of APG on IIRI remain unknown. This study aimed to investigate the effects of APG on IIRI both in vivo and in vitro and identify the potential molecular mechanism. We found that APG could significantly improve intestinal edema and increase Chiu's score. MST analysis suggested that APG could specifically bind to heme oxygenase 1 (HO-1) and monoamine oxidase b (MAO-B). Simultaneously, APG could attenuate ROS generation and Fe2+ accumulation, maintain mitochondria function thus inhibit ferroptosis with a dose-dependent manner. Moreover, we used siRNA silencing technology to confirm that knocking down both HO-1 and MAO-B had a positive effect on intestine. In addition, we found the HO-1 and MAO-B inhibitors also could reduce endothelial cell loss and protect vascular endothelial after reperfusion. We demonstrate that APG plays a protective role on decreasing activation of HO-1 and MAO-B, attenuating IIRI-induced ROS generation and Fe2+ accumulation, maintaining mitochondria function thus inhibiting ferroptosis.
Collapse
Affiliation(s)
- Ying-Da Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Ye
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wen Tian
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Jing-Ru Meng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Meng Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Yang Sun
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Hui-Nan Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shou-Jia Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Ke-Han Wu
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chen-Xu Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Shao-Yuan Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China
| | - Wei Cao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Xi'an, Shaanxi, 710032, China; Shaanxi Key Laboratory of "Qin Medicine" Research and Development, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
7
|
Floresta G, Fallica AN, Patamia V, Sorrenti V, Greish K, Rescifina A, Pittalà V. From Far West to East: Joining the Molecular Architecture of Imidazole-like Ligands in HO-1 Complexes. Pharmaceuticals (Basel) 2021; 14:ph14121289. [PMID: 34959690 PMCID: PMC8704944 DOI: 10.3390/ph14121289] [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: 11/16/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
HO-1 overexpression has been reported in several cases/types of human malignancies. Unfortunately, poor clinical outcomes are reported in most of these cases, and the inhibition of HO-1 is considered a valuable and proven anticancer approach. To identify novel hit compounds suitable as HO-1 inhibitors, we report here a fragment-based approach where ligand joining experiments were used. The two most important parts of the classical structure of the HO-1 inhibitors were used as a starting point, and 1000 novel compounds were generated and then virtually evaluated by structure and ligand-based approaches. The joining experiments led us to a novel series of indole-based compounds. A synthetic pathway for eight selected molecules was designed, and the compounds were synthesized. The biological activity revealed that some molecules reach the micromolar activity, whereas molecule 4d inhibits the HO-1 with an IC50 of 1.03 μM. This study suggested that our joining approach was successful, and a novel hit compound was generated. These results are ongoing for further development.
Collapse
Affiliation(s)
- Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
- Department of Analytics, Environmental & Forensics, King’s College London, London SE1 9NH, UK
| | - Antonino Nicolò Fallica
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
| | - Vincenzo Patamia
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
| | - Khaled Greish
- Department of Molecular Medicine and Nanomedicine Unit, Princess Al-Jawhara Center for Molecular Medicine, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 329, Bahrain;
| | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
- Correspondence: (A.R.); (V.P.)
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy; (G.F.); (A.N.F.); (V.P.); (V.S.)
- Correspondence: (A.R.); (V.P.)
| |
Collapse
|
8
|
Floresta G, Fallica AN, Salerno L, Sorrenti V, Pittalà V, Rescifina A. Growing the molecular architecture of imidazole-like ligands in HO-1 complexes. Bioorg Chem 2021; 117:105428. [PMID: 34710668 DOI: 10.1016/j.bioorg.2021.105428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/27/2021] [Accepted: 10/11/2021] [Indexed: 12/11/2022]
Abstract
Up-regulation of HO-1 had been frequently reported in different cases and types of human malignancies. Since poor clinical outcomes are reported in these cases, this enzyme's inhibition is considered a valuable and proven anticancer approach. To identify novel HO-1 inhibitors suitable for drug development, we report a structure-guided fragment-based approach to identify new lead compounds. Different parts of the selected molecules were analyzed, and the different series of novel compounds were virtually evaluated. The growing experiments of the classical HO-1 inhibitors structure led us to different hit-compounds. A synthetic pathway for six selected molecules was designed, and the compounds were synthesized. The biological activity revealed that molecules 10 and 12 inhibit the HO-1 activity with an IC50 of 1.01 and 0.90 μM, respectively. This study suggested that our growing approach was successful, and these results are ongoing for further development.
Collapse
Affiliation(s)
- Giuseppe Floresta
- Department of Analytical, Environmental and Forensic Sciences, King's College London, London, UK.
| | - Antonino N Fallica
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy.
| | - Antonio Rescifina
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| |
Collapse
|
9
|
Fallica A, Sorrenti V, D’Amico AG, Salerno L, Romeo G, Intagliata S, Consoli V, Floresta G, Rescifina A, D’Agata V, Vanella L, Pittalà V. Discovery of Novel Acetamide-Based Heme Oxygenase-1 Inhibitors with Potent In Vitro Antiproliferative Activity. J Med Chem 2021; 64:13373-13393. [PMID: 34472337 PMCID: PMC8474116 DOI: 10.1021/acs.jmedchem.1c00633] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Indexed: 12/25/2022]
Abstract
Heme oxygenase-1 (HO-1) promotes heme catabolism exercising cytoprotective roles in normal and cancer cells. Herein, we report the design, synthesis, molecular modeling, and biological evaluation of novel HO-1 inhibitors. Specifically, an amide linker in the central spacer and an imidazole were fixed, and the hydrophobic moiety required by the pharmacophore was largely modified. In many tumors, overexpression of HO-1 correlates with poor prognosis and chemoresistance, suggesting the inhibition of HO-1 as a possible antitumor strategy. Accordingly, compounds 7i and 7l-p emerged for their potency against HO-1 and were investigated for their anticancer activity against prostate (DU145), lung (A549), and glioblastoma (U87MG, A172) cancer cells. The selected compounds showed the best activity toward U87MG cells. Compound 7l was further investigated for its in-cell enzymatic HO-1 activity, expression levels, and effects on cell invasion and vascular endothelial growth factor (VEGF) extracellular release. The obtained data suggest that 7l can reduce cell invasivity acting through modulation of HO-1 expression.
Collapse
Affiliation(s)
- Antonino
N. Fallica
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | - Valeria Sorrenti
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | - Agata G. D’Amico
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | - Loredana Salerno
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | - Giuseppe Romeo
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | | | - Valeria Consoli
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | - Giuseppe Floresta
- Department
of Analytics, Environmental & Forensics, King’s College London, Stamford Street, London SE1 9NH, U.K.
| | - Antonio Rescifina
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | - Velia D’Agata
- Sections
of Human Anatomy and Histology, Department of Biomedical and Biotechnological
Sciences, University of Catania, 95123 Catania, Italy
| | - Luca Vanella
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| | - Valeria Pittalà
- Department
of Drug and Health Sciences, University
of Catania, 95125 Catania, Italy
| |
Collapse
|
10
|
Ciaffaglione V, Modica MN, Pittalà V, Romeo G, Salerno L, Intagliata S. Mutual Prodrugs of 5-Fluorouracil: From a Classic Chemotherapeutic Agent to Novel Potential Anticancer Drugs. ChemMedChem 2021; 16:3496-3512. [PMID: 34415107 PMCID: PMC9290623 DOI: 10.1002/cmdc.202100473] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/18/2021] [Indexed: 12/18/2022]
Abstract
The development of potent antitumor agents with a low toxicological profile against healthy cells is still one of the greatest challenges facing medicinal chemistry. In this context, the “mutual prodrug” approach has emerged as a potential tool to overcome undesirable physicochemical features and mitigate the side effects of approved drugs. Among broad‐spectrum chemotherapeutics available for clinical use today, 5‐fluorouracil (5‐FU) is one of the most representative, also included in the World Health Organization model list of essential medicines. Unfortunately, severe side effects and drug resistance phenomena are still the primary limits and drawbacks in its clinical use. This review describes the progress made over the last ten years in developing 5‐FU‐based mutual prodrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues.
Collapse
Affiliation(s)
- Valeria Ciaffaglione
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Maria N Modica
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
11
|
Sharma P, LaRosa C, Antwi J, Govindarajan R, Werbovetz KA. Imidazoles as Potential Anticancer Agents: An Update on Recent Studies. Molecules 2021; 26:molecules26144213. [PMID: 34299488 PMCID: PMC8307698 DOI: 10.3390/molecules26144213] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022] Open
Abstract
Nitrogen-containing heterocyclic rings are common structural components of marketed drugs. Among these heterocycles, imidazole/fused imidazole rings are present in a wide range of bioactive compounds. The unique properties of such structures, including high polarity and the ability to participate in hydrogen bonding and coordination chemistry, allow them to interact with a wide range of biomolecules, and imidazole-/fused imidazole-containing compounds are reported to have a broad spectrum of biological activities. This review summarizes recent reports of imidazole/fused imidazole derivatives as anticancer agents appearing in the peer-reviewed literature from 2018 through 2020. Such molecules have been shown to modulate various targets, including microtubules, tyrosine and serine-threonine kinases, histone deacetylases, p53-Murine Double Minute 2 (MDM2) protein, poly (ADP-ribose) polymerase (PARP), G-quadraplexes, and other targets. Imidazole-containing compounds that display anticancer activity by unknown/undefined mechanisms are also described, as well as key features of structure-activity relationships. This review is intended to provide an overview of recent advances in imidazole-based anticancer drug discovery and development, as well as inspire the design and synthesis of new anticancer molecules.
Collapse
Affiliation(s)
- Pankaj Sharma
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (P.S.); (C.L.)
| | - Chris LaRosa
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (P.S.); (C.L.)
| | - Janet Antwi
- Division of Mathematics, Computer & Natural Sciences Division, Ohio Dominican University, Columbus, OH 43219, USA;
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA;
| | - Karl A. Werbovetz
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (P.S.); (C.L.)
- Correspondence:
| |
Collapse
|
12
|
Salerno L, Vanella L, Sorrenti V, Consoli V, Ciaffaglione V, Fallica AN, Canale V, Zajdel P, Pignatello R, Intagliata S. Novel mutual prodrug of 5-fluorouracil and heme oxygenase-1 inhibitor (5-FU/HO-1 hybrid): design and preliminary in vitro evaluation. J Enzyme Inhib Med Chem 2021; 36:1378-1386. [PMID: 34167427 PMCID: PMC8231349 DOI: 10.1080/14756366.2021.1928111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In this work, the first mutual prodrug of 5-fluorouracil and heme oxygenase1 inhibitor (5-FU/HO-1 hybrid) has been designed, synthesised, and evaluated for its in vitro chemical and enzymatic hydrolysis stability. Predicted in silico physicochemical properties of the newly synthesised hybrid (3) demonstrated a drug-like profile with suitable Absorption, Distribution, Metabolism, and Excretion (ADME) properties and low toxic liabilities. Preliminary cytotoxicity evaluation towards human prostate (DU145) and lung (A549) cancer cell lines demonstrated that 3 exerted a similar effect on cell viability to that produced by the reference drug 5-FU. Among the two tested cancer cell lines, the A549 cells were more susceptible for 3. Of note, hybrid 3 also had a significantly lower cytotoxic effect on healthy human lung epithelial cells (BEAS-2B) than 5-FU. Altogether our results served as an initial proof-of-concept to develop 5-FU/HO-1 mutual prodrugs as potential novel anticancer agents.
Collapse
Affiliation(s)
- Loredana Salerno
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | | - Antonino N Fallica
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Vittorio Canale
- Department of Organic Chemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Paweł Zajdel
- Department of Organic Chemistry, Jagiellonian University Medical College, Kraków, Poland
| | - Rosario Pignatello
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | |
Collapse
|
13
|
Combination of Heme Oxygenase-1 Inhibition and Sigma Receptor Modulation for Anticancer Activity. Molecules 2021; 26:molecules26133860. [PMID: 34202711 PMCID: PMC8270315 DOI: 10.3390/molecules26133860] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is a multifactorial disease that may be tackled by targeting different signaling pathways. Heme oxygenase-1 (HO-1) and sigma receptors (σRs) are both overexpressed in different human cancers, including prostate and brain, contributing to the cancer spreading. In the present study, we investigated whether HO-1 inhibitors and σR ligands, as well a combination of the two, may influence DU145 human prostate and U87MG human glioblastoma cancer cells proliferation. In addition, we synthesized, characterized, and tested a small series of novel hybrid compounds (HO-1/σRs) 1–4 containing the chemical features needed for HO-1 inhibition and σR modulation. Herein, we report for the first time that targeting simultaneously HO-1 and σR proteins may be a good strategy to achieve increased antiproliferative activity against DU145 and U87MG cells, with respect to the mono administration of the parent compounds. The obtained outcomes provide an initial proof of concept useful to further optimize the structure of HO-1/σRs hybrids to develop novel potential anticancer agents.
Collapse
|
14
|
Sorrenti V, D’Amico AG, Barbagallo I, Consoli V, Grosso S, Vanella L. Tin Mesoporphyrin Selectively Reduces Non-Small-Cell Lung Cancer Cell Line A549 Proliferation by Interfering with Heme Oxygenase and Glutathione Systems. Biomolecules 2021; 11:biom11060917. [PMID: 34205698 PMCID: PMC8235249 DOI: 10.3390/biom11060917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/24/2022] Open
Abstract
In order to maintain redox homeostasis, non-small-cell lung cancer (NSCLC) increases the activation of many antioxidant systems, including the heme-oxygenase (HO) system. The overexpression of HO-1 has been often associated with chemoresistance and tumor aggressiveness. Our results clearly showed an overexpression of the HO-1 protein in A549 NSCLC cell lines compared to that in non-cancerous cells. Thus, we hypothesized that "off-label" use of tin mesoporphyrin, a well-known HO activity inhibitor clinically used for neonatal hyperbilirubinemia, has potential use as an anti-cancer agent. The pharmacological inhibition of HO activity caused a reduction in cell proliferation and migration of A549. SnMP treatment caused an increase in oxidative stress, as demonstrated by the upregulation of reactive oxygen species (ROS) and the depletion of glutathione (GSH) content. To support these data, Western blot analysis was performed to analyze glucose-6-phosphate dehydrogenase (G6PD), TP53-induced glycolysis and the apoptosis regulator (TIGAR), and the glutamate cysteine ligase catalytic (GCLC) subunit, as they represent the main regulators of the pentose phosphate pathway (PPP) and glutathione synthesis, respectively. NCI-H292, a subtype of the NSCLC cell line, did not respond to SnMP treatment, possibly due to low basal levels of HO-1, suggesting a cellular-dependent antitumorigenic effect. Altogether, our results suggest HO activity inhibition may represent a potential target for selective chemotherapy in lung cancer subtypes.
Collapse
|
15
|
Nitti M, Ivaldo C, Traverso N, Furfaro AL. Clinical Significance of Heme Oxygenase 1 in Tumor Progression. Antioxidants (Basel) 2021; 10:antiox10050789. [PMID: 34067625 PMCID: PMC8155918 DOI: 10.3390/antiox10050789] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase 1 (HO-1) plays a key role in cell adaptation to stressors through the antioxidant, antiapoptotic, and anti-inflammatory properties of its metabolic products. For these reasons, in cancer cells, HO-1 can favor aggressiveness and resistance to therapies, leading to poor prognosis/outcome. Genetic polymorphisms of HO-1 promoter have been associated with an increased risk of cancer progression and a high degree of therapy failure. Moreover, evidence from cancer biopsies highlights the possible correlation between HO-1 expression, pathological features, and clinical outcome. Indeed, high levels of HO-1 in tumor specimens often correlate with reduced survival rates. Furthermore, HO-1 modulation has been proposed in order to improve the efficacy of antitumor therapies. However, contrasting evidence on the role of HO-1 in tumor biology has been reported. This review focuses on the role of HO-1 as a promising biomarker of cancer progression; understanding the correlation between HO-1 and clinical data might guide the therapeutic choice and improve the outcome of patients in terms of prognosis and life quality.
Collapse
|
16
|
Novel Heme Oxygenase-1 (HO-1) Inducers Based on Dimethyl Fumarate Structure. Int J Mol Sci 2020; 21:ijms21249541. [PMID: 33333908 PMCID: PMC7765375 DOI: 10.3390/ijms21249541] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/31/2022] Open
Abstract
Novel heme oxygenase-1 (HO-1) inducers based on dimethyl fumarate (DMF) structure are reported in this paper. These compounds are obtained by modification of the DMF backbone. Particularly, maintaining the α, β-unsaturated dicarbonyl function as the central chain crucial for HO-1 induction, different substituted or unsubstituted phenyl rings are introduced by means of an ester or amide linkage. Symmetric and asymmetric derivatives are synthesized. All compounds are tested on a human hepatic stellate cell line LX-2 to assay their capacity for modifying HO-1 expression. Compounds 1b, 1l and 1m stand out for their potency as HO-1 inducers, being 2–3 fold more active than DMF, and for their ability to reverse reactive oxygen species (ROS) production mediated using palmitic acid (PA). These properties, coupled with a low toxicity toward LX-2 cell lines, make these compounds potentially useful for treatment of diseases in which HO-1 overexpression may counteract inflammation, such as hepatic fibrosis. Docking studies show a correlation between predicted binding free energy and experimental HO-1 expression data. These preliminary results may support the development of new approaches in the management of liver fibrosis.
Collapse
|
17
|
Identification of a potent heme oxygenase-2 (HO-2) inhibitor by targeting the secondary hydrophobic pocket of the HO-2 western region. Bioorg Chem 2020; 104:104310. [DOI: 10.1016/j.bioorg.2020.104310] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/15/2020] [Accepted: 09/20/2020] [Indexed: 12/12/2022]
|
18
|
Jang HY, Hong OY, Chung EY, Park KH, Kim JS. Roles of JNK/Nrf2 Pathway on Hemin-Induced Heme Oxygenase-1 Activation in MCF-7 Human Breast Cancer Cells. ACTA ACUST UNITED AC 2020; 56:medicina56060268. [PMID: 32485912 PMCID: PMC7353851 DOI: 10.3390/medicina56060268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 11/16/2022]
Abstract
Heme oxygenase-1 (HO-1) is highly induced in various human disease states, including cancer, indicating that HO-1 is an emerging target of cancer therapy. In this study, we investigated that the mechanisms of hemin-induced HO-1 expression and its signaling pathways in human breast cancer cell. We used MCF-7 cells, a human breast cancer cell line. Hemin increased HO-1 expression in MCF-7 cells in a dose- and time-dependent manner. Hemin enhanced HO-1 expression through the activation of c-Jun N-terminal kinases (JNK) signaling pathway. Hemin also induced activation of Nrf2, a major transcription factor of HO-1 expression. These responses in MCF-7 cells were completely blocked by pretreatment with brazilin, a HO-1 regulator. These results indicated that brazilin inhibits hemin-induced HO-1 expressions through inactivation of JNK/Nrf2 in MCF-7 cells. Thus, our findings suggest that HO-1 is an important anticancer-target of brazilin in human breast cancer.
Collapse
Affiliation(s)
- Hye-Yeon Jang
- Department of Biochemistry and Institute of Cardiovascular Research, Chonbuk National University Medical School, Jeonju 54896, Korea; (H.-Y.J.); (O.-Y.H.)
| | - On-Yu Hong
- Department of Biochemistry and Institute of Cardiovascular Research, Chonbuk National University Medical School, Jeonju 54896, Korea; (H.-Y.J.); (O.-Y.H.)
| | - Eun-Yong Chung
- Department of Anesthesiology and Pain Medicine, Bucheon St. Mary’s Hospital, Catholic University of Korea, Bucheon 14647, Korea;
| | - Kwang-Hyun Park
- Department of Emergency Medical Rescue, Nambu University, Gwangju 62271, Korea
- Department of Emergency Medicine, Graduate School of Chonnam National University, Gwangju 61469, Korea
- Correspondence: (K.-H.P.); (J.-S.K.); Tel.: +82-62-970-0220 (K.-H.P.); +82-63-270-3085 (J.-S.K.)
| | - Jong-Suk Kim
- Department of Biochemistry and Institute of Cardiovascular Research, Chonbuk National University Medical School, Jeonju 54896, Korea; (H.-Y.J.); (O.-Y.H.)
- Correspondence: (K.-H.P.); (J.-S.K.); Tel.: +82-62-970-0220 (K.-H.P.); +82-63-270-3085 (J.-S.K.)
| |
Collapse
|
19
|
Heme Oxygenase-1 in Central Nervous System Malignancies. J Clin Med 2020; 9:jcm9051562. [PMID: 32455831 PMCID: PMC7290325 DOI: 10.3390/jcm9051562] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/23/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Central nervous system tumors are the most common pediatric solid tumors and account for 20–25% of all childhood malignancies. Several lines of evidence suggest that brain tumors show altered redox homeostasis that triggers the activation of various survival pathways, leading to disease progression and chemoresistance. Among these pathways, heme oxygenase-1 (HO-1) plays an important role. HO-1 catalyzes the enzymatic degradation of heme with the simultaneous release of carbon monoxide (CO), ferrous iron (Fe2+), and biliverdin. The biological effects of HO-1 in tumor cells have been shown to be cell-specific since, in some tumors, its upregulation promotes cell cycle arrest and cellular death, whereas, in other neoplasms, it is associated with tumor survival and progression. This review focuses on the role of HO-1 in central nervous system malignancies and the possibility of exploiting such a target to improve the outcome of well-established therapeutic regimens. Finally, several studies show that HO-1 overexpression is involved in the development and resistance of brain tumors to chemotherapy and radiotherapy, suggesting the use of HO-1 as an innovative therapeutic target to overcome drug resistance. The following keywords were used to search the literature related to this topic: nuclear factor erythroid 2 p45-related factor 2, heme oxygenase, neuroblastoma, medulloblastoma, meningioma, astrocytoma, oligodendroglioma, glioblastoma multiforme, and gliomas.
Collapse
|