1
|
Meerovich G, Kogan E, Romanishkin I, Zharkov N, Avraamova S, Shchelokova E, Akhlyustina E, Strakhovskaya M, Meerovich I, Demura S, Tiganova I, Romanova Y, Chen ZL, Reshetov I. Potential of photodynamic therapy using polycationic photosensitizers in the treatment of lung cancer patients with SARS-CoV-2 infection and bacterial complications: Our recent experience. Photodiagnosis Photodyn Ther 2025; 51:104447. [PMID: 39681293 DOI: 10.1016/j.pdpdt.2024.104447] [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: 09/30/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
The problem of treating cancer patients with lung cancer has become more difficult due to the SARS-CoV-2 viral infection and concomitant bacterial lesions. The analysis shows that the photodynamic effect of long-wavelength polycationic photosensitizers suppresses the tumor process (including the destruction of cancer stem cells), SARS-CoV-2 coronavirus infection, Gram-positive and Gram-negative bacteria, including those that can cause pneumonia. Therefore, the photodynamic approach using such photosensitizers is promising for the development of an effective treatment method for patients with lung cancer, including those with SARS-CoV-2 infection and bacterial complications.
Collapse
Affiliation(s)
- Gennady Meerovich
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia; Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| | - Evgeniya Kogan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Igor Romanishkin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Moscow 119991, Russia.
| | - Nikolay Zharkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Sofiya Avraamova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Elena Shchelokova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119992, Russia
| | - Ekaterina Akhlyustina
- Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| | | | - Irina Meerovich
- Research Center of Biotechnology of the Russian Academy of Sciences, A.N. Bach Institute of Biochemistry, Moscow 119071, Russia
| | - Sofya Demura
- Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| | - Irina Tiganova
- Lomonosov Moscow State University, Moscow 119991, Russia
| | - Yulia Romanova
- Lomonosov Moscow State University, Moscow 119991, Russia
| | - Zhi-Long Chen
- Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Igor Reshetov
- Institute for Physics and Engineering in Biomedicine, National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), Moscow 115409, Russia
| |
Collapse
|
2
|
Yue P, He Y, Zuo R, Gong W, Wang Y, Chen L, Luo Y, Feng Y, Gao Y, Liu Z, Chen P, Guo H. CCDC34 maintains stemness phenotype through β-catenin-mediated autophagy and promotes EGFR-TKI resistance in lung adenocarcinoma. Cancer Gene Ther 2025; 32:104-121. [PMID: 39587349 DOI: 10.1038/s41417-024-00843-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/22/2024] [Accepted: 09/30/2024] [Indexed: 11/27/2024]
Abstract
Despite recent advances in treatment strategy, lung cancer remains the leading cause of cancer-related mortality worldwide, and it is a serious threat to human health. Lung adenocarcinoma (LUAD) is the most common histological type of lung cancer, and approximately 40-50% of patients with LUAD in Asian populations have epidermal growth factor receptor (EGFR) mutations. The use of EGFR tyrosine kinase inhibitors (EGFR-TKIs) has revolutionarily improved the prognosis of patients with EGFR-mutated LUAD. However, acquired drug resistance is the main cause of treatment failure. Therefore, new therapeutic strategies are necessary to address the resistance to EGFR-TKIs in patients with LUAD. Cancer stemness-related factors lead to multiple-drug resistance in cancer treatment, including EGFR-TKI resistance. Coiled-coil domain-containing 34 (CCDC34) serves as an oncogene in several types of cancer. However, the role and molecular mechanism of CCDC34 in the malignant progression of LUAD have not been reported to date. In the present study, we found that CCDC34 may be associated with LUAD stemness through weighted gene co-expression network analysis (WGCNA). Furthermore, we demonstrated that CCDC34 promoted LUAD stemness properties through β-catenin-mediated regulation of ATG5-induced autophagy, which was conducive to acquired EGFR-TKI resistance in LUAD in vitro and in vivo. Knockdown CCDC34 can synergistically inhibit tumor growth when combined with EGFR-TKIs. This study reveals a positive association between CCDC34 and the stemness phenotype of LUAD, providing new insights into overcoming EGFR-TKI resistance in LUAD by inhibiting CCDC34 expression.
Collapse
Affiliation(s)
- Ping Yue
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuchao He
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Ran Zuo
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Integrated Chinese and Western Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Wenchen Gong
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
| | - Yu Wang
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Liwei Chen
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Yi Luo
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Yuanying Feng
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Yuan Gao
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Zhiyong Liu
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| | - Peng Chen
- Department of Thoracic Oncology, Lung Cancer Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| | - Hua Guo
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| |
Collapse
|
3
|
Lypova N, Dougherty SM, Clem BF, Feng J, Yin X, Zhang X, Li X, Chesney JA, Imbert-Fernandez Y. PFKFB3-dependent redox homeostasis and DNA repair support cell survival under EGFR-TKIs in non-small cell lung carcinoma. Cancer Metab 2024; 12:37. [PMID: 39696407 PMCID: PMC11658331 DOI: 10.1186/s40170-024-00366-y] [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: 04/17/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The efficacy of tyrosine kinase inhibitors (TKIs) targeting the EGFR is limited due to the persistence of drug-tolerant cell populations, leading to therapy resistance. Non-genetic mechanisms, such as metabolic rewiring, play a significant role in driving lung cancer cells into the drug-tolerant state, allowing them to persist under continuous drug treatment. METHODS Our study employed a comprehensive approach to examine the impact of the glycolytic regulator 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB3) on the adaptivity of lung cancer cells to EGFR TKI therapies. We conducted metabolomics to trace glucose rerouting in response to PFKFB3 inhibition during TKI treatment. Live cell imaging and DCFDA oxidation were used to quantify levels of oxidation stress. Immunocytochemistry and Neutral Comet assay were employed to evaluate DNA integrity in response to therapy-driven oxidative stress. RESULTS Our metabolic profiling revealed that PFKFB3 inhibition significantly alters the metabolic profile of TKI-treated cells. It limited glucose utilization in the polyol pathway, glycolysis, and TCA cycle, leading to a depletion of ATP levels. Furthermore, pharmacological inhibition of PFKFB3 overcome TKI-driven redox capacity by diminishing the expression of glutathione peroxidase 4 (GPX4), thereby exacerbating oxidative stress. Our study also unveiled a novel role of PFKFB3 in DNA oxidation and damage by controlling the expression of DNA-glycosylases involved in base excision repair. Consequently, PFKFB3 inhibition improved the cytotoxicity of EGFR-TKIs by facilitating ROS-dependent cell death. CONCLUSIONS Our results suggest that PFKFB3 inhibition reduces glucose utilization and DNA damage repair, limiting the adaptivity of the cells to therapy-driven oxidative stress and DNA integrity insults. Inhibiting PFKFB3 can be an effective strategy to eradicate cancer cells surviving under EGFR TKI therapy before they enter the drug-resistant state. These findings may have potential implications in the development of new therapies for drug-resistant cancer treatment.
Collapse
Affiliation(s)
- Nadiia Lypova
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| | - Susan M Dougherty
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Brian F Clem
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Jing Feng
- Center for Regulatory Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, 40208, USA
- Department of Chemistry, University of Louisville, Louisville, KY, 40208, USA
| | - Xinmin Yin
- Center for Regulatory Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, 40208, USA
- Department of Chemistry, University of Louisville, Louisville, KY, 40208, USA
| | - Xiang Zhang
- Center for Regulatory Environmental Analytical Metabolomics, University of Louisville, Louisville, KY, 40208, USA
- Department of Chemistry, University of Louisville, Louisville, KY, 40208, USA
| | - Xiaohong Li
- Department of Anatomical Sciences and Neurobiology, Bioinformatics Core, University of Louisville, Louisville, KY, 40202, USA
| | - Jason A Chesney
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Yoannis Imbert-Fernandez
- Department of Medicine, School of Medicine, University of Louisville, Louisville, KY, 40202, USA.
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
4
|
Castro MP, Quinn J, Wasserman A, Awawda A, Cole ZD, Shapiro MA, Stuhlmiller TJ, Kesari S. Proton pump inhibitors are detrimental to overall survival of patients with glioblastoma: Results from a nationwide real-world evidence database. Neurooncol Pract 2024; 11:713-722. [PMID: 39554793 PMCID: PMC11567743 DOI: 10.1093/nop/npae044] [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] [Indexed: 11/19/2024] Open
Abstract
Background Proton pump inhibitors (PPIs) are often prescribed to manage corticosteroid-induced gastrointestinal toxicity during glioblastoma (GBM) treatment, but were recently identified as strong inducers of aldehyde dehydrogenase-1A1 (ALDH1A1). ALDH1A1 is a primary metabolic enzyme impacting the outcome of chemotherapy, including temozolomide. High expression of ALDH1A1 is associated with poor prognosis in multiple cancers, suggesting PPIs may have a negative impact on survival. Methods Real-world data on GBM patients was annotated from electronic medical records (EMR) according to the prospective observational study, XCELSIOR (NCT03793088). Patients with known IDH1/2 mutations were excluded. Causal effects on survival were analyzed using a multivariate, time-varying Cox Proportional Hazard (CPH) model with stratifications including MGMT methylation status, age, sex, duration of corticosteroid use, extent of resection, starting standard-of-care, and PPI use. Results EMR data from 554 GBM patients across 225 cancer centers was collected, with 72% of patients receiving care from academic medical centers. Patients treated with PPIs (51%) had numerically lower median overall survival (mOS) and 2-year OS rates in the total population and across most strata, with the greatest difference for MGMT-methylated patients (mOS 29.2 vs. 40.1 months). In a time-varying multivariate CPH analysis of the above strata, PPIs caused an adverse effect on survival (HR 1.67 [95% CI: 1.15-2.44], P = .007). Conclusions Evidence from a nationwide cancer registry has suggested PPIs have a negative impact on OS for GBM patients, particularly those with MGMT promoter methylation. This suggests PPIs should be avoided for prophylactic management of gastrointestinal toxicity in patients with GBM receiving chemoradiotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Santosh Kesari
- xCures Inc., Oakland, California, USA
- Pacific Neuroscience Institute, Providence Saint John’s Health Center, Santa Monica, California, USA
| |
Collapse
|
5
|
Bian Y, Shan G, Bi G, Liang J, Hu Z, Sui Q, Shi H, Zheng Z, Yao G, Wang Q, Fan H, Zhan C. Targeting ALDH1A1 to enhance the efficacy of KRAS-targeted therapy through ferroptosis. Redox Biol 2024; 77:103361. [PMID: 39317105 PMCID: PMC11465744 DOI: 10.1016/j.redox.2024.103361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024] Open
Abstract
KRAS is among the most commonly mutated oncogenes in human malignancies. Although the advent of sotorasib and adagrasib, has lifted the "undruggable" stigma of KRAS, the resistance to KRAS inhibitors quickly becomes a major issue. Here, we reported that aldehyde dehydrogenase 1 family member A1 (ALDH1A1), an enzyme in retinoic acid biosynthesis and redox balance, increases in response to KRAS inhibitors and confers resistance in a range of cancer types. KRAS inhibitors' efficacy is significantly improved in sensitive or drug-resistant cells, patient-derived organoids (PDO), and xenograft models by ALDH1A1 knockout, loss of enzyme function, or inhibitor. Furthermore, we discovered that ALDH1A1 suppresses the efficacy of KRAS inhibitors by counteracting ferroptosis. ALDH1A1 detoxicates deleterious aldehydes, boosts the synthesis of NADH and retinoic acid (RA), and improves RARA function. ALDH1A1 also activates the CREB1/GPX4 pathway, stimulates the production of lipid droplets in a pH-dependent manner, and subsequently prevents ferroptosis induced by KRAS inhibitors. Meanwhile, we established that GTF2I is dephosphorylated at S784 via ERK by KRAS inhibitors, which hinders its nuclear translocation and mediates ALDH1A1's upregulation in response to KRAS inhibitors. In summary, the results offer valuable insights into targeting ALDH1A1 to enhance the effectiveness of KRAS-targeted therapy through ferroptosis in cancer treatment.
Collapse
Affiliation(s)
- Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhengyang Hu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qihai Sui
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haochun Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaolin Zheng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangyu Yao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Thoracic Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China.
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Lin W, Wang X, Diao M, Wang Y, Zhao R, Chen J, Liao Y, Long Q, Meng Y. Promoting reactive oxygen species accumulation to overcome tyrosine kinase inhibitor resistance in cancer. Cancer Cell Int 2024; 24:239. [PMID: 38982494 PMCID: PMC11234736 DOI: 10.1186/s12935-024-03418-x] [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: 09/16/2023] [Accepted: 06/22/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND In tumor treatment, protein tyrosine kinase inhibitors (TKIs) have been extensively utilized. However, the efficacy of TKI is significantly compromised by drug resistance. Consequently, finding an effective solution to overcome TKI resistance becomes crucial. Reactive oxygen species (ROS) are a group of highly active molecules that play important roles in targeted cancer therapy including TKI targeted therapy. In this review, we concentrate on the ROS-associated mechanisms of TKI lethality in tumors and strategies for regulating ROS to reverse TKI resistance in cancer. MAIN BODY Elevated ROS levels often manifest during TKI therapy in cancers, potentially causing organelle damage and cell death, which are critical to the success of TKIs in eradicating cancer cells. However, it is noteworthy that cancer cells might initiate resistance pathways to shield themselves from ROS-induced damage, leading to TKI resistance. Addressing this challenge involves blocking these resistance pathways, for instance, the NRF2-KEAP1 axis and protective autophagy, to promote ROS accumulation in cells, thereby resensitizing drug-resistant cancer cells to TKIs. Additional effective approaches inducing ROS generation within drug-resistant cells and providing exogenous ROS stimulation. CONCLUSION ROS play pivotal roles in the eradication of tumor cells by TKI. Harnessing the accumulation of ROS to overcome TKI resistance is an effective and widely applicable approach.
Collapse
Affiliation(s)
- Wei Lin
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojun Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Mingxin Diao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Yangwei Wang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Rong Zhao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China
| | - Jiaping Chen
- Department of Cardiothoracic Surgery, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming, Yunnan, China
| | - Yongde Liao
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China.
| | - Qinghong Long
- Department of Internal Medicine, Renmin Hospital, Wuhan University, Wuhan, 430022, China.
| | - Yunchong Meng
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue, Jianghan District, Wuhan, Hubei, 430022, P.R. China.
| |
Collapse
|
7
|
Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther 2024; 9:170. [PMID: 38965243 PMCID: PMC11224386 DOI: 10.1038/s41392-024-01851-y] [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: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/28/2024] [Indexed: 07/06/2024] Open
Abstract
Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity. Cancer continues to be a significant global disease burden. In the past, surgery, radiotherapy, and chemotherapy were the main cancer treatments. The technology of cancer treatments continues to develop and advance, and the emergence of targeted therapy, and immunotherapy provides more options for patients to a certain extent. However, the limitations of efficacy and treatment resistance are still inevitable. Our review begins with a brief introduction of the historical discoveries, original hypotheses, and pathways that regulate CSCs, such as WNT/β-Catenin, hedgehog, Notch, NF-κB, JAK/STAT, TGF-β, PI3K/AKT, PPAR pathway, and their crosstalk. We focus on the role of CSCs in various therapeutic outcomes and resistance, including how the treatments affect the content of CSCs and the alteration of related molecules, CSCs-mediated therapeutic resistance, and the clinical value of targeting CSCs in patients with refractory, progressed or advanced tumors. In summary, CSCs affect therapeutic efficacy, and the treatment method of targeting CSCs is still difficult to determine. Clarifying regulatory mechanisms and targeting biomarkers of CSCs is currently the mainstream idea.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunqi Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhuofan Zhai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jie Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
8
|
Kucinska M, Pospieszna J, Tang J, Lisiak N, Toton E, Rubis B, Murias M. The combination therapy using tyrosine kinase receptors inhibitors and repurposed drugs to target patient-derived glioblastoma stem cells. Biomed Pharmacother 2024; 176:116892. [PMID: 38876048 DOI: 10.1016/j.biopha.2024.116892] [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: 03/13/2024] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024] Open
Abstract
The lesson from many studies investigating the efficacy of targeted therapy in glioblastoma (GBM) showed that a future perspective should be focused on combining multiple target treatments. Our research aimed to assess the efficacy of drug combinations against glioblastoma stem cells (GSCs). Patient-derived cells U3042, U3009, and U3039 were obtained from the Human Glioblastoma Cell Culture resource. Additionally, the study was conducted on a GBM commercial U251 cell line. Gene expression analysis related to receptor tyrosine kinases (RTKs), stem cell markers and genes associated with significant molecular targets was performed, and selected proteins encoded by these genes were assessed using the immunofluorescence and flow cytometry methods. The cytotoxicity studies were preceded by analyzing the expression of specific proteins that serve as targets for selected drugs. The cytotoxicity study using the MTS assay was conducted to evaluate the effects of selected drugs/candidates in monotherapy and combinations. The most cytotoxic compounds for U3042 cells were Disulfiram combined with Copper gluconate (DSF/Cu), Dacomitinib, and Foretinib with IC50 values of 52.37 nM, 4.38 µM, and 4.54 µM after 24 h incubation, respectively. Interactions were assessed using SynergyFinder Plus software. The analysis enabled the identification of the most effective drug combinations against patient-derived GSCs. Our findings indicate that the most promising drug combinations are Dacomitinib and Foretinib, Dacomitinib and DSF/Cu, and Foretinib and AZD3759. Since most tested combinations have not been previously examined against glioblastoma stem-like cells, these results can shed new light on designing the therapeutic approach to target the GSC population.
Collapse
Affiliation(s)
- Malgorzata Kucinska
- Department of Toxicology, Poznan University of Medical Sciences 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Julia Pospieszna
- Department of Toxicology, Poznan University of Medical Sciences 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Jing Tang
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki 00290, Finland.
| | - Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Ewa Toton
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Blazej Rubis
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 3 Rokietnicka Street, Poznan 60-806, Poland.
| | - Marek Murias
- Department of Toxicology, Poznan University of Medical Sciences 3 Rokietnicka Street, Poznan 60-806, Poland.
| |
Collapse
|
9
|
Fan W, Xing Y, Yan S, Liu W, Ning J, Tian F, Wang X, Zhan Y, Luo L, Cao M, Huang J, Cai L. DUSP5 regulated by YTHDF1-mediated m6A modification promotes epithelial-mesenchymal transition and EGFR-TKI resistance via the TGF-β/Smad signaling pathway in lung adenocarcinoma. Cancer Cell Int 2024; 24:208. [PMID: 38872157 DOI: 10.1186/s12935-024-03382-6] [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: 02/16/2024] [Accepted: 05/23/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) patients have a dismal survival rate because of cancer metastasis and drug resistance. The study aims to identify the genes that concurrently modulate EMT, metastasis and EGFR-TKI resistance, and to investigate the underlying regulatory mechanisms. METHODS Cox regression and Kaplan-Meier analyses were applied to identify prognostic oncogenes in LUAD. Gene set enrichment analysis (GSEA) was used to indicate the biological functions of the gene. Wound-healing and Transwell assays were used to detect migratory and invasive ability. EGFR-TKI sensitivity was evaluated by assessing the proliferation, clonogenic survival and metastatic capability of cancer cells with treatment with gefitinib. Methylated RNA immunoprecipitation (MeRIP) and RNA immunoprecipitation (RIP) analyses established the level of m6A modification present on the target gene and the protein's capability to interact with RNA, respectively. Single-sample gene set enrichment (ssGSEA) algorithm used to investigate levels of immune cell infiltration. RESULTS Our study identified dual-specificity phosphatase 5 (DUSP5) as a novel and powerful predictor of adverse outcomes for LUAD by using public datasets. Functional enrichment analysis found that DUSP5 was positively enriched in EMT and transforming growth factor-beta (TGF-β) signaling pathway, a prevailing pathway involved in the induction of EMT. As expected, DUSP5 knockdown suppressed EMT via inhibiting the canonical TGF-β/Smad signaling pathway in in vitro experiments. Consistently, knockdown of DUSP5 was first found to inhibit migratory ability and invasiveness of LUAD cells in in vitro and prevent lung metastasis in in vivo. DUSP5 knockdown re-sensitized gefitinib-resistant LUAD cells to gefitinib, accompanying reversion of EMT progress. In LUAD tissue samples, we found 14 cytosine-phosphate-guanine (CpG) sites of DUSP5 that were negatively associated with DUSP5 gene expression. Importantly, 5'Azacytidine (AZA), an FDA-approved DNA methyltransferase inhibitor, restored DUSP5 expression. Moreover, RIP experiments confirmed that YTH N6-methyladenosine RNA binding protein 1 (YTHDF1), a m6A reader protein, could bind DUSP5 mRNA. YTHDF1 promoted DUSP5 expression and the malignant phenotype of LUAD cells. In addition, the DUSP5-derived genomic model revealed the two clusters with distinguishable immune features and tumor mutational burden (TMB). CONCLUSIONS Briefly, our study discovered DUSP5 which was regulated by epigenetic modification, might be a potential therapeutic target, especially in LUAD patients with acquired EGFR-TKI resistance.
Collapse
Affiliation(s)
- Weina Fan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Ying Xing
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Shi Yan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Wei Liu
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Jinfeng Ning
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Fanglin Tian
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Xin Wang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Yuning Zhan
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Lixin Luo
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China
| | - Mengru Cao
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China.
| | - Jian Huang
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China.
| | - Li Cai
- The Fourth Department of Medical Oncology, Harbin Medical University Cancer Hospital, Haping Road 150, Harbin, 150081, China.
| |
Collapse
|
10
|
Ciccone V, Simonis V, Del Gaudio C, Cucini C, Ziche M, Morbidelli L, Donnini S. ALDH1A1 confers resistance to RAF/MEK inhibitors in melanoma cells by maintaining stemness phenotype and activating PI3K/AKT signaling. Biochem Pharmacol 2024; 224:116252. [PMID: 38701866 DOI: 10.1016/j.bcp.2024.116252] [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: 11/24/2023] [Revised: 04/16/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
The mitogen-activated protein kinase (MAPK/ERK) pathway is pivotal in controlling the proliferation and survival of melanoma cells. Several mutations, including those in BRAF, exhibit an oncogenic effect leading to increased cellular proliferation. As a result, the combination therapy of a MEK inhibitor with a BRAF inhibitor demonstrated higher efficacy and lower toxicity than BRAF inhibitor alone. This combination has become the preferred standard of care for tumors driven by BRAF mutations. Aldehyde dehydrogenase 1A1 (ALDH1A1) is a known marker of stemness involved in drug resistance in several type of tumors, including melanoma. This study demonstrates that melanoma cells overexpressing ALDH1A1 displayed resistance to vemurafenib and trametinib through the activation of PI3K/AKT signaling instead of MAPK axis. Inhibition of PI3K/AKT signaling partially rescued sensitivity to the drugs. Consistently, pharmacological inhibition of ALDH1A1 activity downregulated the activation of AKT and partially recovered responsiveness to vemurafenib and trametinib. We propose ALDH1A1 as a new potential target for treating melanoma resistant to MAPK/ERK inhibitors.
Collapse
Affiliation(s)
- Valerio Ciccone
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Vittoria Simonis
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Cinzia Del Gaudio
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Claudio Cucini
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Marina Ziche
- Department of Medicine, Surgery and Neurosciences, University of Siena, Siena I‑53100, Italy
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Siena I-53100, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Siena I-53100, Italy.
| |
Collapse
|
11
|
Duan X, Hu H, Wang L, Chen L. Aldehyde dehydrogenase 1 family: A potential molecule target for diseases. Cell Biol Int 2024. [PMID: 38800962 DOI: 10.1002/cbin.12188] [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: 08/08/2023] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024]
Abstract
Aldehyde dehydrogenase 1 (ALDH1), a crucial aldehyde metabolizing enzyme, has six family members. The ALDH1 family is expressed in various tissues, with a significant presence in the liver. It plays a momentous role in several pathophysiological processes, including aldehyde detoxification, oxidative stress, and lipid peroxidation. Acetaldehyde detoxification is the fundamental function of the ALDH1 family in participating in vital pathological mechanisms. The ALDH1 family can catalyze retinal to retinoic acid (RA) that is a hormone-signaling molecule and plays a vital role in the development and adult tissues. Furthermore, there is a need for further and broader research on the role of the ALDH1 family as a signaling molecule. The ALDH1 family is widely recognized as a cancer stem cell (CSC) marker and plays a significant role in the proliferation, invasion, metastasis, prognosis, and drug resistance of cancer. The ALDH1 family also participates in other human diseases, such as neurodegenerative diseases, osteoarthritis, diabetes, and atherosclerosis. It can inhibit disease progression by inhibiting/promoting the expression/activity of the ALDH1 family. In this review, we comprehensively analyze the tissue distribution, and functions of the ALDH1 family. Additionally, we review the involvement of the ALDH1 family in diseases, focusing on the underlying pathological mechanisms and briefly talk about the current status and development of ALDH1 family inhibitors. The ALDH1 family presents new possibilities for treating diseases, with both its upstream and downstream pathways serving as promising targets for therapeutic intervention. This offers fresh perspectives for drug development in the field of disease research.
Collapse
Affiliation(s)
- Xiangning Duan
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| | - Haoliang Hu
- Changde Research Centre for Artificial Intelligence and Biomedicine, Zoology Key Laboratory of Hunan Higher Education, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
| | - Lingzhi Wang
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou, Hunan, China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| |
Collapse
|
12
|
Li S, Cao C, Huang Z, Tang D, Chen J, Wang A, He Q. SOD2 confers anlotinib resistance via regulation of mitochondrial damage in OSCC. Oral Dis 2024; 30:281-291. [PMID: 36229195 DOI: 10.1111/odi.14404] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/22/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Previous studies had revealed that anlotinib had outstanding anti-tumor efficacy on oral squamous cell carcinoma. However, the underlying mechanism is still unclear. MATERIALS AND METHODS Anlotinib resistant OSCC cells were established and analyzed by RNA-sequencing. The correlations between SOD2 expression and anlotinib resistance were investigated in OSCC cells and PDX models. Functional assays were performed to verify the SOD2 expression and anlotinib resistance in OSCC cells. RESULTS Anlotinib resistant genes were enriched in the biological processes of mitochondrion organization and the gene pathway of reactive oxygen species. SOD2 expression level was positively correlated with the resistance of anlotinib in OSCC cells and PDX models. Higher SOD2 expression of OSCC cells was more resistant to anlotinib. Anlotinib induced ROS generation, apoptosis and mitochondrial damage in OSCC cells, which can be enhanced by SOD2 knockdown and decreased by SOD2 overexpression. Mitochondrial damage was identified as swelling and cristae disappearance morphology under TEM, decreased mitochondrial membrane potential and lower MFN2 expression. CONCLUSIONS SOD2 may be capable of protecting mitochondria by downregulating ROS generation, which contributes to the resistance of anlotinib in OSCC cells. SOD2 can be utilized as a potential therapeutic target to improve the anti-cancer efficacy of anlotinib in OSCC.
Collapse
Affiliation(s)
- Shuai Li
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, China
| | - Congyuan Cao
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhexun Huang
- Center of Oral Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Dongxiao Tang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Stomatology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Chen
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Anxun Wang
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qianting He
- Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
13
|
Duong HQ, Hoang MC, Nguyen TH, Nguyen PT, Le VT, Dao TN, Ngo VL, Dang TH. Aldehyde Dehydrogenase-1A1 (ALDH1A1): The Novel Regulator of Chemoresistance in Pancreatic Cancer Cells. Cancer Control 2024; 31:10732748241305835. [PMID: 39611960 PMCID: PMC11607765 DOI: 10.1177/10732748241305835] [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: 08/15/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Aldehyde dehydrogenase-1A1 (ALDH1A1), a member of a superfamily of 19 isozymes, exhibits various biological functions and is involved in several important physiological and pathological processes, including those associated with various diseases including cancers such as pancreatic cancer. Chemotherapy is one of the most important strategies for the treatment of pancreatic cancer; however, the chemoresistance exhibited by pancreatic cancer cells is a leading cause of chemotherapy failure. It has been reported that overexpression of ALDH1A1 significantly correlates with poor prognosis and tumor aggressiveness, and is clinically associated with chemoresistance. Additionally, ALDH1A1 may serve as a novel regulator for the diagnosis and prognosis of cancer resistance. In particular, ALDH1A1 can promote cancer progression by facilitating the manifestation of cancer stem cell properties. However, the molecular mechanism by which ALDH1A1 clinically regulates the development of chemoresistance, and its role in prognosis and cancer stem cells, including pancreatic cancer stem cells, remain unclear. Therefore, the current review aims to summarize the clinical functions of ALDH1A1 as a novel regulator of chemoresistance, prognosis, and cancer stem cell development in pancreatic cancer.
Collapse
Affiliation(s)
- Hong-Quan Duong
- Laboratory Center, Hanoi University of Public Health, Hanoi, Vietnam
| | - Minh-Cong Hoang
- Laboratory Department, Yenphong Medical Center, Bacninh, Vietnam
| | - Thi-Hue Nguyen
- Laboratory Department, Bacgiang General Hospital, Bacgiang, Vietnam
| | | | - Van-Thu Le
- Laboratory Center, Hanoi University of Public Health, Hanoi, Vietnam
| | - Thi-Nguyet Dao
- Pathology Department, Ducgiang General Hospital, Hanoi, Vietnam
| | - Van-Lang Ngo
- Faculty of Biomedical Sciences, Phenikaa University, Hanoi, Vietnam
| | - The-Hung Dang
- Laboratory Center, Hanoi University of Public Health, Hanoi, Vietnam
| |
Collapse
|
14
|
Kogan EA, Shchelokova EE, Demura TA, Zharkov NV, Kichigina ON, Kovyazina NV, Mordovina AI, Zelenchenkova PI, Meerovich GA, Reshetov IV. [ALDH1, CD133, CD34-positive cancer stem cells in lung adenocarcinoma in patients who had a new coronavirus infection and retained the persistence of viral proteins in the lung tissue]. Arkh Patol 2024; 86:5-14. [PMID: 39434522 DOI: 10.17116/patol2024860515] [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] [Indexed: 10/23/2024]
Abstract
Lung cancer occupies a leading position in the structure of global cancer morbidity and mortality, due to the biological properties of the key pool of tumor cells - cancer stem cells (CSCs). The effects of SARS-CoV2 on tumor CSCs and its niche have not been studied. OBJECTIVE To study CSCs in lung adenocarcinomas after COVID19. MATERIAL AND METHODS Surgical material from lung adenocarcinoma from 12 patients who had a new coronavirus infection and from 12 patients who did not have COVID19 was examined. Analysis of clinical and anamnestic data, macroscopic and microscopic examination of tumor samples and adjacent intact tissue, immunohistochemical reactions using antibodies to virus proteins and CSC markers ALDH1, CD133 and CD34 were performed. RESULTS Adenocarcinoma samples from patients in the main group showed a significant increase in the number of cells expressing the CSCs markers ALDH1, CD133 and CD34 compared to adenocarcinoma samples from control group patients without SARS-CoV2 infection. We found an increase in the number of CSCs in patients with adenocarcinoma metastasis in lymph nodes in both the main and control groups. CSCs of lung adenocarcinomas from SARS-CoV2 survivors contain virus proteins Nucleocapside and Spike protein. CONCLUSIONS We found an increase in the number of CSCs with expression of ALDH1, CD133 and CD34 in lung adenocarcinoma in patients with new coronavirus infection. Increased number of ALDH1+, CD133+ CD34+ CSCs in tumor tissue enhance the metastatic potential of lung adenocarcinoma. The Nucleocapsid and Spike proteins of SARS-CoV2 virus are detectable in lung tissue from patients with new coronavirus infection, both in adenocarcinoma cells, CSCs, and in type II pneumocytes, macrophages, and endothelial cells, suggesting prolonged persistence of the virus proteins and probably the virus.
Collapse
Affiliation(s)
- E A Kogan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - E E Shchelokova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - T A Demura
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - N V Zharkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - O N Kichigina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - A I Mordovina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - P I Zelenchenkova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - G A Meerovich
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- A.M. Prokhorov General Physics Institute Russian Academy of Sciences (GPI RAS), Moscow, Russia
| | - I V Reshetov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
15
|
Li D, Cao Y, Luo CW, Zhang LP, Zou YB. The Clinical Significance and Prognostic Value of ALDH1 Expression in Non-small Cell Lung Cancer: A Systematic Review and Meta-analysis. Recent Pat Anticancer Drug Discov 2024; 19:599-609. [PMID: 37818578 DOI: 10.2174/0115748928265992230925053308] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND The results of the association between aldehyde dehydrogenase 1 (ALDH1) expression and prognosis of non-small cell lung cancer (NSCLC) are contradictory. We conducted this meta-analysis to investigate the clinical significance and prognostic value of ALDH1 in NSCLC. METHODS The databases PubMed, Web of Science, EMBASE, the Cochrane Library, Wanfang, and CNKI were systematically queried to identify eligible studies. The retrieval time was from database establishment to August 2023. We evaluated the correlation between ALDH1 expression and clinical features of NSCLC by employing odds ratios (ORs) and 95% confidence intervals (95% CIs). In addition, we used hazard ratios (HRs) and 95% CIs to evaluate the role of ALDH1 expression in the prognosis of NSCLC. RESULTS Our study included 21 literatures involving 2721 patients. The expression of ALDH1 in NSCLC was higher than that in normal tissues (OR = 6.04, 95% CI: 1.25-29.27, P = 0.026). The expression of ALDH1 was related to TNM stage (OR = 1.81, 95% CI: 1.06-3.09, P = 0.029), tumor grade (OR = 0.29, 95% CI: 0.17-0.48, P < 0.0001), lymph node metastasis (OR = 2.60, 95% CI: 1.52-4.45, P = 0001) and histological subtype (OR = 0.67, 95% CI: 0.52-0.86, P = 0.002). In patients with NSCLC, we found that the over-expression of ALDH1 was significantly associated with poor overall survival (OS) (HR = 1.44, 95% CI: 1.15-1.81, P = 0.002) and disease-free survival (DFS) (HR = 1.74, 95% CI: 1.45-2.10, P < 0.0001). CONCLUSION The expression of ALDH1 is closely associated with the clinicopathologic characteristics and prognosis of NSCLC. ALDH1 may serve as a valuable clinical assessment tool and prognostic predictor in NSCLC.
Collapse
Affiliation(s)
- Dong Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of CQMU (General Hospital), Chongqing, 401120, China
| | - Yu Cao
- Department of Thoracic Surgery, The Third Affiliated Hospital of CQMU (General Hospital), Chongqing, 401120, China
| | - Cheng-Wen Luo
- Department of Thoracic Surgery, The Third Affiliated Hospital of CQMU (General Hospital), Chongqing, 401120, China
| | - Li-Ping Zhang
- Department of Thoracic Surgery, The Third Affiliated Hospital of CQMU (General Hospital), Chongqing, 401120, China
| | - Ying-Bo Zou
- Department of Thoracic Surgery, The Third Affiliated Hospital of CQMU (General Hospital), Chongqing, 401120, China
| |
Collapse
|
16
|
Vogt M, Dienstbier N, Schliehe-Diecks J, Scharov K, Tu JW, Gebing P, Hogenkamp J, Bilen BS, Furlan S, Picard D, Remke M, Yasin L, Bickel D, Kalia M, Iacoangeli A, Lenz T, Stühler K, Pandyra AA, Hauer J, Fischer U, Wagener R, Borkhardt A, Bhatia S. Co-targeting HSP90 alpha and CDK7 overcomes resistance against HSP90 inhibitors in BCR-ABL1+ leukemia cells. Cell Death Dis 2023; 14:799. [PMID: 38057328 PMCID: PMC10700369 DOI: 10.1038/s41419-023-06337-3] [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: 06/06/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/08/2023]
Abstract
HSP90 has emerged as an appealing anti-cancer target. However, HSP90 inhibitors (HSP90i) are characterized by limited clinical utility, primarily due to the resistance acquisition via heat shock response (HSR) induction. Understanding the roles of abundantly expressed cytosolic HSP90 isoforms (α and β) in sustaining malignant cells' growth and the mechanisms of resistance to HSP90i is crucial for exploiting their clinical potential. Utilizing multi-omics approaches, we identified that ablation of the HSP90β isoform induces the overexpression of HSP90α and extracellular-secreted HSP90α (eHSP90α). Notably, we found that the absence of HSP90α causes downregulation of PTPRC (or CD45) expression and restricts in vivo growth of BCR-ABL1+ leukemia cells. Subsequently, chronic long-term exposure to the clinically advanced HSP90i PU-H71 (Zelavespib) led to copy number gain and mutation (p.S164F) of the HSP90AA1 gene, and HSP90α overexpression. In contrast, acquired resistance toward other tested HSP90i (Tanespimycin and Coumermycin A1) was attained by MDR1 efflux pump overexpression. Remarkably, combined CDK7 and HSP90 inhibition display synergistic activity against therapy-resistant BCR-ABL1+ patient leukemia cells via blocking pro-survival HSR and HSP90α overexpression, providing a novel strategy to avoid the emergence of resistance against treatment with HSP90i alone.
Collapse
Affiliation(s)
- Melina Vogt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Niklas Dienstbier
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julian Schliehe-Diecks
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Katerina Scharov
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jia-Wey Tu
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Philip Gebing
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julian Hogenkamp
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Berna-Selin Bilen
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Silke Furlan
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Daniel Picard
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Marc Remke
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Layal Yasin
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - David Bickel
- Interuniversity Institute of Bioinformatics in Brussels, ULB-VUB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Munishikha Kalia
- Department of Biostatistics and Health Informatics, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
| | - Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Thomas Lenz
- Molecular Proteomics Laboratory, Biological Medical Research Center, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Kai Stühler
- Institute for Molecular Medicine, Proteome Research, University Hospital and Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Aleksandra A Pandyra
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Bonn, Germany
| | - Julia Hauer
- Department of Pediatrics and Children's Cancer Research Center, Children's Hospital Munich Schwabing, Technical University of Munich, School of Medicine, Munich, Germany
| | - Ute Fischer
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Rabea Wagener
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany
| | - Sanil Bhatia
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
17
|
Lu X, Wu M, Luo Q. Development of a NIR fluorescent probe for fluorescence-assisted EGFR-TKI applicable patients screening and drug resistance monitoring. Eur J Med Chem 2023; 261:115818. [PMID: 37741183 DOI: 10.1016/j.ejmech.2023.115818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/10/2023] [Accepted: 09/11/2023] [Indexed: 09/25/2023]
Abstract
EGFR tyrosine kinase inhibitor exerts significant benefits to non-small cell lung cancer patient, but was also limited by the applicable patient screening and drug resistance. Here we presented with an EGFR-targeted and reactive oxygen species-responsive NIR probe (LX) to achieve both patient screening and drug resistance monitoring for EGFR-tyrosine kinase inhibitor. LX inherited EGFR selectivity and preference from EGFR-tyrosine kinase inhibitor, which only showed specificity to tumor with EGFR mutation. Meanwhile, the near-infrared fluorescence of LX was initially inhibited and could be turned on by intratumoral reactive oxygen species. When LX could bind to tumor EGFR, reactive oxygen species-responsive specific fluorescence was generated to indicate the applicability of tumors to EGFR-tyrosine kinase inhibitor. However, no specific LX fluorescence could be observed in inapplicable tumors due to the lack of specificity between tumor EGFR and LX. Meanwhile, when drug resistance was developed during treatments, obvious intratumoral reactive species oxygen decrease happened, which was also deemed as a significant signal of the drug resistance. By visualizing intratumoral reactive oxygen species fluctuation by responsive fluorescence, drug resistance could be monitored and reported.
Collapse
Affiliation(s)
- Xinmiao Lu
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China
| | - Muyu Wu
- Department of Nuclear Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Quanyong Luo
- Department of Nuclear Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200235, China.
| |
Collapse
|
18
|
Bagherpoor AJ, Shameem M, Luo X, Seelig D, Kassie F. Inhibition of lung adenocarcinoma by combinations of sulfasalazine (SAS) and disulfiram-copper (DSF-Cu) in cell line models and mice. Carcinogenesis 2023; 44:291-303. [PMID: 37053033 PMCID: PMC10290516 DOI: 10.1093/carcin/bgad020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/20/2023] [Accepted: 04/11/2023] [Indexed: 04/14/2023] Open
Abstract
Sulfasalazine (SAS) is a repurposed antitumor drug which inhibits the proliferation and survival of cancer cells by inhibiting the xCT cellular antioxidant system. Recent clinical studies have shown that, due to poor bioavailability, the antitumor effects of SAS monotherapy are minimal. Therefore, we hypothesized that DSF, another repurposed drug that has demonstrated anticancer effects, or its complex with copper (DSF-copper, DSF-Cu) could potentiate the antilung cancer effects of SAS. Exposure of non-small cell lung cancer cells to therapeutically achievable concentrations of SAS-induced low-to-moderate cytotoxic effects (20-40% reduction in cell viability) and, unexpectedly, induced the antioxidant protein NRF2 and its downstream effectors xCT and ALDH1A1. However, combinations of SAS and DSF-Cu, but not SAS and DSF, induced a significantly higher cytotoxic effect (64-88% reduction in cell viability), apoptosis and generation of mitochondrial reactive oxygen species as compared with SAS or DSF-Cu alone. Moreover, DSF-Cu abrogated SAS-induced NRF2, xCT and ALDH1A1 expression. In a mouse model of lung tumor, SAS + DSF-Cu showed a higher efficacy than the individual drugs in reducing the number and size of tumors as well as the incidence and multiplicity of lung adenocarcinoma. Taken together, our findings indicate that the observed antilung cancer effects of SAS plus DSF-Cu are mediated, at least in part, via impairment of reactive oxygen species defense and -enhancement of oxidative stress and provide evidence for the preventive/therapeutic potential of this combinatorial approach against lung cancer.
Collapse
Affiliation(s)
| | | | - Xianghua Luo
- Masonic Cancer Center, Minneapolis, MN 55455, USA
- Division of Biostatistics, School of Public Health, Minneapolis, MN 55455, USA
| | - Davis Seelig
- Masonic Cancer Center, Minneapolis, MN 55455, USA
- College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - Fekadu Kassie
- Masonic Cancer Center, Minneapolis, MN 55455, USA
- College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
19
|
Han JM, Kim SM, Kim HL, Cho HJ, Jung HJ. Natural Cyclophilin A Inhibitors Suppress the Growth of Cancer Stem Cells in Non-Small Cell Lung Cancer by Disrupting Crosstalk between CypA/CD147 and EGFR. Int J Mol Sci 2023; 24:ijms24119437. [PMID: 37298389 DOI: 10.3390/ijms24119437] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is a fatal malignant tumor with a high mortality rate. Cancer stem cells (CSCs) play pivotal roles in tumor initiation and progression, treatment resistance, and NSCLC recurrence. Therefore, the development of novel therapeutic targets and anticancer drugs that effectively block CSC growth may improve treatment outcomes in patients with NSCLC. In this study, we evaluated, for the first time, the effects of natural cyclophilin A (CypA) inhibitors, including 23-demethyl 8,13-deoxynargenicin (C9) and cyclosporin A (CsA), on the growth of NSCLC CSCs. C9 and CsA more sensitively inhibited the proliferation of epidermal growth factor receptor (EGFR)-mutant NSCLC CSCs than EGFR wild-type NSCLC CSCs. Both compounds suppressed the self-renewal ability of NSCLC CSCs and NSCLC-CSC-derived tumor growth in vivo. Furthermore, C9 and CsA inhibited NSCLC CSC growth by activating the intrinsic apoptotic pathway. Notably, C9 and CsA reduced the expression levels of major CSC markers, including integrin α6, CD133, CD44, ALDH1A1, Nanog, Oct4, and Sox2, through dual downregulation of the CypA/CD147 axis and EGFR activity in NSCLC CSCs. Our results also show that the EGFR tyrosine kinase inhibitor afatinib inactivated EGFR and decreased the expression levels of CypA and CD147 in NSCLC CSCs, suggesting close crosstalk between the CypA/CD147 and EGFR pathways in regulating NSCLC CSC growth. In addition, combined treatment with afatinib and C9 or CsA more potently inhibited the growth of EGFR-mutant NSCLC CSCs than single-compound treatments. These findings suggest that the natural CypA inhibitors C9 and CsA are potential anticancer agents that suppress the growth of EGFR-mutant NSCLC CSCs, either as monotherapy or in combination with afatinib, by interfering with the crosstalk between CypA/CD147 and EGFR.
Collapse
Affiliation(s)
- Jang Mi Han
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Republic of Korea
| | - Sung Min Kim
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Republic of Korea
| | - Hong Lae Kim
- Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan 31460, Republic of Korea
| | - Hee Jeong Cho
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Republic of Korea
| | - Hye Jin Jung
- Department of Life Science and Biochemical Engineering, Graduate School, Sun Moon University, Asan 31460, Republic of Korea
- Department of Pharmaceutical Engineering and Biotechnology, Sun Moon University, Asan 31460, Republic of Korea
- Genome-Based BioIT Convergence Institute, Sun Moon University, Asan 31460, Republic of Korea
| |
Collapse
|
20
|
Dancik GM, Varisli L, Vlahopoulos SA. The Molecular Context of Oxidant Stress Response in Cancer Establishes ALDH1A1 as a Critical Target: What This Means for Acute Myeloid Leukemia. Int J Mol Sci 2023; 24:ijms24119372. [PMID: 37298333 DOI: 10.3390/ijms24119372] [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: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The protein family of aldehyde dehydrogenases (ALDH) encompasses nineteen members. The ALDH1 subfamily consists of enzymes with similar activity, having the capacity to neutralize lipid peroxidation products and to generate retinoic acid; however, only ALDH1A1 emerges as a significant risk factor in acute myeloid leukemia. Not only is the gene ALDH1A1 on average significantly overexpressed in the poor prognosis group at the RNA level, but its protein product, ALDH1A1 protects acute myeloid leukemia cells from lipid peroxidation byproducts. This capacity to protect cells can be ascribed to the stability of the enzyme under conditions of oxidant stress. The capacity to protect cells is evident both in vitro, as well as in mouse xenografts of those cells, shielding cells effectively from a number of potent antineoplastic agents. However, the role of ALDH1A1 in acute myeloid leukemia has been unclear in the past due to evidence that normal cells often have higher aldehyde dehydrogenase activity than leukemic cells. This being true, ALDH1A1 RNA expression is significantly associated with poor prognosis. It is hence imperative that ALDH1A1 is methodically targeted, particularly for the acute myeloid leukemia patients of the poor prognosis risk group that overexpress ALDH1A1 RNA.
Collapse
Affiliation(s)
- Garrett M Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Spiros A Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
| |
Collapse
|
21
|
Xia J, Li S, Liu S, Zhang L. Aldehyde dehydrogenase in solid tumors and other diseases: Potential biomarkers and therapeutic targets. MedComm (Beijing) 2023; 4:e195. [PMID: 36694633 PMCID: PMC9842923 DOI: 10.1002/mco2.195] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 01/18/2023] Open
Abstract
The family of aldehyde dehydrogenases (ALDHs) contains 19 isozymes and is involved in the oxidation of endogenous and exogenous aldehydes to carboxylic acids, which contributes to cellular and tissue homeostasis. ALDHs play essential parts in detoxification, biosynthesis, and antioxidants, which are of important value for cell proliferation, differentiation, and survival in normal body tissues. However, ALDHs are frequently dysregulated and associated with various diseases like Alzheimer's disease, Parkinson's disease, and especially solid tumors. Notably, the involvement of the ALDHs in tumor progression is responsible for the maintenance of the stem-cell-like phenotype, triggering rapid and aggressive clinical progressions. ALDHs have captured increasing attention as biomarkers for disease diagnosis and prognosis. Nevertheless, these require further longitudinal clinical studies in large populations for broad application. This review summarizes our current knowledge regarding ALDHs as potential biomarkers in tumors and several non-tumor diseases, as well as recent advances in our understanding of the functions and underlying molecular mechanisms of ALDHs in disease development. Finally, we discuss the therapeutic potential of ALDHs in diseases, especially in tumor therapy with an emphasis on their clinical implications.
Collapse
Affiliation(s)
- Jie Xia
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Siqin Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer MedicineNanjing Medical UniversityNanjingChina
| | - Lixing Zhang
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering, Cancer Institutes, Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Shanghai Key Laboratory of Radiation Oncology, The International Co‐laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
22
|
Yuan G, Hu B, Ma J, Zhang C, Xie H, Wei T, Yang Y, Ni B. Histone lysine methyltransferase
SETDB2
suppresses
NRF2
to restrict tumor progression and modulates chemotherapy sensitivity in lung adenocarcinoma. Cancer Med 2022; 12:7258-7272. [PMID: 36504353 PMCID: PMC10067124 DOI: 10.1002/cam4.5451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Aberrant epigenetic remodeling represents a molecular hallmark in lung adenocarcinoma (LUAD). We aim to investigate the biological roles of SETDB2 and its underlying associations with oxidative stress, providing therapeutic targets for individualized treatment of LUAD. METHODS Differential analysis was conducted via Limma package, and Kaplan-Meier analysis was performed with survival package. CCK-8, cell proliferation assay, transwell assay, and in vivo assays were conducted to assess the function of SETDB2. Western blot assay, RT-qPCR, and immunohistochemistry (IHC) were conducted to assess the expression levels of SETDB2/NRF2. Chromatin immunoprecipitation (ChIP) assay and ChIP-qPCR were conducted to assess the epigenetic roles of SETDB2. RESULTS We found that SETDB2 expression is decreased in tumor samples versus normal tissues in TCGA-LUAD cohort, LUAD-EAS cohort, GSE72094 dataset, and independent Soochow-LUAD dataset. Patients with low SETDB2 levels had a worse prognosis relative to those with high SETDB2. SETDB2 inhibition could significantly promote cell growth, migration ability, and stemness maintenance. Gene set enrichment analysis (GSEA) suggested that SETDB2 correlated with oxidative stress crosstalk and regulated NRF2 mRNA levels. ChIP assay suggested that SETDB2 mainly recruited the H3K9me3 enrichment at the NRF2 promoter region to suppress the mRNA levels of NRF2. Downregulated SETDB2 could activate NRF2 transcription and expression, thereby promoting its downstream targets, like NQO1, FTH1, and ME1. Functional experiments demonstrated that low SETDB2 allowed NRF2 to drive malignant processes of LUAD. SETDB2 overexpression attenuated the ability of NRF2 signaling to neutralize cellular reactive oxygen species (ROS) levels, leading to enhanced cell apoptosis. Overexpressed SETDB2 could inhibit tumor progression in vivo and further render LUAD cells sensitive to chemotherapy. CONCLUSIONS In conclusion, these findings uncovered the suppressive role of SETDB2 in LUAD. SETDB2 negatively regulates NRF2 signaling to modulate tumor progression, which creates a therapeutic vulnerability in LUAD.
Collapse
Affiliation(s)
- Guangda Yuan
- Department of Thoracic Surgery The First Affiliated Hospital of Soochow University Suzhou China
| | - Bowen Hu
- Department of Thoracic Surgery The First Affiliated Hospital of Soochow University Suzhou China
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Jun Ma
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Chuanyu Zhang
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Hongya Xie
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Tengteng Wei
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Yong Yang
- Department of Thoracic Surgery The Affiliated Suzhou Hospital of Nanjing Medical University Suzhou China
| | - Bin Ni
- Department of Thoracic Surgery The First Affiliated Hospital of Soochow University Suzhou China
| |
Collapse
|
23
|
Wei Y, Li Y, Chen Y, Liu P, Huang S, Zhang Y, Sun Y, Wu Z, Hu M, Wu Q, Wu H, Liu F, She T, Ning Z. ALDH1: A potential therapeutic target for cancer stem cells in solid tumors. Front Oncol 2022; 12:1026278. [PMID: 36387165 PMCID: PMC9650078 DOI: 10.3389/fonc.2022.1026278] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
Solid tumors can be divided into benign solid tumors and solid malignant tumors in the academic community, among which malignant solid tumors are called cancers. Cancer is the second leading cause of death in the world, and the global incidence of cancer is increasing yearly New cancer patients in China are always the first. After the concept of stem cells was introduced in the tumor community, the CSC markers represented by ALDH1 have been widely studied due to their strong CSC cell characteristics and potential to be the driving force of tumor metastasis. In the research results in the past five years, it has been found that ALDH1 is highly expressed in various solid cancers such as breast cancer, lung cancer, colorectal cancer, liver cancer, gastric cancer, cervical cancer, esophageal cancer, ovarian cancer, head,and neck cancer. ALDH1 can activate and transform various pathways (such as the USP28/MYC signaling pathway, ALDH1A1/HIF-1α/VEGF axis, wnt/β-catenin signaling pathway), as well as change the intracellular pH value to promote formation and maintenance, resulting in drug resistance in tumors. By targeting and inhibiting ALDH1 in tumor stem cells, it can enhance the sensitivity of drugs and inhibit the proliferation, differentiation, and metastasis of solid tumor stem cells to some extent. This review discusses the relationship and pathway of ALDH1 with various solid tumors. It proposes that ALDH1 may serve as a diagnosis and therapeutic target for CSC, providing new insights and new strategies for reliable tumor treatment.
Collapse
Affiliation(s)
- Yaolu Wei
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yan Li
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yenan Chen
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Pei Liu
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Sheng Huang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yuping Zhang
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanling Sun
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhe Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Hongnian Wu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Fuxing Liu
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Tonghui She
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| | - Zhifeng Ning
- School of Basic Medicine Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Fuxing Liu, ; Tonghui She, ; Zhifeng Ning,
| |
Collapse
|
24
|
Luo MY, Zhou Y, Gu WM, Wang C, Shen NX, Dong JK, Lei HM, Tang YB, Liang Q, Zou JH, Xu L, Ma P, Zhuang G, Bi L, Xu L, Zhu L, Chen HZ, Shen Y. Metabolic and Nonmetabolic Functions of PSAT1 Coordinate Signaling Cascades to Confer EGFR Inhibitor Resistance and Drive Progression in Lung Adenocarcinoma. Cancer Res 2022; 82:3516-3531. [PMID: 36193649 DOI: 10.1158/0008-5472.can-21-4074] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 05/03/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022]
Abstract
Emerging evidence demonstrates that the dysregulated metabolic enzymes can accelerate tumorigenesis and progression via both metabolic and nonmetabolic functions. Further elucidation of the role of metabolic enzymes in EGFR inhibitor resistance and metastasis, two of the leading causes of death in lung adenocarcinoma, could help improve patient outcomes. Here, we found that aberrant upregulation of phosphoserine aminotransferase 1 (PSAT1) confers erlotinib resistance and tumor metastasis in lung adenocarcinoma. Depletion of PSAT1 restored sensitivity to erlotinib and synergistically augmented the tumoricidal effect. Mechanistically, inhibition of PSAT1 activated the ROS-dependent JNK/c-Jun pathway to induce cell apoptosis. In addition, PSAT1 interacted with IQGAP1, subsequently activating STAT3-mediated cell migration independent of its metabolic activity. Clinical analyses showed that PSAT1 expression positively correlated with the progression of human lung adenocarcinoma. Collectively, these findings reveal the multifunctionality of PSAT1 in promoting tumor malignancy through its metabolic and nonmetabolic activities. SIGNIFICANCE Metabolic and nonmetabolic functions of PSAT1 confer EGFR inhibitor resistance and promote metastasis in lung adenocarcinoma, suggesting therapeutic targeting of PSAT1 may attenuate the malignant features of lung cancer.
Collapse
Affiliation(s)
- Ming-Yu Luo
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Ye Zhou
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Wei-Ming Gu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Cheng Wang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Ning-Xiang Shen
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Jiang-Kai Dong
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Hui-Min Lei
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Ya-Bin Tang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Qian Liang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Jing-Hua Zou
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Lu Xu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Pengfei Ma
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglei Zhuang
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Bi
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Xu
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| | - Hong-Zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai, China
| |
Collapse
|
25
|
Zhang W, Wan S, Qu Z, Ge J, Zhang C, Li C, Jiang Y. Establishment of a prognostic signature for lung adenocarcinoma by integration of 7 pyroptosis-related genes and cross-validation between the TCGA and GEO cohorts: A comprehensive bioinformatics analysis. Medicine (Baltimore) 2022; 101:e29710. [PMID: 35866781 PMCID: PMC9302251 DOI: 10.1097/md.0000000000029710] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Pyroptosis-related genes (PRGs) have been reported to be associated with prognosis of lung adenocarcinoma (LUAD). Until now, the relationship of PRGs to the prognosis of LUAD patients and its underlying mechanisms have been poorly elucidated. Using The Cancer Genome Atlas (TCGA) LUAD cohort, a prior bioinformatics analysis constructed a prognostic signature incorporating 5 PRGs (NLRP7, NLRP1, NLRP2, NOD1, and CASP6) for predicting prognosis of LUAD patients. However, it has not been validated by the Gene Expression Omnibus (GEO) LUAD cohort yet. We implemented a modified bioinformatics analysis to, respectively, construct one prognostic signature with the TCGA cohort and with the GEO cohort and attempted to perform cross-validations by the GEO cohort and the TCGA cohort alternately in turn. Univariate and multivariate Cox regression analysis screened PRGs and constructed 2 prognostic signatures with the TCGA and GEO cohorts. All LUAD samples were classified into high- and low-risk groups according to the median risk score that was generated by regression formula. Kaplan-Meier survival analysis compared the overall survival rate between the 2 risk groups, and receiver operating characteristic curve analysis evaluated predictive performance of the 2 signatures. Additionally, risk score, combined with clinicopathological features, was subjected to multivariate Cox regression analysis, to evaluate independent prognostic value of the 2 signatures. Finally, the 2 signatures received cross-validations by the GEO and TCGA cohorts, alternately. The TCGA cohort yielded a 3-gene signature (PYCARD, NLRP1, and NLRC4), whereas the GEO cohort built a 7-gene signature (SCAF11, NOD1, NLRP2, NLRP1, GPX4, CASP8, and AIM2) for predicting the prognosis of LUAD patients. Multivariate analysis proved independent prognostic value of risk score in the TCGA cohort (hazard ratio, = 1.939,; P = 8.43 × 10-4) and the GEO cohort (hazard ratio, = 2.291,; P = 4.34 × 10-9). Cross-validations confirmed prognostic value for the 7-gene signature from the GEO cohort by the TCGA cohort but not for the 3-gene signature from the TCGA cohort by the GEO cohort. We develop and validate a 7-gene prognostic signature (SCAF11, NOD1, NLRP2, NLRP1, GPX4, CASP8, and AIM2) with independent prognostic value for patients with LUAD.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Critical Care Medicine, Wuhan Jinyintan Hospital, Wuhan, Hubei, People’s Republic of China
| | - Shiqian Wan
- Department of Infectious Diseases, Wuhan Jinyintan Hospital, Wuhan, Hubei, People’s Republic of China
| | - Zhaohui Qu
- Department of Critical Care Medicine, Wuhan Jinyintan Hospital, Wuhan, Hubei, People’s Republic of China
| | - Jing Ge
- Department of Critical Care Medicine, Wuhan Jinyintan Hospital, Wuhan, Hubei, People’s Republic of China
| | - Chunxia Zhang
- Department of Critical Care Medicine, Wuhan Jinyintan Hospital, Wuhan, Hubei, People’s Republic of China
| | - Chunfang Li
- Department of Critical Care Medicine, Wuhan Jinyintan Hospital, Wuhan, Hubei, People’s Republic of China
| | - Yingchun Jiang
- Department of Ultrasound Diagnostics, Wuhan Jinyintan Hospital, Wuhan, Hubei, People’s Republic of China
- * Correspondence: Yingchun Jiang, Department of Ultrasound Diagnostics, Wuhan Jinyintan Hospital, 1 Yintan Rd., Wuhan, Hubei 430023, People’s Republic of China (e-mail: )
| |
Collapse
|
26
|
Zheng Y, Wang L, Yin L, Yao Z, Tong R, Xue J, Lu Y. Lung Cancer Stem Cell Markers as Therapeutic Targets: An Update on Signaling Pathways and Therapies. Front Oncol 2022; 12:873994. [PMID: 35719973 PMCID: PMC9204354 DOI: 10.3389/fonc.2022.873994] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer stem cells, a relatively small group of self-renewing cancer cells, were first isolated from acute myeloid leukemia. These cells can play a crucial role in tumor metastasis, relapse, and therapy resistance. The cancer stem cell theory may be applied to lung cancer and explain the inefficiency of traditional treatments and eventual recurrence. However, because of the unclear accuracy and illusive biological function of cancer stem cells, some researchers remain cautious about this theory. Despite the ongoing controversy, cancer stem cells are still being investigated, and their biomarkers are being discovered for application in cancer diagnosis, targeted therapy, and prognosis prediction. Potential lung cancer stem cell markers mainly include surface biomarkers such as CD44, CD133, epithelial cell adhesion molecule, and ATP-binding cassette subfamily G member 2, along with intracellular biomarkers such as aldehyde dehydrogenase, sex-determining region Y-box 2, NANOG, and octamer-binding transcription factor 4. These markers have different structures and functions but are closely associated with the stem potential and uncontrollable proliferation of tumor cells. The aberrant activation of major signaling pathways, such as Notch, Hedgehog, and Wnt, may be associated with the expression and regulation of certain lung cancer stem cell markers, thus leading to lung cancer stem cell maintenance, chemotherapy resistance, and cancer promotion. Treatments targeting lung cancer stem cell markers, including antibody drugs, nanoparticle drugs, chimeric antigen receptor T-cell therapy, and other natural or synthetic specific inhibitors, may provide new hope for patients who are resistant to conventional lung cancer therapies. This review provides comprehensive and updated data on lung cancer stem cell markers with regard to their structures, functions, signaling pathways, and promising therapeutic target approaches, aiming to elucidate potential new therapies for lung cancer.
Collapse
Affiliation(s)
- Yue Zheng
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Laduona Wang
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Limei Yin
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuoran Yao
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ruizhan Tong
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - You Lu
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Hsieh PL, Chao SC, Chu PM, Yu CC. Regulation of Ferroptosis by Non-Coding RNAs in Head and Neck Cancers. Int J Mol Sci 2022; 23:3142. [PMID: 35328568 PMCID: PMC8950679 DOI: 10.3390/ijms23063142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/09/2022] [Accepted: 03/12/2022] [Indexed: 02/06/2023] Open
Abstract
Ferroptosis is a newly identified mode of programmed cell death characterized by iron-associated accumulation of lipid peroxides. Emerging research on ferroptosis has suggested its implication in tumorigenesis and stemness of cancer. On the other hand, non-coding RNAs have been shown to play a pivotal role in the modulation of various genes that affect the progression of cancer cells and ferroptosis. In this review, we summarize recent advances in the theoretical modeling of ferroptosis and its relationship between non-coding RNAs and head and neck cancers. Aside from the significance of ferroptosis-related non-coding RNAs in prognostic relevance, we also review how these non-coding RNAs participate in the regulation of iron, lipid metabolism, and reactive oxygen species accumulation. We aim to provide a thorough grounding in the function of ferroptosis-related non-coding RNAs based on current knowledge in an effort to develop effective therapeutic strategies for head and neck cancers.
Collapse
Affiliation(s)
- Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404333, Taiwan; (P.-L.H.); (P.-M.C.)
| | - Shih-Chi Chao
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan;
- Department of Medical Research and Education, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan 265, Taiwan
| | - Pei-Ming Chu
- Department of Anatomy, School of Medicine, China Medical University, Taichung 404333, Taiwan; (P.-L.H.); (P.-M.C.)
| | - Cheng-Chia Yu
- Institute of Oral Sciences, Chung Shan Medical University, Taichung 40201, Taiwan;
- School of Dentistry, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
28
|
Pan Z, Liu H, Chen J. [Lung Cancer Stem-like Cells and Drug Resistance]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:111-117. [PMID: 35224964 PMCID: PMC8913289 DOI: 10.3779/j.issn.1009-3419.2022.102.02] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lung cancer remains the leading cause of cancer-related death world-wide. Therapy resistance and relapse are considered major reasons contributing to the poor survival rates of lung cancer. Accumulated evidences have demonstrated that a small subpopulation of stem-like cells existed within lung cancer tissues and cell lines, possessing the abilities of self-renewal, multipotent differentiation and unlimited proliferation. These lung cancer stem-like cells (LCSCs) can generate tumors with high effeciency in vivo, survive cytotoxic therapies, and eventually lead to therapy resistance and recurrence. In this review, we would like to present recent knowledges on LCSCs, including the origins where they come from, the molecular features to identify them, and key mechanisms for them to survive and develop resistance, in order to provide a better view for targeting them in future clinic.
.
Collapse
Affiliation(s)
- Zhenhua Pan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin 300052, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin 300052, China
| | - Jun Chen
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin 300052, China.,Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
29
|
Gkountakos A, Centonze G, Vita E, Belluomini L, Milella M, Bria E, Milione M, Scarpa A, Simbolo M. Identification of Targetable Liabilities in the Dynamic Metabolic Profile of EGFR-Mutant Lung Adenocarcinoma: Thinking beyond Genomics for Overcoming EGFR TKI Resistance. Biomedicines 2022; 10:biomedicines10020277. [PMID: 35203491 PMCID: PMC8869286 DOI: 10.3390/biomedicines10020277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 01/27/2023] Open
Abstract
The use of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) as first-line treatment in patients with lung adenocarcinoma (LUAD) harboring EGFR-activating mutations has resulted in a dramatic improvement in the management of the disease. However, the long-term clinical benefit is inevitably compromised by multiple resistance mechanisms. Accumulating evidence suggests that metabolic landscape remodeling is one of the mechanisms that EGFR-mutant LUAD cells activate, thus acquiring higher plasticity, tolerating EGFR TKI-mediated cytotoxic stress, and sustaining their oncogenic phenotype. Several metabolic pathways are upregulated in EGFR TKI-resistant models modulating the levels of numerous metabolites such as lipids, carbohydrates, and metabolic enzymes which have been suggested as potential mediators of resistance to EGFR TKIs. Moreover, metabolites have been shown to carry signals and stimulate oncogenic pathways and tumor microenvironment (TME) components such as fibroblasts, facilitating resistance to EGFR TKIs in various ways. Interestingly, metabolic signatures could function as predictive biomarkers of EGFR TKI efficacy, accurately classifying patients with EGFR-mutant LUAD. In this review, we present the identified metabolic rewiring mechanisms and how these act either independently or in concert with epigenetic or TME elements to orchestrate EGFR TKI resistance. Moreover, we discuss potential nutrient dependencies that emerge, highlighting them as candidate druggable metabolic vulnerabilities with already approved drugs which, in combination with EGFR TKIs, might counteract the solid challenge of resistance, hopefully prolonging the clinical benefit.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
| | - Giovanni Centonze
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (G.C.); (M.M.)
| | - Emanuele Vita
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.V.); (E.B.)
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Belluomini
- Medical Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy; (L.B.); (M.M.)
| | - Michele Milella
- Medical Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy; (L.B.); (M.M.)
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.V.); (E.B.)
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Massimo Milione
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (G.C.); (M.M.)
| | - Aldo Scarpa
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Michele Simbolo
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
- Correspondence:
| |
Collapse
|
30
|
The Epithelial-Mesenchymal Transition at the Crossroads between Metabolism and Tumor Progression. Int J Mol Sci 2022; 23:ijms23020800. [PMID: 35054987 PMCID: PMC8776206 DOI: 10.3390/ijms23020800] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/12/2022] Open
Abstract
The transition between epithelial and mesenchymal phenotype is emerging as a key determinant of tumor cell invasion and metastasis. It is a plastic process in which epithelial cells first acquire the ability to invade the extracellular matrix and migrate into the bloodstream via transdifferentiation into mesenchymal cells, a phenomenon known as epithelial–mesenchymal transition (EMT), and then reacquire the epithelial phenotype, the reverse process called mesenchymal–epithelial transition (MET), to colonize a new organ. During all metastatic stages, metabolic changes, which give cancer cells the ability to adapt to increased energy demand and to withstand a hostile new environment, are also important determinants of successful cancer progression. In this review, we describe the complex interaction between EMT and metabolism during tumor progression. First, we outline the main connections between the two processes, with particular emphasis on the role of cancer stem cells and LncRNAs. Then, we focus on some specific cancers, such as breast, lung, and thyroid cancer.
Collapse
|
31
|
Resveratrol Analog 4-Bromo-Resveratrol Inhibits Gastric Cancer Stemness through the SIRT3-c-Jun N-Terminal Kinase Signaling Pathway. Curr Issues Mol Biol 2021; 44:63-72. [PMID: 35723384 PMCID: PMC8929134 DOI: 10.3390/cimb44010005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/24/2022] Open
Abstract
Chemotherapy is the treatment of choice for gastric cancer, but the currently available therapeutic drugs have limited efficacy. Studies have suggested that gastric cancer stem cells may play a key role in drug resistance in chemotherapy. Therefore, new agents that selectively target gastric cancer stem cells in gastric tumors are urgently required. Sirtuin-3 (SIRT3) is a deacetylase that regulates mitochondrial metabolic homeostasis to maintain stemness in glioma stem cells. Targeting the mitochondrial protein SIRT3 may provide a novel therapeutic option for gastric cancer treatment. However, the mechanism by which stemness is regulated through SIRT3 inhibition in gastric cancer remains unknown. We evaluated the stemness inhibition ability of the SIRT3 inhibitor 4′-bromo-resveratrol (4-BR), an analog of resveratrol in human gastric cancer cells. Our results suggested that 4-BR inhibited gastric cancer cell stemness through the SIRT3-c-Jun N-terminal kinase pathway and may aid in gastric cancer stem-cell–targeted therapy.
Collapse
|
32
|
Qiu F, Jin Y, Pu J, Huang Y, Hou J, Zhao X, Lu Y. Aberrant FBXW7-mediated ubiquitination and degradation of ZMYND8 enhances tumor progression and stemness in bladder cancer. Exp Cell Res 2021; 407:112807. [PMID: 34487730 DOI: 10.1016/j.yexcr.2021.112807] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022]
Abstract
ZMYND8, an epigenetic regulator, was identified as a common oncogene across various tumors. However, little was reported about the association between ZMYND8 and bladder cancer. Besides, aberrant mechanisms that contribute to abnormal ZMYND8 expressions still remain unclear. In the current study, we first found that ZMYND8 protein levels were significantly elevated in Bca samples versus normal tissues, but not the mRNA levels. We then utilized the Cell Counting Kit-8 (CCK-8) assay, clone formation assay and transwell analysis to confirm that ZMYND8 could remarkably promote the tumor progression in vitro, including growth capacity and migration. Bioinformatic predictive analysis revealed that E3 ubiquitin ligase FBXW7 interacts directly with ZMYND8 and degrades ZMYND8 in a polyubiquitination manner. Low FBXW7 was a hazard factor for promoting and depending on accumulated ZMYND8 proteins to promote Bca progression. Gene set enrichment analysis (GSEA) further indicated that ZMYND8 was notably associated with stemness process, which was well functionally validated. Lastly, ZMYND8 deficiency was observed to inhibit tumor growth of Bca in vivo, revealing a promising translational significance in Bca treatment. In conclusion, our study for the first time provided evidence for a novel mechanism of FBXW7/ZMYND8 axis in Bca, providing therapeutic vulnerability for individualized cancer treatment.
Collapse
Affiliation(s)
- Feng Qiu
- Department of Urology, The First Affiliated Hospital of Soochow University, China
| | - Yichen Jin
- Department of Urology, The First Affiliated Hospital of Soochow University, China
| | - Jinxian Pu
- Department of Urology, The First Affiliated Hospital of Soochow University, China
| | - Yuhua Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, China
| | - Xiaojun Zhao
- Department of Urology, The First Affiliated Hospital of Soochow University, China
| | - Yong Lu
- Department of Urology, The First Affiliated Hospital of Soochow University, China.
| |
Collapse
|
33
|
Zhang KR, Zhang YF, Lei HM, Tang YB, Ma CS, Lv QM, Wang SY, Lu LM, Shen Y, Chen HZ, Zhu L. Targeting AKR1B1 inhibits glutathione de novo synthesis to overcome acquired resistance to EGFR-targeted therapy in lung cancer. Sci Transl Med 2021; 13:eabg6428. [PMID: 34613810 DOI: 10.1126/scitranslmed.abg6428] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ke-Ren Zhang
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu-Fei Zhang
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hui-Min Lei
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ya-Bin Tang
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chun-Shuang Ma
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian-Ming Lv
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shi-Yi Wang
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li-Ming Lu
- Central Laboratory, Shanghai Chest Hospital and Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong-Zhuan Chen
- Department of Clinical Pharmacy, Institute of Interdisciplinary Integrative Biomedical Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences and Respiratory Department, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
34
|
Apatinib suppresses lung cancer stem-like cells by complex interplay between β-catenin signaling and mitochondrial ROS accumulation. Cell Death Discov 2021; 7:102. [PMID: 33980809 PMCID: PMC8115647 DOI: 10.1038/s41420-021-00480-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/21/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023] Open
Abstract
The abnormal activation of Wnt/β-catenin signaling plays a critical role in the development of lung cancer, which is also important in the generation and maintenance of lung cancer stem cell (CSC). CSCs have unique capabilities to resist anticancer therapy, seed recurrent tumors, and disseminate to and colonize distant tissues. Apatinib, a small-molecule VEGFR2-tyrosine kinase inhibitor, shows highly efficient antitumor activity in heavily treated, chemoresistant, and metastatic lung cancer. We speculated that inhibition of Wnt/β-catenin signaling and targeting lung CSCs could be one of the anti-tumor mechanisms of apatinib. In the present study we demonstrated that apatinib repressed lung CSC-like traits by hindering sphere formation ability, lung CSC-related marker expression and decreasing chemoresistance derived stemness. Mechanistically, apatinib exerted its anti-CSC effects by inhibiting β-catenin and its downstream targets. Moreover, apatinib induced the production of reactive oxyen species (ROS), which participated in the inhibitory effects of apatinib on lung CSCs. It was found that β-catenin regulated apatinib-induced production of ROS. Inhibition or promotion of ROS production with N-acetyl-L-cysteine or H2O2 not only upregulated or downregulated β-catenin expression, but also prevented or promoted DNA damage, rescued or impeded sphere formation, respectively. Collectively, our findings reveal that apatinib directly inhibits β-catenin signaling and promotes ROS generation to suppress lung CSC-like characteristics. A clearer understanding of the anti-cancer mechanisms of apatinib is required for its better application in combating advanced and refractory/recurrent lung cancer when combined with conventional chemotherapy.
Collapse
|
35
|
Wang Z, Zhang X, Tian X, Yang Y, Ma L, Wang J, Yu Y. CREB stimulates GPX4 transcription to inhibit ferroptosis in lung adenocarcinoma. Oncol Rep 2021; 45:88. [PMID: 33846793 PMCID: PMC8042667 DOI: 10.3892/or.2021.8039] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis is a new form of regulated cell death and closely related to cancer. However, the mechanism underlying the regulation of ferroptosis in lung adenocarcinoma (LUAD) remains unclear. IB, IHC and ELISA were performed to analyze protein expression. RT-qPCR was used to analyze mRNA expression. Cell viability, 3D cell growth, MDA, the generation of lipid ROS and the Fe2+ concentration were measured to evaluate the responses to the induction of ferroptosis. Measurement of luciferase activity and ChIP were used to analyze the promoter activity regulated by the transcriptional regulator. Co-IP assays were performed to identify protein-protein interactions. In the present study, it was revealed that cAMP response element-binding protein (CREB) was highly expressed in LUAD, and knockdown of CREB inhibited cell viability and growth by promoting apoptosis- and ferroptosis-like cell death, concurrently. It was observed that CREB suppressed lipid peroxidation by binding the promoter region of glutathione peroxidase 4 (GPX4), and this binding could be enhanced by E1A binding protein P300 (EP300). The bZIP domain in CREB and the CBP/p300-HAT domain in EP300 were essential for CREB-EP300 binding in LUAD cells. Finally, it was revealed that CREB, GPX4, EP300 and 4-HNE were closely related to tumor size and stage, and tumors with a higher degree of malignancy were more likely to have a low degree of lipid peroxidation. Therefore, targeting this CREB/EP300/GPX4 axis may provide new strategies for treating LUAD.
Collapse
Affiliation(s)
- Zhixian Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Xiao Zhang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Xiaoting Tian
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yueyue Yang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, P.R. China
| | - Lifang Ma
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Jiayi Wang
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Yongchun Yu
- Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
36
|
Ma CS, Lv QM, Zhang KR, Tang YB, Zhang YF, Shen Y, Lei HM, Zhu L. NRF2-GPX4/SOD2 axis imparts resistance to EGFR-tyrosine kinase inhibitors in non-small-cell lung cancer cells. Acta Pharmacol Sin 2021; 42:613-623. [PMID: 32704041 PMCID: PMC8115089 DOI: 10.1038/s41401-020-0443-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022]
Abstract
Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have achieved satisfactory clinical effects in the therapy of non-small cell lung cancer (NSCLC), but acquired resistance limits their clinical application. NRF2 has been shown to enhance the resistance to apoptosis induced by radiotherapy and some chemotherapy. In this study, we investigated the role of NRF2 in resistance to EGFR-TKIs. We showed that NRF2 protein levels were markedly increased in a panel of EGFR-TKI-resistant NSCLC cell lines due to slow degradation of NRF2 protein. NRF2 knockdown overcame the resistance to EGFR-TKIs in HCC827ER and HCC827GR cells. Furthermore, we demonstrated that NRF2 imparted EGFR-TKIs resistance in HCC827 cells via upregulation of GPX4 and SOD2, and suppression of GPX4 and SOD2 reversed resistance to EGFR-TKIs. Thus, we conclude that targeting NRF2-GPX4/SOD2 pathway is a potential strategy for overcoming resistance to EGFR-TKIs.
Collapse
|
37
|
Bao S, Zheng H, Ye J, Huang H, Zhou B, Yao Q, Lin G, Zhang H, Kou L, Chen R. Dual Targeting EGFR and STAT3 With Erlotinib and Alantolactone Co-Loaded PLGA Nanoparticles for Pancreatic Cancer Treatment. Front Pharmacol 2021; 12:625084. [PMID: 33815107 PMCID: PMC8017486 DOI: 10.3389/fphar.2021.625084] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most common malignancies and also a leading cause of cancer-related mortality worldwide. Many studies have shown that epidermal growth factor receptor (EGFR) is highly expressed in PC, which provides a potential target for PC treatment. However, EGFR inhibitors use alone was proven ineffective in clinical trials, due to the persistence of cellular feedback mechanisms which foster therapeutic resistance to single targeting of EGFR. Specifically, the signal transducer and activator of transcription 3 (STAT3) is over-activated when receiving an EGFR inhibitor and is believed to be highly involved in the failure and resistance of EGFR inhibitor treatment. Therein, we hypothesized that dual inhibition of EGFR and STAT3 strategy could address the STAT3 induced resistance during EGFR inhibitor treatment. To this end, we tried to develop poly (lactic-co-glycolic acid) (PLGA) nanoparticles to co-load Alantolactone (ALA, a novel STAT3 inhibitor) and Erlotinib (ERL, an EGFR inhibitor) for pancreatic cancer to test our guess. The loading ratio of ALA and ERL was firstly optimized in vitro to achieve a combined cancer-killing effect. Then, the ALA- and ERL-co-loaded nanoparticles (AE@NPs) were successfully prepared and characterized, and the related anticancer effects and cellular uptake of AE@NPs were studied. We also further detailly explored the underlying mechanisms. The results suggested that AE@NPs with uniform particle size and high drug load could induce significant pancreatic cancer cell apoptosis and display an ideal anticancer effect. Mechanism studies showed that AE@NPs inhibited the phosphorylation of both EGFR and STAT3, indicating the dual suppression of these two signaling pathways. Additionally, AE@NPs could also activate the ROS-p38 axis, which is not observed in the single drug treatments. Collectively, the AE@NPs prepared in this study possess great potential for pancreatic cancer treatment by dual suppressing of EGFR and STAT3 pathways and activating ROS-responsive p38 MAPK pathway.
Collapse
Affiliation(s)
- Shihui Bao
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
| | - Hailun Zheng
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
| | - Jinyao Ye
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huirong Huang
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Bin Zhou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Guangyong Lin
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
| | - Hailin Zhang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
- Department of Children’s Respiration Disease, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
| | - Ruijie Chen
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Wenzhou, China
| |
Collapse
|
38
|
Targeting NAD-dependent dehydrogenases in drug discovery against infectious diseases and cancer. Biochem Soc Trans 2021; 48:693-707. [PMID: 32311017 DOI: 10.1042/bst20191261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 12/24/2022]
Abstract
Dehydrogenases are oxidoreductase enzymes that play a variety of fundamental functions in the living organisms and have primary roles in pathogen survival and infection processes as well as in cancer development. We review here a sub-set of NAD-dependent dehydrogenases involved in human diseases and the recent advancements in drug development targeting pathogen-associated NAD-dependent dehydrogenases. We focus also on the molecular aspects of the inhibition process listing the structures of the most relevant molecules targeting this enzyme family. Our aim is to review the most impacting findings regarding the discovery of novel inhibitory compounds targeting the selected NAD-dependent dehydrogenases involved in cancer and infectious diseases.
Collapse
|
39
|
Grimley E, Cole AJ, Luong TT, McGonigal SC, Sinno S, Yang D, Bernstein KA, Buckanovich RJ. Aldehyde dehydrogenase inhibitors promote DNA damage in ovarian cancer and synergize with ATM/ATR inhibitors. Am J Cancer Res 2021; 11:3540-3551. [PMID: 33664846 PMCID: PMC7914353 DOI: 10.7150/thno.51885] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022] Open
Abstract
Rationale: Aldehyde dehydrogenase (ALDH) enzymes are often upregulated in cancer cells and associated with therapeutic resistance. ALDH enzymes protect cells by metabolizing toxic aldehydes which can induce DNA double stand breaks (DSB). We recently identified a novel ALDH1A family inhibitor (ALDHi), 673A. We hypothesized that 673A, via inhibition of ALDH1A family members, could induce intracellular accumulation of genotoxic aldehydes to cause DSB and that ALDHi could synergize with inhibitors of the ATM and ATR, proteins which direct DSB repair. Methods: We used immunofluorescence to directly assess levels of the aldehyde 4-hydroxynonenal and comet assays to evaluate DSB. Western blot was used to evaluate activation of the DNA damage response pathways. Cell counts were performed in the presence of 673A and additional aldehydes or aldehyde scavengers. ALDH inhibition results were confirmed using ALDH1A3 CRISPR knockout. Synergy between 673A and ATM or ATR inhibitors was evaluated using the Chou-Talalay method and confirmed in vivo using cell line xenograft tumor studies. Results: The ALDHi 673A cellular accumulation of toxic aldehydes which induce DNA double strand breaks. This is exacerbated by addition of exogenous aldehydes such as vitamin-A (retinaldehyde) and ameliorated by aldehyde scavengers such as metformin and hydralazine. Importantly, ALDH1A3 knockout cells demonstrated increased sensitivity to ATM/ATR inhibitors. And, ALDHi synergized with inhibitors of ATM and ATR, master regulators of the DSB DNA damage response, both in vitro and in vivo. This synergy was evident in homologous recombination (HR) proficient cell lines. Conclusions: ALDHi can be used to induce DNA DSB in cancer cells and synergize with inhibitors the ATM/ATR pathway. Our data suggest a novel therapeutic approach to target HR proficient ovarian cancer cells.
Collapse
|
40
|
Liang Q, Gu WM, Huang K, Luo MY, Zou JH, Zhuang GL, Lei HM, Chen HZ, Zhu L, Zhou L, Shen Y. HKB99, an allosteric inhibitor of phosphoglycerate mutase 1, suppresses invasive pseudopodia formation and upregulates plasminogen activator inhibitor-2 in erlotinib-resistant non-small cell lung cancer cells. Acta Pharmacol Sin 2021; 42:115-119. [PMID: 32404981 DOI: 10.1038/s41401-020-0399-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/15/2020] [Indexed: 12/23/2022]
Abstract
Acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), such as erlotinib, remains a major challenge in the targeted therapy of non-small cell lung cancer (NSCLC). HKB99 is a novel allosteric inhibitor of phosphoglycerate mutase 1 (PGAM1) that preferentially suppresses cell proliferation and induces more apoptosis in acquired erlotinib-resistant HCC827ER cells compared with its parental HCC827 cells. In this study we identified the molecular biomarkers for HKB99 response in erlotinib-resistant HCC827ER cells. We showed that HCC827ER cells displayed enhanced invasive pseudopodia structures as well as downregulated plasminogen activator inhibitor-2 (PAI-2). Meanwhile, PAI-2 knockdown by siPAI-2 candidates decreased the sensitivity of HCC827 parental cells to erlotinib. Moreover, HKB99 (5 μM) preferentially inhibited the invasive pseudopodia formation and increased the level of PAI-2 in HCC827ER cells. Collectively, this study provides new insight into the role of PAI-2 in regulating the sensitivity of erlotinib resistant NSCLC cells to PGAM1 inhibitor. Furthermore, PAI-2 level might be considered as a potential biomarker for predicting the efficacy of the PGAM1 allosteric inhibitor on the erlotinib resistant NSCLC cells.
Collapse
|
41
|
Aramini B, Masciale V, Bianchi D, Manfredini B, Banchelli F, D'Amico R, Bertolini F, Dominici M, Morandi U, Maiorana A. ALDH Expression in Angiosarcoma of the Lung: A Potential Marker of Aggressiveness? Front Med (Lausanne) 2020; 7:544158. [PMID: 33195295 PMCID: PMC7662079 DOI: 10.3389/fmed.2020.544158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/09/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Primary angiosarcoma of the lung is a very aggressive rare malignant disease resulting in a severe prognosis (1). This type of cancer represents about 2% of all soft tissue sarcomas and has a high rate of metastasis through the hematogenous route. For the rarity of this malignant vascular tumor it is still challenging to set a diagnosis (1). The diagnostic features that have thus far been considered include primarily clinical and radiological findings. In some cases, immunohistochemical characteristics based on the most common markers used in pathology have been described. The aim of this report is to present two cases of angiosarcoma of the lung in which the aldehyde dehydrogenase (ALDH) marker was analyzed by immunohistochemistry. Methods: We report two cases of angiosarcoma of the lung in patients underwent lung surgery at our Unit. In addition to the standard histopathological analysis for this disease, immunohistochemistry using an ALDH1A1 antibody was performed in both of the cases. For ALDH quantification, a semi-quantitative method based on the positivity of the tumor cells was used: 0 (<5%), 1 (5–25%), 2 (>25–50%), 3 (>50–75%), 4 (>75%). Results: One patient with recurrent lung disease survived, achieving complete remission after chemo- and radiotherapy. The second patient died of recurrent disease within 5 years of diagnosis. ALDH1A1 was evaluated in both of these cases using an immunohistochemistry scoring system based on the positivity for this marker. The scores were consistent with the patients' clinical outcomes, as the lower (score 1) was observed in the patient with the better clinical outcome, while the higher (score 3) was seen in the patient with the worse outcome. Conclusion: Our data suggest that ALDH may be an important clinical marker in angiosarcoma of the lung. Although further studies need to be performed in a larger cohort of patients, we believe that, if the results will be confirmed, ALDH1A1 may be used to stratify patients in terms of prognosis and for targeted therapy.
Collapse
Affiliation(s)
- Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Daniel Bianchi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Manfredini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Department of Medical and Surgical Sciences, Center of Statistic, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D'Amico
- Department of Medical and Surgical Sciences, Center of Statistic, University of Modena and Reggio Emilia, Modena, Italy
| | - Federica Bertolini
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Department of Medical and Surgical Sciences, Institute of Pathology, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
42
|
Li B, Yang L, Peng X, Fan Q, Wei S, Yang S, Li X, Jin H, Wu B, Huang M, Tang S, Liu J, Li H. Emerging mechanisms and applications of ferroptosis in the treatment of resistant cancers. Biomed Pharmacother 2020; 130:110710. [PMID: 33568263 DOI: 10.1016/j.biopha.2020.110710] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/09/2023] Open
Abstract
The development of chemotherapy drugs has promoted anticancer treatment, but the effect on tumours is not clear because of treatment resistance; thus, it is necessary to further understand the mechanism of cell death to explore new therapeutic targets. As a new type of programmed cell death, ferroptosis is increasingly being targeted in the treatment of many cancers with clinical drugs and experimental compounds. Ferroptosis is stimulated in tumours with inherently high levels of ferrous ions by a reaction with abundant polyunsaturated fatty acids and the inhibition of antioxidant enzymes, which can overcome treatment resistance in cancers mainly through GPX4. In this review, we focus on the intrinsic cellular regulators against ferroptosis in cancer resistance, such as GPX4, NRF2 and the thioredoxin system. We summarize the application of novel compounds and drugs to circumvent treatment resistance. We also introduce the application of nanoparticles for the treatment of resistant cancers. In conclusion, targeting ferroptosis represents a considerable strategy for resistant cancer treatment.
Collapse
Affiliation(s)
- Bowen Li
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Liang Yang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Xueqiang Peng
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Qin Fan
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Shibo Wei
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Shuo Yang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Xinyu Li
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Hongyuan Jin
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Bo Wu
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Mingyao Huang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Shilei Tang
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Jingang Liu
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China
| | - Hangyu Li
- Department of General Surgery, the Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, Liaoning, China.
| |
Collapse
|
43
|
Ramesh V, Brabletz T, Ceppi P. Targeting EMT in Cancer with Repurposed Metabolic Inhibitors. Trends Cancer 2020; 6:942-950. [PMID: 32680650 DOI: 10.1016/j.trecan.2020.06.005] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/31/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) determines the most lethal features of cancer, metastasis formation and chemoresistance, and therefore represents an attractive target in oncology. However, direct targeting of EMT effector molecules is, in most cases, pharmacologically challenging. Since emerging research has highlighted the distinct metabolic circuits involved in EMT, we propose the use of metabolism-specific inhibitors, FDA approved or under clinical trials, as a drug repurposing approach to target EMT in cancer. Metabolism-inhibiting drugs could be coupled with standard chemo- or immunotherapy to combat EMT-driven resistant and aggressive cancers.
Collapse
Affiliation(s)
- Vignesh Ramesh
- Interdisciplinary Centre for Clinical Research, University Hospital Erlangen, FAU-Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine-I and Comprehensive Cancer Center Erlangen-EMN, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Paolo Ceppi
- Interdisciplinary Centre for Clinical Research, University Hospital Erlangen, FAU-Erlangen-Nuremberg, Erlangen, Germany; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
44
|
Wang X, Li M, Peng L, Tang N. SOD2 promotes the expression of ABCC2 through lncRNA CLCA3p and improves the detoxification capability of liver cells. Toxicol Lett 2020; 327:9-18. [PMID: 32201199 DOI: 10.1016/j.toxlet.2020.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/17/2022]
Abstract
Superoxide dismutase 2 (SOD2) is a key enzyme for scavenging reactive oxygen species produced by mitochondria, which plays an important role in maintaining cellular homeostasis. However, its effects on the detoxification capability of liver cells have not been reported. In this study, we found that change in SOD2 expression affects the proliferation of liver cells. Genome-wide microarray analysis showed that SOD2 positively regulates the drug transporter ABCC2, and co-expression analysis suggested that lncRNA CLCA3P participates in the process. Further experiments showed that SOD2 can promote the expression of CLCA3P, which increases the transcription of ABCC2 by interacting with the transcription factor IRF1. By increasing ABCC2 expression SOD2 facilitates drugs efflux of liver cells and thus promotes their survival under a drug-toxic environment. This study elucidates the improvement of the detoxification of liver cells by a regulatory axis, SOD2-CLCA3P-IRF1-ABCC2, and provides novel insight into the modification of human liver cells that can be applied to bioartificial liver system or the study of SOD2 in drug metabolism.
Collapse
Affiliation(s)
- Xiaoqian Wang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Man Li
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lirong Peng
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Nanhong Tang
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Research Center for Molecular Medicine, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
45
|
Chin TM, Boopathy GTK, Man EP, Clohessy JG, Csizmadia E, Quinlan MP, Putti T, Wan SC, Xie C, Ali A, Wai FC, Ong YS, Goh BC, Settleman J, Hong W, Levantini E, Tenen DG. Targeting microtubules sensitizes drug resistant lung cancer cells to lysosomal pathway inhibitors. Am J Cancer Res 2020; 10:2727-2743. [PMID: 32194831 PMCID: PMC7052910 DOI: 10.7150/thno.38729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/24/2019] [Indexed: 12/20/2022] Open
Abstract
Oncogene-addicted cancers are predominantly driven by specific oncogenic pathways and display initial exquisite sensitivity to designer therapies, but eventually become refractory to treatments. Clear understanding of lung tumorigenic mechanisms is essential for improved therapies. Methods: Lysosomes were analyzed in EGFR-WT and mutant cells and corresponding patient samples using immunofluorescence or immunohistochemistry and immunoblotting. Microtubule organization and dynamics were studied using immunofluorescence analyses. Also, we have validated our findings in a transgenic mouse model that contain EGFR-TKI resistant mutations. Results: We herein describe a novel mechanism that a mutated kinase disrupts the microtubule organization and results in a defective endosomal/lysosomal pathway. This prevents the efficient degradation of phosphorylated proteins that become trapped within the endosomes and continue to signal, therefore amplifying downstream proliferative and survival pathways. Phenotypically, a distinctive subcellular appearance of LAMP1 secondary to microtubule dysfunction in cells expressing EGFR kinase mutants is seen, and this may have potential diagnostic applications for the detection of such mutants. We demonstrate that lysosomal-inhibitors re-sensitize resistant cells to EGFR tyrosine-kinase inhibitors (TKIs). Identifying the endosome-lysosome pathway and microtubule dysfunction as a mechanism of resistance allows to pharmacologically intervene on this pathway. Conclusions: We find that the combination of microtubule stabilizing agent and lysosome inhibitor could reduce the tumor progression in EGFR TKI resistant mouse models of lung cancer.
Collapse
|