1
|
Lv B, Xing S, Wang Z, Zhang A, Wang Q, Bian Y, Pei Y, Sun H, Chen Y. NRF2 inhibitors: Recent progress, future design and therapeutic potential. Eur J Med Chem 2024; 279:116822. [PMID: 39241669 DOI: 10.1016/j.ejmech.2024.116822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a crucial transcription factor involved in oxidative stress response, which controls the expression of various cytoprotective genes. Recent research has indicated that constitutively activated NRF2 can enhance patients' resistance to chemotherapy drugs, resulting in unfavorable prognosis. Therefore, the development of NRF2 inhibitors has emerged as a promising approach for overcoming drug resistance in cancer treatment. However, there are limited reports and reviews focusing on NRF2 inhibitors. This review aims to provide a comprehensive analysis of the structure and regulation of the NRF2 signaling pathway, followed by a comprehensive review of reported NRF2 inhibitors. Moreover, the current design strategies and future prospects of NRF2 inhibitors will be discussed, aiming to establish a foundation for the development of more effective NRF2 inhibitors.
Collapse
Affiliation(s)
- Bingbing Lv
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Zhiqiang Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Ao Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qinjie Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yaoyao Bian
- Jiangsu Provincial Engineering Center of TCM External Medication Researching and Industrializing, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
2
|
Kudo Y, Nakamura K, Tsuzuki H, Hirota K, Kawai M, Takaya D, Fukuzawa K, Honma T, Yoshino Y, Nakamura M, Shiota M, Fujimoto N, Ikari A, Endo S. Docosahexaenoic acid enhances the treatment efficacy for castration-resistant prostate cancer by inhibiting autophagy through Atg4B inhibition. Arch Biochem Biophys 2024; 760:110135. [PMID: 39181384 DOI: 10.1016/j.abb.2024.110135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Autophagy induction in cancer is involved in cancer progression and the acquisition of resistance to anticancer agents. Therefore, autophagy is considered a potential therapeutic target in cancer therapy. In this study, we found that long-chain fatty acids (LCFAs) have inhibitory effects on Atg4B, which is essential for autophagosome formation, through screening based on the pharmacophore of 21f, a recently developed Atg4B inhibitor. Among these fatty acids, docosahexaenoic acid (DHA), a polyunsaturated fatty acid, exhibited the most potent Atg4B inhibitory activity. DHA inhibited autophagy induced by androgen receptor signaling inhibitors (ARSI) in LNCaP and 22Rv1 prostate cancer cells and significantly increased apoptotic cell death. Furthermore, we investigated the effect of DHA on resistance to ARSI by establishing darolutamide-resistant prostate cancer 22Rv1 (22Rv1/Dar) cells, which had developed resistance to darolutamide, a novel ARSI. At baseline, 22Rv1/Dar cells showed a higher autophagy level than parental 22Rv1 cells. DHA significantly suppressed Dar-induced autophagy and sensitized 22Rv1/Dar cells by inducing apoptotic cell death through mitochondrial dysfunction. These results suggest that DHA supplementation may improve prostate cancer therapy with ARSI.
Collapse
Affiliation(s)
- Yudai Kudo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Kana Nakamura
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Honoka Tsuzuki
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Kotaro Hirota
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Mina Kawai
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Daisuke Takaya
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Kaori Fukuzawa
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Teruki Honma
- Center for Biosystems Dynamics Research, RIKEN, Kanagawa, 230-0045, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Mitsuhiro Nakamura
- Laboratories of Drug Informatics, Department of Pharmacy Practice and Science, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Fukuoka, 807-8555, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Satoshi Endo
- The United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, 501-1194, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
3
|
Jebanesan DZP, Illangeswaran RSS, Rajamani BM, Vidhyadharan RT, Das S, Bijukumar NK, Balakrishnan B, Mathews V, Velayudhan SR, Balasubramanian P. Inhibition of NRF2 signaling overcomes acquired resistance to arsenic trioxide in FLT3-mutated Acute Myeloid Leukemia. Ann Hematol 2024; 103:1919-1929. [PMID: 38630133 DOI: 10.1007/s00277-024-05742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/01/2024] [Indexed: 05/14/2024]
Abstract
De novo acute myeloid leukemia (AML) patients with FMS-like tyrosine kinase 3 internal tandem duplications (FLT3-ITD) have worse treatment outcomes. Arsenic trioxide (ATO) used in the treatment of acute promyelocytic leukemia (APL) has been reported to be effective in degrading the FLT3 protein in AML cell lines and sensitizing non-APL AML patient samples in-vitro. We have previously reported that primary cells from FLT3-ITD mutated AML patients were sensitive to ATO in-vitro compared to other non-M3 AML and molecular/pharmacological inhibition of NF-E2 related factor 2 (NRF2), a master regulator of antioxidant response improved the chemosensitivity to ATO and daunorubicin even in non FLT3-ITD mutated cell lines and primary samples. We examined the effects of molecular/pharmacological suppression of NRF2 on acquired ATO resistance in the FLT3-ITD mutant AML cell line (MV4-11-ATO-R). ATO-R cells showed increased NRF2 expression, nuclear localization, and upregulation of bonafide NRF2 targets. Molecular inhibition of NRF2 in this resistant cell line improved ATO sensitivity in vitro. Digoxin treatment lowered p-AKT expression, abrogating nuclear NRF2 localization and sensitizing cells to ATO. However, digoxin and ATO did not sensitize non-ITD AML cell line THP1 with high NRF2 expression. Digoxin decreased leukemic burden and prolonged survival in MV4-11 ATO-R xenograft mice. We establish that altering NRF2 expression may reverse acquired ATO resistance in FLT3-ITD AML.
Collapse
Affiliation(s)
- Daniel Zechariah Paul Jebanesan
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
- Manipal Academy of Higher Education, Manipal, India
| | | | - Bharathi M Rajamani
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | | | - Saswati Das
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | - Nayanthara K Bijukumar
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | - Balaji Balakrishnan
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Vikram Mathews
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | - Shaji R Velayudhan
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
- Adjunct Scientist, Centre for Stem Cell Research, A Unit of InStem Bengaluru, CMC Campus, Vellore, India
| | - Poonkuzhali Balasubramanian
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India.
| |
Collapse
|
4
|
Ohya S, Kito H, Kajikuri J, Yamaguchi Y, Matsui M. Transcriptional Up-Regulation of FBXW7 by K Ca1.1 K + Channel Inhibition through the Nrf2 Signaling Pathway in Human Prostate Cancer LNCaP Cell Spheroid Model. Int J Mol Sci 2024; 25:6019. [PMID: 38892210 PMCID: PMC11172474 DOI: 10.3390/ijms25116019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 05/25/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The tumor suppressor gene F-box and WD repeat domain-containing (FBXW) 7 reduces cancer stemness properties by promoting the protein degradation of pluripotent stem cell markers. We recently demonstrated the transcriptional repression of FBXW7 by the three-dimensional (3D) spheroid formation of several cancer cells. In the present study, we found that the transcriptional activity of FBXW7 was promoted by the inhibition of the Ca2+-activated K+ channel, KCa1.1, in a 3D spheroid model of human prostate cancer LNCaP cells through the Akt-Nrf2 signaling pathway. The transcriptional activity of FBXW7 was reduced by the siRNA-mediated inhibition of the CCAAT-enhancer-binding protein C/EBP δ (CEBPD) after the transfection of miR223 mimics in the LNCaP spheroid model, suggesting the transcriptional regulation of FBXW7 through the Akt-Nrf2-CEBPD-miR223 transcriptional axis in the LNCaP spheroid model. Furthermore, the KCa1.1 inhibition-induced activation of FBXW7 reduced (1) KCa1.1 activity and protein levels in the plasma membrane and (2) the protein level of the cancer stem cell (CSC) markers, c-Myc, which is a molecule degraded by FBXW7, in the LNCaP spheroid model, indicating that KCa1.1 inhibition-induced FBXW7 activation suppressed CSC conversion in KCa1.1-positive cancer cells.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan; (H.K.); (J.K.); (Y.Y.); (M.M.)
| | | | | | | | | |
Collapse
|
5
|
Liu Q, Zhou H, Wang Y, Gui J, Yang D, Sun J, Ge D, Wu S, Liu Q, Zhu L, Mi Y. H3K27 acetylation activated-PDLIM7 promotes castration-resistant prostate cancer progression by inducing O-Glycosylation of YAP1 protein. Transl Oncol 2024; 40:101830. [PMID: 38056280 PMCID: PMC10714362 DOI: 10.1016/j.tranon.2023.101830] [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: 07/19/2023] [Revised: 10/17/2023] [Accepted: 11/11/2023] [Indexed: 12/08/2023] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a fatal disease that evolves from prostate cancer due to drug resistance after long-term androgen deprivation therapy. In this study, we aimed to find novel molecular targets for treating CRPC. Through peptidome, we screened out polypeptides dysregulated in the serum of CRPC patients. According to RT-qPCR analysis and cell viability detection, we chose PDZ and LIM Domain 7 (PDLIM7) as the research object. As demonstrated by loss-of-function assays, silencing of PDLIM7 could suppress CRPC cell proliferation, migration, and angiogenesis. Moreover, PDLIM7 knockdown enhanced the sensitivity of CRPC cells to docetaxel treatment. Subsequently, we found that CBP/p300 increases the H3K27ac level in the PDLIM7 promoter to activate PDLIM7. Mechanism experiments such as IP and western blot revealed that PDLIM7 interacted with YAP1 to induce O-Glycosylation of YAP1 and thus stabilize YAP1 protein. Rescue assays demonstrated that PDLIM7 promoted the malignant processes of CRPC cells through YAP1. Finally, an animal study validated that PDLIM7 aggravated tumor growth. In conclusion, our findings highlighted the oncogenic role of PDLIM7 upregulated by CBP/p300-induced H3K27ac enhancement in CRPC by stabilizing YAP1.
Collapse
Affiliation(s)
- Qing Liu
- Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China; Department of Health and Wellness, Huadong Sanatorium, Wuxi, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Yanjuan Wang
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Dongjie Yang
- Department of Pathology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China
| | - Jian Sun
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Dongsheng Ge
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Sheng Wu
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Qin Liu
- Department of Health and Wellness, Huadong Sanatorium, Wuxi, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi, Jiangsu 214122, China; Wuxi Medical College, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
6
|
Himura R, Kawano S, Nagata Y, Kawai M, Ota A, Kudo Y, Yoshino Y, Fujimoto N, Miyamoto H, Endo S, Ikari A. Inhibition of aldo-keto reductase 1C3 overcomes gemcitabine/cisplatin resistance in bladder cancer. Chem Biol Interact 2024; 388:110840. [PMID: 38122923 DOI: 10.1016/j.cbi.2023.110840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/27/2023] [Accepted: 12/17/2023] [Indexed: 12/23/2023]
Abstract
Systemic chemotherapy with gemcitabine and cisplatin (GC) has been used for the treatment of bladder cancer in which androgen receptor (AR) signaling is suggested to play a critical role. However, its efficacy is often limited, and the prognosis of patients who develop resistance is extremely poor. Aldo-keto reductase 1C3 (AKR1C3), which is responsible for the production of a potent androgen, 5α-dihydrotestosterone (DHT), by the reduction of 5α-androstane-3α,17β-dione (5α-Adione), has been attracting attention as a therapeutic target for prostate cancer that shows androgen-dependent growth. By contrast, the role of AKR1C3 in bladder cancer remains unclear. In this study, we examined the effect of an AKR1C3 inhibitor on androgen-dependent proliferation and GC sensitivity in bladder cancer cells. 5α-Adione treatment induced the expression of AR and its downstream factor ETS-domain transcription factor (ELK1) in both T24 cells and newly established GC-resistant T24GC cells, while it did not alter AKR1C3 expression. AKR1C3 inhibitor 2j significantly suppressed 5α-Adione-induced AR and ELK1 upregulation, as did an AR antagonist apalutamide. Moreover, the combination of GC and 2j in T24GC significantly induced apoptotic cell death, suggesting that 2j could enhance GC sensitivity. Immunohistochemical staining in surgical specimens further revealed that strong expression of AKR1C3 was associated with significantly higher risks of tumor progression and cancer-specific mortality in patients with muscle-invasive bladder cancer. These results suggest that AKR1C3 inhibitors as adjunctive agents enhance the efficacy of GC therapy for bladder cancer.
Collapse
Affiliation(s)
- Rin Himura
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Shinya Kawano
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Yujiro Nagata
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Mina Kawai
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Atsumi Ota
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Yudai Kudo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Hiroshi Miyamoto
- Departments of Pathology & Laboratory Medicine and Urology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Satoshi Endo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu Pharmaceutical University, Gifu, 501-1193, Japan.
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Japan
| |
Collapse
|
7
|
Hou Y, Wei D, Zhang Z, Lei T, Li S, Bao J, Guo H, Tan L, Xie X, Zhuang Y, Lu Z, Zhao Y. Downregulation of nutrition sensor GCN2 in macrophages contributes to poor wound healing in diabetes. Cell Rep 2024; 43:113658. [PMID: 38175755 DOI: 10.1016/j.celrep.2023.113658] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/27/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
Poor skin wound healing, which is common in patients with diabetes, is related to imbalanced macrophage polarization. Here, we find that nutrition sensor GCN2 (general control nonderepressible 2) and its downstream are significantly upregulated in human skin wound tissue and mouse skin wound macrophages, but skin wound-related GCN2 expression and activity are significantly downregulated by diabetes and hyperglycemia. Using wound healing models of GCN2-deleted mice, bone marrow chimeric mice, and monocyte-transferred mice, we show that GCN2 deletion in macrophages significantly delays skin wound healing compared with wild-type mice by altering M1 and M2a/M2c polarization. Mechanistically, GCN2 inhibits M1 macrophages via OXPHOS-ROS-NF-κB pathway and promotes tissue-repairing M2a/M2c macrophages through eukaryotic translation initiation factor 2 (eIF2α)-hypoxia-inducible factor 1α (HIF1α)-glycolysis pathway. Importantly, local supplementation of GCN2 activator halofuginone efficiently restores wound healing in diabetic mice with re-balancing M1 and M2a/2c polarization. Thus, the decreased macrophage GCN2 expression and activity contribute to poor wound healing in diabetes and targeting GCN2 improves wound healing in diabetes.
Collapse
Affiliation(s)
- Yangxiao Hou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Dong Wei
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China; CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Shenzhen, China
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China; CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Shenzhen, China
| | - Tong Lei
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regeneration, Beijing, China
| | - Sihong Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jiaming Bao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Han Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Liang Tan
- Kidney Transplantation Department, Second Xiangya Hospital of Central South University, Changsha, China
| | - Xubiao Xie
- Kidney Transplantation Department, Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuan Zhuang
- Department of Blood Transfusion, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhongbing Lu
- University of Chinese Academy of Sciences, Beijing, China.
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China; CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Shenzhen, China; Beijing Institute for Stem Cell and Regeneration, Beijing, China.
| |
Collapse
|
8
|
Ben-Eltriki M, Shankar G, Tomlinson Guns ES, Deb S. Pharmacokinetics and pharmacodynamics of Rh2 and aPPD ginsenosides in prostate cancer: a drug interaction perspective. Cancer Chemother Pharmacol 2023; 92:419-437. [PMID: 37709921 DOI: 10.1007/s00280-023-04583-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Ginsenoside Rh2 and its aglycon (aPPD) are one of the major metabolites from Panax ginseng. Preclinical studies suggest that Rh2 and aPPD have antitumor effects in prostate cancer (PCa). Our aims in this review are (1) to describe the pharmacokinetic (PK) properties of Rh2 and aPPD ginsenosides; 2) to provide an overview of the preclinical findings on the use of Rh2 and aPPD in the treatment of PCa; and (3) to highlight the mechanisms of its PK and pharmacodynamic (PD) drug interactions. Increasing evidence points to the potential efficacy of Rh2 or aPPD for PCa treatment. Based on the laboratory studies, Rh2 or aPPD combinations revealed an additive or synergistic interaction or enhanced sensitivity of anticancer drugs toward PCa. This review reveals that enhanced anticancer activities were demonstrated in preclinical studies through interactions of Rh2 and/or aPPD with the proteins related to PK (e.g., cytochrome P450 enzymes, transporters) or PD of the other anticancer drugs or PCa signaling pathways. In conclusion, combining Rh2 or aPPD with anti-prostate cancer drugs leads to PK or PD interactions which could facilitate either therapeutically beneficial or toxic effects.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC, Canada.
- Community Pharmacist, Vancouver Area, BC, Canada.
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Gehana Shankar
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA.
| |
Collapse
|
9
|
Tossetta G, Fantone S, Marzioni D, Mazzucchelli R. Role of Natural and Synthetic Compounds in Modulating NRF2/KEAP1 Signaling Pathway in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15113037. [PMID: 37296999 DOI: 10.3390/cancers15113037] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Prostate cancer is the second most common cancer in men worldwide. Prostate cancer can be treated by surgery or active surveillance when early diagnosed but, when diagnosed at an advanced or metastatic stage, radiation therapy or androgen-deprivation therapy is needed to reduce cancer progression. However, both of these therapies can cause prostate cancer resistance to treatment. Several studies demonstrated that oxidative stress is involved in cancer occurrence, development, progression and treatment resistance. The nuclear factor erythroid 2-related factor 2 (NRF2)/KEAP1 (Kelch-Like ECH-Associated Protein 1) pathway plays an important role in protecting cells against oxidative damage. Reactive oxygen species (ROS) levels and NRF2 activation can determine cell fate. In particular, toxic levels of ROS lead physiological cell death and cell tumor suppression, while lower ROS levels are associated with carcinogenesis and cancer progression. On the contrary, a high level of NRF2 promotes cell survival related to cancer progression activating an adaptive antioxidant response. In this review, we analyzed the current literature regarding the role of natural and synthetic compounds in modulating NRF2/KEAP1 signaling pathway in prostate cancer.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Roberta Mazzucchelli
- Department of Biomedical Sciences and Public Health, Section of Pathological Anatomy, Università Politecnica delle Marche, 60126 Ancona, Italy
| |
Collapse
|
10
|
Ben-Eltriki M, Gayle EJ, Walker N, Deb S. Pharmacological Significance of Heme Oxygenase 1 in Prostate Cancer. Curr Issues Mol Biol 2023; 45:4301-4316. [PMID: 37232742 DOI: 10.3390/cimb45050273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Heme oxygenase 1 (HO-1) is a detoxifying antioxidant microsomal enzyme that regulates inflammation, apoptosis, cell proliferation, and angiogenesis in prostate cancer (PCa). This makes HO-1 a promising target for therapeutic prevention and treatment due to its anti-inflammatory properties and ability to control redox homeostasis. Clinical evidence highlights the possible correlation between HO-1 expression and PCa growth, aggressiveness, metastasized tumors, resistance to therapy, and poor clinical outcomes. Interestingly, studies have reported anticancer benefits mediated by both HO-1 induction and inhibition in PCa models. Contrasting evidence exists on the role of HO-1 in PCa progression and possible treatment targets. Herein, we provide an overview of available evidence on the clinical significance of HO-1 signaling in PCa. It appears that the beneficial effects of HO-1 induction or inhibition are dependent on whether it is a normal versus malignant cell as well as the intensity (major vs. minor) of the increase in HO-1 enzymatic activity. The current literature evidence indicates that HO-1 has dual effects in PCa. The amount of cellular iron and reactive oxygen species (ROS) can determine the role of HO-1 in PCa. A major increase in ROS enforces HO-1 to a protective role. HO-1 overexpression may provide cryoprotection to normal cells against oxidative stress via suppressing the expression of proinflammatory genes, and thus offer therapeutic prevention. In contrast, a moderate increase in ROS can lead to the perpetrator role of HO-1, which is associated with PCa progression and metastasis. HO-1 inhibition by xenobiotics in DNA-damaged cells tilts the balance to promote apoptosis and inhibit PCa proliferation and metastasis. Overall, the totality of the evidence revealed that HO-1 may play a dual role in the therapeutic prevention and treatment of PCa.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- Department of Pharmacology and Therapeutics, Clinical Pharmacology Lab, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Erysa J Gayle
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Noah Walker
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL 33169, USA
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA
| |
Collapse
|
11
|
Ota A, Kawai M, Kudo Y, Segawa J, Hoshi M, Kawano S, Yoshino Y, Ichihara K, Shiota M, Fujimoto N, Matsunaga T, Endo S, Ikari A. Artepillin C overcomes apalutamide resistance through blocking androgen signaling in prostate cancer cells. Arch Biochem Biophys 2023; 735:109519. [PMID: 36642262 DOI: 10.1016/j.abb.2023.109519] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/07/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Prostate cancer has a relatively good prognosis, but most cases develop resistance to hormone therapy, leading to castration-resistant prostate cancer (CRPC). Androgen receptor (AR) antagonists and a cytochrome P450 17A1 inhibitor have been used to treat CRPC, but cancer cells readily develop resistance to these drugs. In this study, to improve the therapy of CRPC, we searched for natural compounds which block androgen signaling. Among cinnamic acid derivatives contained in Brazilian green propolis, artepillin C (ArtC) suppressed expressions of androgen-induced prostate-specific antigen and transmembrane protease serine 2 in a dose-dependent manner. Reporter assays revealed that ArtC displayed AR antagonist activity, albeit weaker than an AR antagonist flutamide. In general, aberrant activation of the androgen signaling is involved in the resistance of prostate cancer cells to hormone therapy. Recently, apalutamide, a novel AR antagonist, has been in clinical use, but its drug-resistant cases have been already reported. To search for compounds which overcome the resistance to apalutamide, we established apalutamide-resistant prostate cancer 22Rv1 cells (22Rv1/APA). The 22Rv1/APA cells showed higher AR expression and androgen sensitivity than parental 22Rv1 cells. ArtC inhibited androgen-induced proliferation of 22Rv1/APA cells by suppressing the enhanced androgen signaling through blocking the nuclear translocation of AR. In addition, ArtC potently sensitized the resistant cells to apalutamide by inducing apoptotic cell death due to mitochondrial dysfunction. These results suggest that the intake of Brazilian green propolis containing ArtC improves prostate cancer therapy.
Collapse
Affiliation(s)
- Atsumi Ota
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| | - Mina Kawai
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| | - Yudai Kudo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| | - Jin Segawa
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| | - Manami Hoshi
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| | - Shinya Kawano
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| | - Kenji Ichihara
- Nagaragawa Research Center, API Co., Ltd., Gifu, 502-0071, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu, 807-8555, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu, 502-8585, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan.
| | - Akira Ikari
- Laboratory of Biochemistry, Department of Biopharmaceutical Sciences, Gifu Pharmaceutical University, Gifu, 501-1196, Gifu, Japan
| |
Collapse
|
12
|
Kudo Y, Endo S, Tanio M, Saka T, Himura R, Abe N, Takeda M, Yamaguchi E, Yoshino Y, Arai Y, Kashiwagi H, Oyama M, Itoh A, Shiota M, Fujimoto N, Ikari A. Antiandrogenic Effects of a Polyphenol in Carex kobomugi through Inhibition of Androgen Synthetic Pathway and Downregulation of Androgen Receptor in Prostate Cancer Cell Lines. Int J Mol Sci 2022; 23:ijms232214356. [PMID: 36430833 PMCID: PMC9696374 DOI: 10.3390/ijms232214356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Prostate cancer (PC) represents the most common cancer disease in men. Since high levels of androgens increase the risk of PC, androgen deprivation therapy is the primary treatment; however this leads to castration-resistant PC (CRPC) with a poor prognosis. The progression to CRPC involves ectopic androgen production in the adrenal glands and abnormal activation of androgen signaling due to mutations and/or amplification of the androgen receptor (AR) as well as activation of androgen-independent proliferative pathways. Recent studies have shown that adrenal-derived 11-oxygenated androgens (11-ketotestosterone and 11-ketodihydrotestosterone) with potencies equivalent to those of traditional androgens (testosterone and dihydrotestosterone) are biomarkers of CRPC. Additionally, dehydrogenase/reductase SDR family member 11 (DHRS11) has been reported to be a 17β-hydroxysteroid dehydrogenase that catalyzes the production of the 11-oxygenated and traditional androgens. This study was conducted to evaluate the pathophysiological roles of DHRS11 in PC using three LNCaP, C4-2 and 22Rv1 cell lines. DHRS11 silencing and inhibition resulted in suppression of the androgen-induced expression of AR downstream genes and decreases in the expression of nuclear AR and the proliferation marker Ki67, suggesting that DHRS11 is involved in androgen-dependent PC cell proliferation. We found that 5,7-dihydroxy-8-methyl-2-[2-(4-hydroxyphenyl)ethenyl]-4H-1-benzopyran-4-one (Kobochromone A, KC-A), an ingredient in the flowers of Carex kobomugi, is a novel potent DHRS11 inhibitor (IC50 = 0.35 μM). Additionally, KC-A itself decreased the AR expression in PC cells. Therefore, KC-A suppresses the androgen signaling in PC cells through both DHRS11 inhibition and AR downregulation. Furthermore, KC-A enhanced the anticancer activity of abiraterone, a CRPC drug, suggesting that it may be a potential candidate for the development of drugs for the prevention and treatment of CRPC.
Collapse
Affiliation(s)
- Yudai Kudo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
- Correspondence: ; Tel.: +81-58-230-8100; Fax: +81-58-230-8105
| | - Masatoshi Tanio
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tomofumi Saka
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Rin Himura
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Naohito Abe
- Laboratory of Pharmacognosy, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Mitsumi Takeda
- Laboratory of Pharmaceutical Synthetic Chemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Eiji Yamaguchi
- Laboratory of Pharmaceutical Synthetic Chemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Yuki Arai
- Universal Corporation Co., Ltd., Gifu 502-0931, Japan
| | - Hirohito Kashiwagi
- Laboratory of Pharmacognosy, Gifu Pharmaceutical University, Gifu 501-1196, Japan
- Universal Corporation Co., Ltd., Gifu 502-0931, Japan
| | - Masayoshi Oyama
- Laboratory of Pharmacognosy, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Akichika Itoh
- Laboratory of Pharmaceutical Synthetic Chemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| |
Collapse
|
13
|
Tian L, Peng Y, Yang K, Cao J, Du X, Liang Z, Shi J, Zhang J. The ERα-NRF2 signalling axis promotes bicalutamide resistance in prostate cancer. Cell Commun Signal 2022; 20:178. [PMID: 36376959 PMCID: PMC9661764 DOI: 10.1186/s12964-022-00979-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Bicalutamide is a nonsteroidal antiandrogen widely used as a first-line clinical treatment for advanced prostate cancer (PCa). Although patients initially show effective responses to bicalutamide treatment, resistance to bicalutamide frequently occurs and leads to the development of castration-resistant PCa (CRPC). This research investigated the roles of the oestrogen receptor α (ERα)-nuclear factor E2-related factor 2 (NRF2) signalling pathway in bicalutamide resistance in PCa cells. METHODS We performed bioinformatic analysis and immunohistochemical staining on normal and cancerous prostate tissue to evaluate ERα and NRF2 expression and their correlation. Gene expression and localization in PCa cell lines were further investigated using real-time reverse transcription PCR/Western blotting and immunofluorescence staining. We treated PCa cells with the ER inhibitor tamoxifen and performed luciferase reporter assays and chromatin immunoprecipitation (ChIP) assays to understand ERα-dependent NRF2 expression. Overexpression and knockdown of ERα and NRF2 were used to explore the potential role of the ERα-NRF2 signalling axis in bicalutamide resistance in PCa cells. RESULTS We found that the expression of ERα and NRF2 was positively correlated and was higher in human CRPC tissues than in primary PCa tissues. Treatment with oestrogen or bicalutamide increased the expression of ERα and NRF2 as well as NRF2 target genes in PCa cell lines. These effects were blocked by pretreatment with tamoxifen. ChIP assays demonstrated that ERα directly binds to the oestrogen response element (ERE) in the NRF2 promoter. This binding led to increased transcriptional activity of NRF2 in a luciferase reporter assay. Activation of the ERα-NRF2 signalling axis increased the expression of bicalutamide resistance-related genes. Inhibition of this signalling axis by knockdown of ERα or NRF2 downregulated the expression of bicalutamide resistance-related genes and inhibited the proliferation and migration of PCa cells. CONCLUSIONS We demonstrated the transcriptional interaction between ERα and NRF2 in CRPC tissues and cell lines by showing the direct binding of ERα to the ERE in the NRF2 promoter under oestrogen treatment. Activation of the ERα-NRF2 signalling axis contributes to bicalutamide resistance in PCa cells, suggesting that the ERα-NRF2 signalling axis is a potential therapeutic target for CRPC. Video Abstract.
Collapse
Affiliation(s)
- Lei Tian
- grid.216938.70000 0000 9878 7032Department of Biochemistry and Molecular Biology, College of Life Sciences, Bioactive Materials Key Lab of the Ministry of Education, Nankai University, Tianjin, 300071 China
| | - Yanfei Peng
- grid.410648.f0000 0001 1816 6218School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617 China
| | - Kuo Yang
- grid.412648.d0000 0004 1798 6160Department of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211 China
| | - Jiasong Cao
- grid.216938.70000 0000 9878 7032Department of Biochemistry and Molecular Biology, College of Life Sciences, Bioactive Materials Key Lab of the Ministry of Education, Nankai University, Tianjin, 300071 China
| | - Xiaoling Du
- grid.216938.70000 0000 9878 7032Department of Biochemistry and Molecular Biology, College of Life Sciences, Bioactive Materials Key Lab of the Ministry of Education, Nankai University, Tianjin, 300071 China
| | - Zhixian Liang
- grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, 999077 China
| | - Jiandang Shi
- grid.216938.70000 0000 9878 7032Department of Biochemistry and Molecular Biology, College of Life Sciences, Bioactive Materials Key Lab of the Ministry of Education, Nankai University, Tianjin, 300071 China
| | - Ju Zhang
- grid.216938.70000 0000 9878 7032Department of Biochemistry and Molecular Biology, College of Life Sciences, Bioactive Materials Key Lab of the Ministry of Education, Nankai University, Tianjin, 300071 China
| |
Collapse
|
14
|
Mi L, Zhang Y, Su A, Tang M, Xing Z, He T, Wu W, Li Z. Halofuginone for cancer treatment: A systematic review of efficacy and molecular mechanisms. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
15
|
Buttari B, Arese M, Oberley-Deegan RE, Saso L, Chatterjee A. NRF2: A crucial regulator for mitochondrial metabolic shift and prostate cancer progression. Front Physiol 2022; 13:989793. [PMID: 36213236 PMCID: PMC9540504 DOI: 10.3389/fphys.2022.989793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/31/2022] [Indexed: 12/05/2022] Open
Abstract
Metabolic alterations are a common survival mechanism for prostate cancer progression and therapy resistance. Oxidative stress in the cellular and tumor microenvironment dictates metabolic switching in the cancer cells to adopt, prosper and escape therapeutic stress. Therefore, regulation of oxidative stress in tumor cells and in the tumor-microenvironment may enhance the action of conventional anticancer therapies. NRF2 is the master regulator for oxidative stress management. However, the overall oxidative stress varies with PCa clinical stage, metabolic state and therapy used for the cancer. In agreement, the blanket use of NRF2 inducers or inhibitors along with anticancer therapies cause adverse effects in some preclinical cancer models. In this review, we have summarized the levels of oxidative stress, metabolic preferences and NRF2 activity in the different stages of prostate cancer. We also propose condition specific ways to use NRF2 inducers or inhibitors along with conventional prostate cancer therapies. The significance of this review is not only to provide a detailed understanding of the mechanism of action of NRF2 to regulate oxidative stress-mediated metabolic switching by prostate cancer cells to escape the radiation, chemo, or hormonal therapies, and to grow aggressively, but also to provide a potential therapeutic method to control aggressive prostate cancer growth by stage specific proper use of NRF2 regulators.
Collapse
Affiliation(s)
- Brigitta Buttari
- Department of Cardiovascular and Endocrine-metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Marzia Arese
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Rome, Italy
| | - Rebecca E. Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Luciano Saso
- Department of Physiology and Pharmacology ‘‘Vittorio Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
- *Correspondence: Arpita Chatterjee,
| |
Collapse
|
16
|
mRNA-Modified FUS/NRF2 Signalling Inhibits Ferroptosis and Promotes Prostate Cancer Growth. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8509626. [PMID: 36035281 PMCID: PMC9410928 DOI: 10.1155/2022/8509626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/08/2022] [Accepted: 07/08/2022] [Indexed: 11/18/2022]
Abstract
Objective. Regarding the imperfect mechanism of occurrence and development of prostate adenocarcinoma (PRAD), this study investigated mRNA-modified FUS/NRF2 signalling to inhibit ferroptosis and promote prostate adenocarcinoma growth. Methods. Bioinformatics analysis was used to obtain the expression of FUS and its mRNA modification in PRAD. The expression of FUS in prostate cells (CRPC) and the level of m6A methylation modification, ferroptosis (P53 and GPX4), apoptosis (Caspase3), ferroptosis (P53 and GPX4), and apoptosis (Caspase3) in CRPC after ferroptosis inducer Erastin, ferroptosis inhibitor, and FUS knockdown were detected. Autophagy (LC3B), oxidative stress (GSH and ROS), and expression of NRF2/HO-1 pathway are indicators. Results. FUS was highly expressed in PRAD and phenomenally reduced the survival rate of patients. After knocking down FUS, the level of m6A methylation was significantly reduced, and the expressions of ferroptosis markers P53 and GPX4 were phenomenally reduced, while the levels of apoptosis and autophagy markers Caspase3 and LC3B remained unchanged. Upregulated and NRF2/HO-1 pathway indicators were upregulated. It shows that m6A methylation modification is reduced when FUS is the low expression, inhibits the expression of P53 and GPX4, downregulates GSH, upregulates ROS, activates the NRF2/HO-1 pathway, and promotes ferroptosis to inhibit the occurrence of RPAD. Conclusions. The increase of m6A methylation modification can increase the expression of FUS, thereby promoting the expression of P53 and GPX4, upregulating GSH, downregulating ROS, inhibiting the NRF2/HO-1 pathway, inhibiting ferroptosis, and promoting the growth of PRAD.
Collapse
|
17
|
Eryilmaz IE, Egeli U, Cecener G. An in vitro redox adaptation model for metastatic prostate cancer: Establishing, characterizing, and Cabazitaxel response evaluating. Clin Exp Pharmacol Physiol 2022; 49:1094-1104. [PMID: 35751096 DOI: 10.1111/1440-1681.13694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/09/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022]
Abstract
Little is known about the redox-adapted cancer cells for understanding their pharmacologically targetable features and chemotherapeutic responses. Thus, we presented the first in vitro redox adaptation model for metastatic prostate cancer (mPC), LNCaP-HPR, with enhanced oxidative stress resistance accompanying poor Cabazitaxel response. After establishing, the cells were characterized by comparing the viability, death, oxidative stress, total GSH levels, and the mRNA and protein levels of the redox-sensitive transcription factors responsible for the adaptation, Nrf-2, NF-κB, and HIF-1α. Then, the apoptotic effect of Cabazitaxel was evaluated in LNCaP mPC, LNCaP-HPR, and C4-2 metastatic castration-resistant (mCRPC) cells. In response to H2 O2 , viability, oxidative stress, and the total GSH levels of LNCaP-HPR cells have confirmed the oxidative stress resistance. Nrf-2, NF-κB, and HIF-1α were upregulated in LNCaP-HPR cells, not in LNCaP, confirming that resistant cells were much less affected by exogenous oxidative stress. Unlike LNCaP, LNCaP-HPR cells were less sensitive to Cabazitaxel, as closer to the response of C4-2 mCRPC cells, indicating that redox adaptation decreased Cabazitaxel response. This is the first evaluated association between redox adaptation and poor Cabazitaxel response, suggesting that in vitro Cabazitaxel efficiency is affected by PC cells' endogenous oxidative stress tolerance. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Isil Ezgi Eryilmaz
- Bursa Uludag University, Faculty of Medicine, Medical Biology Department, Bursa, Turkey
| | - Unal Egeli
- Bursa Uludag University, Faculty of Medicine, Medical Biology Department, Bursa, Turkey
| | - Gulsah Cecener
- Bursa Uludag University, Faculty of Medicine, Medical Biology Department, Bursa, Turkey
| |
Collapse
|
18
|
Lv T, Huang J, Wu M, Wang H, Zeng Q, Wang X. Halofuginone enhances the anti-tumor effect of ALA-PDT by suppressing NRF2 signaling in cSCC. Photodiagnosis Photodyn Ther 2022; 37:102572. [PMID: 34628069 DOI: 10.1016/j.pdpdt.2021.102572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND 5-aminolevulinic acid-mediated PDT (ALA-PDT) has been used in a variety of skin diseases including cSCC (cutaneous squamous cell carcinoma). Halofuginone (HL) is a less-toxic febrifugine derivative and has inhibitory effects on a variety of cancer cells. For now, there are no published study focusing on the combination use of ALA-PDT with HL to improve clinical efficacy of cSCC. OBJECTIVE In this study, we will examine the effectiveness of combined treatment of ALA-PDT and HL in cSCC as well as its underlying mechanism. METHODS The human epidermoid carcinoma cell line SCL-1 was treated with ALA-PDT or/ and HL, and cell viability, cell migration, ROS production, apoptosis were evaluated by CCK-8, colony formation, scratch assay, DCFH-DA probe, flow cytometry, respectively. The protein expression of NRF2 signaling was examined by western blot. RESULTS HL strengthened ALA-PDT's inhibition of SCL-1 cell viability, migration, as well as NRF2 related β-catenin, p-Erk1/2, p-Akt and p-S6K1 expression. Overexpression of NRF2 conferred resistance to co-treatment's effects on c-Myc, Cyclin D1, Bcl-2, as well as cell proliferation. HL also strengthened ALA-PDT's inhibition of tumor volume in cSCC mouse model and elevated ROS generation of ALA-PDT. CONCLUSION HL enhances the anti-tumor effect of ALA-PDT in vitro and in vivo. HL has the potential to enhance the anti-tumor effect of ALA-PDT in cSCC via inhibiting NRF2 signaling.
Collapse
Affiliation(s)
- Ting Lv
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jianhua Huang
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Minfeng Wu
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Hongwei Wang
- Department of Dermatology, Huadong Hospital, Fudan University, Shanghai 200040, China.
| | - Qingyu Zeng
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Xiuli Wang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|