1
|
Karmakar S, Dettmer SJ, Hooper CAJ, Hodges NJ, Hannon MJ. Switching on Supramolecular DNA Junction Binding Using a Human Enzyme. Angew Chem Int Ed Engl 2025; 64:e202503683. [PMID: 40088149 PMCID: PMC12087844 DOI: 10.1002/anie.202503683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Non-canonical DNA junction structures are important in human disease and in nucleic acid nanoscience and there is a growing interest in how to bind and modulate them. A key next step is to exert "on command" control over such binding. Herein we develop a new metallo-supramolecular triple-helicate cylinder agent that is inert to DNA junction binding until activated by human enzyme NAD(P)H:quinone oxidoreductase 1 (NQO1) and its cofactor nicotinamide adenine dinucleotide phosphate (NADPH). This inactive cylinder bears six flexible arms each with a quinone group at the termini. Reduction by the enzyme leads to all six arms being removed, transforming the inert cylinder into a new and active metallo-supramolecular agent that binds junctions. This gives the ability to "switch-on" DNA junction formation and binding in response to the presence of two external stimuli - a human enzyme overexpressed in many disease states, and NADPH - and absence of inhibitor, giving NAND logic control. Modelling indicates the binding activation originates not in steric unblocking but changes in conformational flexibility. The work provides the foundation for and a route map toward future designs of sophisticated, inert, and supramolecular structures which are transformed by enzymes into new, active, and supramolecular structures for a variety of potential applications.
Collapse
Affiliation(s)
- Subhendu Karmakar
- School of ChemistryUniversity of BirminghamEdgbastonBirminghamB15 2TTUK
| | - Samuel J. Dettmer
- School of ChemistryUniversity of BirminghamEdgbastonBirminghamB15 2TTUK
| | | | - Nikolas J. Hodges
- School of BiosciencesUniversity of BirminghamEdgbastonBirminghamB15 2TTUK
| | - Michael J. Hannon
- School of ChemistryUniversity of BirminghamEdgbastonBirminghamB15 2TTUK
| |
Collapse
|
2
|
Li X, Gong Q, Yu J, Liang J, Yao R, Zhou J, Chen Y, Lei Z, Yu Z, Zhang X, Qiu X. Identification of effective anti-osteosarcoma agents via screening of an in-house NQO1-targeted compound library. Bioorg Med Chem 2025; 122:118162. [PMID: 40117702 DOI: 10.1016/j.bmc.2025.118162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
Osteosarcoma is one of the most prevalent malignant bone tumors, and despite advances in treatment, significant improvements in survival rates for osteosarcoma patients remain elusive. There is an urgent need for developing novel molecules for the targeted treatment of osteosarcoma. NAD(P)H:quinone oxidoreductase 1 (NQO1) is highly overexpressed in osteosarcoma. Here, we evaluated a series of in-house NQO1-targeting compounds, including NQO1 substrates and β-lap prodrugs, through phenotypic screening using NQO1-positive methylnitronitrosoguanidine-induced human osteosarcoma cells (MNNG) and NQO1-negative normal human umbilical vein endothelial cells (HUVEC), aiming to identify novel candidate compounds for osteosarcoma therapy. As a result, compound 21, an NQO1 substrate, was identified as a potent anti-osteosarcoma agent that promotes apoptosis and induces cell cycle arrest in osteosarcoma cells in vitro, while significantly inhibiting tumor growth in vivo. These findings suggest that compound 21 holds promise as a candidate for osteosarcoma treatment. Moreover, NQO1-targeting substrates present a promising pathway for the discovery of novel anti-osteosarcoma agents.
Collapse
Affiliation(s)
- Xiang Li
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Qijie Gong
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jianglin Yu
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing 210008, China
| | - Jiaqi Liang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Rui Yao
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Jian Zhou
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yaxin Chen
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Zhijie Lei
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing 210008, China
| | - Zhan Yu
- Departemt of Emergency, The Affiliated Jiangning Hospital of NJMU, Nanjing Medical University (NJMU), Nanjing 211100, China
| | - Xiaojin Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Xusheng Qiu
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing 210008, China.
| |
Collapse
|
3
|
Morataya-Reyes M, Villacorta A, Gutiérrez-García J, Egea R, Martín-Pérez J, Barguilla I, Marcos R, Hernández A. The long-term in vitro co-exposure of polyethylene terephthalate (PET) nanoplastics and cigarette smoke condensate exacerbates the induction of carcinogenic traits. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138359. [PMID: 40267709 DOI: 10.1016/j.jhazmat.2025.138359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/03/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
This study examines the long-term impact of polyethylene terephthalate nanoplastics (PET-NPLs) and cigarette smoke condensate (CSC) on human lung BEAS-2B cells, focusing on key biological hallmarks of carcinogenesis. True-to-life PET-NPLs were generated from plastic water bottles and characterized to simulate environmental exposure conditions; and a comprehensive battery of assays was employed to assess genotoxicity, cellular transformation, and invasiveness. It was observed that, compared to passage control and individual exposures, co-exposure to PET-NPLs and CSC exacerbates oxidative stress, genotoxicity, and tumorigenic transformation, as evidenced by increased DNA damage, colony formation in soft agar, and enhanced cell migration and invasion. Transcriptomic analysis revealed a shift in cellular stress regulation including the upregulation of stress-response genes, including SLC7A11, NQO1, and HSPA1A, which are linked to oxidative stress adaptation and tumor survival. At the same time, key tumor-suppressor genes, such as LOX, and FN1, were significantly downregulated, promoting cellular transformation and invasiveness. These results provide compelling evidence that the combination of PET-NPLs and CSC enhances carcinogenic traits through oxidative stress, genomic instability, and disruption of tumor-suppressive pathways. This study underscores the importance of evaluating the synergistic effects of combined environmental exposures and their implications for human health.
Collapse
Affiliation(s)
- Michelle Morataya-Reyes
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Aliro Villacorta
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain; Facultad de Recursos Naturales Renovables, Universidad Arturo Prat, Iquique, Chile
| | - Javier Gutiérrez-García
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Raquel Egea
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Joan Martín-Pérez
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Irene Barguilla
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Ricard Marcos
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| | - Alba Hernández
- Group of Mutagenesis, Department of Genetics and Microbiology, Faculty of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
4
|
Yang Z, Han T, Yang R, Zhang Y, Qin Y, Hou J, Huo F, Feng Z, Ding Y, Yang J, Zhou G, Wang S, Xie X, Lin P, Chen ZN, Wu J. Dicoumarol sensitizes hepatocellular carcinoma cells to ferroptosis induced by imidazole ketone erastin. Front Immunol 2025; 16:1531874. [PMID: 40007539 PMCID: PMC11852437 DOI: 10.3389/fimmu.2025.1531874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Introduction Ferroptosis, an iron-dependent form of regulated cell death, is characterized by the lethal accumulation of lipid peroxides on cellular membranes. It not only inhibits tumor growth but also enhances immunotherapy responses and overcomes drug resistance in cancer therapy. The inhibition of the cystine-glutamate antiporter, system Xc-, induces ferroptosis. Imidazole ketone erastin (IKE), an inhibitor of the system Xc- functional subunit solute carrier family 7 member 11 (SLC7A11), is an effective and metabolically stable inducer of ferroptosis with potential in vivo applications. However, tumor cells exhibited differential sensitivity to IKE-induced ferroptosis. The intrinsic factors determining sensitivity to IKE-induced ferroptosis remain to be explored to improve its efficacy. Methods Bulk RNA-sequencing data from hepatocellular carcinoma (HCC) and normal liver tissues were collected from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. Differentially expressed genes were identified and intersected with the ferroptosis-related genes (FRGs) listed in the FerrDb database, yielding the identification of 13 distinct FRGs. Results A ferroptosis signature index model (Risk Score) was developed to predict HCC prognosis. And SLC7A11 and NAD(P)H quinone dehydrogenase 1 (NQO1) were identified as candidate FRGs indicating poor prognosis of HCC. Dicoumarol (DIC), an inhibitor of NQO1, was subsequently employed to assess its sensitizing effects on IKE in HCC treatment. In HCC cell lines and the subcutaneous xenograft model, the combined suppression of SLC7A11 and NQO1 significantly enhanced the inhibitory effect on tumor growth by inducing ferroptosis. Discussion In conclusion, our findings demonstrate that DIC sensitized HCC cells to IKE-induced ferroptosis in HCC. Moreover, the identification of potential drugs that enhance the susceptibility of HCC cells to ferroptosis could provide novel therapeutic strategies for the treatment of HCC.
Collapse
Affiliation(s)
- Ziwei Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Tixin Han
- Shaanxi Key Laboratory of Bio-electromagnetic Detection and Intelligent Sensing, Military Biomedical Engineering School, Fourth Military Medical University, Xi'an, China
| | - Ruibin Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yinuo Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yifei Qin
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China
| | - Jialu Hou
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Fei Huo
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhuan Feng
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yaxin Ding
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Jiali Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Gang Zhou
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Shijie Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Xiaohang Xie
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Peng Lin
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Jiao Wu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
5
|
Luo M, Shen N, Shang L, Fang Z, Xin Y, Ma Y, Du M, Yuan Y, Hu C, Tang Y, Huang J, Wei W, Lee MR, Hergenrother PJ, Wicha MS. Simultaneous Targeting of NQO1 and SOD1 Eradicates Breast Cancer Stem Cells via Mitochondrial Futile Redox Cycling. Cancer Res 2024; 84:4264-4282. [PMID: 39264695 PMCID: PMC11647209 DOI: 10.1158/0008-5472.can-24-0800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 07/24/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Triple-negative breast cancer (TNBC) contains the highest proportion of cancer stem-like cells (CSC), which display intrinsic resistance to currently available cancer therapies. This therapeutic resistance is partially mediated by an antioxidant defense coordinated by the transcription factor NRF2 and its downstream targets that include NAD(P)H quinone oxidoreductase 1 (NQO1). In this study, we identified the antioxidant enzymes NQO1 and superoxide dismutase 1 (SOD1) as therapeutic vulnerabilities of ALDH+ epithelial-like CSCs and CD24-/loCD44+/hi mesenchymal-like CSCs in TNBC. Effective targeting of these CSC states was achieved by using isobutyl-deoxynyboquinone (IB-DNQ), a potent and specific NQO1-bioactivatable futile redox cycling molecule, which generated large amounts of reactive oxygen species including superoxide and hydrogen peroxide. Furthermore, the CSC killing effect was specifically enhanced by genetic or pharmacologic inhibition of SOD1, a copper-containing superoxide dismutase highly expressed in TNBC. Mechanistically, a significant portion of NQO1 resides in the mitochondrial intermembrane space, catalyzing futile redox cycling from IB-DNQ to generate high levels of mitochondrial superoxide, and SOD1 inhibition markedly potentiated this effect, resulting in mitochondrial oxidative injury, cytochrome c release, and activation of the caspase-3-mediated apoptotic pathway. Treatment with IB-DNQ alone or together with SOD1 inhibition effectively suppressed tumor growth, metastasis, and tumor-initiating potential in xenograft models of TNBC expressing different levels of NQO1. This futile oxidant-generating strategy, which targets CSCs across the epithelial-mesenchymal continuum, could be a promising therapeutic approach for treating patients with TNBC. Significance: Combining NQO1-bioactivatable futile oxidant generators with SOD1 inhibition eliminates breast cancer stem cells, providing a therapeutic strategy that may have wide applicability, as NQO1 and SOD1 are overexpressed in several cancers.
Collapse
Affiliation(s)
- Ming Luo
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Na Shen
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Breast and Thyroid Surgery, Union Hospital, Tonji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Shang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Zeng Fang
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Ying Xin
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Yuxi Ma
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Cancer Center, Union Hospital, Tonji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Du
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuan Yuan
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chenchen Hu
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yun Tang
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jing Huang
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wei Wei
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Myung Ryul Lee
- Department of Chemistry, Cancer Center at Illinois, and Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois
| | - Paul J. Hergenrother
- Department of Chemistry, Cancer Center at Illinois, and Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois
| | - Max S. Wicha
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
6
|
Wattanathavorn W, Buranapraditkun S, Kitkumthorn N, Bhattarakosol P, Chaiwongkot A. Effect of NQO1 Downregulation on the Migration and Invasion of HPV16-Positive Cervical Cancer Cells. Asian Pac J Cancer Prev 2024; 25:4189-4200. [PMID: 39733409 PMCID: PMC12008345 DOI: 10.31557/apjcp.2024.25.12.4189] [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: 05/08/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024] Open
Abstract
OBJECTIVE This study aimed to identify upregulated genes in HPV16-positive cervical cancer cells and investigate the impact of downregulating NAD(P) H:quinone oxidoreductase 1 (NQO1) on the survival of these cells. METHODS Transcriptomic sequencing (RNA-seq) was utilized to pinpoint upregulated genes and associated cancer-related pathways in HPV16-positive cervical cancer cells, comparing them to HPV-negative cervical cancer cells. NQO1 gene knockdown was performed in HPV16-positive cervical cancer cell lines to assess its effect on cell survival, including parameters such as cell proliferation, migration, invasion, cell cycle progression, apoptosis, and the expression of key proteins in the PI3K/AKT pathway, p53, and RECK. RESULTS Genes with a fold change ≥4.0 in HPV16-positive cervical cancer cell lines were predominantly localized to the extracellular region and plasma membrane. These genes were involved in protein binding and cell adhesion, influencing cellular responses to stimuli and tissue development. KEGG pathway analysis identified the most significant pathways, including metabolic pathways, cancer pathways, MAPK signaling, and PI3K-AKT signaling. Knockdown of NQO1 significantly decreased cell proliferation, migration, and invasion, while increasing apoptosis in HPV16-positive cervical cancer cells (p ≤ 0.01). Additionally, proteins associated with the PI3K-AKT pathway were downregulated, while p53 and RECK protein levels were elevated. CONCLUSION Our findings suggest that NQO1 plays a crucial role in promoting migration and invasion in HPV16-positive cervical cancer cells, highlighting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Warattaya Wattanathavorn
- Interdisciplinary Program, Graduate school, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Applied Medical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Supranee Buranapraditkun
- Division of Allergy and Clinical Immunology, Department of Medicine, King Chulalongkorn Memorial Hospital, Faculty of Medicine, Chulalongkorn University, Thai Red Cross Society, Bangkok, 10330, Thailand.
- Center of Excellence in Thai Pediatric Gastroenterology, Hepatology and Immunology (TPGHAI), Faculty of Medicine, Chulalongkon University, Bangkok, 10330, Thailand.
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center- Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Nakarin Kitkumthorn
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand.
| | - Parvapan Bhattarakosol
- Center of Excellence in Applied Medical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| | - Arkom Chaiwongkot
- Center of Excellence in Applied Medical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
7
|
Wang X, Qu Y, Xu Q, Jiang Z, Wang H, Lin B, Cao Z, Pan Y, Li S, Hu Y, Yang H, He L, Chang H, Hang B, Wen H, Wu H, Mao JH. NQO1 Triggers Neutrophil Recruitment and NET Formation to Drive Lung Metastasis of Invasive Breast Cancer. Cancer Res 2024; 84:3538-3555. [PMID: 39073320 DOI: 10.1158/0008-5472.can-24-0291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Metastasis to the lungs is a leading cause of death for patients with breast cancer. Therefore, effective therapies are urgently needed to prevent and treat lung metastasis. In this study, we uncovered a mechanism by which NAD(P)H:quinone oxidoreductase 1 (NQO1) orchestrates lung metastasis. NQO1 stabilized and upregulated peptidyl-prolyl cis-trans isomerase A (PPIA), a chaperone that regulates protein conformation and activity, by preventing its oxidation at a critical cysteine residue C161. PPIA subsequently activated CD147, a membrane protein that facilitates cell invasion. Moreover, NQO1-induced secretion of PPIA modulated the immune landscape of both primary and lung metastatic sites. Secreted PPIA engaged CD147 on neutrophils and triggered the release of neutrophil extracellular traps (NET) and neutrophil elastase, which enhanced tumor progression, invasiveness, and lung colonization. Pharmacological targeting of PPIA effectively inhibited NQO1-mediated breast cancer lung metastasis. These findings reveal a previously unrecognized NQO1-PPIA-CD147-NET axis that drives breast cancer lung metastasis. Inhibiting this axis is a potential therapeutic strategy to limit lung metastasis in patients with breast cancer. Significance: NQO1 stabilizes and promotes the secretion of PPIA to activate CD147 in neutrophils and stimulate NET formation, promoting breast cancer lung metastasis and providing therapeutic targets for this fatal condition.
Collapse
Affiliation(s)
- Xinzhi Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Yi Qu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Research and Development in Marine Bio-Resource Pharmaceutics, Nanjing, China
| | - Qianqian Xu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Research and Development in Marine Bio-Resource Pharmaceutics, Nanjing, China
| | - Zeyu Jiang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Research and Development in Marine Bio-Resource Pharmaceutics, Nanjing, China
| | - Hang Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Research and Development in Marine Bio-Resource Pharmaceutics, Nanjing, China
| | - Binyan Lin
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zehong Cao
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuqi Pan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Research and Development in Marine Bio-Resource Pharmaceutics, Nanjing, China
| | - Sheng Li
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yili Hu
- Experiment Center for Science and Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Yang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Li He
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hang Chang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Bo Hang
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| | - Hongmei Wen
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jian-Hua Mao
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, California
| |
Collapse
|
8
|
Luo F, Yang J, Yang X, Mi J, Ye T, Li G, Xie Y. Saikosaponin D potentiates the antineoplastic effects of doxorubicin in drug-resistant breast cancer through perturbing NQO1-mediated intracellular redox balance. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155945. [PMID: 39146878 DOI: 10.1016/j.phymed.2024.155945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/09/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Drug resistance to doxorubicin (DOX) significantly limits its therapeutic efficacy in breast cancer (BC) patients. Saikosaponin D (SSD), a triterpene saponin derived from the traditional herb Radix Bupleuri, has shown promise as a chemotherapeutic sensitizer in preclinical studies due to its notable antitumor activity. However, the role and mechanism of SSD in DOX-resistant BC cells remain largely unexplored. PURPOSE This study aimed to investigate the chemosensitizing effect of SSD on DOX-resistant BC and the underlying molecular mechanisms both in vitro and in vivo. METHODS In vitro assays, including cell viability, clone formation, three-dimensional tumor spheroid growth, and apoptosis analysis, were conducted to evaluate the synergistic effect of SSD and DOX on resistant BC cells. Reactive oxygen species (ROS), GSH/GSSG, NADPH/NADP+, and NADH/NAD+ detections were employed to assess the impact of SSD on cellular redox homeostasis. Western blotting, cell cycle distribution assay, and DOX uptake assay were performed to further elucidate the possible antineoplastic mechanism of SSD. Finally, a subcutaneous MCF7/DOX cell xenografted model in nude mice was established to identify the in vivo anticarcinogenic effect of SSD combined with DOX. RESULTS SSD significantly inhibited cell viability, proliferation, and clone formation, enhancing DOX's anticancer efficacy in vitro and in vivo. Mechanistically, SSD reduced STAT1, NQO1, and PGC-1α protein levels, leading to cellular redox imbalance, excessive ROS generation, and depletion of GSH, NADPH, and NADH. SSD induced DNA damage by disrupting redox homeostasis, resulting in G0/G1 phase cell cycle arrest. Additionally, SSD increased DOX accumulation in BC cells via inhibiting P-gp protein expression and efflux activity. CONCLUSION We demonstrated for the first time that SSD enhances the sensitivity of chemoresistant BC cells to DOX by disrupting cellular redox homeostasis through inactivation of the STAT1/NQO1/PGC-1α signaling pathway. This study provides evidence for SSD as an adjuvant agent in drug-resistant BC treatment.
Collapse
Affiliation(s)
- Fazhen Luo
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China; Pharmacy Department, Shanghai Integrated Traditional Chinese and Western Medicine Hospital, 184 Baoding Road, Shanghai 200082, China
| | - Juan Yang
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Xiuru Yang
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Jinxia Mi
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Taiwei Ye
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Guowen Li
- Pharmacy Department, Shanghai Integrated Traditional Chinese and Western Medicine Hospital, 184 Baoding Road, Shanghai 200082, China.
| | - Yan Xie
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
9
|
Hou Y, Wang H, Wu J, Guo H, Chen X. Dissecting the pleiotropic roles of reactive oxygen species (ROS) in lung cancer: From carcinogenesis toward therapy. Med Res Rev 2024; 44:1566-1595. [PMID: 38284170 DOI: 10.1002/med.22018] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024]
Abstract
Lung cancer is a major cause of morbidity and mortality. The specific pulmonary structure to directly connect with ambient air makes it more susceptible to damage from airborne toxins. External oxidative stimuli and endogenous reactive oxygen species (ROS) play a crucial role in promoting lung carcinogenesis and development. The biological properties of higher ROS levels in tumor cells than in normal cells make them more sensitive and vulnerable to ROS injury. Therefore, the strategy of targeting ROS has been proposed for cancer therapy for decades. However, it is embarrassing that countless attempts at ROS-based therapies have had very limited success, and no FDA approval in the anticancer list was mechanistically based on ROS manipulation. Even compared with the untargetable proteins, such as transcription factors, ROS are more difficult to be targeted due to their chemical properties. Thus, the pleiotropic roles of ROS provide therapeutic potential for anticancer drug discovery, while a better dissection of the mechanistic action and signaling pathways is a prerequisite for future breakthroughs. This review discusses the critical roles of ROS in cancer carcinogenesis, ROS-inspired signaling pathways, and ROS-based treatment, exemplified by lung cancer. In particular, an eight considerations rule is proposed for ROS-targeting strategies and drug design and development.
Collapse
Affiliation(s)
- Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Heng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
- Department of Pharmaceutical Sciences, University of Macau, Taipa, Macao, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, China
| |
Collapse
|
10
|
Chen Y, Wu R, Li X, Cao M, Yang M, Fu B, Xuan C, Chen C, Zhou Y, Hu R. β-Lapachone, an NQO1 bioactivatable drug, prevents lung tumorigenesis in mice. Eur J Pharmacol 2024; 973:176511. [PMID: 38604545 DOI: 10.1016/j.ejphar.2024.176511] [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: 10/17/2023] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 04/13/2024]
Abstract
Lung cancer is one of the most lethal cancers with high incidence worldwide. The prevention of lung cancer is of great significance to reducing the social harm caused by this disease. An in-depth understanding of the molecular changes underlying precancerous lesions is essential for the targeted chemoprevention against lung cancer. Here, we discovered an increased NQO1 level over time within pulmonary premalignant lesions in both the KrasG12D-driven and nicotine-derived nitrosamine ketone (NNK)-induced mouse models of lung cancer, as well as in KrasG12D-driven and NNK-induced malignant transformed human bronchial epithelial cells (BEAS-2B and 16HBE). This suggests a potential correlation between the NQO1 expression and lung carcinogenesis. Based on this finding, we utilized β-Lapachone (β-Lap), an NQO1 bioactivatable drug, to suppress lung tumorigenesis. In this study, the efficacy and safety of low-dose β-Lap were demonstrated in preventing lung tumorigenesis in vivo. In conclusion, our study suggests that long-term consumption of low-dose β-Lap could potentially be an effective therapeutic strategy for the prevention of lung premalignant lesions. However, further studies and clinical trials are necessary to validate our findings, determine the safety of long-term β-Lap usage in humans, and promote the use of β-Lap in high-risk populations.
Collapse
Affiliation(s)
- Yaxin Chen
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Ruoyu Wu
- Jinling High School, 210005, Nanjing, China
| | - Xingyan Li
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Mengran Cao
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Mengdi Yang
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Bin Fu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Chenyuan Xuan
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Chi Chen
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China
| | - Yang Zhou
- Henan Provincial Clinical Research Center for Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou University, 450018, Zhengzhou, China.
| | - Rong Hu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
11
|
Liu J, Ma R, Chen S, Lai Y, Liu G. Anoikis patterns via machine learning strategy and experimental verification exhibit distinct prognostic and immune landscapes in melanoma. Clin Transl Oncol 2024; 26:1170-1186. [PMID: 37989822 DOI: 10.1007/s12094-023-03336-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/10/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Anoikis is a cell death programmed to eliminate dysfunctional or damaged cells induced by detachment from the extracellular matrix. Utilizing an anoikis-based risk stratification is anticipated to understand melanoma's prognostic and immune landscapes comprehensively. METHODS Differential expression genes (DEGs) were analyzed between melanoma and normal skin tissues in The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression data sets. Next, least absolute shrinkage and selection operator, support vector machine-recursive feature elimination algorithm, and univariate and multivariate Cox analyses on the 308 DEGs were performed to build the prognostic signature in the TCGA-melanoma data set. Finally, the signature was validated in GSE65904 and GSE22155 data sets. NOTCH3, PIK3R2, and SOD2 were validated in our clinical samples by immunohistochemistry. RESULTS The prognostic model for melanoma patients was developed utilizing ten hub anoikis-related genes. The overall survival (OS) of patients in the high-risk subgroup, which was classified by the optimal cutoff value, was remarkably shorter in the TCGA-melanoma, GSE65904, and GSE22155 data sets. Low-risk patients exhibited low immune cell infiltration and high expression of immunophenoscores and immune checkpoints. They also demonstrated increased sensitivity to various drugs, including dasatinib and dabrafenib. NOTCH3, PIK3R2, and SOD2 were notably associated with OS by univariate Cox analysis in the GSE65904 data set. The clinical melanoma samples showed remarkably higher protein expressions of NOTCH3 (P = 0.003) and PIK3R2 (P = 0.009) than the para-melanoma samples, while the SOD2 protein expression remained unchanged. CONCLUSIONS In this study, we successfully established a prognostic anoikis-connected signature using machine learning. This model may aid in evaluating patient prognosis, clinical characteristics, and immune treatment modalities for melanoma.
Collapse
Affiliation(s)
- Jinfang Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, China
| | - Rong Ma
- School of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Siyuan Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, China
| | - Yongxian Lai
- Department of Dermatologic Surgery, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, No. 1278 Baode Road, Shanghai, China.
| | - Guangpeng Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, No. 301 Middle Yanchang Road, Shanghai, China.
| |
Collapse
|
12
|
Sharma A, Verwilst P, Li M, Ma D, Singh N, Yoo J, Kim Y, Yang Y, Zhu JH, Huang H, Hu XL, He XP, Zeng L, James TD, Peng X, Sessler JL, Kim JS. Theranostic Fluorescent Probes. Chem Rev 2024; 124:2699-2804. [PMID: 38422393 PMCID: PMC11132561 DOI: 10.1021/acs.chemrev.3c00778] [Citation(s) in RCA: 86] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The ability to gain spatiotemporal information, and in some cases achieve spatiotemporal control, in the context of drug delivery makes theranostic fluorescent probes an attractive and intensely investigated research topic. This interest is reflected in the steep rise in publications on the topic that have appeared over the past decade. Theranostic fluorescent probes, in their various incarnations, generally comprise a fluorophore linked to a masked drug, in which the drug is released as the result of certain stimuli, with both intrinsic and extrinsic stimuli being reported. This release is then signaled by the emergence of a fluorescent signal. Importantly, the use of appropriate fluorophores has enabled not only this emerging fluorescence as a spatiotemporal marker for drug delivery but also has provided modalities useful in photodynamic, photothermal, and sonodynamic therapeutic applications. In this review we highlight recent work on theranostic fluorescent probes with a particular focus on probes that are activated in tumor microenvironments. We also summarize efforts to develop probes for other applications, such as neurodegenerative diseases and antibacterials. This review celebrates the diversity of designs reported to date, from discrete small-molecule systems to nanomaterials. Our aim is to provide insights into the potential clinical impact of this still-emerging research direction.
Collapse
Affiliation(s)
- Amit Sharma
- Amity
School of Chemical Sciences, Amity University
Punjab, Sector 82A, Mohali 140 306, India
| | - Peter Verwilst
- Rega
Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49, Box 1041, 3000 Leuven, Belgium
| | - Mingle Li
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
| | - Dandan Ma
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Nem Singh
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Jiyoung Yoo
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Yujin Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
| | - Ying Yang
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Jing-Hui Zhu
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Haiqiao Huang
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Xi-Le Hu
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xiao-Peng He
- Key
Laboratory for Advanced Materials and Joint International Research
Laboratory of Precision Chemistry and Molecular Engineering, Feringa
Nobel Prize Scientist Joint Research Center, School of Chemistry and
Molecular Engineering, East China University
of Science and Technology, 130 Meilong Road, Shanghai 200237, China
- National
Center for Liver Cancer, the International Cooperation Laboratory
on Signal Transduction, Eastern Hepatobiliary
Surgery Hospital, Shanghai 200438, China
| | - Lintao Zeng
- School of
Light Industry and Food Engineering, Guangxi
University, Nanning, Guangxi 530004, China
| | - Tony D. James
- Department
of Chemistry, University of Bath, Bath BA2 7AY, United Kingdom
- School
of Chemistry and Chemical Engineering, Henan
Normal University, Xinxiang 453007, China
| | - Xiaojun Peng
- College
of Materials Science and Engineering, Shenzhen
University, Shenzhen 518060, China
- State
Key Laboratory of Fine Chemicals, Dalian
University of Technology, Dalian 116024, China
| | - Jonathan L. Sessler
- Department
of Chemistry, The University of Texas at
Austin, Texas 78712-1224, United
States
| | - Jong Seung Kim
- Department
of Chemistry, Korea University, Seoul 02841, Korea
- TheranoChem Incorporation, Seongbuk-gu, Seoul 02841, Korea
| |
Collapse
|
13
|
Zhong S, Borlak J. Sex differences in the tumor promoting effects of tobacco smoke in a cRaf transgenic lung cancer disease model. Arch Toxicol 2024; 98:957-983. [PMID: 38245882 PMCID: PMC10861769 DOI: 10.1007/s00204-023-03671-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024]
Abstract
Tobacco smoke (TS) is the leading cause for lung cancer (LC), and female smokers are at a greater risk for LC. Yet, the underlying causes are unknown. We performed whole genome scans in TS exposed wild type and histologically characterized tumor lesions of cRaf transgenic mice. We constructed miRNA-gene and transcription factor-miRNA/gene regulatory networks and determined sex-specific gene regulations by evaluating hormone receptor activities. We validated the findings from TS exposed cRaf mice in a large cohort of smoking and never-smoking LC patients. When compared to males, TS prompted a sevenfold increase in tumor multiplicity in cRaf females. Genome-wide scans of tumor lesions identified 161 and 53 genes and miRNAs, which code for EGFR/MAPK signaling, cell proliferation, oncomirs and oncogenes, and 50% of DEGs code for immune response and tumor evasion. Outstandingly, in transgenic males, TS elicited upregulation of 20 tumor suppressors, some of which are the targets of the androgen and estrogen receptor. Conversely, in females, 18 tumor suppressors were downregulated, and five were specifically repressed by the estrogen receptor. We found TS to perturb the circadian clock in a sex-specific manner and identified a female-specific regulatory loop that consisted of the estrogen receptor, miR-22-3p and circadian genes to support LC growth. Finally, we confirmed sex-dependent tumor promoting effects of TS in a large cohort of LC patients. Our study highlights the sex-dependent genomic responses to TS and the interplay of circadian clock genes and hormone receptors in the regulation of oncogenes and oncomirs in LC growth.
Collapse
Affiliation(s)
- Shen Zhong
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
14
|
Wang J, Su X, Jiang L, Boudreau MW, Chatkewitz LE, Kilgore JA, Zahid KR, Williams NS, Chen Y, Liu S, Hergenrother PJ, Huang X. Augmented Concentration of Isopentyl-Deoxynyboquinone in Tumors Selectively Kills NAD(P)H Quinone Oxidoreductase 1-Positive Cancer Cells through Programmed Necrotic and Apoptotic Mechanisms. Cancers (Basel) 2023; 15:5844. [PMID: 38136388 PMCID: PMC10741405 DOI: 10.3390/cancers15245844] [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: 10/18/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Lung and breast cancers rank as two of the most common and lethal tumors, accounting for a substantial number of cancer-related deaths worldwide. While the past two decades have witnessed promising progress in tumor therapy, developing targeted tumor therapies continues to pose a significant challenge. NAD(P)H quinone oxidoreductase 1 (NQO1), a two-electron reductase, has been reported as a promising therapeutic target across various solid tumors. β-Lapachone (β-Lap) and deoxynyboquinone (DNQ) are two NQO1 bioactivatable drugs that have demonstrated potent antitumor effects. However, their curative efficacy has been constrained by adverse effects and moderate lethality. To enhance the curative potential of NQO1 bioactivatable drugs, we developed a novel DNQ derivative termed isopentyl-deoxynyboquinone (IP-DNQ). Our study revealed that IP-DNQ treatment significantly increased reactive oxygen species generation, leading to double-strand break (DSB) formation, PARP1 hyperactivation, and catastrophic energy loss. Notably, we discovered that this novel drug induced both apoptosis and programmed necrosis events, which makes it entirely distinct from other NQO1 bioactivatable drugs. Furthermore, IP-DNQ monotherapy demonstrated significant antitumor efficacy and extended mice survival in A549 orthotopic xenograft models. Lastly, we identified that in mice IP-DNQ levels were significantly elevated in the plasma and tumor compared with IB-DNQ levels. This study provides novel preclinical evidence supporting IP-DNQ efficacy in NQO1+ NSCLC and breast cancer cells.
Collapse
Affiliation(s)
- Jiangwei Wang
- Department of Radiation Oncology, Indianapolis, IN 46202, USA
| | - Xiaolin Su
- Department of Biochemistry and Molecular Biology, Indianapolis, IN 46202, USA
| | - Lingxiang Jiang
- Department of Radiation Oncology, Indianapolis, IN 46202, USA
| | - Matthew W. Boudreau
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Lindsay E. Chatkewitz
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jessica A. Kilgore
- Department of Biochemistry, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA (N.S.W.)
| | | | - Noelle S. Williams
- Department of Biochemistry, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA (N.S.W.)
| | - Yaomin Chen
- Indiana University Health Pathology Laboratory, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Shaohui Liu
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul J. Hergenrother
- Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiumei Huang
- Department of Radiation Oncology, Indianapolis, IN 46202, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
15
|
Wu L, Chen Z, Yu Q, Fang J, Chen Y, Chen Y, Wang K, Xu Y, Song W, Peng Z. NAD (P)H Quinone Dehydrogenase 1-Targeting Triptolide Analogue Causes Tumor Regression and Sensitizes Cisplatin-Resistant Lung Cancer to Chemotherapy. ACS Pharmacol Transl Sci 2023; 6:1508-1517. [PMID: 37854615 PMCID: PMC10580380 DOI: 10.1021/acsptsci.3c00144] [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: 07/08/2023] [Indexed: 10/20/2023]
Abstract
Cisplatin (DDP) is a first-line chemotherapeutic drug against lung cancer. Nonetheless, the effectiveness of this drug is hampered by drug resistance. Overcoming drug resistance is crucial for improving the outcomes of lung cancer treatment. Here, we reported the effect of CX-23, an activated triptolide analogue that targets NAD (P)H quinone dehydrogenase 1 (NQO1), on DDP-resistant lung cancer and sensitizes DDP-resistant lung cancer to chemotherapy. Our findings unveiled the antiproliferative activity of CX-23 against both A549- and DDP-resistant A549 (A549/DDP) cells while enhancing the chemosensitivity of these cells to DDP. Notably, CX-23 demonstrated no toxicity toward normal lung cells. Further investigations revealed that CX-23 exerts its effects by blocking AKT phosphorylation, leading to reduced expression of Mcl-1 and Bcl-2, and upregulating cleaved-caspase-3 levels, ultimately inducing apoptosis in cancer cells. CX-23 can be rapidly transformed in both A549 and A549/DDP cell lysates while remaining stable in mouse plasma and normal lung tissues. Pharmacokinetic analysis showed that CX-23 can distribute to lung tissues. Moreover, in vivo studies showed that CX-23 can prevent DDP-resistant lung cancer progression without causing any toxicity in the liver, kidneys, or lungs after 6 weeks of treatment. The combination of CX-23 and DDP not only significantly inhibited tumor progression compared to DDP alone but also attenuated DDP-induced kidney toxicity. These findings suggest that CX-23 alone or in combination with DDP may provide an alternative therapeutic option for DDP-resistant lung cancer.
Collapse
Affiliation(s)
- Liuying Wu
- National
& Local Joint Engineering Research Center of High-Throughput Drug
Screening Technology, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Zhelin Chen
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350004, China
| | - Qing Yu
- National
& Local Joint Engineering Research Center of High-Throughput Drug
Screening Technology, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Jinxin Fang
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350004, China
| | - Yong Chen
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350004, China
| | - Yuhan Chen
- National
& Local Joint Engineering Research Center of High-Throughput Drug
Screening Technology, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Kai Wang
- National
& Local Joint Engineering Research Center of High-Throughput Drug
Screening Technology, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
- Hubei
Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Yan Xu
- Department
of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350004, China
| | - Wei Song
- National
& Local Joint Engineering Research Center of High-Throughput Drug
Screening Technology, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| | - Zhihong Peng
- National
& Local Joint Engineering Research Center of High-Throughput Drug
Screening Technology, College of Health Science and Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
16
|
Zheng L, Yang S, Xu R, Yang Y, Quan J, Lin Z, Quan C. NQO1 drives glioblastoma cell aggressiveness through EMT induction via the PI3K/Akt/mTOR/Snail pathway. Int J Oncol 2023; 63:110. [PMID: 37594082 PMCID: PMC10552716 DOI: 10.3892/ijo.2023.5558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 06/20/2023] [Indexed: 08/19/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequent and lethal cancer derived from the central nervous system, of which the mesenchymal (MES) subtype seriously influences the survival and prognosis of patients. NAD(P)H: quinone acceptor oxidoreductase 1 (NQO1) serves an important role in the carcinogenesis and progression of various types of cancer; however, the specific mechanism underlying the regulatory effects of NQO1 on GBM is unclear. Thus, the present study aimed to explore the role and mechanism of NQO1 in GBM progression. The results of bioinformatics analysis and immunohistochemistry showed that high expression of NQO1 was significantly related to the MES phenotype of GBM and shorter survival. In addition, MTT, colony formation, immunofluorescence and western blot analyses, and lung metastasis model experiments suggested that silencing NQO1 inhibited the proliferation and metastasis of GBM cells in vitro and in vivo. Furthermore, western blotting showed that the activity of the PI3K/Akt/mTOR signaling pathway was revealed to be inhibited by downregulation of NQO1 expression, whereas it was enhanced by overexpression of NQO1. Notably, co‑immunoprecipitation and ubiquitination experiments suggested that Snail was considered an important downstream target of NQO1 in GBM cells. Snail knockdown could eliminate the promoting effect of ectopic NQO1 on the proliferation and invasion of GBM cells, and reduce its effects on the activity of PI3K/Akt/mTOR signaling pathway. These results indicated that NQO1 could promote GBM aggressiveness by activating the PI3K/Akt/mTOR signaling pathway in a Snail‑dependent manner, and NQO1 and its relevant pathways may be considered novel targets for GBM therapy.
Collapse
Affiliation(s)
- Lan Zheng
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Department of Obstetrics and Gynecology, Taizhou Hospital of Zhejiang Province Affiliated with Wenzhou Medical University, Linhai, Zhejiang 317000
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
| | - Shipeng Yang
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Ran Xu
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Yang Yang
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Jishu Quan
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Zhenhua Lin
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
- Department of Pathology, Yanbian University Medical College, Yanji, Jilin 133000, P.R. China
| | - Chunhua Quan
- Central Laboratory, Affiliated Hospital of Yanbian University, Yanji, Jilin 133002
- Key Laboratory of Pathobiology, State Ethnic Affairs Commission, Yanbian University, Yanji, Jilin 133000
| |
Collapse
|
17
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
18
|
Roy NJ, Save SN, Sharma VK, Abraham B, Kuttanamkuzhi A, Sharma S, Lahiri M, Talukdar P. NAD(P)H:Quinone Acceptor Oxidoreductase 1 (NQO1) Activatable Salicylamide H + /Cl - Transporters. Chemistry 2023; 29:e202301412. [PMID: 37345998 DOI: 10.1002/chem.202301412] [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: 05/03/2023] [Revised: 06/10/2023] [Accepted: 06/22/2023] [Indexed: 06/23/2023]
Abstract
NAD(P)H:quinone acceptor oxidoreductase 1 (NQO1), a detoxifying enzyme overexpressed in tumors, plays a key role in protecting cancer cells against oxidative stress and thus has been considered an attractive candidate for activating prodrug(s). Herein, we report the first use of NQO1 for the selective activation of 'protransporter' systems in cancer cells leading to the induction of apoptosis. Salicylamides, easily synthesizable small molecules, have been effectively used for efficient H+ /Cl- symport across lipid membranes. The ion transport activity of salicylamides was efficiently abated by caging the OH group with NQO1 activatable quinones via either ether or ester linkage. The release of active transporters, following the reduction of quinone caged 'protransporters' by NQO1, was verified. Both the transporters and protransporters exhibited significant toxicity towards the MCF-7 breast cancer line, mediated via the induction of oxidative stress, mitochondrial membrane depolarization, and lysosomal deacidification. Induction of cell death via intrinsic apoptotic pathway was verified by monitoring PARP1 cleavage.
Collapse
Affiliation(s)
- Naveen J Roy
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Shreyada N Save
- Department of Biotechnology, Savitribai Phule Pune University (Formerly University of Pune), Pune, 411007, Maharashtra, India
| | - Virender Kumar Sharma
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Benchamin Abraham
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Abhijith Kuttanamkuzhi
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Shilpy Sharma
- Department of Biotechnology, Savitribai Phule Pune University (Formerly University of Pune), Pune, 411007, Maharashtra, India
| | - Mayurika Lahiri
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| | - Pinaki Talukdar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, 411008, Maharashtra, India
| |
Collapse
|
19
|
He Z, Shen X, Wang B, Xu L, Ling Q. CT radiomics for noninvasively predicting NQO1 expression levels in hepatocellular carcinoma. PLoS One 2023; 18:e0290900. [PMID: 37695786 PMCID: PMC10495018 DOI: 10.1371/journal.pone.0290900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023] Open
Abstract
Using noninvasive radiomics to predict pathological biomarkers is an innovative work worthy of exploration. This retrospective cohort study aimed to analyze the correlation between NAD(P)H quinone oxidoreductase 1 (NQO1) expression levels and the prognosis of patients with hepatocellular carcinoma (HCC) and to construct radiomic models to predict the expression levels of NQO1 prior to surgery. Data of patients with HCC from The Cancer Genome Atlas (TCGA) and the corresponding arterial phase-enhanced CT images from The Cancer Imaging Archive were obtained for prognosis analysis, radiomic feature extraction, and model development. In total, 286 patients with HCC from TCGA were included. According to the cut-off value calculated using R, patients were divided into high-expression (n = 143) and low-expression groups (n = 143). Kaplan-Meier survival analysis showed that higher NQO1 expression levels were significantly associated with worse prognosis in patients with HCC (p = 0.017). Further multivariate analysis confirmed that high NQO1 expression was an independent risk factor for poor prognosis (HR = 1.761, 95% CI: 1.136-2.73, p = 0.011). Based on the arterial phase-enhanced CT images, six radiomic features were extracted, and a new bi-regional radiomics model was established, which could noninvasively predict higher NQO1 expression with good performance. The area under the curve (AUC) was 0.9079 (95% CI 0.8127-1.0000). The accuracy, sensitivity, and specificity were 0.86, 0.88, and 0.84, respectively, with a threshold value of 0.404. The data verification of our center showed that this model has good predictive efficiency, with an AUC of 0.8791 (95% CI 0.6979-1.0000). In conclusion, there existed a significant correlation between the CT image features and the expression level of NQO1, which could indirectly reflect the prognosis of patients with HCC. The predictive model based on arterial phase CT imaging features has good stability and diagnostic efficiency and is a potential means of identifying the expression level of NQO1 in HCC tissues before surgery.
Collapse
Affiliation(s)
- Zenglei He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Xiaoyong Shen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Bin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Li Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Qi Ling
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| |
Collapse
|
20
|
Shen L, Jiang S, Yang Y, Yang H, Fang Y, Tang M, Zhu R, Xu J, Jiang H. Pan-cancer and single-cell analysis reveal the prognostic value and immune response of NQO1. Front Cell Dev Biol 2023; 11:1174535. [PMID: 37583897 PMCID: PMC10424457 DOI: 10.3389/fcell.2023.1174535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 07/21/2023] [Indexed: 08/17/2023] Open
Abstract
Background: Overexpression of the NAD(P)H: Quinone Oxidoreductase 1 (NQOI) gene has been linked with tumor progression, aggressiveness, drug resistance, and poor patient prognosis. Most research has described the biological function of the NQO1 in certain types and limited samples, but a comprehensive understanding of the NQO1's function and clinical importance at the pan-cancer level is scarce. More research is needed to understand the role of NQO1 in tumor infiltration, and immune checkpoint inhibitors in various cancers are needed. Methods: The NQO1 expression data for 33 types of pan-cancer and their association with the prognosis, pathologic stage, gender, immune cell infiltration, the tumor mutation burden, microsatellite instability, immune checkpoints, enrichment pathways, and the half-maximal inhibitory concentration (IC50) were downloaded from public databases. Results: Our findings indicate that the NQO1 gene was significantly upregulated in most cancer types. The Cox regression analysis showed that overexpression of the NQO1 gene was related to poor OS in Glioma, uveal melanoma, head and neck squamous cell carcinoma, kidney renal papillary cell carcinoma, and adrenocortical carcinoma. NQO1 mRNA expression positively correlated with infiltrating immune cells and checkpoint molecule levels. The single-cell analysis revealed a potential relationship between the NQO1 mRNA expression levels and the infiltration of immune cells and stromal cells in bladder urothelial carcinoma, invasive breast carcinoma, and colorectal cancer. Conversely, a negative association was noted between various drugs (17-AAG, Lapatinib, Trametinib, PD-0325901) and the NQO1 mRNA expression levels. Conclusion: NQO1 expression was significantly associated with prognosis, immune infiltrates, and drug resistance in multiple cancer types. The inhibition of the NQO1-dependent signaling pathways may provide a promising strategy for developing new cancer-targeted therapies.
Collapse
Affiliation(s)
- Liping Shen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical, Taizhou, Zhejiang, China
| | - Shan Jiang
- Department of Radiology, Jining No. 1 People’s Hospital, Jining, Shandong, China
| | - Yu Yang
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical, Taizhou, Zhejiang, China
| | - Hongli Yang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanchun Fang
- Department of Ultrasonography, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical, Taizhou, Zhejiang, China
| | - Meng Tang
- Department of Ultrasonography, Jining No. 1 People’s Hospital, Jining, Shandong, China
| | - Rangteng Zhu
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical, Taizhou, Zhejiang, China
| | - Jiaqin Xu
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical, Taizhou, Zhejiang, China
| | - Hantao Jiang
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical, Taizhou, Zhejiang, China
| |
Collapse
|
21
|
Gong Q, Wang P, Li T, Yu Z, Yang L, Wu C, Hu J, Yang F, Zhang X, Li X. Novel NQO1 substrates bearing two nitrogen redox centers: Design, synthesis, molecular dynamics simulations, and antitumor evaluation. Bioorg Chem 2023; 134:106480. [PMID: 36958178 DOI: 10.1016/j.bioorg.2023.106480] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/20/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023]
Abstract
By analyzing the crystal structure of NQO1, an additional binding region for the ligand was discovered. In this study, a series of derivatives with a novel skeleton bearing two nitrogen redox centers were designed by introducing amines or hydrazines to fit with the novel binding region of NQO1. Compound 24 with a (4-fluorophenyl)hydrazine substituent was identified as the most efficient substrate for NQO1 with the reduction rate and catalytic efficiency of 1972 ± 82 μmol NADPH/min/μmol NQO1 and 6.4 ± 0.4 × 106 M-1s-1, respectively. Molecular dynamics (MD) simulation revealed that the distances between the nitrogen atom of the redox centers and the key Tyr128 and Tyr126 residues were 3.5 Å (N1-Tyr128) and 3.4 Å (N2-Tyr126), respectively. Compound 24 (IC50/A549 = 0.69 ± 0.09 μM) showed potent antitumor activity against A549 cells both in vitro and in vivo through ROS generation via NQO1-mediated redox cycling, leading to a promising NQO1-targeting antitumor candidate.
Collapse
Affiliation(s)
- Qijie Gong
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Pengfei Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China; Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Tian Li
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China; Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Zhan Yu
- The Affiliated Jiangning Hospital of NJMU, Nanjing Medical University (NJMU), Nanjing 211199, China; Jiangning Clinical Medical College of Jiangsu University, Nanjing 211100, China.
| | - Le Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Chenyang Wu
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jiabao Hu
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Fulai Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiaojin Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiang Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
22
|
Liang C, Yi K, Zhou X, Li X, Zhong C, Cao H, Xie C, Zhu J. Destruction of the cellular antioxidant pool contributes to resveratrol-induced senescence and apoptosis in lung cancer. Phytother Res 2023. [PMID: 36866538 DOI: 10.1002/ptr.7795] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Resveratrol (RES) has various pharmacological bioactivities and its anticancer effects in lung cancer have been proven. However, the underlying mechanisms of action of RES in lung cancer remain unclear. This study focused on Nrf2-mediated antioxidant systems in RES-treated lung cancer cells. A549 and H1299 cells were treated with various concentrations of RES at different times. RES decreased cell viability, inhibited cell proliferation, and increased the number of senescent and apoptotic cells in a concentration- and time-dependent manner. Moreover, RES-induced lung cancer cell arrest at the G1 phase was accompanied by changes in apoptotic proteins (Bax, Bcl-2, and cleaved caspase 3). Furthermore, RES induced a senescent phenotype along with changes in senescence-related markers (senescence-associated β-galactosidase activity, p21, and p-γH2AX). More importantly, with prolonged exposure time and increased exposure concentration, intracellular reactive oxygen species (ROS) continuously accumulated, resulting in a decrease in Nrf2 and its downstream antioxidant response elements, including CAT, HO-1, NQO1, and SOD1. Meanwhile, RES-induced ROS accumulation and cell apoptosis were reversed by N-acetyl-l-cysteine treatment. Taken together, these results suggest that RES disturb lung cancer cellular homeostasis by destroying the intracellular antioxidant pool to increase ROS production. Our findings provide a new perspective on RES intervention in lung cancer.
Collapse
Affiliation(s)
- Chunhua Liang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Kefan Yi
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xu Zhou
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoting Li
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China.,Cancer Research Division, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hui Cao
- Department of Thoracic Surgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chunfeng Xie
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jianyun Zhu
- Department of Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
23
|
Kaghazchi B, Um IH, Elshani M, Read OJ, Harrison DJ. Spatial Analysis of NQO1 in Non-Small Cell Lung Cancer Shows Its Expression Is Independent of NRF1 and NRF2 in the Tumor Microenvironment. Biomolecules 2022; 12:1652. [PMID: 36359002 PMCID: PMC9687417 DOI: 10.3390/biom12111652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 07/22/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 1 (NFE2L1, NRF1) and nuclear factor erythroid 2-related factor 2 (NFE2L2, NRF2) are distinct oxidative stress response transcription factors, both of which have been shown to perform cytoprotective functions, modulating cell stress response and homeostasis. NAD(P)H:quinone oxidoreductase (NQO1) is a mutual downstream antioxidant gene target that catalyzes the two-electron reduction of an array of substrates, protecting against reactive oxygen species (ROS) generation. NQO1 is upregulated in non-small cell lung cancer (NSCLC) and is proposed as a predictive biomarker and therapeutic target. Antioxidant protein expression of immune cells within the NSCLC tumor microenvironment (TME) remains undetermined and may affect immune cell effector functions and survival outcomes. Multiplex immunofluorescence was performed to examine the co-localization of NQO1, NRF1 and NRF2 within the tumor and TME of 162 chemotherapy-naïve, early-stage NSCLC patients treated by primary surgical resection. This study demonstrates that NQO1 protein expression is high in normal, tumor-adjacent tissue and that NQO1 expression varies depending on the cell type. Inter and intra-patient heterogenous NQO1 expression was observed in lung cancer. Co-expression analysis showed NQO1 is independent of NRF1 and NRF2 in tumors. Density-based co-expression analysis demonstrated NRF1 and NRF2 double-positive expression in cancer cells is associated with improved overall survival.
Collapse
Affiliation(s)
- Boback Kaghazchi
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - In Hwa Um
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
| | - Mustafa Elshani
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
- NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - Oliver J. Read
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
- NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| | - David J. Harrison
- School of Medicine, University of St Andrews, St Andrews KY16 9TF, UK
- NuCana plc, 3 Lochside Way, Edinburgh EH12 9DT, UK
| |
Collapse
|
24
|
Chen Y, Wu L, Bao M. MiR-485-5p Suppress the Malignant Characteristics of the Lung Adenocarcinoma via Targeting NADPH Quinone Oxidoreductase-1 to Inhibit the PI3K/Akt. Mol Biotechnol 2022; 65:794-806. [PMID: 36219369 DOI: 10.1007/s12033-022-00577-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/22/2022] [Indexed: 10/17/2022]
Abstract
Lung adenocarcinoma (LUAD), a prevalent form of non-small cell lung cancer (NSCLC), has a high incidence and mortality rate. However, its molecular regulatory mechanisms have yet to be fully understood. The purpose of this study was to look into how NADPH quinone oxidoreductase-1 (NQO1) and it miR-485-5p and affected LUAD cells. The levels of miR-485-5p and NQO1 expression in LUAD cells and tissues were determined by means of quantitative reverse transcription polymerase chain reaction. The viability, proliferation, migration, and apoptosis of LUAD cells were assessed using cell counting Kit-8, 5-bromo-2'-deoxyuridine, transwell, and caspase-3 assays, respectively. Western blot experiments were used to examine the relative protein expression of matrix metallopeptidase 2 and matrix metallopeptidase 9, as well as the phosphorylation of phosphoinositide 3-kinase (PI3K) and protein kinase B (Akt) in LUAD cells. Luciferase and RNA pull-down experiments were also conducted for the verification of miR-485-5p's underlying relationship with NQO1. In our study, we found that LUAD cells and tissues had miR-485-5p downregulation and NQO1 upregulation. The experimental outcomes indicated that miR-485-5p overexpression in LUAD cells reduced their malignant behaviors, suppressed PI3K and Akt phosphorylation, and facilitated apoptosis. The results also revealed that NQO1 was a direct miR-485-5p target, and that NQO1 could reverse miR-485-5p's inhibitory effect on the malignant phenotype of LUAD cells. Furthermore, it was also observed that through targeting NQO1, miR-485-5p could suppress LUAD cell migration and proliferation, further blocking the phosphorylation of PI3K and Akt and inducing apoptosis among LUAD cells. In conclusion, the miR-485-5/NQO1 axis regulates LUAD progression through the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Yupeng Chen
- Thoracic Surgery, Wuhan No.1 Hospital, Wuhan, 430022, Hubei, China
| | - Lin Wu
- Department of Oncology, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan, 430014, Hubei, China
| | - Min Bao
- Department of Respiratory Medicine, Wuhan Third Hospital, No. 241 Pengliuyang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| |
Collapse
|
25
|
Zhang X, Jiang K, Jiang S, Zhao F, Chen P, Huang P, Lin J. In Vivo Near-Infrared Fluorescence/Ratiometric Photoacoustic Duplex Imaging of Lung Cancer-Specific hNQO1. Anal Chem 2022; 94:13770-13776. [PMID: 36173742 DOI: 10.1021/acs.analchem.2c02153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Overexpressing human NAD(P)H:quinone oxidoreductase 1 (hNQO1) in lung cancer tissues is deemed to be an attractive biomarker, which is directly connected to cancerous pathological processes. Monitoring of hNQO1 activity is crucial to early diagnosis and prognosis of lung cancer. In this study, an activatable hemi-cyanine dye-based probe (denoted as the LET-10 probe) was synthesized for near-infrared fluorescence (NIRF) and ratiometric photoacoustic (RPA) imaging of hNQO1. LET-10 can realize the NIRF and PA signal opening in the presence of hNQO1. Taking the octabutoxy naphthalocyanine in the LET-10 probe as a built-in reference signal, the LET-10 probe further demonstrated a double-signal self-calibration process for RPA imaging. Finally, the LET-10 probe was successfully applied for NIRF/RPA duplex imaging in the hNQO1-positive A549 lung cancer model, which suggests that the LET-10 probe is a promising tool for in vivo hNQO1 detection, especially for lung cancer diagnosis.
Collapse
Affiliation(s)
- Xinming Zhang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Kejia Jiang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Shanshan Jiang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Feng Zhao
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Penghang Chen
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering, International Cancer Center, Laboratory of Evolutionary Theranostics (LET), School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| |
Collapse
|
26
|
EROĞLU C, KİRAZ E, KONTAŞ O, ÖZTÜRK A. Eşzamanlı Kemoradyoterapi ile Tedavi Edilen Evre III Küçük Hücreli Dışı Akciğer Kanserli Hastalarda NQO1’in Prognostik Önemi. KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2022. [DOI: 10.17517/ksutfd.1063749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Amaç: Bu çalışmada da eşzamanlı kemoradyoterapi ile tedavi edilen evre III küçük hücreli dışı akciğer kanseri (KHDAK)’li hastalarda NQO1’in prediktif ve prognostik öneminin tespiti amaçlandı.
Gereç ve yöntemler: Bu çalışmaya eşzamanlı cisplatin + dosetaksel kemoradyoterapisi ile tedavi edilen evre III 64 KHDAK’li olgu dahil edildi. Hastaların patoloji preparatlarında NQO1 immünohistokimyasal (İHK) boyama yapılarak negatif ve pozitiflik açısından karşılaştırıldı.
Bulgular: Medyan yaş 63 (sınırlar, 35-83) olup olguların %86 (n=55)’sı erkek idi. Olguların %27 (n=17)’si evre IIIA, %56 (n=36)’sı evre IIIB ve %17 (n=11)’si evre IIIC idi. Olgular histopatolojik olarak %47 (n=30) adenokarsinom, %47 (n=47)’si epidermoid karsinom ve %6 (n=4)’sı alt tipi belirlenemeyen KHDAK şeklinde sınıflandı. Medyan takip süresi 20 ay (sınırlar, 3-7 ay) bulundu. NQO1’in İHK boyaması sonucunda olguların 7 (%11)’si negatif, 11 (%17)’i (+) pozitif, 14 (%22)’ü (++) pozitif ve 32 (%50)’si (+++) pozitif olarak bulundu. Olguların NQO1’e göre medyan genel sağkalımı; (-)’lerde 21 ay, (+)’lerde 19 ay, (++)’lerde 16 ay ve (+++)’lerde 19 ay bulundu. Pozitif NQO1 olan olgularda medyan genel sağkalım sayısal olarak negatif olan olgulardan daha düşük olmasına rağmen istatistiksel olarak anlamlı bulunmadı (p=0,801).
Sonuç: Eşzamanlı kemoradyoterapi ile tedavi edilen evre III KHDAK’li hastalarda NQO1 prognostik faktör olabilir ancak çok hasta sayılı çalışmalara ihtiyaç vardır.
Collapse
|
27
|
Luo Y, Wang W, Zeng Y, Wang S, Guo X, Hu R, Yang G. A bioluminescent probe for NQO1 overexpressing cancer cell imaging in vitro and in vivo. Analyst 2022; 147:5264-5268. [DOI: 10.1039/d2an01435a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A bioluminescent probe NQO1-Luc toward NQO1 was constructed, which exhibits high selectivity and sensitivity toward NQO1 in vitro and adequate capability of distinguishing NQO1-overexpressing tumors in vivo.
Collapse
Affiliation(s)
- Yuhong Luo
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Wentao Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Yi Zeng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Shuangqing Wang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Xudong Guo
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Rui Hu
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| | - Guoqiang Yang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
28
|
Reactive Oxygen Species in Acute Lymphoblastic Leukaemia: Reducing Radicals to Refine Responses. Antioxidants (Basel) 2021; 10:antiox10101616. [PMID: 34679751 PMCID: PMC8533157 DOI: 10.3390/antiox10101616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 12/27/2022] Open
Abstract
Acute lymphoblastic leukaemia (ALL) is the most common cancer diagnosed in children and adolescents. Approximately 70% of patients survive >5-years following diagnosis, however, for those that fail upfront therapies, survival is poor. Reactive oxygen species (ROS) are elevated in a range of cancers and are emerging as significant contributors to the leukaemogenesis of ALL. ROS modulate the function of signalling proteins through oxidation of cysteine residues, as well as promote genomic instability by damaging DNA, to promote chemotherapy resistance. Current therapeutic approaches exploit the pro-oxidant intracellular environment of malignant B and T lymphoblasts to cause irreversible DNA damage and cell death, however these strategies impact normal haematopoiesis and lead to long lasting side-effects. Therapies suppressing ROS production, especially those targeting ROS producing enzymes such as the NADPH oxidases (NOXs), are emerging alternatives to treat cancers and may be exploited to improve the ALL treatment. Here, we discuss the roles that ROS play in normal haematopoiesis and in ALL. We explore the molecular mechanisms underpinning overproduction of ROS in ALL, and their roles in disease progression and drug resistance. Finally, we examine strategies to target ROS production, with a specific focus on the NOX enzymes, to improve the treatment of ALL.
Collapse
|
29
|
Lee KM, Gwon MR, Lee HW, Seong SJ, Yoon YR. The possibility of low isomerization of β-lapachone in the human body. Transl Clin Pharmacol 2021; 29:160-170. [PMID: 34621708 PMCID: PMC8492396 DOI: 10.12793/tcp.2021.29.e16] [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: 08/25/2021] [Revised: 09/16/2021] [Accepted: 09/22/2021] [Indexed: 11/19/2022] Open
Abstract
β-Lapachone has been reported to have anticancer and various other therapeutic effects, but is limited in clinical applications by its low bioavailability. pH-Dependent isomerization can be suggested as one plausible factor influencing its low bioavailability. Since it is known that β-lapachone is converted to its isomer, α-lapachone in hydrochloric acid (HCl) solution, isomerization in the human body may be driven by HCl in the gastric fluid. The purpose of this study was to evaluate the possibility of isomerization of β-lapachone in the human body. Chemical reactions were conducted using simulated gastric fluid (SGF, pH 1.2) and simulated intestinal fluid (SIF, pH 7.5) at 37°C. β-Lapachone was observed in SGF at 37°C for 1 hour and SIF for 3 hours. In addition, biofluid analysis was performed on plasma samples 1 hour and 4 hours, and on urine sample 12 hours after oral administration of 100 mg MB12066, a synthetic β-lapachone, in healthy adult male. All samples were analyzed using liquid chromatography-tandem mass spectrometry. Only β-lapachone peaks existed in the spectra obtained from SGF and SIF. No isomerization of β-lapachone was observed in the analysis of any of the human samples. In the current study, the possibility of pH-dependent isomerization of β-lapachone in the human body was not confirmed.
Collapse
Affiliation(s)
- Kyung Min Lee
- Department of Clinical Pharmacology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,BK21 Plus KNU Bio-Medical Convergence Program for Creative Talent, Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Mi-Ri Gwon
- Department of Clinical Pharmacology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Clinical Omics Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hae Won Lee
- Department of Clinical Pharmacology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Sook Jin Seong
- Department of Clinical Pharmacology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Young-Ran Yoon
- Department of Clinical Pharmacology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,BK21 Plus KNU Bio-Medical Convergence Program for Creative Talent, Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea.,Clinical Omics Institute, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
30
|
Zhao W, Jiang L, Fang T, Fang F, Liu Y, Zhao Y, You Y, Zhou H, Su X, Wang J, Liu S, Chen Y, Wan J, Huang X. β-Lapachone Selectively Kills Hepatocellular Carcinoma Cells by Targeting NQO1 to Induce Extensive DNA Damage and PARP1 Hyperactivation. Front Oncol 2021; 11:747282. [PMID: 34676172 PMCID: PMC8523939 DOI: 10.3389/fonc.2021.747282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death globally. Currently there is a lack of tumor-selective and efficacious therapies for hepatocellular carcinoma. β-Lapachone (ARQ761 in clinical form) selectively kill NADPH: quinone oxidoreductase 1 (NQO1)-overexpressing cancer cells. However, the effect of β-Lapachone on HCC is virtually unknown. In this study, we found that relatively high NQO1 and low catalase levels were observed in both clinical specimens collected from HCC patients and HCC tumors from the TCGA database. β-Lapachone treatment induced NQO1-selective killing of HCC cells and caused ROS formation and PARP1 hyperactivation, resulting in a significant decrease in NAD+ and ATP levels and a dramatic increase in double-strand break (DSB) lesions over time in vitro. Administration of β-Lapachone significantly inhibited tumor growth and prolonged survival in a mouse xenograft model in vivo. Our data suggest that NQO1 is an ideal potential biomarker, and relatively high NQO1:CAT ratios in HCC tumors but low ratios in normal tissues offer an optimal therapeutic window to use β-Lapachone. This study provides novel preclinical evidence for β-Lapachone as a new promising chemotherapeutic agent for use in NQO1-positive HCC patients.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Lingxiang Jiang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ting Fang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Fei Fang
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Yingchun Liu
- Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Ye Zhao
- Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yuting You
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Hao Zhou
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaolin Su
- Departments of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jiangwei Wang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Yaomin Chen
- Indiana University Health Pathology Laboratory, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
- Center for Computational Biology and Bioinformatics, Indiana University, School of Medicine, Indianapolis, IN, United States
| | - Xiumei Huang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
31
|
Torrente L, DeNicola GM. Targeting NRF2 and Its Downstream Processes: Opportunities and Challenges. Annu Rev Pharmacol Toxicol 2021; 62:279-300. [PMID: 34499527 DOI: 10.1146/annurev-pharmtox-052220-104025] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The transcription factor NRF2 coordinates the expression of a vast array of cytoprotective and metabolic genes in response to various stress inputs to restore cellular homeostasis. Transient activation of NRF2 in healthy tissues has been long recognized as a cellular defense mechanism and is critical to prevent cancer initiation by carcinogens. However, cancer cells frequently hijack the protective capability of NRF2 to sustain the redox balance and meet their metabolic requirements for proliferation. Further, aberrant activation of NRF2 in cancer cells confers resistance to commonly used chemotherapeutic agents and radiotherapy. During the last decade, many research groups have attempted to block NRF2 activity in tumors to counteract the survival and proliferative advantage of cancer cells and reverse resistance to treatment. In this review, we highlight the role of NRF2 in cancer progression and discuss the past and current approaches to disable NRF2 signaling in tumors. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Laura Torrente
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA;
| | - Gina M DeNicola
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA;
| |
Collapse
|
32
|
Abstract
The gene expression program induced by NRF2 transcription factor plays a critical role in cell defense responses against a broad variety of cellular stresses, most importantly oxidative stress. NRF2 stability is fine-tuned regulated by KEAP1, which drives its degradation in the absence of oxidative stress. In the context of cancer, NRF2 cytoprotective functions were initially linked to anti-oncogenic properties. However, in the last few decades, growing evidence indicates that NRF2 acts as a tumor driver, inducing metastasis and resistance to chemotherapy. Constitutive activation of NRF2 has been found to be frequent in several tumors, including some lung cancer sub-types and it has been associated to the maintenance of a malignant cell phenotype. This apparently contradictory effect of the NRF2/KEAP1 signaling pathway in cancer (cell protection against cancer versus pro-tumoral properties) has generated a great controversy about its functions in this disease. In this review, we will describe the molecular mechanism regulating this signaling pathway in physiological conditions and summarize the most important findings related to the role of NRF2/KEAP1 in lung cancer. The focus will be placed on NRF2 activation mechanisms, the implication of those in lung cancer progression and current therapeutic strategies directed at blocking NRF2 action.
Collapse
|
33
|
Gong Q, Yang F, Hu J, Li T, Wang P, Li X, Zhang X. Rational designed highly sensitive NQO1-activated near-infrared fluorescent probe combined with NQO1 substrates in vivo: An innovative strategy for NQO1-overexpressing cancer theranostics. Eur J Med Chem 2021; 224:113707. [PMID: 34303080 DOI: 10.1016/j.ejmech.2021.113707] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/05/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022]
Abstract
Since NQO1 is overexpressed in many cancer cells, it can be used as a biomarker for cancer diagnosis and targeted therapy. NQO1 substrates show potent anticancer activity through the redox cycle mediated by NQO1, while the NQO1 probes can monitor NQO1 levels in cancers. High sensitivity of probes is needed for diagnostic imaging in clinic. In this study, based on the analysis of NQO1 catalytic pocket, the naphthoquinone trigger group 13 rationally designed by expanding the aromatic plane of the benzoquinone trigger group 10 shows significantly increased sensitivity to NQO1. The sensitivity of the naphthoquinone trigger group-based probe A was eight times higher than that of benzoquinone trigger group-based probe B in vivo. Probe A was selectively and efficiently sensitive to NQO1 with good safety profile and plasma stability, enabling its combination with NQO1 substrates in vivo for NQO1-overexpressing cancer theranostics for the first time.
Collapse
Affiliation(s)
- Qijie Gong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Fulai Yang
- Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiabao Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China
| | - Tian Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Pengfei Wang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiang Li
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing, 211198, China.
| | - Xiaojin Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Laboratory of Drug Design and Discovery, Department of Chemistry, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
34
|
Walia HK, Singh N, Sharma S. Association of NQO1Pro187Ser polymorphism with clinical outcomes and survival of lung cancer patients treated with platinum chemotherapy. Per Med 2021; 18:333-346. [PMID: 33973803 DOI: 10.2217/pme-2020-0119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 03/23/2021] [Indexed: 02/02/2023]
Abstract
Background: The study was carried out to evaluate the association of NQO1 P187S polymorphism in North Indian lung cancer (LC) patients. We determined the effect of this polymorphic variant on the survival of LC patients. Patients & methods/results: This case-control study comprised a total of 1100 subjects. The genotyping was carried out using PCR-RFLP and statistical analysis was carried out. The variant TT genotype exhibited 3.5-fold higher odds in subjects with stage III (p = 0.0006), fivefold higher odds of lymph-node invasion (p = 0.007) and an odd of <1 in case of metastasis (p = 0.0028). Patients possessing TT genotype and administered with paclitaxel, exhibited a poor survival (3.57 vs 12.20 months; hazard ratio = 7.95; p = 0.0098). Conclusion: These results suggest that NQO1 variant genotype was not found to modulate risk toward LC. However, the variant genotype was found to be strongly correlated with stage III LC, lymph node invasion and was found to be positively correlating with metastasis.
Collapse
Affiliation(s)
- Harleen Kaur Walia
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| | - Navneet Singh
- Department of Pulmonary Medicine, Post Graduate Institute of Medical Education & Research (PGIMER), Chandigarh, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, India
| |
Collapse
|
35
|
Jiang ZN, Ahmed SMU, Wang QC, Shi HF, Tang XW. Quinone oxidoreductase 1 is overexpressed in gastric cancer and associated with outcome of adjuvant chemotherapy and survival. World J Gastroenterol 2021; 27:3085-3096. [PMID: 34168410 PMCID: PMC8192289 DOI: 10.3748/wjg.v27.i22.3085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/28/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Quinine oxidoreductase 1 (NQO1) plays a vital role in protecting normal cells against oxidative damage and electrophilic attack. It is highly expressed in many solid tumors, suggesting a role in cancer development and progression. However, the role of NQO1 in gastric cancer and its effect on cancer development and prognosis have not been fully investigated.
AIM To investigate the clinical relevance of NQO1 protein expression in gastric cancer and to explore the potential of NQO1 to serve as a prognostic biomarker and therapeutic target.
METHODS In this retrospective study, gastric cancer specimens of 175 patients who were treated between 1995 and 2011 were subjected to immunohistochemistry analyses for NQO1. The correlation of NQO1 expression with gastric cancer prognosis and clinical and pathological parameters was investigated.
RESULTS NQO1 protein was overexpressed in 59.43% (104/175) of the analyzed samples. Overexpression of NQO1 was associated with a significantly inferior prognosis. In addition, multivariate analysis suggested that NQO1 overexpression, along with tumor stage and patient age, are prominent prognostic biomarkers for gastric cancer. Moreover, NQO1 overexpression was correlated to a better response to 5-fluorouracil (5-FU)-based adjuvant chemotherapy.
CONCLUSION NQO1 overexpression is associated with a significantly poor prognosis and better response to 5-FU in patients with gastric cancer. These findings are relevant for improving therapeutic approaches for gastric cancer patients.
Collapse
Affiliation(s)
- Zhi-Nong Jiang
- Department of Pathology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang Province, China
| | - Syed Minhaj Uddin Ahmed
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang Province, China
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qin-Chuan Wang
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, Zhejiang Province, China
| | - Hong-Fei Shi
- Department of Pathology, The Forth Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 322000, Zhejiang Province, China
| | - Xiu-Wen Tang
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang Province, China
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
36
|
STAT1 potentiates oxidative stress revealing a targetable vulnerability that increases phenformin efficacy in breast cancer. Nat Commun 2021; 12:3299. [PMID: 34083537 PMCID: PMC8175605 DOI: 10.1038/s41467-021-23396-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 04/28/2021] [Indexed: 12/14/2022] Open
Abstract
Bioenergetic perturbations driving neoplastic growth increase the production of reactive oxygen species (ROS), requiring a compensatory increase in ROS scavengers to limit oxidative stress. Intervention strategies that simultaneously induce energetic and oxidative stress therefore have therapeutic potential. Phenformin is a mitochondrial complex I inhibitor that induces bioenergetic stress. We now demonstrate that inflammatory mediators, including IFNγ and polyIC, potentiate the cytotoxicity of phenformin by inducing a parallel increase in oxidative stress through STAT1-dependent mechanisms. Indeed, STAT1 signaling downregulates NQO1, a key ROS scavenger, in many breast cancer models. Moreover, genetic ablation or pharmacological inhibition of NQO1 using β-lapachone (an NQO1 bioactivatable drug) increases oxidative stress to selectively sensitize breast cancer models, including patient derived xenografts of HER2+ and triple negative disease, to the tumoricidal effects of phenformin. We provide evidence that therapies targeting ROS scavengers increase the anti-neoplastic efficacy of mitochondrial complex I inhibitors in breast cancer. Complex I inhibition induces oxidative stress leading to cancer cell cytotoxicity. Here, the authors show, in breast cancer models, that inflammatory mediators can potentiate complex I inhibitor phenformin cytotoxicity through STAT1-mediated downregulation of the reactive oxygen species scavenger NQO1.
Collapse
|
37
|
Wu Z, Wang Q, Yang H, Wang J, Li W, Liu G, Yang Y, Zhao Y, Tang Y. Discovery of Natural Products Targeting NQO1 via an Approach Combining Network-Based Inference and Identification of Privileged Substructures. J Chem Inf Model 2021; 61:2486-2498. [PMID: 33955748 DOI: 10.1021/acs.jcim.1c00260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
NAD(P)H:quinone oxidoreductase 1 (NQO1) has been shown to be a potential therapeutic target for various human diseases, such as cancer and neurodegenerative disorders. Recent advances in computational methods, especially network-based methods, have made it possible to identify novel compounds for a target with high efficiency and low cost. In this study, we designed a workflow combining network-based methods and identification of privileged substructures to discover new compounds targeting NQO1 from a natural product library. According to the prediction results, we purchased 56 compounds for experimental validation. Without the assistance of privileged substructures, 31 compounds (31/56 = 55.4%) showed IC50 < 100 μM, and 11 compounds (11/56 = 19.6%) showed IC50 < 10 μM. With the assistance of privileged substructures, the two success rates were increased to 61.8 and 26.5%, respectively. Seven natural products further showed inhibitory activity on NQO1 at the cellular level, which may serve as lead compounds for further development. Moreover, network analysis revealed that osthole may exert anticancer effects against multiple cancer types by inhibiting not only carbonic anhydrases IX and XII but also NQO1. Our workflow and computational methods can be easily applied in other targets and become useful tools in drug discovery and development.
Collapse
Affiliation(s)
- Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Qiaohui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jiye Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yi Yang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuzheng Zhao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China.,Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
38
|
Tan S, Zhang M, Shi X, Ding K, Zhao Q, Guo Q, Wang H, Wu Z, Kang Y, Zhu T, Sun J, Zhao X. CPSF6 links alternative polyadenylation to metabolism adaption in hepatocellular carcinoma progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:85. [PMID: 33648552 PMCID: PMC7923339 DOI: 10.1186/s13046-021-01884-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/16/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Alternative polyadenylation (APA) is an important mechanism of gene expression regulation through generation of RNA isoforms with distinct 3' termini. Increasing evidence has revealed that APA is actively involved in development and disease, including hepatocellular carcinoma (HCC). However, how APA functions in tumor formation and progression remains elusive. In this study, we investigated the role of cleavage factor I (CFIm) subunit CPSF6 in human hepatocellular carcinoma (HCC). METHODS Expression levels of CPSF6 in clinical tissues and cell lines were determined by qRT-PCR and western blot. Functional assays, including the cell number, MTT, colony formation and transwell, were used to determine the oncogenic role of CPSF6 in HCC. Animal experiments were used to determine the role of CPSF6 in HCC tumorigenicity in vivo. Deep sequencing-based 3 T-seq was used to profile the transcriptome-wide APA sites in both HCC cells and CPSF6 knockdown HCC cells. The function of CPSF6-affected target NQO1 with distinct 3'UTRs was characterized by metabolism assays. RESULTS We observed CPSF6 was upregulated in HCC and the high expression of CPSF6 was associated with poor prognosis in patients. Overexpression of CPSF6 promoted proliferation, migration and invasion of HCC cells in vitro and in vivo. Transcriptome-wide APA profiling analysis indicated that high expression of CPSF6 promoted the favorable usage of the proximal poly(A) site in the 3'UTR of NQO1. We demonstrated CPSF6-induced tumorigenic activities were mediated by the NQO1 isoform with short 3'UTR. Furthermore, we found that CPSF6 induced metabolic alterations in liver cells through NQO1. CONCLUSION CPSF6 plays a critical role in HCC progression by upregulating NQO1 expression through APA. These findings provide evidence to demonstrate that APA of NQO1 contributes to HCC progression and may have implications for developing new therapeutic strategy against this disease.
Collapse
Affiliation(s)
- Sheng Tan
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ming Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xinglong Shi
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Keshuo Ding
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Qiang Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qianying Guo
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Hao Wang
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Zhengsheng Wu
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yani Kang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| | - Jielin Sun
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Xiaodong Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
39
|
Huang J, Wang C, Lin M, Zeng F, Wu S. Synthesis of NQO1-activatable Optoacoustic Probe and Its Imaging of Breast Cancer. ACTA CHIMICA SINICA 2021. [DOI: 10.6023/a20100459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Su D, Chen X, Zhang Y, Gao X. Activatable imaging probes for cancer-linked NAD(P)H:quinone oxidoreductase-1 (NQO1): Advances and future prospects. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.116112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Han J, Cheng L, Zhu Y, Xu X, Ge C. Covalent-Assembly Based Fluorescent Probes for Detection of hNQO1 and Imaging in Living Cells. Front Chem 2020; 8:756. [PMID: 33005608 PMCID: PMC7479225 DOI: 10.3389/fchem.2020.00756] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/21/2020] [Indexed: 12/12/2022] Open
Abstract
Human NAD(P)H: quinone oxidoreductase (hNQO1) is an important biomarker for human malignant tumors. Detection of NQO1 accurately is of great significance to improve the early diagnosis of cancer and prognosis of cancer patients. In this study, based on the covalent assembly strategy, hNQO1-activated fluorescent probes 1 and 2 are constructed by introducing coumarin precursor 2-cyano-3-(4-(diethylamino)-2-hydroxyphenyl) acrylic acid and self-immolative linkers. Under reaction with hNQO1 and NADH, turn-on fluorescence appears due to in-situ formation of the organic fluorescent compound 7-diethylamino-3-cyanocoumarin, and fluorescent intensity changes significantly. Probe 1 and 2 for detection of hNQO1 are not interfered by other substances and have low toxicity in cells. In addition to quantitative detection of hNQO1 in vitro, they have also been successfully applied to fluorescent imaging in living cells.
Collapse
Affiliation(s)
| | - Longhao Cheng
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ya Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaowei Xu
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chaoliang Ge
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
42
|
An HBT-based fluorescent dye with enhanced quantum yield in water system and its application for constructing NQO1 fluorescent probe. Talanta 2020; 216:120982. [DOI: 10.1016/j.talanta.2020.120982] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/23/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022]
|
43
|
Therapeutic Strategies and Biomarkers to Modulate PARP Activity for Targeted Cancer Therapy. Cancers (Basel) 2020; 12:cancers12040972. [PMID: 32295316 PMCID: PMC7226473 DOI: 10.3390/cancers12040972] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/31/2020] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
Poly-(ADP-ribose) polymerase 1 (PARP1) is commonly known for its vital role in DNA damage response and repair. However, its enzymatic activity has been linked to a plethora of physiological and pathophysiological transactions ranging from cellular proliferation, survival and death. For instance, malignancies with BRCA1/2 mutations heavily rely on PARP activity for survival. Thus, the use of PARP inhibitors is a well-established intervention in these types of tumors. However, recent studies indicate that the therapeutic potential of attenuating PARP1 activity in recalcitrant tumors, especially where PARP1 is aberrantly overexpressed and hyperactivated, may extend its therapeutic utility in wider cancer types beyond BRCA-deficiency. Here, we discuss treatment strategies to expand the tumor-selective therapeutic application of PARP inhibitors and novel approaches with predictive biomarkers to perturb NAD+ levels and hyperPARylation that inactivate PARP in recalcitrant tumors. We also provide an overview of genetic alterations that transform non-BRCA mutant cancers to a state of "BRCAness" as potential biomarkers for synthetic lethality with PARP inhibitors. Finally, we discuss a paradigm shift for the use of novel PARP inhibitors outside of cancer treatment, where it has the potential to rescue normal cells from severe oxidative damage during ischemia-reperfusion injury induced by surgery and radiotherapy.
Collapse
|
44
|
Torrente L, Prieto-Farigua N, Falzone A, Elkins CM, Boothman DA, Haura EB, DeNicola GM. Inhibition of TXNRD or SOD1 overcomes NRF2-mediated resistance to β-lapachone. Redox Biol 2020; 30:101440. [PMID: 32007910 PMCID: PMC6997906 DOI: 10.1016/j.redox.2020.101440] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/10/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Alterations in the NRF2/KEAP1 pathway result in the constitutive activation of NRF2, leading to the aberrant induction of antioxidant and detoxification enzymes, including NQO1. The NQO1 bioactivatable agent β-lapachone can target cells with high NQO1 expression but relies in the generation of reactive oxygen species (ROS), which are actively scavenged in cells with NRF2/KEAP1 mutations. However, whether NRF2/KEAP1 mutations influence the response to β-lapachone treatment remains unknown. To address this question, we assessed the cytotoxicity of β-lapachone in a panel of NSCLC cell lines bearing either wild-type or mutant KEAP1. We found that, despite overexpression of NQO1, KEAP1 mutant cells were resistant to β-lapachone due to enhanced detoxification of ROS, which prevented DNA damage and cell death. To evaluate whether specific inhibition of the NRF2-regulated antioxidant enzymes could abrogate resistance to β-lapachone, we systematically inhibited the four major antioxidant cellular systems using genetic and/or pharmacologic approaches. We demonstrated that inhibition of the thioredoxin-dependent system or copper-zinc superoxide dismutase (SOD1) could abrogate NRF2-mediated resistance to β-lapachone, while depletion of catalase or glutathione was ineffective. Interestingly, inhibition of SOD1 selectively sensitized KEAP1 mutant cells to β-lapachone exposure. Our results suggest that NRF2/KEAP1 mutational status might serve as a predictive biomarker for response to NQO1-bioactivatable quinones in patients. Further, our results suggest SOD1 inhibition may have potential utility in combination with other ROS inducers in patients with KEAP1/NRF2 mutations.
Collapse
Affiliation(s)
- Laura Torrente
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Nicolas Prieto-Farigua
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Aimee Falzone
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Cody M Elkins
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - David A Boothman
- Department of Biochemistry and Molecular Biology, Simon Cancer Center Indiana, University School of Medicine, Indianapolis, IN, 46202, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Gina M DeNicola
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
45
|
Dimri M, Humphries A, Laknaur A, Elattar S, Lee TJ, Sharma A, Kolhe R, Satyanarayana A. NAD(P)H Quinone Dehydrogenase 1 Ablation Inhibits Activation of the Phosphoinositide 3-Kinase/Akt Serine/Threonine Kinase and Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Pathways and Blocks Metabolic Adaptation in Hepatocellular Carcinoma. Hepatology 2020; 71:549-568. [PMID: 31215069 PMCID: PMC6920612 DOI: 10.1002/hep.30818] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/13/2019] [Indexed: 12/12/2022]
Abstract
Cancer cells undergo metabolic adaptation to sustain uncontrolled proliferation. Aerobic glycolysis and glutaminolysis are two of the most essential characteristics of cancer metabolic reprogramming. Hyperactivated phosphoinositide 3-kinase (PI3K)/Akt serine/threonine kinase (Akt) and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathways play central roles in cancer cell metabolic adaptation given that their downstream effectors, such as Akt and c-Myc, control most of the glycolytic and glutaminolysis genes. Here, we report that the cytosolic flavoprotein, NAD(P)H quinone dehydrogenase 1 (Nqo1), is strongly overexpressed in mouse and human hepatocellular carcinoma (HCC). Knockdown of Nqo1 enhanced activity of the serine/threonine phosphatase, protein phosphatase 2A, which operates at the intersection of the PI3K/Akt and MAPK/ERK pathways and dephosphorylates and inactivates pyruvate dehydrogenase kinase 1, Akt, Raf, mitogen-activated protein kinase kinase, and ERK1/2. Nqo1 ablation also induced the expression of phosphatase and tensin homolog, a dual protein/lipid phosphatase that blocks PI3K/Akt signaling, through the ERK/cAMP-responsive element-binding protein/c-Jun pathway. Together, Nqo1 ablation triggered simultaneous inhibition of the PI3K/Akt and MAPK/ERK pathways, suppressed the expression of glycolysis and glutaminolysis genes and blocked metabolic adaptation in liver cancer cells. Conversely, Nqo1 overexpression caused hyperactivation of the PI3K/Akt and MAPK/ERK pathways and promoted metabolic adaptation. Conclusion: In conclusion, Nqo1 functions as an upstream activator of both the PI3K/Akt and MAPK/ERK pathways in liver cancer cells, and Nqo1 ablation blocked metabolic adaptation and inhibited liver cancer cell proliferation and HCC growth in mice. Therefore, our results suggest that Nqo1 may function as a therapeutic target to inhibit liver cancer cell proliferation and inhibit HCC.
Collapse
Affiliation(s)
- Manali Dimri
- Department of Biochemistry and Molecular Biology, Molecular Oncology & Biomarkers Program, Georgia Cancer Center, Augusta University, Room-CN3150, 1410 Laney Walker Blvd., Augusta, GA 30912
| | - Ashley Humphries
- Department of Biochemistry and Molecular Biology, Molecular Oncology & Biomarkers Program, Georgia Cancer Center, Augusta University, Room-CN3150, 1410 Laney Walker Blvd., Augusta, GA 30912
| | - Archana Laknaur
- Department of Biochemistry and Molecular Biology, Molecular Oncology & Biomarkers Program, Georgia Cancer Center, Augusta University, Room-CN3150, 1410 Laney Walker Blvd., Augusta, GA 30912
| | - Sawsan Elattar
- Department of Biochemistry and Molecular Biology, Molecular Oncology & Biomarkers Program, Georgia Cancer Center, Augusta University, Room-CN3150, 1410 Laney Walker Blvd., Augusta, GA 30912
| | - Tae Jin Lee
- Center for Biotechnology and Genomic Medicine, Department of Population Health Sciences, Augusta University, GA, 30912
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Department of Population Health Sciences, Augusta University, GA, 30912
| | - Ravindra Kolhe
- Department of Pathology, Section of Anatomic Pathology, Augusta University, Augusta, GA 30912
| | - Ande Satyanarayana
- Department of Biochemistry and Molecular Biology, Molecular Oncology & Biomarkers Program, Georgia Cancer Center, Augusta University, Room-CN3150, 1410 Laney Walker Blvd., Augusta, GA 30912
| |
Collapse
|
46
|
Attenuation of NAD[P]H:quinone oxidoreductase 1 aggravates prostate cancer and tumor cell plasticity through enhanced TGFβ signaling. Commun Biol 2020; 3:12. [PMID: 31909204 PMCID: PMC6941961 DOI: 10.1038/s42003-019-0720-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/04/2019] [Indexed: 12/30/2022] Open
Abstract
NAD[P]H:quinone oxidoreductase 1 (NQO1) regulates cell fate decisions in response to stress. Oxidative stress supports cancer maintenance and progression. Previously we showed that knockdown of NQO1 (NQO1low) prostate cancer cells upregulate pro-inflammatory cytokines and survival under hormone-deprived conditions. Here, we tested the ability of NQO1low cells to form tumors. We found NQO1low cells form aggressive tumors compared with NQO1high cells. Biopsy specimens and circulating tumor cells showed biochemical recurrent prostate cancer was associated with low NQO1. NQO1 silencing was sufficient to induce SMAD-mediated TGFβ signaling and mesenchymal markers. TGFβ treatment decreased NQO1 levels and induced molecular changes similar to NQO1 knockdown cells. Functionally, NQO1 depletion increased migration and sensitivity to oxidative stress. Collectively, this work reveals a possible new gatekeeper role for NQO1 in counteracting cellular plasticity in prostate cancer cells. Further, combining NQO1 with TGFβ signaling molecules may serve as a better signature to predict biochemical recurrence. Thapa et al find that depletion of the antioxidant enzyme NAD[P]H:Quinone Oxidoreductase 1 (NQO1) accelerates prostate tumorigenesis and induces the epithelial-to-mesenchymal transition by activating TGFβ signaling. They also find that low NQO1 is associated with mesenchymal signature and biochemical recurrence in clinical samples.
Collapse
|
47
|
NQO1 potentiates apoptosis evasion and upregulates XIAP via inhibiting proteasome-mediated degradation SIRT6 in hepatocellular carcinoma. Cell Commun Signal 2019; 17:168. [PMID: 31842909 PMCID: PMC6915971 DOI: 10.1186/s12964-019-0491-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/26/2019] [Indexed: 12/11/2022] Open
Abstract
Background Our previous study has demonstrated that NAD(P)H: quinone oxidoreductase 1 (NQO1) is significantly upregulated in human liver cancer where it potentiates the apoptosis evasion of liver cancer cell. However, the underlying mechanisms of the oncogenic function of NQO1 in HCC have not been fully elucidated. Methods Expression of NQO1, SIRT6, AKT and X-linked inhibitor of apoptosis protein (XIAP) protein were measured by western blotting and immunohistochemistry. Additionally, the interaction between NQO1 and potential proteins were determined by immunoprecipitation assays. Furthermore, the effect of NQO1 and SIRT6 on tumor growth was determined in cell model and orthotopic tumor implantation model. Results We found that NQO1 overexpression in HCC enhanced SIRT6 protein stability via inhibiting ubiquitin-mediated 26S proteasome degradation. High level of SIRT6 reduced acetylation of AKT which resulted in increased phosphorylation and activity of AKT. Activated AKT subsequently phosphorylated anti-apoptotic protein XIAP at Ser87 which determined its protein stability. Reintroduction of SIRT6 or AKT efficiently rescued NQO1 knock-out-mediated inhibition of growth and induction of apoptosis. In orthotopic mouse model, NQO1 knock-out inhibited tumor growth and induced apoptosis while this effect was effectively rescued by SIRT6 overexpression or MG132 treatment partially. Conclusions Collectively, these results reveal an oncogenic function of NQO1 in sustaining HCC cell proliferation through SIRT6/AKT/XIAP signaling pathway.
Collapse
|
48
|
Zaboli M, Zaboli M, Torkzadeh-Mahani M. From in vitro to in silico: Modeling and recombinant production of DT-Diaphorase enzyme. Int J Biol Macromol 2019; 143:213-223. [PMID: 31812741 DOI: 10.1016/j.ijbiomac.2019.12.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/20/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
DT-Diaphorase (DTD) belonging to the oxidoreductase family, is among the most important enzymes and is of great significance in present-day biotechnology. Also, it has potential applications in glucose and pyruvate biosensors. Another important role of the DTD enzyme is in the detection of Phenylketonuria disease. According to the above demands, at first, we tried to study molecular cloning and production of recombinant DTD in E. coli BL21 strain. We have successfully cloned, expressed, and purified functionally active diaphorase. The amount of enzyme was increased in 10-h using IPTG induction, and the recombinant protein was purified by Ni-NTA agarose affinity chromatography. After that, the kinetic and thermodynamic parameters of the enzyme, optimum temperature and pH were also investigated to find more in-depth information. In the end, to represent the connections between the structures and function of this enzyme, the molecular dynamics simulations have been considered at two temperatures in which DTD had maximum and minimum activity (310 and 293 K, respectively). The results of MD simulations indicated that the interaction between NADH with phenylalanine 232 residue at 310 K is more severe than other residues. So, to investigate the interaction details of NADH/PHE 232 the DFT calculations were done.
Collapse
Affiliation(s)
- Mahdiye Zaboli
- Department of Biotechnology, Institute of Science, High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | - Maryam Zaboli
- Department of Chemistry, Faculty of Science, University of Birjand, Birjand, Iran
| | - Masoud Torkzadeh-Mahani
- Department of Biotechnology, Institute of Science, High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran.
| |
Collapse
|
49
|
Chen T, Zhu J, Cai T, Du W, Zhang Y, Zhu Q, Liu Z, Huang JA. Suppression of non-small cell lung cancer migration and invasion by hsa-miR-486-5p via the TGF-β/SMAD2 signaling pathway. J Cancer 2019; 10:6014-6024. [PMID: 31762811 PMCID: PMC6856587 DOI: 10.7150/jca.35017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/18/2019] [Indexed: 12/13/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide. SMAD family member 2 (SMAD2) is a key element downstream of the transforming growth factor beta (TGF-β) signaling pathway that regulates cancer metastasis by promoting the epithelial-mesenchyme transition (EMT). MicroRNA miR-486-5p is a tumor suppressor in NSCLC progression. However, it remains unclear whether miR-486-5p is implicated in TGF-β signaling and EMT in NSCLC. In the present study, high expression of SMAD2 mRNA was detected in NSCLC tissues and cell lines, and was associated with poor survival of patients with NSCLC. By contrast, miR-486-5p was downregulated in NSCLC tissues and cell lines. In silico prediction showed that SMAD2 was a potential target of miR-486-5p. The prediction was verified using a dual-luciferase reporter assay. Transwell assays showed that knockdown of SMAD2 inhibited TGF-β-induced EMT and migration and invasion in NSCLC cells. Similarly, miR-486-5p overexpression suppressed TGF-β-induced EMT and migration and invasion of NSCLC cells. The present study provides a new insight into the role of miR-486-5p in regulating TGF-β-mediated EMT and invasion in NSCLC.
Collapse
Affiliation(s)
- Tao Chen
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Jianjie Zhu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China.,Institute of Respiratory Diseases, Soochow University, Suzhou 215006, China
| | - Tingting Cai
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Wenwen Du
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Yang Zhang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Qingqing Zhu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China
| | - Zeyi Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China.,Institute of Respiratory Diseases, Soochow University, Suzhou 215006, China
| | - Jian-An Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou 215006, China.,Institute of Respiratory Diseases, Soochow University, Suzhou 215006, China
| |
Collapse
|
50
|
Wang Q, Xin Y, Pa B, He X, Bao W. Synthesis and antitumor activity of sacroflavonoside. Nat Prod Res 2019; 35:1292-1298. [PMID: 31359788 DOI: 10.1080/14786419.2019.1645660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Sacroflavonoside, a new derivative of diphenylethene, was isolated from Artemisia sacrorum, which have been found to possess the inhibitory effect on the proliferation of gastric carcinoma cells (MKN-45) in vitro in our previous studies. With anisaldehyde (SM-A) as starting material, the sacroflavonoside was synthesized by nucleophilic addition, electrophilic substitution and dehydration cyclization. The structure of sacroflavonoside was established by 1 D (1H NMR and 13C NMR) and 2 D-NMR (HSQC and HMBC) spectral analysis. The antitumor activity and potential mechanism against MKN-45 cells of sacroflavonoside were evaluated in vitro. The results showed that sacroflavonoside could significantly induce MKN-45 cells apoptosis and autophagy by increasing the expression of Bax, Caspase-3, Beclin1 and LC3-II proteins and decreasing the expression of Bcl-2 protein at low micromole level. This investigation provided a valuable lead structure for the development of antitumor drugs.
Collapse
Affiliation(s)
- Qinghu Wang
- College of Traditional Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China
| | - Ying Xin
- College of Traditional Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China
| | - Biligetu Pa
- College of Traditional Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China
| | - Xiang He
- College of Traditional Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China
| | - Wenqiang Bao
- College of Traditional Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia, China
| |
Collapse
|