1
|
Hu Q, Tang Z, Lynch A, Freeman B, Fujioka N, Salloum RG, Malaty J, Orlando FA, Langaee T, Huo Z, Xing C. One-Week Kava Dietary Supplementation Increases Both Urinary N- and O-Glucuronides of NNAL, a Lung Carcinogen Major Metabolite, among Smokers. Chem Res Toxicol 2024; 37:1515-1523. [PMID: 39001862 DOI: 10.1021/acs.chemrestox.4c00109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Abstract
4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone (commonly known as NNK) is one of the most prevalent and potent pulmonary carcinogens in tobacco products that increases the human lung cancer risk. Kava has the potential to reduce NNK and tobacco smoke-induced lung cancer risk by enhancing urinary excretion of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL, the major metabolite of NNK) and thus reducing NNK-induced DNA damage. In this study, we quantified N-glucuronidated NNAL (NNAL-N-gluc), O-glucuronidated NNAL (NNAL-O-gluc), and free NNAL in the urine samples collected before and after 1-week kava dietary supplementation. The results showed that kava increased both NNAL-N-glucuronidation and O-glucuronidation. Since NNAL-N-glucuronidation is dominantly catalyzed by UGT2B10, its representative single-nucleotide polymorphisms (SNPs) were analyzed among the clinical trial participants. Individuals with any of the four analyzed SNPs appear to have a reduced basal capacity in NNAL-N-glucuronidation. Among these individuals, kava also resulted in a smaller extent of increases in NNAL-N-glucuronidation, suggesting that participants with those UGT2B10 SNPs may not benefit as much from kava with respect to enhancing NNAL-N-glucuronidation. In summary, our results provide further evidence that kava enhances NNAL urinary detoxification via an increase in both N-glucuronidation and O-glucuronidation. UGT2B10 genetic status has not only the potential to predict the basal capacity of the participants in NNAL-N-glucuronidation but also potentially the extent of kava benefits.
Collapse
Affiliation(s)
- Qi Hu
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Zhixin Tang
- Department of Biostatistics, Colleges of Public Health & Health Professions and Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Allison Lynch
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Breanne Freeman
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Naomi Fujioka
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ramzi G Salloum
- Department of Health Outcomes & Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - John Malaty
- Department of Community Health & Family Medicine, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Frank A Orlando
- Department of Community Health & Family Medicine, College of Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Taimour Langaee
- Center for Pharmacogenomics and Precision Medicine, Department of Pharmacotherapy & Translational Research, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Zhiguang Huo
- Department of Biostatistics, Colleges of Public Health & Health Professions and Medicine, University of Florida, Gainesville, Florida 32610, United States
| | - Chengguo Xing
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
2
|
Hushmandi K, Saadat SH, Mirilavasani S, Daneshi S, Aref AR, Nabavi N, Raesi R, Taheriazam A, Hashemi M. The multifaceted role of SOX2 in breast and lung cancer dynamics. Pathol Res Pract 2024; 260:155386. [PMID: 38861919 DOI: 10.1016/j.prp.2024.155386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/09/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Breast and lung cancers are leading causes of death among patients, with their global mortality and morbidity rates increasing. Conventional treatments often prove inadequate due to resistance development. The alteration of molecular interactions may accelerate cancer progression and treatment resistance. SOX2, known for its abnormal expression in various human cancers, can either accelerate or impede cancer progression. This review focuses on examining the role of SOX2 in breast and lung cancer development. An imbalance in SOX2 expression can promote the growth and dissemination of these cancers. SOX2 can also block programmed cell death, affecting autophagy and other cell death mechanisms. It plays a significant role in cancer metastasis, mainly by regulating the epithelial-to-mesenchymal transition (EMT). Additionally, an imbalanced SOX2 expression can cause resistance to chemotherapy and radiation therapy in these cancers. Genetic and epigenetic factors may affect SOX2 levels. Pharmacologically targeting SOX2 could improve the effectiveness of breast and lung cancer treatments.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, the Islamic Republic of Iran.
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Seyedalireza Mirilavasani
- Campus Venlo, Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, The Netherlands
| | - Salman Daneshi
- Department of Public Health,School of Health,Jiroft University of Medical Sciences,Jiroft, the Islamic Republic of Iran
| | - Amir Reza Aref
- Department of Translational Sciences, Xsphera Biosciences Inc. Boston, MA, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6 Canada
| | - Rasoul Raesi
- Department of Health Services Management, School of Health, Mashhad University of Medical Sciences, Mashhad, Iran.; Department of Nursing, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran.
| |
Collapse
|
3
|
Meyer ML, Hirsch FR, Bunn PA, Ujhazy P, Fredrickson D, Berg CD, Carbone DP, Halmos B, Singh H, Borghaei H, Ferris A, Langer C, Dacic S, Mok TS, Peters S, Johnson BE. Calls to action on lung cancer management and research. Oncologist 2024:oyae169. [PMID: 39002167 DOI: 10.1093/oncolo/oyae169] [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: 03/18/2024] [Accepted: 05/24/2024] [Indexed: 07/15/2024] Open
Abstract
Lung cancer, the leading cause of cancer-related deaths globally, remains a pressing health issue despite significant medical advances. The New York Lung Cancer Foundation brought together experts from academia, the pharmaceutical and biotech industries as well as organizational leaders and patient advocates, to thoroughly examine the current state of lung cancer diagnosis, treatment, and research. The goal was to identify areas where our understanding is incomplete and to develop collaborative public health and scientific strategies to generate better patient outcomes, as highlighted in our "Calls to Action." The consortium prioritized 8 different calls to action. These include (1) develop strategies to cure more patients with early-stage lung cancer, (2) investigate carcinogenesis leading to lung cancers in patients without a history of smoking, (3) harness precision medicine for disease interception and prevention, (4) implement solutions to deliver prevention measures and effective therapies to individuals in under-resourced countries, (5) facilitate collaborations with industry to collect and share data and samples, (6) create and maintain open access to big data repositories, (7) develop new immunotherapeutic agents for lung cancer treatment and prevention, and (8) invest in research in both the academic and community settings. These calls to action provide guidance to representatives from academia, the pharmaceutical and biotech industries, organizational and regulatory leaders, and patient advocates to guide ongoing and planned initiatives.
Collapse
Affiliation(s)
- May-Lucie Meyer
- Hematology and Oncology Department, Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine and Thoracic Oncology Center, New York, NY, United States
| | - Fred R Hirsch
- Hematology and Oncology Department, Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine and Thoracic Oncology Center, New York, NY, United States
| | - Paul A Bunn
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Peter Ujhazy
- Translational Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, United States
| | | | | | - David P Carbone
- Division of Medical Oncology, The Ohio State University-James Comprehensive Cancer Center, Columbus, OH, United States
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Harpreet Singh
- US Food and Drug Administration (FDA), Washington, DC, United States
| | | | | | - Corey Langer
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sanja Dacic
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Tony S Mok
- State Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Solange Peters
- Department of Oncology, University Hospital CHUV, Lausanne, Switzerland
| | - Bruce E Johnson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Guo S, Ding R, Zhao Q, Wang X, Lv S, Ji XY. Recent Insights into the Roles of PEST-Containing Nuclear Protein. Mol Biotechnol 2024:10.1007/s12033-024-01188-5. [PMID: 38762838 DOI: 10.1007/s12033-024-01188-5] [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: 11/05/2023] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
PEST-containing nuclear protein (PCNP), a short-lived small nuclear protein with 178 amino acids, is a nuclear protein containing two PEST sequences. PCNP is highly expressed in several malignant tumors such as cervical cancer, rectal cancer, and lung cancer. It is also associated with cell cycle regulation and the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and Wnt signaling pathways during tumor growth. The present article discuss how PCNP regulates the PI3K/AKT/mTOR and Wnt signaling pathways and related proteins, and the ubiquitination of PCNP regulates tumor cell cycle as well as the progress of the application of PCNP in the pathophysiology and treatment of colon cancer, human ovarian cancer, thyroid cancer, lung adenocarcinoma and oral squamous cell carcinoma. The main relevant articles were retrieved from PubMed, with keywords such as PEST-containing nuclear protein (PCNP), cancer (tumor), and signaling pathways as inclusion/exclusion criteria. Relevant references has been included and cited in the manuscript.
Collapse
Affiliation(s)
- Shiyun Guo
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Ruidong Ding
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Qian Zhao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Xu Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Shuangyu Lv
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China.
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, Kaifeng, 475004, Henan, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Mazhai, Erqi District, Zhengzhou, 450064, Henan, China.
| |
Collapse
|
5
|
Tian J, Zhao W, Wu Y, Shi Y, Yu J, Zhang W, Xing C, Zhuang C, Qu Z. Diallyl Disulfide Blocks Cigarette Carcinogen 4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanone-Induced Lung Tumorigenesis via Activation of the Nrf2 Antioxidant System and Suppression of NF-κB Inflammatory Response. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:17763-17774. [PMID: 37956253 DOI: 10.1021/acs.jafc.3c02007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Chemoprevention is a potential strategy to reduce lung cancer incidence and death. Recently, we reported that garlic oil significantly inhibits 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis. Diallyl disulfide (DADS) is a bioactive ingredient in garlic. Our goal was to examine the chemopreventive effectiveness and mechanism of DADS on NNK-triggered lung cancer in vivo and in vitro in the current investigation. The results indicated that DADS significantly reduced the number of lung nodules in the NNK-induced A/J mice. Consistent with the in vivo results, DADS markedly inhibited NNK-induced decrease of MRC-5 cells' viability. Mechanistically, DADS could promote Nrf2 dissociated from the Keap1-Nrf2 complex and accelerate Nrf2 nuclear translocation, which in turn upregulates its downstream target genes. Besides, DADS further inhibited the NF-κB signaling cascade, thus reducing the accumulation of inflammatory factors. Collectively, these discoveries supported the potential of DADS as a novel candidate for the chemoprevention of tobacco-carcinogen-induced lung cancer.
Collapse
Affiliation(s)
- Jiahui Tian
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Wenli Zhao
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Yanran Wu
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Ying Shi
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Jianqiang Yu
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Wannian Zhang
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Chengguo Xing
- Department of Medicinal Chemistry, University of Florida, 1345 Center Drive, Gainesville, Florida 32610, United States
| | - Chunlin Zhuang
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhuo Qu
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| |
Collapse
|
6
|
Moutabian H, Radi UK, Saleman AY, Adil M, Zabibah RS, Chaitanya MNL, Saadh MJ, Jawad MJ, Hazrati E, Bagheri H, Pal RS, Akhavan-Sigari R. MicroRNA-155 and cancer metastasis: Regulation of invasion, migration, and epithelial-to-mesenchymal transition. Pathol Res Pract 2023; 250:154789. [PMID: 37741138 DOI: 10.1016/j.prp.2023.154789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 09/25/2023]
Abstract
Among the leading causes of death globally has been cancer. Nearly 90% of all cancer-related fatalities are attributed to metastasis, which is the growing of additional malignant growths out of the original cancer origin. Therefore, a significant clinical need for a deeper comprehension of metastasis exists. Beginning investigations are being made on the function of microRNAs (miRNAs) in the metastatic process. Tiny non-coding RNAs called miRNAs have a crucial part in controlling the spread of cancer. Some miRNAs regulate migration, invasion, colonization, cancer stem cells' properties, the epithelial-mesenchymal transition (EMT), and the microenvironment, among other processes, to either promote or prevent metastasis. One of the most well-conserved and versatile miRNAs, miR-155 is primarily distinguished by overexpression in a variety of illnesses, including malignant tumors. It has been discovered that altered miR-155 expression is connected to a number of physiological and pathological processes, including metastasis. As a result, miR-155-mediated signaling pathways were identified as possible cancer molecular therapy targets. The current research on miR-155, which is important in controlling cancer cells' invasion, and metastasis as well as migration, will be summarized in the current work. The crucial significance of the lncRNA/circRNA-miR-155-mRNA network as a crucial regulator of carcinogenesis and a player in the regulation of signaling pathways or related genes implicated in cancer metastasis will be covered in the final section. These might provide light on the creation of fresh treatment plans for controlling cancer metastasis.
Collapse
Affiliation(s)
- Hossein Moutabian
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran
| | - Usama Kadem Radi
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Mv N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan; Applied Science Research Center. Applied Science Private University, Amman, Jordan
| | | | - Ebrahi Hazrati
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Hamed Bagheri
- Radiation Sciences Research Center (RSRC), AJA University of Medical Sciences, Tehran, Iran; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rashmi Saxena Pal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144402, India
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany; Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
7
|
Shaikh ZM, Bhatia N, Aradhye P, Anbarasu A, Gawde J, Thorat R, Mahimkar MB. Thearubigins/Polymeric black tea polyphenols (PBPs) do not prevent benzo[a]pyrene (B[a]P) induced lung tumors in A/J mice. Am J Transl Res 2023; 15:5826-5834. [PMID: 37854224 PMCID: PMC10579030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/15/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVES Previously we have demonstrated the chemopreventive effect of Thearubigins/Polymeric Black-tea Polyphenols (PBPs) upon pre-treatment to a combination of carcinogens, that is, Benzo[a]pyrene (B[a]P) and 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) which are present in Tobacco smoke (TS). However, the chemopreventive effect in response to B[a]P as a single carcinogen remains unexplored. B[a]P is a universal carcinogen and an important constituent of particulate matter 2.5 (PM2.5) found in the environment and in TS. METHODS We investigated the pre-treatment of Thearubigins/PBPs as a chemopreventive agent at three doses (1.5, 5, 10%) against B[a]P-induced lung carcinogenesis at early & late time points. We also investigated the effect of PBPs at early time points to understand molecular changes by employing western blotting in xenobiotic metabolism pathways, DNA damage, inflammation, apoptosis, and proliferation as they are modulated in response to carcinogens. We used 6-8 weeks male A/J mice for tumorigenicty and western blotting to probe the molecular biomarkers. RESULTS We report no decrease in tumor incidence and multiplicity upon pre and concurrent treatment of Thearubigins/PBPs. Further, we also report no changes in molecular markers at early time points, in agreement with former observations. CONCLUSION Our results suggest that chemopreventive agents need to be tested with different combinations of carcinogens and regimens to fully understand the complex interplay between carcinogenesis and the efficacy of chemopreventive agents. Studies like these will provide meaningful data before initiating large-scale clinical trials.
Collapse
Affiliation(s)
- Zaid M Shaikh
- Mahimkar Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial CentreNavi Mumbai 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School ComplexAnushakti Nagar, Mumbai 400094, Maharashtra, India
| | - Nirav Bhatia
- Mahimkar Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial CentreNavi Mumbai 410210, Maharashtra, India
| | - Prasad Aradhye
- Mahimkar Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial CentreNavi Mumbai 410210, Maharashtra, India
| | - Amarnath Anbarasu
- Mahimkar Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial CentreNavi Mumbai 410210, Maharashtra, India
| | - Jitendra Gawde
- Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC)Kharghar, Navi Mumbai 410210, Maharashtra, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC)Kharghar, Navi Mumbai 410210, Maharashtra, India
| | - Manoj B Mahimkar
- Mahimkar Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial CentreNavi Mumbai 410210, Maharashtra, India
- Homi Bhabha National Institute, Training School ComplexAnushakti Nagar, Mumbai 400094, Maharashtra, India
| |
Collapse
|
8
|
Tang H, Kuang Y, Wu W, Peng B, Fu Q. Quercetin inhibits the metabolism of arachidonic acid by inhibiting the activity of CYP3A4, thereby inhibiting the progression of breast cancer. Mol Med 2023; 29:127. [PMID: 37710176 PMCID: PMC10502985 DOI: 10.1186/s10020-023-00720-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/25/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND Recent years have witnessed impressive growth in applying natural medicine in tumor treatment. Saffron is reported to elicit an inhibitory property against BC. Herein, we sought to explore the specific components and mechanistic basis of saffron's anti-breast carcinoma (BC) function. METHODS Bioinformatics analysis was employed to analyze saffron components' anti-BC activity and screen the corresponding target genes involved in BC. Then, the roles of the main saffron ingredient quercetin in the activity of BC cells were examined using CCK-8, MTS, flow cytometry, colony formation, Transwell, and Gelatin zymogram assays. Additionally, the interactions among Quercetin, EET, and Stat3 were assessed by immunofluorescence and Western blot, and LC-MS/MS determined the levels of AA, EETs, and CYP3A. Finally, BC xenograft mouse models were established to verify the anti-BC function of Quercetin in vivo. RESULTS Quercetin, the main active component of saffron, inhibited BC progression. Quercetin suppressed BC cell growth, migration, and invasion and inhibited CYP3A4 expression and activity in BC. Mechanistically, Quercetin down-regulated CYP3A4 to block the nuclear translocation of Stat3 by decreasing the metabolization of AA to EETs, thereby alleviating BC. Moreover, exogenously added EETs counteracted the anti-tumor effect of Quercetin on BC. Quercetin also inhibited the tumor growth of tumor-bearing nude mice. CONCLUSION Quercetin could inhibit the activity of CYP3A to down-regulate AA metabolites EETs, consequently hampering p-Stat3 and nuclear translocation, thus impeding BC development.
Collapse
Affiliation(s)
- Huaming Tang
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Sichuan Province, Chengdu, 610000, People's Republic of China
| | - Yuanli Kuang
- Department of General Surgery, Chongqing Kaizhou District People's Hospital, Chongqing, 400700, People's Republic of China
| | - Wan Wu
- Department of General Surgery, Chongqing Kaizhou District People's Hospital, Chongqing, 400700, People's Republic of China
| | - Bing Peng
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Wuhou District, Sichuan Province, Chengdu, 610000, People's Republic of China.
| | - Qianmei Fu
- Department of Oncology, Chongqing Kaizhou District People's Hospital, No. 8, Ankang Road, Hanfeng Street, Kaizhou District, Chongqing, 400700, People's Republic of China.
| |
Collapse
|
9
|
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: 2.0] [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
|
10
|
Zhao Z, Wang Y, Gong Y, Wang X, Zhang L, Zhao H, Li J, Zhu J, Huang X, Zhao C, Yang L, Wang L. Celastrol elicits antitumor effects by inhibiting the STAT3 pathway through ROS accumulation in non-small cell lung cancer. J Transl Med 2022; 20:525. [PMID: 36371217 PMCID: PMC9652895 DOI: 10.1186/s12967-022-03741-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/30/2022] [Indexed: 11/14/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is the most common lung cancer with high mortality across the world, but it is challenging to develop an effective therapy for NSCLC. Celastrol is a natural bioactive compound, which has been found to possess potential antitumor activity. However, the underlying molecular mechanisms of celastrol activity in NSCLC remain elusive. Methods Cellular function assays were performed to study the suppressive role of celastrol in human NSCLC cells (H460, PC-9, and H520) and human bronchial epithelial cells BEAS-2B. Cell apoptosis levels were analyzed by flow cytometry, Hoechst 33342, caspase-3 activity analysis, and western blot analysis. Intracellular reactive oxygen species (ROS) were analyzed by flow cytometry and fluorescence microscope. Expression levels of endoplasmic reticulum (ER) stress-related proteins and phosphorylated signal transducer and activator of transcription 3 (P-STAT3) were identified via western blot analysis. A heterograft model in nude mice was employed to evaluate the effect of celastrol in vivo. Results Celastrol suppressed the growth, proliferation, and metastasis of NSCLC cells. Celastrol significantly increased the level of intracellular ROS; thus, triggering the activation of the ER stress pathway and inhibition of the P-STAT3 pathway, and eventually leading to cell apoptosis, and the effects were reversed by the pre-treatment with N-Acetyl-l-cysteine (NAC). Celastrol also suppressed tumor growth in vivo. Conclusion The outcomes revealed that celastrol plays a potent suppressive role in NSCLC in vitro and in vivo. Celastrol induces apoptosis via causing mitochondrial ROS accumulation to suppress the STAT3 pathway. Celastrol may have potential application prospects in the therapy of NSCLC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03741-9.
Collapse
|
11
|
Zhao H, Wu S, Luo Z, Liu H, Sun J, Jin X. The association between circulating docosahexaenoic acid and lung cancer: A Mendelian randomization study. Clin Nutr 2022; 41:2529-2536. [PMID: 36223714 DOI: 10.1016/j.clnu.2022.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Lung cancer is a malignant tumor with a high incidence, it is vital to identify modifiable and avoidable risk factors for primary prevention, which can significantly lower the risk of cancer by preventing exposure to hazards and altering risky behavior. Some observational studies suggest that an increase in docosahexaenoic acid (DHA) consumption can reduce lung cancer risk. However, interpretation of these observational findings is difficult due to residual confounding or reverse causality. To evaluate the link between DHA and lung cancer, we have undertaken this analysis to examine the causal association between DHA and the risk of lung cancer using a two-sample Mendelian randomization (MR) framework. METHODS We performed a two-sample MR analysis to evaluate the causal effect of plasma DHA levels on lung cancer risk. For the exposure data, we extracted genetic variants as instrumental variables (IVs) that are strongly associated with DHA from a large-scale genome-wide association study (GWAS). We obtained the corresponding effect estimates for IVs on the risk of lung cancer with 11,348 cases and 15,861 controls. Finally, we applied Mendelian randomization analysis to obtain preliminary MR results and performed sensitivity analyses to verify the robustness of our results. RESULTS According to the primary MR estimates and further sensitivity analyses, a higher serum DHA level was associated with a higher risk of lung cancer [OR = 1.159, 95% CI (1.04-1.30), P = 0.01]. For lung adenocarcinoma, the results also showed a close correlation between the DHA level and lung adenocarcinoma [OR = 1.277, 95% CI (1.09-1.50), P = 0.003], but it was not statistically significant for squamous cell carcinoma [OR = 1.071, 95% CI (0.89-1.29), P = 0.467]. CONCLUSIONS Our study revealed that plasma DHA is positively associated with the risk of lung cancer overall, especially for lung adenocarcinoma. This study provides new information to develop dietary guidelines for primary lung cancer prevention.
Collapse
Affiliation(s)
- Hang Zhao
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China; China-Japan Friendship Hospital, Yinghuadong Road, Beijing 100029, Chaoyang District, China
| | - Shengnan Wu
- The First Affiliated Hospital of China Medical University, Shengyang, China
| | - Zhenkai Luo
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China; Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hailong Liu
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou Guangdong, China
| | - Junwei Sun
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xiaolin Jin
- The First Affiliated Hospital of China Medical University, Shengyang, China; Department of International Physical Examination Center, The First Affiliated Hospital of China Medical University, Shengyang, China.
| |
Collapse
|
12
|
Shao M, Jiang Q, Shen C, Liu Z, Qiu L. Sinapine induced ferroptosis in non-small cell lung cancer cells by upregulating transferrin/transferrin receptor and downregulating SLC7A11. Gene 2022; 827:146460. [PMID: 35358657 DOI: 10.1016/j.gene.2022.146460] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/09/2022] [Accepted: 03/25/2022] [Indexed: 11/04/2022]
Abstract
Sinapine (SI) is a naturally occurring product with biological properties, but its activity against non-small cell lung cancer (NSCLC) remains unclear. This research examined the anti-tumour effects of SI in NSCLC cells and the underlying mechanisms of any effects. SI induced ferroptosis, a novel form of cell death, by increasing intracellular ferrous iron, lipid peroxidation, and reactive oxygen species (ROS) in NSCLC cells. SI treatment resulted in transferrin and transferrin receptor upregulation, and inhibition of transferrin or the transferrin receptor reduced the ferroptosis caused by SI. SI treatment also resulted in a p-53 dependent downregulation of SLC7A11. Finally, we evaluated the effects of SI in vivo and it was found that SI also successfully inhibited the growth of NSCLC in vivo. In summary, our data demonstrated that SI triggered ferroptosis in NSCLC cells and may be a promising therapeutic agent for this condition.
Collapse
Affiliation(s)
- Min Shao
- Health Management Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qi Jiang
- Health Management Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chao Shen
- Health Management Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhong Liu
- Health Management Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lihong Qiu
- Health Management Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Shanshan W, Hongying M, Jingjing F, Yiming Y, Yu R, Rui Y. CircDTL Functions as an Oncogene and Regulates Both Apoptosis and Ferroptosis in Non-small Cell Lung Cancer Cells. Front Genet 2021; 12:743505. [PMID: 34621297 PMCID: PMC8490767 DOI: 10.3389/fgene.2021.743505] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 08/18/2021] [Indexed: 01/31/2023] Open
Abstract
Background: Circular RNAs (circRNA) play an essential role in the tumorigenesis of non-small cell lung cancer (NSCLC). CircDTL is a novel identified circRNA with little information regarding its biological role. However, the role of circDTL in NSCLC has not been investigated yet. Method: In this study, the levels of circDTL in tissues and cells were measured by RT-PCR. Cell viability was measured by the CCK-8 assay. Cell migration and invasion were evaluated using the wound healing assay and transwell assay, respectively. Cell death was measured by the cell death ELISA kit. The levels of Fe2+, ROS, MDA and GSH were measured using the commercial kits. The interactions between miR-1287-5p and circDTL/3'UTR GPX4 were verified by dual-luciferase activity assay. The effects of circDTL on tumor growth were evaluated in vivo. Results: CircDTL was found to be upregulated and acted as an oncogene in NSCLC cells. Knockdown of circDTL promoted both apoptosis and ferroptosis of NSCLC cells. It was identified that circDTL exerts its oncogenic effects via the circDTL/miR-1287-5p/GPX4 axis and GPX4 inhibits both ferroptosis and apoptosis. Finally, this study showed that silencing of circDTL promoted the sensitivity of NSCLC cells to chemotherapeutic agents and inhibited the growth of tumors in vivo. Conclusion: CircDTL acts as an oncogene and exerts its effects via the miR-1287-5p/GPX4 axis in NSCLC, providing a potential therapeutic target for NSCLC cancer therapy.
Collapse
Affiliation(s)
- Wang Shanshan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Ma Hongying
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Fang Jingjing
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Yu Yiming
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Ren Yu
- Department of Urologic Surgery, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Yu Rui
- Department of Biochemistry and Molecular Biology, Medical School of Ningbo University, Ningbo, China
| |
Collapse
|
14
|
Maoz A, Merenstein C, Koga Y, Potter A, Gower AC, Liu G, Zhang S, Liu H, Stevenson C, Spira A, Reid ME, Campbell JD, Mazzilli SA, Lenburg ME, Beane J. Elevated T cell repertoire diversity is associated with progression of lung squamous cell premalignant lesions. J Immunother Cancer 2021; 9:jitc-2021-002647. [PMID: 34580161 PMCID: PMC8477334 DOI: 10.1136/jitc-2021-002647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2021] [Indexed: 11/21/2022] Open
Abstract
Objective The immune response to invasive carcinoma has been the focus of published work, but little is known about the adaptive immune response to bronchial premalignant lesions (PMLs), precursors of lung squamous cell carcinoma. This study was designed to characterize the T cell receptor (TCR) repertoire in PMLs and its association with clinical, pathological, and molecular features. Methods Endobronchial biopsies (n=295) and brushings (n=137) from high-risk subjects (n=50), undergoing lung cancer screening at approximately 1-year intervals via autofluorescence bronchoscopy and CT, were profiled by RNA-seq. We applied the TCR Repertoire Utilities for Solid Tissue/Tumor tool to the RNA-seq data to identify TCR CDR3 sequences across all samples. In the biopsies, we measured the correlation of TCR diversity with previously derived immune-associated PML transcriptional signatures and PML outcome. We also quantified the spatial and temporal distribution of shared and clonally expanded TCRs. Using the biopsies and brushes, the ratio of private (ie, found in one patient only) and public (ie, found in two or more patients) TCRs was quantified, and the CDR3 sequences were compared with those found in curated databases with known antigen specificities. Results We detected 39,303 unique TCR sequences across all samples. In PML biopsies, TCR diversity was negatively associated with a transcriptional signature of T cell mediated immune activation (p=4e-4) associated with PML outcome. Additionally, in lesions of the proliferative molecular subtype, TCR diversity was decreased in regressive versus progressive/persistent PMLs (p=0.045). Within each patient, TCRs were more likely to be shared between biopsies sampled at the same timepoint than biopsies sampled at the same anatomic location at different times. Clonally expanded TCRs, within a biopsied lesion, were more likely to be expanded at future time points than non-expanded clones. The majority of TCR sequences were found in a single sample, with only 3396 (8.6%) found in more than one sample and 1057 (2.7%) found in two or more patients (ie, public); however, when compared with a public database of CDR3 sequences, 4543 (11.6%) of TCRs were identified as public. TCRs with known antigen specificities were enriched among public TCRs (p<0.001). Conclusions Decreased TCR diversity may reflect nascent immune responses that contribute to PML elimination. Further studies are needed to explore the potential for immunoprevention of PMLs.
Collapse
Affiliation(s)
- Asaf Maoz
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA.,Boston Medical Center, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Carter Merenstein
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA.,Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Yusuke Koga
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Austin Potter
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Adam C Gower
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Gang Liu
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Sherry Zhang
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Hanqiao Liu
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | | | - Avrum Spira
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA.,The Lung Cancer Initiative at Johnson and Johnson, Cambridge, Massachusetts, USA
| | - Mary E Reid
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Joshua D Campbell
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Sarah A Mazzilli
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Marc E Lenburg
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Jennifer Beane
- Department of Medicine, Secion of Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
15
|
Tennis MA, Smith AJ, Dwyer-Nield LD, Keith RL. Intranasal iloprost prevents tumors in a murine lung carcinogenesis model. Cancer Prev Res (Phila) 2021; 15:11-16. [PMID: 34556494 DOI: 10.1158/1940-6207.capr-21-0086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/12/2021] [Accepted: 09/09/2021] [Indexed: 11/16/2022]
Abstract
Lung cancer chemoprevention with the prostacyclin analogue iloprost is the most promising approach to date for intercepting progression of premalignant lung lesions in former smokers. Previous pre-clinical studies of iloprost used oral delivery, but a study modeling delivery directly to the target organ was needed. In vivo and in vitro studies have identified gene expression changes following iloprost treatment, including increased E-cadherin and PPAR𝛄 and decreased COX2 and Vimentin. We used tumor counts and gene expression to demonstrate the effectiveness of intranasal delivery of iloprost in a murine model of premalignant adenomas. Intranasal delivery of iloprost reduced adenoma multiplicity14 weeks after urethane exposure in FVB mice compared to untreated urethane controls. Intranasal iloprost reversed urethane-induced gene expression changes in tumors and whole lung. These results correspond to previous studies of oral iloprost and in vitro treatment of human bronchial epithelial cells. This study demonstrates that intranasal delivery of iloprost in a mouse model of lung premalignant lesions is effective chemoprevention. This will be an essential tool for exploring mechanisms and outcomes of iloprost chemoprevention, along with supporting ongoing clinical trials of inhaled iloprost chemoprevention.
Collapse
Affiliation(s)
- Meredith A Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus
| | - Alex J Smith
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus
| | | | - Robert L Keith
- Medicine/Division of Pulmonary Sciences, Eastern Colorado Veterans Affairs Medical Center
| |
Collapse
|
16
|
Xie X, Cai X, Tang Y, Jiang C, Zhou F, Yang L, Liu Z, Wang L, Zhao H, Zhao C, Huang X. Flubendazole Elicits Antitumor Effects by Inhibiting STAT3 and Activating Autophagy in Non-small Cell Lung Cancer. Front Cell Dev Biol 2021; 9:680600. [PMID: 34513827 PMCID: PMC8427440 DOI: 10.3389/fcell.2021.680600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/26/2021] [Indexed: 01/16/2023] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is a major neoplastic disease with a high mortality worldwide; however, effective treatment of this disease remains a challenge. Flubendazole, a traditional anthelmintic drug, possesses potent antitumor properties; however, the detailed molecular mechanism of flubendazole activity in NSCLC needs to be further explored. In the present study, flubendazole was found to exhibit valid antitumor activity in vitro as well as in vivo. Flubendazole blocked phosphorylation of STAT3 in a dose- and time-dependent manner and regulated the transcription of STAT3 target genes encoding apoptotic proteins. Further, flubendazole inhibited STAT3 activation by inhibiting its phosphorylation and nuclear localization induced by interleukin-6 (IL-6). Notably, the autophagic flux of NSCLC cell lines was increased after flubendazole treatment. Furthermore, flubendazole downregulated the expression of BCL2, P62, and phosphorylated-mTOR, but it upregulated LC3-I/II and Beclin-1 expression, which are the main genes associated with autophagy. Collectively, these data contribute to elucidating the efficacy of flubendazole as an anticancer drug, demonstrating its potential as a therapeutic agent via its suppression of STAT3 activity and the activation of autophagy in NSCLC.
Collapse
Affiliation(s)
- Xiaona Xie
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xueding Cai
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yemeng Tang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chunhui Jiang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Feng Zhou
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Lehe Yang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Zhiguo Liu
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Liangxing Wang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Haiyang Zhao
- The Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Chengguang Zhao
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaoying Huang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
17
|
Garrison GW, Cho JL, Deng JC, Camac E, Oh S, Sundar K, Baptiste JV, Cheng GS, De Cardenas J, Fitzgerald C, Garfield J, Ha NT, Holden VK, O’Corragain O, Patel S, Wayne MT, McSparron JI, Wang T, Çoruh B, Hayes MM, Guzman E, Channick CL. ATS Core Curriculum 2021. Adult Pulmonary Medicine: Thoracic Oncology. ATS Sch 2021; 2:468-483. [PMID: 34667994 PMCID: PMC8518653 DOI: 10.34197/ats-scholar.2021-0032re] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/25/2021] [Indexed: 11/18/2022] Open
Abstract
The American Thoracic Society Core Curriculum updates clinicians annually in adult and pediatric pulmonary disease, medical critical care, and sleep medicine at the annual international conference. The 2021 Pulmonary Core Curriculum focuses on lung cancer and include risks and prevention, screening, nodules, therapeutics and associated pulmonary toxicities, and malignant pleural effusions. Although tobacco smoking remains the primary risk factor for developing lung cancer, exposure to other environmental and occupational substances, including asbestos, radon, and burned biomass, contribute to the global burden of disease. Randomized studies have demonstrated that routine screening of high-risk smokers with low-dose chest computed tomography results in detection at an earlier stage and reduction in lung cancer mortality. On the basis of these trials and other lung cancer risk tools, screening recommendations have been developed. When evaluating lung nodules, clinical and radiographic features are used to estimate the probability of cancer. Management guidelines take into account the nodule size and cancer risk estimates to provide recommendations at evaluation. Newer lung cancer therapies, including immune checkpoint inhibitors and molecular therapies, cause pulmonary toxicity more frequently than conventional chemotherapy. Treatment-related toxicity should be suspected in patients receiving these medications who present with respiratory symptoms. Evaluation is aimed at excluding other etiologies, and treatment is based on the severity of symptoms. Malignant pleural effusions can be debilitating. The diagnosis is made by using simple pleural drainage and/or pleural biopsies. Management depends on the clinical scenario and the patient's preferences and includes the use of serial thoracentesis, a tunneled pleural catheter, or pleurodesis.
Collapse
Affiliation(s)
- Garth W. Garrison
- Divison of Pulmonary Disease and Critical Care Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Josalyn L. Cho
- Division of Pulmonary, Critical Care and Occupational Medicine, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Jane C. Deng
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Erin Camac
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Scott Oh
- Division of Pulmonary, Critical Care Medicine, Clinical Immunology, and Allergy, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Krishna Sundar
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, Utah
| | - Janelle V. Baptiste
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center–Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Guang-Shing Cheng
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Jose De Cardenas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
- Section of Thoracic Surgery, Department of Surgery, School of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Codi Fitzgerald
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Jamie Garfield
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Ngoc-Tram Ha
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Maryland, Baltimore, Maryland; and
| | - Van K. Holden
- Division of Pulmonary and Critical Care Medicine, School of Medicine, University of Maryland, Baltimore, Maryland; and
| | - Oisin O’Corragain
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Sahil Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center–Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Max T. Wayne
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Jakob I. McSparron
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Tisha Wang
- Division of Pulmonary, Critical Care Medicine, Clinical Immunology, and Allergy, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Başak Çoruh
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Margaret M. Hayes
- Division of Pulmonary, Critical Care and Sleep Medicine, Beth Israel Deaconess Medical Center–Harvard Medical School, Harvard University, Boston, Massachusetts
| | | | - Colleen L. Channick
- Division of Pulmonary, Critical Care Medicine, Clinical Immunology, and Allergy, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
18
|
Otsuka K, Ochiya T. Possible connection between diet and microRNA in cancer scenario. Semin Cancer Biol 2021; 73:4-18. [DOI: 10.1016/j.semcancer.2020.11.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/30/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
|
19
|
Ramani VK, Ganesha DV, Naik R. A Narrative Review of the Risk Factors for Cancer and the Preventive Opportunities: Current Status, Future Perspectives, and Implications for India. ASIAN JOURNAL OF ONCOLOGY 2021. [DOI: 10.1055/s-0041-1731092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Abstract
Introduction Clinical cancer can arise from heterogenous pathways through various genetic mutations. Although we cannot predict the timeline by which an individual will develop cancer, certain risk assessment tools can be used among high-risk groups for focusing the preventive activities. As primary level of cancer prevention, healthy lifestyle approach is being promoted. The etiological factors for lung cancer include by-products of industrialization and air pollution. We need to factor the increase in household air pollution as well.
Methods “PubMed” database and Google search engines were used for searching the relevant articles. Search terms with Boolean operators used include “Cancer prevention,” “Missed opportunities in cancer causation,” and “incidence of risk factors.” This review includes 20 studies and other relevant literature that address the opportunities for cancer prevention.
Body The narrative describes the association between many of the risk factors and development of cancer. This includes tobacco, alcohol, infections, air pollution, physical inactivity, diet, obesity, screening and preventive strategies, chemoprevention, biomarkers of carcinogenesis, and factors that prolong the diagnosis of cancer.
Discussion Reports from basic science research provide evidence on the potential of biologically active food components and pharmacological agents for mitigating the risk of cancer and its progression. However, some reports from observational studies and randomized trials have been inconsistent. We need to recognize the impact of sociodemographic factors such as age, sex, ethnicity, culture, and comorbid illness on preventive interventions. Spiral computed tomographic scan is a robust tool for early detection of lung cancer.
Conclusion Infectious etiology for specific cancers provides opportunities for prevention and treatment. The complex interplay between man and microbial flora needs to be dissected, for understanding the pathogenesis of relevant malignancies. For reducing the morbidity of cancer, we need to focus on prevention as a priority strategy and intervene early during the carcinogenic process.
Collapse
Affiliation(s)
- Vinod K. Ramani
- Department of Preventive Oncology, Healthcare Global Enterprise Ltd., Bangalore, Karnataka, India
- Department of Public health, Ramaiah University of Applied Sciences, Bangalore, Karnataka, India
| | - D. V. Ganesha
- Department of Medical Oncology, St. John’s Medical College and Hospital, Bangalore, Karnataka, India
| | - Radheshyam Naik
- Department of Medical Oncology, Healthcare Global Enterprise Ltd., Bangalore, India
| |
Collapse
|
20
|
Hang T, Yang L, Zhang X, Li J, Long F, Zhu N, Li Y, Xia J, Zhang Y, Zhang P, Zhang Y, Wei X, Li S. Peroxisome proliferator-activated receptor γ improves pemetrexed therapeutic efficacy in non-squamous non-small cell lung cancer. Am J Transl Res 2021; 13:2296-2307. [PMID: 34017390 PMCID: PMC8129360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/20/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE The folic acid analog pemetrexed (PMX) is recommended for the first-line chemotherapy for advanced non-squamous non-small cell lung cancer (NSCLC). However, the mechanisms underlying PMX cytotoxicity in NSCLC remain to be fully explored. METHODS PMX effect was evaluated in a urethane-induced lung adenocarcinoma mouse model. The interaction between PMX and intracellular proteins, particularly peroxisome proliferator-activated receptor γ (PPARγ), was investigated. The role of PPARγ in mediating pemetrexed cytotoxicity was investigated using NSCLC cell lines, mouse models and clinical specimens. RESULTS This study found that PPARγ expression was correlated with prolonged progression-free survival in NSCLC patients. PPARγ downregulated hypoxanthine-guanine phosphoribosyl transferase (HGPRT), a key enzyme for nucleotide salvage synthesis, thereby sensitizing cells to PMX inhibition on nucleotide de novo synthesis. PMX was also a candidate partial agonist of PPARγ, and PMX-activated PPARγ bound to NF-κB and transcriptionally suppressed the NF-κB target gene, c-Myc. PMX inhibited tumor growth by activating PPARγ in a urethane-induced lung cancer model characterized by elevated NF-κB activity. CONCLUSION PPARγ improves pemetrexed therapeutic efficacy in non-squamous NSCLC. The cytotoxicity effect of PMX can be synergized by activating PPARγ and thereby inhibiting NF-κB pathway.
Collapse
Affiliation(s)
- Tianxing Hang
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese MedicineNanjing, China
| | - Li Yang
- Department of Anesthesiology, Changzheng Hospital, Navy Medical University of Chinese PLAShanghai, China
| | - Xiujuan Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Jing Li
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Feng Long
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Ning Zhu
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Ying Li
- Department of Respiratory Medicine, Shaanxi Provincial Second People’s HospitalXi’an, China
| | - Jingwen Xia
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Youzhi Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Peng Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Yuanyuan Zhang
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Xiaomin Wei
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| | - Shengqing Li
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan UniversityShanghai, China
| |
Collapse
|
21
|
AMPK activation by ASP4132 inhibits non-small cell lung cancer cell growth. Cell Death Dis 2021; 12:365. [PMID: 33824293 PMCID: PMC8024326 DOI: 10.1038/s41419-021-03655-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 12/25/2022]
Abstract
Activation of adenosine monophosphate-activated protein kinase (AMPK) is able to produce significant anti-non-small cell lung cancer (NSCLC) cell activity. ASP4132 is an orally active and highly effective AMPK activator. The current study tested its activity against NSCLC cells. In primary NSCLC cells and established cell lines (A549 and NCI-H1944) ASP4132 potently inhibited cell growth, proliferation and cell cycle progression as well as cell migration and invasion. Robust apoptosis activation was detected in ASP4132-treated NSCLC cells. Furthermore, ASP4132 treatment in NSCLC cells induced programmed necrosis, causing mitochondrial p53-cyclophilin D (CyPD)-adenine nucleotide translocase 1 (ANT1) association, mitochondrial depolarization and medium lactate dehydrogenase release. In NSCLC cells ASP4132 activated AMPK signaling, induced AMPKα1-ACC phosphorylation and increased AMPK activity. Furthermore, AMPK downstream events, including mTORC1 inhibition, receptor tyrosine kinases (PDGFRα and EGFR) degradation, Akt inhibition and autophagy induction, were detected in ASP4132-treated NSCLC cells. Importantly, AMPK inactivation by AMPKα1 shRNA, knockout (using CRISPR/Cas9 strategy) or dominant negative mutation (T172A) almost reversed ASP4132-induced anti-NSCLC cell activity. Conversely, a constitutively active AMPKα1 (T172D) mimicked and abolished ASP4132-induced actions in NSCLC cells. In vivo, oral administration of a single dose of ASP4132 largely inhibited NSCLC xenograft growth in SCID mice. AMPK activation, mTORC1 inhibition and EGFR-PDGFRα degradation as well as Akt inhibition and autophagy induction were detected in ASP4132-treated NSCLC xenograft tumor tissues. Together, activation of AMPK by ASP4132 potently inhibits NSCLC cell growth in vitro and in vivo.
Collapse
|
22
|
Alam A, Ansari MA, Badrealam KF, Pathak S. Molecular approaches to lung cancer prevention. Future Oncol 2021; 17:1793-1810. [PMID: 33653087 DOI: 10.2217/fon-2020-0789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Lung cancer is generally diagnosed at advanced stages when surgical resection is not possible. Late diagnosis, along with development of chemoresistance, results in high mortality. Preventive approaches, including smoking cessation, chemoprevention and early detection are needed to improve survival. Smoking cessation combined with low-dose computed tomography screening has modestly improved survival. Chemoprevention has also shown some promise. Despite these successes, most lung cancer cases remain undetected until advanced stages. Additional early detection strategies may further improve survival and treatment outcome. Molecular alterations taking place during lung carcinogenesis have the potential to be used in early detection via noninvasive methods and may also serve as biomarkers for success of chemopreventive approaches. This review focuses on the utilization of molecular biomarkers to increase the efficacy of various preventive approaches.
Collapse
Affiliation(s)
- Asrar Alam
- Department of Preventive Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| | - Mohammad A Ansari
- Department of Epidemic Disease Research, Institute of Research & Medical Consultation, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Khan F Badrealam
- Cardiovascular & Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Sujata Pathak
- Department of Preventive Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, 110029, India
| |
Collapse
|
23
|
Zhao HS, Tao XM, Wang Q, Fang YY, Zhang HY, Wang HQ, Zhang GJ. Silencing SIX1 by miR-7160 inhibits non-small cell lung cancer cell growth. Aging (Albany NY) 2021; 13:8055-8067. [PMID: 33686961 PMCID: PMC8034971 DOI: 10.18632/aging.202398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/15/2020] [Indexed: 01/20/2023]
Abstract
The homeoprotein SIX1 is upregulated in non-small cell lung cancer (NSCLC) and associated with NSCLC tumorigenesis and progression. We identified microRNA-7160 (miR-7160) as a SIX1-targeting miRNA. RNA immunoprecipitation results confirmed a direct binding between miR-7160 and SIX1 mRNA in NSCLC cells. In the primary and established NSCLC cells, forced overexpression of miR-7160 downregulated SIX1 and inhibited cancer cell growth, proliferation, migration and invasion. Furthermore, miR-7160 overexpression induced apoptosis activation in NSCLC cells. Conversely, miR-7160 inhibition elevated SIX1 expression and enhanced NSCLC cell progression in vitro. Restoring SIX1 expression, by an untranslated region-depleted SIX1 expression construct, reversed miR-7160-induced anti-NSCLC cell activity. CRISPR/Cas9-inudced knockout of SIX1 mimicked miR-7160-induced actions and produced anti-NSCLC cell activity. In vivo, intratumoral injection of miR-7160-expressing lentivirus downregulated SIX1 mRNA and inhibited NSCLC xenograft growth in severe combined immunodeficient mice. Significantly, miR-7160 expression is downregulated in human NSCLC tissues and is correlated with SIX1 mRNA upregulation. Collectively, miR-7160 silenced SIX1 and inhibited NSCLC cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Hua-Si Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Min Tao
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qun Wang
- Department of Respiratory Medicine, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Yuan-Yuan Fang
- Department of Endocrinology, Henan Provincial People’s Hospital; People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Hong-Yu Zhang
- Department of Infectious Disease, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hua-Qi Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guo-Jun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Exploration in the mechanism of fucosterol for the treatment of non-small cell lung cancer based on network pharmacology and molecular docking. Sci Rep 2021; 11:4901. [PMID: 33649481 PMCID: PMC7921686 DOI: 10.1038/s41598-021-84380-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/15/2021] [Indexed: 12/23/2022] Open
Abstract
Fucosterol, a sterol isolated from brown algae, has been demonstrated to have anti-cancer properties. However, the effects and underlying molecular mechanism of fucosterol on non-small cell lung cancer remain to be elucidated. In this study, the corresponding targets of fucosterol were obtained from PharmMapper, and NSCLC related targets were gathered from the GeneCards database, and the candidate targets of fucosterol-treated NSCLC were predicted. The mechanism of fucosterol against NSCLC was identified in DAVID6.8 by enrichment analysis of GO and KEGG, and protein–protein interaction data were collected from STRING database. The hub gene GRB2 was further screened out and verified by molecular docking. Moreover, the relationship of GRB2 expression and immune infiltrates were analyzed by the TIMER database. The results of network pharmacology suggest that fucosterol acts against candidate targets, such as MAPK1, EGFR, GRB2, IGF2, MAPK8, and SRC, which regulate biological processes including negative regulation of the apoptotic process, peptidyl-tyrosine phosphorylation, positive regulation of cell proliferation. The Raf/MEK/ERK signaling pathway initiated by GRB2 showed to be significant in treating NSCLC. In conclusion, our study indicates that fucosterol may suppress NSCLC progression by targeting GRB2 activated the Raf/MEK/ERK signaling pathway, which laying a theoretical foundation for further research and providing scientific support for the development of new drugs.
Collapse
|
25
|
Li X, Chen Z, Ni Y, Bian C, Huang J, Chen L, Xie X, Wang J. Tumor-associated macrophages secret exosomal miR-155 and miR-196a-5p to promote metastasis of non-small-cell lung cancer. Transl Lung Cancer Res 2021; 10:1338-1354. [PMID: 33889514 PMCID: PMC8044469 DOI: 10.21037/tlcr-20-1255] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Understanding the molecular basis underlying metastasis of non-small cell lung cancer (NSCLC) may provide a new therapeutic modality for the treatment of NSCLC. However, the mechanisms by which tumor-associated macrophages (TAMs) affect NSCLC metastasis remain undefined. In this study, we aimed to discover a novel regulatory pathway involved in NSCLC metastasis. Methods Cell Counting Kit-8 (CCK-8), Transwell, western blot assays were used to assess cell viability, migration, invasion and epithelial-mesenchymal transition (EMT). Exosomes from macrophages medium were characterized, and in vitro cell coculture was further conducted to investigate M2 derived exosomes mediated crosstalk between TAMs and tumor cells. Besides, miRNA microarray was used to analyze miRNA expression profiles of M0 and M2 derived exosomes. Luciferase reporter assay was used to verify the potential binding between miRNA and mRNA. Moreover, 6-week-old male BALB/c nude mice were performed to establish transplantation tumor model using tail vein injection. Hematoxylin & eosin staining was used to detect the metastasis of tumor tissues. Results We found that M2 TAMs were the main TAMs in metastatic tissues of NSCLC patients and exosomes derived from M2 TAMs were able to promote cell viability, cell migration, cell invasion and EMT in NSCLC. We demonstrated that miR-155 and miR-196a-5p were abundant in M2 TAMs and exosomes secreted by M2 TAMs. Functional experiments demonstrated that the deletion of miR-155 and miR-196a-5p in M2 TAMs significantly prevented NSCLC metastasis in vitro and in vivo. To clarify the mechanism governing miR-155 and miR-196a-5p from M2 TAMs, we carried out bioinformatics analysis to predict potential target genes. Mechanistically, miR-155 and miR-196a-5p directly bound to the 3'-UTR of Ras association domain family member 4 (RASSF4), and negatively regulating RASSF4 expression. At last, rescue assays demonstrated that miR-155 and miR-196a-5p exerted its performance by RASSF4. Conclusions Overall, we revealed a new regulatory pathway that was M2 TAMs secreted exosomal miR-155 and miR-196a-5p to promote NSCLC metastasis. This dynamic and reciprocal cross-talk between NSCLC and macrophages innovatively provided a potential opportunity for diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Xiang Li
- Department of Thoracic Surgery, Jiangsu Province People's Hospital and the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhipeng Chen
- Department of Thoracic Surgery, Jiangsu Province People's Hospital and the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yaojun Ni
- Department of Thoracic Surgery, Jiangsu Province People's Hospital and the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Cardiothoracic Surgery, Huai'an First People's Hospital Affiliated to Nanjing Medical University, Huai'an, China
| | - Chengyu Bian
- Department of Thoracic Surgery, Jiangsu Province People's Hospital and the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Huang
- Department of Thoracic Surgery, Jiangsu Province People's Hospital and the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Thoracic Surgery, Jiangsu Province People's Hospital and the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xueying Xie
- State Key Laboratory of Bioelectronics, School of Biological Sciences and Medical Engineering, Southeast University, Nanjing, China
| | - Jun Wang
- Department of Thoracic Surgery, Jiangsu Province People's Hospital and the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Zhao Y, Bilal M, Raza A, Khan MI, Mehmood S, Hayat U, Hassan STS, Iqbal HMN. Tyrosine kinase inhibitors and their unique therapeutic potentialities to combat cancer. Int J Biol Macromol 2021; 168:22-37. [PMID: 33290765 DOI: 10.1016/j.ijbiomac.2020.12.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 02/05/2023]
Abstract
Cancer is one of the leading causes of death with a mortality rate of 12%. Although significant progress has been achieved in cancer research, the effective treatment of cancer remains the greatest global challenge in medicine. Dysregulation of tyrosine kinases (TK) is one of the characteristics of several types of cancers. Thus, drugs that target and inhibit these enzymes, known as TK inhibitors (TKIs), are considered vital chemotherapeutics to combat various types of cancer. The oral bioavailability of available TKIs and their targeted therapy are their potential benefits. Based on these characteristics, most TKIs are included in first/second-line therapy for the treatment of different cancers. This review aims to shed light on orally-active TKIs (natural and synthetic molecules) and their promising implication in the therapy of numerous types of tumors along with their mechanisms of action. Further, recent progress in the development of synthetic and isolation of natural TKIs is reviewed. A significant growth in research regarding the development of new-generation TKIs is made with time (23 FDA-approved TKIs from 2018) due to their better therapeutic response. Oral bioavailability should be considered as an important parameter while developing of new-generation TKIs; however, drug delivery systems can also be used to address issue of poor bioavailability to a certain extent. Moreover, clinical trials should be designed in consideration of the development of resistance and tumor heterogeneity.
Collapse
Affiliation(s)
- Yuping Zhao
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Ali Raza
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Muhammad Imran Khan
- Hefei National Lab for Physical Sciences at the Microscale and the Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shahid Mehmood
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Uzma Hayat
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Sherif T S Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 6-Suchdol, 165 21 Prague, Czech Republic
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, 64849, Mexico.
| |
Collapse
|
27
|
Treekitkarnmongkol W, Hassane M, Sinjab A, Chang K, Hara K, Rahal Z, Zhang J, Lu W, Sivakumar S, McDowell TL, Kantrowitz J, Zhou J, Lang W, Xu L, Ochieng JK, Nunomura-Nakamura S, Deng S, Behrens C, Raso MG, Fukuoka J, Reuben A, Ostrin EJ, Parra E, Solis LM, Spira AE, McAllister F, Cascone T, Wistuba II, Moghaddam SJ, Scheet PA, Fujimoto J, Kadara H. Augmented Lipocalin-2 Is Associated with Chronic Obstructive Pulmonary Disease and Counteracts Lung Adenocarcinoma Development. Am J Respir Crit Care Med 2021; 203:90-101. [PMID: 32730093 DOI: 10.1164/rccm.202004-1079oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rationale: Early pathogenesis of lung adenocarcinoma (LUAD) remains largely unknown. We found that, relative to wild-type littermates, the innate immunomodulator Lcn2 (lipocalin-2) was increased in normal airways from mice with knockout of the airway lineage gene Gprc5a (Gprc5a-/-) and that are prone to developing inflammation and LUAD. Yet, the role of LCN2 in lung inflammation and LUAD is poorly understood.Objectives: Delineate the role of Lcn2 induction in LUAD pathogenesis.Methods: Normal airway brushings, uninvolved lung tissues, and tumors from Gprc5a-/- mice before and after tobacco carcinogen exposure were analyzed by RNA sequencing. LCN2 mRNA was analyzed in public and in-house data sets of LUAD, lung squamous cancer (LUSC), chronic obstructive pulmonary disease (COPD), and LUAD/LUSC with COPD. LCN2 protein was immunohistochemically analyzed in a tissue microarray of 510 tumors. Temporal lung tumor development, gene expression programs, and host immune responses were compared between Gprc5a-/- and Gprc5a-/-/Lcn2-/- littermates.Measurements and Main Results: Lcn2 was progressively elevated during LUAD development and positively correlated with proinflammatory cytokines and inflammation gene sets. LCN2 was distinctively elevated in human LUADs, but not in LUSCs, relative to normal lungs and was associated with COPD among smokers and patients with LUAD. Relative to Gprc5a-/- mice, Gprc5a-/-/Lcn2-/- littermates exhibited significantly increased lung tumor development concomitant with reduced T-cell abundance (CD4+) and richness, attenuated antitumor immune gene programs, and increased immune cell expression of protumor inflammatory cytokines.Conclusions: Augmented LCN2 expression is a molecular feature of COPD-associated LUAD and counteracts LUAD development in vivo by maintaining antitumor immunity.
Collapse
Affiliation(s)
| | - Maya Hassane
- Department of Biochemistry and Molecular Genetics and
| | | | | | - Kieko Hara
- Department of Translational Molecular Pathology
| | - Zahraa Rahal
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jiexin Zhang
- Department of Bioinformatics and Computer Biology
| | - Wei Lu
- Department of Translational Molecular Pathology
| | | | - Tina L McDowell
- Department of Translational Molecular Pathology.,Department of Epidemiology
| | - Jacob Kantrowitz
- Section of Computational Biomedicine, School of Medicine, Boston University, Boston, Massachusetts; and
| | | | - Wenhua Lang
- Department of Translational Molecular Pathology
| | - Li Xu
- Department of Translational Molecular Pathology
| | | | | | | | | | | | - Junya Fukuoka
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | | | | | - Edwin Parra
- Department of Translational Molecular Pathology
| | | | - Avrum E Spira
- Section of Computational Biomedicine, School of Medicine, Boston University, Boston, Massachusetts; and
| | - Florencia McAllister
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tina Cascone
- Department of Thoracic Head and Neck Medical Oncology
| | | | | | | | | | | |
Collapse
|
28
|
Bars-Cortina D, Sakhawat A, Piñol-Felis C, Motilva MJ. Chemopreventive effects of anthocyanins on colorectal and breast cancer: A review. Semin Cancer Biol 2021; 81:241-258. [DOI: 10.1016/j.semcancer.2020.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
|
29
|
Yang Z, Zhao Y, Hao D, Wang H, Li S, Jia L, Yuan X, Zhang L, Meng L, Zhang S. Computational identification of potential chemoprophylactic agents according to dynamic behavior of peroxisome proliferator-activated receptor gamma. RSC Adv 2020; 11:147-159. [PMID: 35423024 PMCID: PMC8690233 DOI: 10.1039/d0ra09059j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/12/2020] [Indexed: 12/14/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is an attractive target for chemoprevention of lung carcinoma, however its highly dynamic nature has plagued drug development for decades, with difficulties in receptor modeling for structure-based design. In this work, an integrated receptor-based virtual screening (VS) strategy was applied to identify PPARγ agonists as chemoprophylactic agents by using extensive docking and conformational sampling methods. Our results showed that the conformational plasticity of PPARγ, especially the H2 & S245 loop, H2' & Ω loop and AF-2 surface, is markedly affected by binding of full/partial agonists. To fully take the dynamic behavior of PPARγ into account, the VS approach effectively sorts out five commercial agents with reported antineoplastic properties. Among them, ZINC03775146 (gusperimus) and ZINC14087743 (miltefosine) might be novel PPARγ agonists with the potential for chemoprophylaxis, that simultaneously take part in a flexible switch of the AF-2 surface and state change of the Ω loop. Furthermore, the dynamic structural coupling between the H2 & S245 and H2' & Ω loops offers enticing hope for PPARγ-targeted therapeutics, by blocking kinase accessibility to PPARγ. These results might aid the development of chemopreventive drugs, and the integrated VS strategy could be conducive to drug design for highly flexible biomacromolecules.
Collapse
Affiliation(s)
- Zhiwei Yang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University Xi'an 710049 China +86-29-82660915 +86-29-82660915
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Chemistry, Xi'an Jiaotong University Xi'an 710049 China
- School of Life Science and Technology, Xi'an Jiaotong University Xi'an 710049 China
| | - Yizhen Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University Xi'an 710049 China +86-29-82660915 +86-29-82660915
| | - Dongxiao Hao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University Xi'an 710049 China +86-29-82660915 +86-29-82660915
| | - He Wang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University Xi'an 710049 China +86-29-82660915 +86-29-82660915
| | - Shengqing Li
- Department of Pulmonary and Critical Care Medicine, Huashan Hospital, Fudan University Shanghai 200041 China
| | - Lintao Jia
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University Xi'an 710032 China
| | - Xiaohui Yuan
- Institute of Biomedicine, Jinan University Guangzhou 510632 China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University Xi'an 710049 China +86-29-82660915 +86-29-82660915
| | - Lingjie Meng
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Chemistry, Xi'an Jiaotong University Xi'an 710049 China
- Instrumental Analysis Center, Xi'an Jiao Tong University Xi'an 710049 China
| | - Shengli Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University Xi'an 710049 China +86-29-82660915 +86-29-82660915
| |
Collapse
|
30
|
Diclofenac Enhances Docosahexaenoic Acid-Induced Apoptosis in Vitro in Lung Cancer Cells. Cancers (Basel) 2020; 12:cancers12092683. [PMID: 32962236 PMCID: PMC7564004 DOI: 10.3390/cancers12092683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Polyunsaturated fatty acids (PUFAs) and non-steroidal anti-inflammatory drugs (NSAIDs) have limited anticancer capacities when used alone. We examined whether combining NSAIDs with docosahexaenoic (DHA) would increase their anticancer activity on lung cancer cell lines. Our results indicate that combining DHA and NSAIDs increased their anticancer activities by altering the expression of critical proteins in the RAS/MEK/ERK and PI3K/Akt pathways. The data suggest that DHA combined with low dose diclofenac provides more significant anticancer potential, which can be further developed for chemoprevention and adjunct therapy in lung cancer. Abstract Polyunsaturated fatty acids (PUFAs) and non-steroidal anti-inflammatory drugs (NSAIDs) show anticancer activities through diverse molecular mechanisms. However, the anticancer capacities of either PUFAs or NSAIDs alone is limited. We examined whether combining NSAIDs with docosahexaenoic (DHA), commonly derived from fish oils, would possibly synergize their anticancer activity. We determined the viability of lung cancer cell lines (NCI-H1573, A549, NCI-H1299, and NCI-H1975) after exposure to DHA and various NSAIDs. We further conducted cell apoptosis assays and analyzed apoptosis-associated proteins and some key proteins in the RAS/MEK/ERK and PI3K/Akt pathways using western blot analysis. We also determined the impact of the treatment on the expression of inducible cancer-related genes using nCounter PanCancer Pathways gene expression analysis. The results showed that the combination of DHA and NSAIDs increased suppression of cell viability in all the lung cancer cell lines tested compared to each of the compounds used alone, with diclofenac being the most potent NSAID tested. This synergistic effect is especially significant in A549 and NCI-H1573 cells. The combination treatment was more effective at inhibiting clonogenic cell growth and anchorage-independent growth in soft agar, inducing caspase-dependent apoptosis, and altering expression of critical proteins in the RAS/MEK/ERK and PI3K/Akt pathways. The data from this study demonstrate that DHA combined with low dose diclofenac provides greater anticancer potential, which can be further developed for chemoprevention and adjunct therapy in lung cancer.
Collapse
|
31
|
Hu Q, Corral P, Narayanapillai SC, Leitzman P, Upadhyaya P, O’Sullivan MG, Hecht SS, Lu J, Xing C. Oral Dosing of Dihydromethysticin Ahead of Tobacco Carcinogen NNK Effectively Prevents Lung Tumorigenesis in A/J Mice. Chem Res Toxicol 2020; 33:1980-1988. [PMID: 32476407 PMCID: PMC8178726 DOI: 10.1021/acs.chemrestox.0c00161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Our early studies demonstrated an impressive chemopreventive efficacy of dihydromethysticin (DHM), unique in kava, against tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis in A/J mice in which DHM was supplemented in the diet. The current work was carried out to validate the efficacy, optimize the dosing schedule, and further elucidate the mechanisms using oral bolus dosing of DHM. The results demonstrated a dose-dependent chemopreventive efficacy of DHM (orally administered 1 h before each of the two NNK intraperitoneal injections, 1 week apart) against NNK-induced lung adenoma formation. Temporally, DHM at 0.8 mg per dose (∼32 mg per kg body weight) exhibited 100% lung adenoma inhibition when given 3 and 8 h before each NNK injection and attained >93% inhibition when dosed at either 1 or 16 h before each NNK injection. The simultaneous treatment (0 h) or 40 h pretreatment (-40 h) decreased lung adenoma burden by 49.8% and 52.1%, respectively. However, post-NNK administration of DHM (1-8 h after each NNK injection) was ineffective against lung tumor formation. In short-term experiments for mechanistic exploration, DHM treatment reduced the formation of NNK-induced O6-methylguanine (O6-mG, a carcinogenic DNA adduct in A/J mice) in the target lung tissue and increased the urinary excretion of NNK detoxification metabolites as judged by the ratio of urinary NNAL-O-gluc to free NNAL, generally in synchrony with the tumor prevention efficacy outcomes in the dose scheduling time-course experiment. Overall, these results suggest DHM as a potential chemopreventive agent against lung tumorigenesis in smokers, with O6-mG and NNAL detoxification as possible surrogate biomarkers.
Collapse
Affiliation(s)
- Qi Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Pedro Corral
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610
| | - Sreekanth C. Narayanapillai
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455
| | - Pablo Leitzman
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455
| | - Pramod Upadhyaya
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - M. Gerard O’Sullivan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108
| | - Stephen S. Hecht
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455
| | - Junxuan Lu
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
32
|
Ye X, Gao Q, Wu J, Zhou L, Tao M. Identification of significant genes in non-small cell lung cancer by bioinformatics analyses. Transl Cancer Res 2020; 9:4330-4340. [PMID: 35117799 PMCID: PMC8799091 DOI: 10.21037/tcr-19-2596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 05/28/2020] [Indexed: 01/10/2023]
Abstract
Background Lung cancer is the most malignant cancer featured with undesirable prognosis. It is urgent to identify novel biomarkers to improve both diagnosis and prognosis. The purpose of the study was to identify significant genes involved in lung cancer through bioinformatic methods and reveal potential underlying mechanisms. Methods Three datasets GSE19188, GSE27262, GSE118375, containing 122 lung cancer and 96 normal tissues, were available from GEO database. GEO2R and Venn diagram online software were applied to pick out differentially expressed genes (DEGs). Next, we used the Database for Annotation, Visualization and Integrated Discovery (DAVID) to analyze Kyoto Encyclopedia of Gene and Genome (KEGG) pathway and gene ontology (GO) enrichment, followed by protein-protein interaction (PPI) of these DEGs visualized by cytoscape. The MCODE plug-in was performed to construct a module complex of DEGs. In addition, Kaplan-Meier analysis was implemented for analysis of overall survival. To further validate the expression of these genes, Gene Expression Profiling Interactive Analysis (GEPIA) was used. Results A total of 149 DEGs were identified, including 127 downregulated genes and 22 upregulated genes. KEGG analysis revealed that the DEGs were mainly enriched in ECM-receptor interaction, Vascular smooth muscle contraction, and PPAR signaling pathway. GO analysis of DEGs showed that significant functional enrichment of angiogenesis, cell adhesion, and vasculogenesis. 13 genes were selected as hub genes based on MCODE, and 11 of 13 genes had a significance. The results of GEPIA were consistent with survival analysis. Furthermore, reanalysis of these genes found they were significantly enriched in ECM-receptor interaction and PI3K-Akt signaling pathway. Conclusions We have identified several key genes, which could be potential diagnostic and prognostic biomarker as well as therapy targets.
Collapse
Affiliation(s)
- Xia Ye
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qian Gao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lin Zhou
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
33
|
Luo Z, Ye X, Shou F, Cheng Y, Li F, Wang G. RNF115-mediated ubiquitination of p53 regulates lung adenocarcinoma proliferation. Biochem Biophys Res Commun 2020; 530:425-431. [PMID: 32553631 DOI: 10.1016/j.bbrc.2020.05.061] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 01/20/2023]
Abstract
Lung adenocarcinoma (LAC) represents approximately 40% of all lung cancer cases and is the leading cause of cancer-associated mortality worldwide. Although combined treatment, including radiotherapy, chemotherapy, surgical treatment and immunotherapy, has been used in treating LAC, the five-year survival rate of patients with LAC has not significantly improved. Therefore, it is vital for cancer research to investigate novel prognostic markers and new targets for molecular targeted therapy in LAC. TP53 is an important tumor suppressor gene and is frequently inactivated in lung cancer, thus upregulation or activation of p53 may be a novel targeted therapy for LAC. The present study found that RNF115 mediates ubiquitination of p53 and predicts poor prognosis of patients with LAC. Functionally, it was demonstrated that disruption of RNF115 significantly inhibited cell viability in vitro through inducing G1 phase arrest of LAC cells, which reduced tumor growth in an xenograft model. Taken together, these results suggest that RNF115 could be a novel prognostic biomarker and the RNF115-p53 axis may be a potential target for LAC therapy.
Collapse
Affiliation(s)
- Zhigang Luo
- Department of Oncology, the People's Hospital of Jianyang, No.180 Hospital Road, Jianyang City, Sichuan Province, China.
| | - Xin Ye
- Department of Oncology, the People's Hospital of Deyang, No.173 Taishangbei Road, Deyang City, Sichuan Province, China
| | - Feng Shou
- Department of Oncology, the People's Hospital of Jianyang, No.180 Hospital Road, Jianyang City, Sichuan Province, China
| | - Yang Cheng
- Department of Oncology, the People's Hospital of Jianyang, No.180 Hospital Road, Jianyang City, Sichuan Province, China
| | - Fugang Li
- Department of Oncology, the People's Hospital of Jianyang, No.180 Hospital Road, Jianyang City, Sichuan Province, China
| | - Gang Wang
- Department of Oncology, the People's Hospital of Jianyang, No.180 Hospital Road, Jianyang City, Sichuan Province, China
| |
Collapse
|
34
|
Li B, Zhou D, Li S, Feng Y, Li X, Chang W, Zhang J, Sun Y, Qing D, Chen G, Li N. Licochalcone A reverses NNK-induced ectopic miRNA expression to elicit in vitro and in vivo chemopreventive effects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153245. [PMID: 32505917 DOI: 10.1016/j.phymed.2020.153245] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/14/2020] [Accepted: 05/10/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Chemoprevention is the best cost-effective way regarding cancers. MicroRNAs (miRNAs) have been reported to be differentially expressed during the development of lung cancer. However, if lung cancer prevention can be achieved through modulating miRNAs expression so far remains unknown. PURPOSE To discover ectopically expressed miRNAs in NNK-induced lung cancer and clarify whether Licochalcone A (lico A) can prevent NNK-induced lung cancer by modulating miRNA expression. STUDY DESIGN AND METHODS A/J mice were used to construct a lung cancer model by intraperitoneal injection with physiological saline NNK (100 mg/kg). Chemopreventive effects of lico A against lung cancer at 2 mg/kg and 20 mg/kg doses were evaluated in vivo. MicroRNA array and RT-qPCR were used to assess the expression levels of miRNAs. MLE-12 cells were treated with 0.1 mg/ml NNK, stimulating the ectopic expression pattern of miR-144-3p, miR-20a-5p, miR-29c-3p, let-7d-3p, and miR-328-3p. miR-144-3p mimics and inhibitors were used to manipulate miR-144-3p levels. The effects of lico A (10 μM) on cell cycle distribution, apoptosis, and the expression of CK19, RASA1, miR-144-3p, miR-20a-5p, miR-29c-3p, let-7d-3p, and miR-328-3p in NNK-treated MLE-12 cells were studied. RESULTS The expression levels of miR-144-3p, miR-20a-5p, and miR-29c-3p increased, while those of let-7d-3p and miR-328-3p decreased in both NNK-induced A/J mice and MLE-12 cells. Lico A could reverse the NNK-induced ectopic miRNA (miR-144-3p, miR-20a-5p, miR-29c-3p, let-7d-3p, and miR-328-3p) expression both in vivo and in vitro and elicit in vivo lung cancer chemopreventive effect against NNK. In MLE-12 cells, the overexpression of miR-144-3p elicited the same effect as NNK regarding the expression of lung cancer biomarker CK19; the silencing of miR-144-3p reversed the effect of NNK on cell cycle distribution and apoptosis. Lico A could reverse the effect of NNK on the expression of miR-144-3p, CK19, and RASA1 (predicted target of miR-144-3p). CONCLUSION The present study suggests that miR-144-3p, miR-20a-5p, miR-29c-3p, let-7d-3p, and miR-328-3p were involved in the in vivo pathogenesis of NNK-induced lung cancer, and lico A could reverse the effect of NNK both in vivo and in vitro to elicit lung cancer chemopreventive effects through, at least partially, these five ectopically expressed miRNAs, especially miR-144-3p.
Collapse
Affiliation(s)
- Bingxin Li
- School of Traditional Chinese Materia Medica; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Di Zhou
- School of Traditional Chinese Materia Medica; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Shuang Li
- Physical Education College, Guangzhou University, Guangzhou 510006, China
| | - Yuan Feng
- School of Traditional Chinese Materia Medica; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Xingyu Li
- School of Traditional Chinese Materia Medica; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Wenhui Chang
- School of Traditional Chinese Materia Medica; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China
| | - Juan Zhang
- XinJiang Institute of Chinese Materia Medica and Ethnodrug, Urumqi 830002, China
| | - Yu Sun
- XinJiang Institute of Chinese Materia Medica and Ethnodrug, Urumqi 830002, China
| | - Degang Qing
- XinJiang Institute of Chinese Materia Medica and Ethnodrug, Urumqi 830002, China
| | - Gang Chen
- School of Traditional Chinese Materia Medica; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China.
| | - Ning Li
- School of Traditional Chinese Materia Medica; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Shenyang Pharmaceutical University, Shenyang 110016, P.R. China.
| |
Collapse
|
35
|
Park HJ, Chi GY, Choi YH, Park SH. Lupeol suppresses plasminogen activator inhibitor-1-mediated macrophage recruitment and attenuates M2 macrophage polarization. Biochem Biophys Res Commun 2020; 527:889-895. [PMID: 32430175 DOI: 10.1016/j.bbrc.2020.04.160] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/30/2020] [Indexed: 12/25/2022]
Abstract
Tumor-associated macrophages (TAMs) are closely related with poor prognosis of cancers. The current study investigated whether lupeol regulates TAMs by focusing on the recruitment and polarization of macrophages. We found that lupeol suppressed the recruitment of THP-1 macrophages (THP-1 cells differentiated into macrophages) towards H1299 lung carcinoma cells by inhibiting plasminogen activator inhibitor-1 (PAI-1) production from H1299 cells. The reduced migration of THP-1 macrophages by lupeol was recovered by adding recombinant human PAI-1 as a chemoattractant. Knockdown of PAI-1 or treatment of tiplaxtinin, a PAI-1 inhibitor, in H1299 cells abrogated the chemotaxis of macrophages. Furthermore, lupeol suppressed the interleukin (IL)-4- and IL-13-induced M2 macrophage polarization. The mRNA expression of M2 macrophage markers and the phosphorylation of signal transducer and activator of transcription 6 (STAT6) were commonly decreased by lupeol in RAW264.7 cells. In addition, lupeol-suppressed M2 macrophage polarization led to the reduced migration of Lewis lung carcinoma (LLC) cells. Taken together, our results suggest that lupeol attenuates PAI-1-mediated macrophage recruitment towards cancer cells and inhibits M2 macrophage polarization.
Collapse
Affiliation(s)
- Hyun-Ji Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan, 47227, Republic of Korea
| | - Gyoo-Yong Chi
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan, 47227, Republic of Korea
| | - Yung-Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan, 47227, Republic of Korea
| | - Shin-Hyung Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan, 47227, Republic of Korea.
| |
Collapse
|
36
|
Gao C, Sun X, Wu Z, Yuan H, Han H, Huang H, Shu Y, Xu M, Gao R, Li S, Zhang J, Tian J. A Novel Benzofuran Derivative Moracin N Induces Autophagy and Apoptosis Through ROS Generation in Lung Cancer. Front Pharmacol 2020; 11:391. [PMID: 32477104 PMCID: PMC7235196 DOI: 10.3389/fphar.2020.00391] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/16/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction The leaves of Morus alba L is a traditional Chinese medicine widely applied in lung diseases. Moracin N (MAN), a secondary metabolite extracted form the leaves of Morus alba L, is a potent anticancer agent. But its molecular mechanism remains unveiled. Objective In this study, we aimed to examine the effect of MAN on human lung cancer and reveal the underlying molecular mechanism. Methods MTT assay was conducted to measure cell viability. Annexin V-FITC/PI staining was used to detect cell apoptosis. Confocal microscope was performed to determine the formation of autophagosomes and autolysosomes. Flow cytometry was performed to quantify cell death. Western blotting was used to determine the related-signaling pathway. Results In the present study, we demonstrated for the first time that MAN inhibitd cell proliferation and induced cell apoptosis in human non-small-cell lung carcinoma (NSCLC) cells. We found that MAN treatment dysregulated mitochondrial function and led to mitochondrial apoptosis in A549 and PC9 cells. Meanwhile, MAN enhanced autophagy flux by the increase of autophagosome formation, the fusion of autophagsomes and lysosomes and lysosomal function. Moreover, mTOR signaling pathway, a classical pathway regualting autophagy, was inhibited by MAN in a time- and dose-dependent mannner, resulting in autophagy induction. Interestingly, autophagy inhibition by CQ or Atg5 knockdown attenuated cell apoptosis by MAN, indicating that autophagy serves as cell death. Furthermore, autophagy-mediated cell death by MAN can be blocked by reactive oxygen species (ROS) scavenger NAC, indicating that ROS accumulation is the inducing factor of apoptosis and autophagy. In summary, we revealed the molecular mechanism of MAN against lung cancer through apoptosis and autophagy, suggesting that MAN might be a novel therapeutic agent for NSCLC treatment.
Collapse
Affiliation(s)
- Chengcheng Gao
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.,Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individual Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xin Sun
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individual Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Zhipan Wu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Huahua Yuan
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Haote Han
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Hongliang Huang
- School of Biosciences & Biopharmaceutics and Center for Bioresources & Drug Discovery, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuhan Shu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Mengting Xu
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Ruilan Gao
- Institution of Hematology Research, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shouxin Li
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou, China
| | - Jianbin Zhang
- Clinical Research Institute, Key Laboratory of Tumor Molecular Diagnosis and Individual Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jingkui Tian
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China.,Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang-Malaysia Joint Research Center for Traditional Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
37
|
Yang H, Zhao M, Zhao L, Li P, Duan Y, Li G. CircRNA BIRC6 promotes non-small cell lung cancer cell progression by sponging microRNA-145. Cell Oncol (Dordr) 2020; 43:477-488. [PMID: 32297303 DOI: 10.1007/s13402-020-00503-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/01/2020] [Accepted: 03/12/2020] [Indexed: 02/01/2023] Open
Abstract
PURPOSE Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related mortality world-wide. Recently, a number of circular RNAs (circRNAs) has been found to be differentially expressed in human NSCLCs, correlating with clinico-pathological features. As yet, the expression and potential role of circRNA BIRC6 (circBIRC6) in NSCLC have not been studied. METHODS Expression of circBIRC6 and its target microRNA-145 (miR-145) in human NSCLC cells and tissues was assessed using qRT-PCR. In vitro genetic strategies were used to exogenously alter circBIRC6 and miR-145 expression. Their impact on in vitro and in vivo NSCLC cell behavior was studied. RESULTS We found that circBIRC6 was upregulated in primary human NSCLC tissues and NSCLC cells, whereas its potential target, miR-145, was downregulated. In A549 NSCLC cells and primary human NSCLC cells, shRNA-induced silencing of circBIRC6 potently inhibited their growth, proliferation, migration and invasion. Conversely, we found that exogenous overexpression of circBIRC6 promoted these characteristics. Using RNA immunoprecipitation (RIP) in A549 cells, we found that Argonaute 2 (Ago2) immunoprecipitated together with both circBIRC6 and miR-145. Additional studies revealed that the miR-145 level increased after circBIRC6 silencing in A549 cells, but decreased after circBIRC6 overexpression. Of note, we found that the circBIRC6 silencing-induced anti-A549 activity could be attenuated by a miR-145 inhibitor. Lastly, we found that circBIRC6 silencing inhibited the growth of NSCLC xenografts in severe combined immunodeficient mice. CONCLUSIONS From our data we conclude that circBIRC6 overexpression promotes NSCLC cell progression, possibly by sponging miR-145.
Collapse
Affiliation(s)
- Han Yang
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengjing Zhao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, South Bai-xiang street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Lihao Zhao
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ping Li
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Yuxia Duan
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, South Bai-xiang street, Ouhai District, Wenzhou, 325000, Zhejiang, China.
| | - Gang Li
- Department of Chemoradiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
38
|
Dong L, Zheng J, Gao Y, Zhou X, Song W, Huang J. The circular RNA NT5E promotes non-small cell lung cancer cell growth via sponging microRNA-134. Aging (Albany NY) 2020; 12:3936-3949. [PMID: 32096481 PMCID: PMC7066882 DOI: 10.18632/aging.102861] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
The current study tested expression and potential function of circular RNA ecto-5’-nucleotidase (circNT5E) in human non-small cell lung cancer (NSCLC). We show that circNT5E levels are significantly elevated in human NSCLC tissues and cells, correlating with downregulation of its potential targets, miR-134, miR-422a and miR-338. In A549 and primary NSCLC cells, circNT5E shRNA inhibited cancer cell growth, proliferation and migration, whiling inducing apoptosis activation. Conversely, ectopic circNT5E overexpression promoted A549 cell progression in vitro. miR-134 is the primary target of circNT5E in lung cancer cells. RNA-Pull down assay in A549 cells confirmed the direct association between biotinylated-miR-134 and circNT6E. miR-134 levels were significantly increased in circNT5E-silenced A549 cells, but reduced with circNT5E overexpression. Forced overexpression of miR-134 mimicked circNT5E shRNA-induced actions, inhibiting NSCLC cell growth and proliferation. In contrast, miR-134 inhibition largely attenuated circNT5E shRNA-induced anti-NSCLC cell activity. Importantly, circNT5E shRNA was ineffective in miR-134-overexpressed A549 cells. Collectively, circNT5E promotes human NSCLC cell progression possibly by sponging miR-134.
Collapse
Affiliation(s)
- Lingyun Dong
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Respiratory Medicine, Affiliated Wujiang Hospital of Nantong University, Suzhou, China
| | - Jiangnan Zheng
- Department of Respiratory Medicine, Affiliated Wujiang Hospital of Nantong University, Suzhou, China
| | - Yun Gao
- Department of General Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaoting Zhou
- Department of Respiratory Medicine, Affiliated Wujiang Hospital of Nantong University, Suzhou, China
| | - Weizhen Song
- Department of Respiratory Medicine, Affiliated Wujiang Hospital of Nantong University, Suzhou, China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
39
|
Zheng J, Zhang Y, Cai S, Dong L, Hu X, Chen MB, Zhu YH. MicroRNA-4651 targets bromodomain-containing protein 4 to inhibit non-small cell lung cancer cell progression. Cancer Lett 2020; 476:129-139. [PMID: 32081805 DOI: 10.1016/j.canlet.2020.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/13/2022]
Abstract
Bromodomain-containing protein 4 (BRD4) overexpression in non-small cell lung cancer (NSCLC) promotes cancer progression. Here, we show that miR-4651 selectively targets and negatively regulates BRD4 in A549 and primary human NSCLC cells. RNA pull-down experiments confirmed that miR-4651 directly binds to BRD4 mRNA. Further, ectopic overexpression of miR-4651 in A549 cells and primary NSCLC cells decreased BRD4 3'-UTR luciferase reporter activity and its expression, whereas miR-4651 inhibition elevated both. Functional studies demonstrated that NSCLC cell growth, proliferation, and migration were suppressed with ectopic miR-4651 overexpression but enhanced with miR-4651 inhibition. BRD4 re-expression using a 3'-UTR mutant BRD4 reversed A549 cell inhibition induced by miR-4651 overexpression. Further, miR-4651 overexpression or inhibition failed to alter the functions of BRD4-KO A549 cells. In vivo, miR-4651-overexpressing A549 xenografts grew slowly than control A549 xenografts in severe combined immunodeficient mice. Finally, miR-4651 was downregulated in human NSCLC tissues, correlating with BRD4 elevation. Together, miR-4651 targets BRD4 to inhibit NSCLC cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Jiangnan Zheng
- Department of Respiratory Medicine, Affiliated Wujiang Hospital of Nantong University, Suzhou, China; Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
| | - Shang Cai
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingyun Dong
- Department of Respiratory Medicine, Affiliated Wujiang Hospital of Nantong University, Suzhou, China
| | - Xiaoyun Hu
- Department of Respiratory Medicine, Affiliated Wujiang Hospital of Nantong University, Suzhou, China
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China.
| | - Ye-Han Zhu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
40
|
Shi Y, Zhu S, Yang J, Shao M, Ding W, Jiang W, Sun X, Yao N. Investigation of Potential Mechanisms Associated with Non-small Cell Lung Cancer. J Comput Biol 2020; 27:1433-1442. [PMID: 32048869 DOI: 10.1089/cmb.2019.0081] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This study aimed at investigating the crucial mechanisms underlying non-small cell lung cancer (NSCLC). NSCLC-related microarray data GSE27262 were downloaded from Gene Expression Omnibus, including 7 NSCLC 1a samples, 18 NSCLC 1b samples, and their matched normal samples. The common differentially expressed genes (DEGs) between NSCLC 1a and NSCLC 1b samples were identified, followed by protein-protein interaction (PPI) network construction, functional enrichment analysis, and weighted gene co-expression network analysis (WGCNA). Further, the key DEGs were confirmed based on the lung adenocarcinoma (LUAD) data from the Cancer Genome Atlas (TCGA) database, followed by clinical prognostic analysis. There were 802 (NSCLC 1a) and 734 (NSCLC 1b) DEGs identified. By intersection analysis, we obtained 255 upregulated and 97 downregulated common DEGs. Upregulated DEGs were significantly enriched in the plasma membrane and extracellular region, whereas the downregulated DEGs were significantly enriched in the cytoskeleton and cell cycle process. Topoisomerase (DNA) II alpha (TOP2A) and cyclin B1 (CCNB1) were hub nodes in the PPI network. Based on WGCNA, 5 modules were obtained. In the module MEgreen, DEGs were significantly enriched in cytokine-cytokine receptor interaction and focal adhesion. Notably, 1797 DEGs were identified based on the LUAD data from the TCGA database; among them, 285 DEGs were common DEGs identified from GSE27262 data. Upregulation of TOP2A and CCNB1 was correlated with poor survival of patients. The hub genes and key pathways identified in this study are helpful for a comprehensive knowledge of the molecular mechanisms of NSCLC.
Collapse
Affiliation(s)
- Yu Shi
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong, China
| | - Shan Zhu
- Department of Anesthesiology, Nantong Tumor Hospital, Nantong, China
| | - Jianxin Yang
- Department of General Surgery, Qidong People's Hospital, Qidong Liver Cancer Institute, Nantong, China
| | - Minghai Shao
- Department of Radiotherapy, Taizhou Hospital of Zhejiang Province, Taizhou, China
| | - Wenxiu Ding
- Department of Radiotherapy, Taixing People's Hospital, Taizhou, China
| | - Wanrong Jiang
- Department of Radiotherapy, People's Liberation Army (PLA) 81 Hospital, Nanjing, China
| | - Xinchen Sun
- Department of Radiotherapy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ninghua Yao
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
41
|
Wang F, Meng F, Wong SCC, Cho WC, Yang S, Chan LW. Combination therapy of gefitinib and miR-30a-5p may overcome acquired drug resistance through regulating the PI3K/AKT pathway in non-small cell lung cancer. Ther Adv Respir Dis 2020; 14:1753466620915156. [PMID: 32552611 PMCID: PMC7303773 DOI: 10.1177/1753466620915156] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/28/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) patients with an epidermal growth factor receptor (EGFR) mutation often initially respond to EGFR tyrosine kinase inhibitor (EGFR-TKI) treatment but may acquire drug resistance due to multiple factors. MicroRNAs are a class of small noncoding and endogenous RNA molecules that may play a role in overcoming the resistance. MATERIALS AND METHODS In this study, we explored and validated, through in vitro experiments and in vivo models, the ability of a combination treatment of EGFR-TKI, namely gefitinib, and a microRNA mimic, miR-30a-5p, to overcome drug resistance through regulation of the insulin-like growth factor receptor-1 (IGF1R) and hepatocyte growth factor receptor signaling pathways, which all converge on phosphatidylinositol 3 kinase (PI3K), in NSCLC. First, we examined the hypothesized mechanisms of drug resistance in H1650, H1650-acquired gefitinib-resistance (H1650GR), H1975, and H460 cell lines. Next, we investigated a potential combination treatment approach to overcome acquired drug resistance in the H1650GR cell line and an H1650GR cell implanted mouse model. RESULTS Dual inhibitors of EGFR and IGF1R significantly lowered the expression levels of phosphorylated protein kinase B (p-AKT) and phosphorylated mitogen-activated protein kinase (p-ERK) compared with the control group in all cell lines. With the ability to repress PI3K expression, miR-30a-5p mimics induced cell apoptosis, and inhibited cell invasion and migration in the treated H1650GR cell line. CONCLUSION Gefitinib, combined with miR-30a-5p mimics, effectively suppressed the growth of H1650GR-induced tumor in xenografts. Hence, a combination therapy of gefitinib and miR-30a-5p may play a critical role in overcoming acquired resistance to EGFR-TKIs. The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Fengfeng Wang
- Department of Health Technology and Informatics,
The Hong Kong Polytechnic University, Hong Kong, P.R. China
| | - Fei Meng
- Department of Health Technology and Informatics,
The Hong Kong Polytechnic University, Hong Kong, P.R. China
| | - Sze Chuen Cesar Wong
- Department of Health Technology and Informatics,
The Hong Kong Polytechnic University, Hong Kong, P.R. China
| | - William C.S. Cho
- Department of Clinical Oncology, Queen Elizabeth
Hospital, Hong Kong, P.R. China
| | - Sijun Yang
- ABSL-3 Laboratory at the Center for Animal
Experiment and Institute of Animal Model for Human Disease, Wuhan University
School of Medicine, Wuhan, P.R. China
| | - Lawrence W.C. Chan
- Department of Health Technology and Informatics,
The Hong Kong Polytechnic University, Y902, 9/F, Lee Shau Kee Building,
Kowloon, Hong Kong, P.R. China
| |
Collapse
|
42
|
Synergistic Effect of Network-Based Multicomponent Drugs: An Investigation on the Treatment of Non-Small-Cell Lung Cancer with Compound Liuju Formula. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9854047. [PMID: 31949474 PMCID: PMC6948348 DOI: 10.1155/2019/9854047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/25/2019] [Accepted: 11/12/2019] [Indexed: 11/22/2022]
Abstract
Lung cancer is the most common cause of cancer death with high morbidity and mortality, which non-small-cell lung cancer (NSCLC) accounting for the majority. Traditional Chinese Medicine (TCM) is effective in the treatment of complex diseases, especially cancer. However, TCM is still in the conceptual stage. The interaction between different components remains unknown due to its multicomponent and multitarget characteristics. In this study, compound Liuju formula was taken as an example to isolate compounds with synergistic biological activity through systems pharmacology strategy. Through pharmacokinetic evaluation, 37 potentially active compounds were screened out. Meanwhile, 116 targets of these compounds were obtained by combing with the target prediction model. Through network analysis, we found that multicomponent drugs can present a synergistic effect through regulating inflammatory signaling pathway, invasion pathway, proliferation, and apoptosis pathway. Finally, it was confirmed that the bioactive compounds of compound Liuju formula have not only a killing effect on NSCLC tumor cells but also a synergistic effect on inhibiting the secretion of correlative inflammatory mediators, including TNF-α and IL-1β. The systems pharmacology method was applied in this study, which provides a new direction for analyzing the mechanism of TCM.
Collapse
|
43
|
Sun X, Cui S, Fu X, Liu C, Wang Z, Liu Y. MicroRNA-146-5p promotes proliferation, migration and invasion in lung cancer cells by targeting claudin-12. Cancer Biomark 2019; 25:89-99. [PMID: 31006666 DOI: 10.3233/cbm-182374] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
MicroRNAs (miRNAs) have been regarded as important regulators in different pathological processes of cells. Abnormal expression of miRNAs is frequently associated with cell proliferation, metastasis and apoptosis in various cancers. This study aimed to explore the effect of miR-146-5p on cell growth, metastasis and its mechanism in lung cancer cells. The expressions of miR-146-5p and claudin-12 in A549 and WI-38 cells were altered by transient transfection. Cisplatin was used to develop cells for regulation of cisplatin sensitivity. Cell viability, migration, invasion, and apoptosis were analyzed by CCK-8, Transwell and flow cytometry assays. The protein expressions of Wnt/β-catenin and PI3K/AKT/MAPK pathway-related factors were detected. miR-146-5p suppression inhibited cell viability, migration and invasion but promoted apoptosis in A549 cells. Moreover, overexpression of miR-146-5p reduced the sensitivity of A549 cells and WI-38 cells to cisplatin. In addition, claudin-12 was a direct target of miR-146-5p and was negatively regulated by miR-146-5p. Claudin-12 silence significantly reversed miR-146-5p suppression-mediated anti-tumor effects in A549 cells. Furthermore, miR-146-5p overexpression activated Wnt/β-catenin and PI3K/AKT/MAPK signal pathways via down-regulation of claudin-12. The results indicated that miR-146-5p promoted cell viability, migration and invasion, inhibited apoptosis and activated Wnt/β-catenin and PI3K/AKT/MAPK signal pathways by regulating claudin-12 expression in lung cancer cells.
Collapse
Affiliation(s)
- Xianghong Sun
- Department of Outpatient (Tumor), The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Shichao Cui
- Department of Respiratory Medicine, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Xiaofeng Fu
- Department of Outpatient, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Chuan Liu
- Department of Surgery, Qingdao West Coast New District No.2 People's Hospital, Qingdao, Shandong, China
| | - Zhi Wang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University (Shinan Branch), Qingdao, Shandong, China
| | - Yuanwei Liu
- Department of Cancer Comprehensive Therapy, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| |
Collapse
|
44
|
Imatinib modulates pro-inflammatory microenvironment with angiostatic effects in experimental lung carcinogenesis. Inflammopharmacology 2019; 28:231-252. [PMID: 31676982 DOI: 10.1007/s10787-019-00656-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 10/10/2019] [Indexed: 10/25/2022]
Abstract
Lung cancer has second highest rate of incidence and mortality around the world. Smoking cigarettes is the main stream cause of lung carcinogenesis along with other factors such as spontaneous mutations, inactivation of tumor suppressor genes. The present study was aimed to identify the mechanistic role of Imatinib in the chemoprevention of experimental lung carcinogenesis in rat model. Gross morphological observations for tumor formation, histological examinations, RT-PCR, Western blotting, fluorescence spectroscopy and molecular docking studies were performed to elucidate the chemopreventive effects of Imatinib and support our hypothesis by various experiments. It is evident that immuno-compromised microenvironment inside solid tumors is responsible for tumor progression and drug resistance. Therefore, it is inevitable to modulate the pro-inflammatory signaling inside solid tumors to restrict neoangiogenesis. In the present study, we observed that Imatinib could downregulate the inflammatory signaling and also attributed angiostatic effects. Moreover, Imatinib also altered the biophysical properties of BAL cells such as plasma membrane potential, fluidity and microviscosity to restrict their infiltration and thereby accumulation to mount immuno-compromised environment inside the solid tumors during angiogenesis. Our molecular docking studies suggest that immunomodulatory and angiostatic properties of Imatinib could be either independent of each other or just a case of synergistic pleiotropy. Imatinib was observed to activate the intrinsic or mitochondrial pathway of apoptosis to achieve desired effects in cancer cell killings. Interestingly, binding of Imatinib inside the catalytic domain of PARP-1 also suggests that it has caspase-independent properties in promoting cancer cell deaths.
Collapse
|
45
|
Isorhamnetin inhibited migration and invasion via suppression of Akt/ERK-mediated epithelial-to-mesenchymal transition (EMT) in A549 human non-small-cell lung cancer cells. Biosci Rep 2019; 39:BSR20190159. [PMID: 31467176 PMCID: PMC6753323 DOI: 10.1042/bsr20190159] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 08/06/2019] [Accepted: 08/16/2019] [Indexed: 12/31/2022] Open
Abstract
In the present study, we investigated the potential effects of Isorhamnetin on the growth and metastasis of A549 human lung cancer cells, as well as the underlying mechanism. Treatment with Isorhamnetin exhibited a dose- and time-dependent inhibition on A549 cell proliferation. Furthermore, the cell adhesion and Transwell assay showed that treatment with Isorhamnetin (2.5, 5, and 10 μM) for 48 h resulted in a significant inhibition effect on cell adhesion, invasion and migration of A549 cells, depending on concentration, which was associated with the suppression of matrix metalloproteinase (MMP)-2 and MMP-9 activity and protein expression. Moreover, Isorhamnetin effectively suppressed the expressions of epithelial-to-mesenchymal transition (EMT) markers, as evidenced by the down-regulation of N-cadherin, vimentin and snail, as well as up-regulation of E-cadherin protein expression. Additionally, these inhibitions were mediated by interrupting AKT/ERK1/2 signaling pathways. Taken together, the results of the current study demonstrated that Isorhamnetin may become a good anti-metastastic agent against lung cancer A549 cell line by the suppression of EMT via interrupting Akt/ERK1/2 signaling pathway.
Collapse
|
46
|
Circular RNA PRKCI promotes glioma cell progression by inhibiting microRNA-545. Cell Death Dis 2019; 10:616. [PMID: 31409777 PMCID: PMC6692337 DOI: 10.1038/s41419-019-1863-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 12/12/2022]
Abstract
We here tested expression and potential functions of circular RNA PRKCI (circPRKCI) in human glioma. Our results show that circPRKCI is upregulated in human glioma tissues and glioma cells, correlating with downregulation of its potential target, microRNA-545 (miR-545). In A172 and primary human glioma cells, shRNA-mediated silencing of circPRKCI inhibited cancer cell growth, survival, proliferation, and migration. Conversely, ectopic circPRKCI overexpression promoted A172 cell progression. miR-545 is the primary target of circPRKCI in glioma cells. Forced overexpression of miR-545 mimicked circPRKCI shRNA-induced actions, inhibiting glioma cell survival and proliferation. In contrast, miR-545 inhibition, by a lentiviral antagomiR-545 construct, reversed circPRKCI shRNA-induced anti-A172 cell activity. Importantly, neither circPRKCI shRNA nor circPRKCI overexpression was effective in miR-545-knockout (Cas9 method) A172 cells. Importantly, the subcutaneous and orthotopic A172 xenograft growth was significantly inhibited by circPRKCI silencing. Collectively, circPRKCI promotes human glioma cell progression possibly by inhibiting miR-545. Targeting circPRKCI-miR-545 cascade could efficiently inhibit human glioma cells.
Collapse
|
47
|
Wang C, Li X, Jin L, Zhao Y, Zhu G, Shen W. Dieckol inhibits non-small-cell lung cancer cell proliferation and migration by regulating the PI3K/AKT signaling pathway. J Biochem Mol Toxicol 2019; 33:e22346. [PMID: 31291034 PMCID: PMC6771741 DOI: 10.1002/jbt.22346] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/18/2019] [Accepted: 04/08/2019] [Indexed: 01/02/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is one of the most prevalent type of lung cancers with an increased mortality rate in both developed and developing countries worldwide. Dieckol is one such polyphenolic drug extracted from brown algae which has proven antioxidant and anti-inflammatory properties. In the present study, we evaluated the anticancer property of dieckol against NSCLC cell line A549. The LC50 value of dieckol was found to be 25 µg/mL by performing 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the antiapoptotic property of dieckol was analyzed by dual staining technique with acridine orange/propidium iodide (AO/PI) stains. It was further confirmed with flow cytometry analysis with Annexin FITC and JC-1 staining and the anti-invasive property was assessed by Transwell assay. The molecular mechanism of dieckol anticancer activity was confirmed by estimating the levels of caspases and by estimating the signaling proteins of Pi3K/AKT/mTOR signaling pathway using the immunoblotting technique. Our data suggest that dieckol is potent anticancer agent, it effectively inhibits the invasive and migratory property A549 cells and it also induces apoptosis via inhibiting Pi3K/AKT/mTOR signaling, activating the tumor suppressor protein E-cadherin signifying that dieckol is potent natural anticancer drug to treat NSCLC.
Collapse
Affiliation(s)
- Chun‐Hong Wang
- Department of Oncology and HematologyThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Xiao‐Feng Li
- Department of Oncology and HematologyThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Li‐Fang Jin
- Department of Oncology and HematologyThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Yan Zhao
- Department of Oncology and HematologyThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Geng‐Jun Zhu
- Department of Oncology and HematologyThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Wei‐Zhang Shen
- Department of Oncology and HematologyThe Second Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
48
|
Sun S, Wu C, Yang C, Chen J, Wang X, Nan Y, Huang Z, Ma L. Prognostic roles of mitochondrial transcription termination factors in non-small cell lung cancer. Oncol Lett 2019; 18:3453-3462. [PMID: 31516563 PMCID: PMC6732965 DOI: 10.3892/ol.2019.10680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 05/02/2019] [Indexed: 12/13/2022] Open
Abstract
Mitochondrial transcription termination factors (MTERFs) regulate mitochondrial gene transcription and metabolism in numerous types of cells. Previous studies have indicated that MTERFs serve pivotal roles in the pathogenesis of various cancer types. However, the expression and prognostic roles of MTERFs in patients with non-small cell lung cancer (NSCLC) remain elusive. The present study investigated the gene alteration frequency and expression level using Gene Expression Omnibus datasets and reverse transcription-quantitative polymerase chain reaction, and evaluated the prognostic roles of MTERFs in patients with NSCLC using the Kaplan-Meier plotter database. In human lung cancer tissues, it was observed that the mRNA levels of MTERF1, 2, 3 and 4 were positively associated with the copy number of these genes. The mRNA expression levels of MTERF1 and 3 were significantly increased in NSCLC tissues compared with adjacent non-tumor tissues; however, the mRNA expression of MTERF2 was significantly decreased in NSCLC tissues. High mRNA expression levels of MTERF1, 2, 3 and 4 were strongly associated with an improved overall survival rate (OS) in patients with lung adenocarcinoma. Additionally, high mRNA expression levels of MTERF1, 2, 3 and 4 were also strongly associated with an improved OS of patients with NSCLC in the earlier stages of disease (stage I) or patients with negative surgical margins. These results indicate the critical prognostic values of MTERF expression levels in NSCLC. The findings of the present study may be beneficial for understanding the molecular biology mechanism of NSCLC and for generating effective therapeutic approaches for patients with NSCLC.
Collapse
Affiliation(s)
- Shuangyan Sun
- Department of Radiology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| | - Chunjiao Wu
- Department of Thoracic Oncology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| | - Changliang Yang
- Department of Thoracic Oncology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| | - Jian Chen
- Department of Interventional Radiology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| | - Xiu Wang
- Department of Interventional Radiology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| | - Yingji Nan
- Department of Radiology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| | - Zhicheng Huang
- Department of Radiology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| | - Lixia Ma
- Department of Thoracic Oncology, Jilin Province Cancer Hospital, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
49
|
Min TR, Park HJ, Ha KT, Chi GY, Choi YH, Park SH. Suppression of EGFR/STAT3 activity by lupeol contributes to the induction of the apoptosis of human non‑small cell lung cancer cells. Int J Oncol 2019; 55:320-330. [PMID: 31115519 DOI: 10.3892/ijo.2019.4799] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/07/2019] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to investigate the underlying mechanisms responsible for the anticancer effects of lupeol on human non‑small cell lung cancer (NSCLC). MTT assay and Trypan blue exclusion assay were used to evaluate the cell viability. DAPI staining and flow cytometric analysis were used to detect apoptosis. Molecular docking and western blot analysis were performed to determine the target of lupeol. We found that lupeol suppressed the proliferation and colony formation of NSCLC cells in a dose‑dependent manner. In addition, lupeol increased chromatin condensation, poly(ADP‑ribose) polymerase (PARP) cleavage, sub‑G1 cell populations, and the proportion of Annexin V‑positive cells, indicating that lupeol triggered the apoptosis of NSCLC cells. Notably, lupeol inhibited the phosphorylation of epithelial growth factor receptor (EGFR). A docking experiment revealed that lupeol directly bound to the tyrosine kinase domain of EGFR. We observed that the signal transducer and activator of transcription 3 (STAT3), a downstream molecule of EGFR, was also dephosphorylated by lupeol. Lupeol suppressed the nuclear translocation and transcriptional activity of STAT3 and downregulated the expression of STAT3 target genes. The constitutive activation of STAT3 by STAT3 Y705D overexpression suppressed lupeol‑induced apoptosis, demonstrating that the inhibition of STAT3 activity contributed to the induction of apoptosis. The anticancer effects of lupeol were consistently observed in EGFR tyrosine kinase inhibitor (TKI)‑resistant H1975 cells (EGFR L858R/T790M). Taken together, the findings of this study suggest that lupeol may be used, not only for EGFR TKI‑naïve NSCLC, but also for advanced NSCLC with acquired resistance to EGFR TKIs.
Collapse
Affiliation(s)
- Tae-Rin Min
- Department of Pathology, College of Korean Medicine, Dong‑Eui University, Busan 47227, Republic of Korea
| | - Hyun-Ji Park
- Department of Pathology, College of Korean Medicine, Dong‑Eui University, Busan 47227, Republic of Korea
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medicine Research Center, Busan National University, Yangsan, Gyeongsangnam‑do 50612, Republic of Korea
| | - Gyoo-Yong Chi
- Department of Pathology, College of Korean Medicine, Dong‑Eui University, Busan 47227, Republic of Korea
| | - Yung-Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong‑Eui University, Busan 47227, Republic of Korea
| | - Shin-Hyung Park
- Department of Pathology, College of Korean Medicine, Dong‑Eui University, Busan 47227, Republic of Korea
| |
Collapse
|
50
|
Min TR, Park HJ, Park MN, Kim B, Park SH. The Root Bark of Morus alba L. Suppressed the Migration of Human Non-Small-Cell Lung Cancer Cells through Inhibition of Epithelial⁻Mesenchymal Transition Mediated by STAT3 and Src. Int J Mol Sci 2019; 20:ijms20092244. [PMID: 31067694 PMCID: PMC6539721 DOI: 10.3390/ijms20092244] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/02/2019] [Accepted: 05/05/2019] [Indexed: 12/28/2022] Open
Abstract
The root bark of Morus alba L. (MA) has been traditionally used for the treatment of various lung diseases in Korea. Although recent research has demonstrated its anticancer effects in several cancer cells, it is still unclear whether MA inhibits the migratory ability of lung cancer cells. The present study investigated the effects of MA on the migration of lung cancer cells and explored the underlying mechanism. Results from a transwell assay and wound-healing assay demonstrated that methylene chloride extracts of MA (MEMA) suppressed the migration and invasion of H1299, H460, and A549 human non-small-cell lung cancer (NSCLC) cells in a concentration-dependent manner. Results from Western blot analyses showed that MEMA reduced the phosphorylation of STAT3 and Src. In addition, MEMA downregulated the expression of epithelial–mesenchymal transition (EMT) marker proteins including Slug, Snail, Vimentin, and N-cadherin, while upregulating the expression of Occludin—a tight-junction protein. The regulation of EMT markers and the decrease of migration by MEMA treatment were reversed once phospho-mimetic STAT3 (Y705D) or Src (Y527F) was transfected into H1299 cells. In conclusions, MEMA inhibited the migratory activity of human NSCLC cells through blocking Src/STAT3-mediated EMT.
Collapse
Affiliation(s)
- Tae-Rin Min
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan 47227, Korea.
| | - Hyun-Ji Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan 47227, Korea.
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea.
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea.
| | - Shin-Hyung Park
- Department of Pathology, College of Korean Medicine, Dong-eui University, Busan 47227, Korea.
| |
Collapse
|