1
|
Mahajan S, Aalhate M, Chatterjee E, Singh H, Sharma A, Maji I, Gupta U, Guru SK, Singh PK. Harnessing the targeting potential of hyaluronic acid for augmented anticancer activity and safety of duvelisib-loaded nanoparticles in hematological malignancies. Int J Biol Macromol 2024; 282:136600. [PMID: 39427787 DOI: 10.1016/j.ijbiomac.2024.136600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/03/2024] [Accepted: 10/12/2024] [Indexed: 10/22/2024]
Abstract
Duvelisib (DUV) is effective against numerous hematological malignancies; however, it suffers from numerous setbacks like poor aqueous solubility, low cellular uptake and adverse effects. Hyaluronic acid is an excellent ligand for CD44 receptors that are overexpressed on cancer cell surfaces. Thus, for the targeted delivery of DUV in hematological malignancies, we have fabricated hyaluronic acid-coated polylactide-co-glycolide nanoparticles (DUV-P/CH/HA-NPs) through electrostatic interactions. DUV-P/CH/HA-NPs exhibited optimum characteristics such as mean particle size of 183.63 ± 0.23 nm, polydispersity index of 0.261 ± 0.02 and drug loading capacity of 5.75 ± 0.05 %. An in-vitro release study demonstrated sustained release behavior of DUV-P/CH/HA-NPs (77.65 ± 2.89 % release in 48 h). The flow cytometry experiments revealed 1.62-fold and 1.50-fold enhanced uptake of DUV-P/CH/HA-NPs compared to non-coated nanoparticles in MOLT-4 and HH cells, respectively. The DUV-P/CH/HA-NPs showed higher cytotoxicity, arrested the cell cycle in G0/G1 phase and showed increased apoptosis compared to non-coated nanoparticles and free DUV. An in-vivo pharmacokinetic study revealed 2.9-fold and 3.6-fold enhancement in AUC0-t and MRT with the DUV-P/CH/HA-NPs compared to free DUV. Further, toxicity evaluation and hemolysis assessment of DUV-P/CH/HA-NPs indicated good safety for intravenous administration. Conclusively, DUV-P/CH/HA-NPs are an excellent option for selectively targeting hematological malignant cells.
Collapse
Affiliation(s)
- Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad, India
| | - Essha Chatterjee
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Hoshiyar Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Indrani Maji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad, India
| | - Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad, India.
| |
Collapse
|
2
|
Elshazly AM, Xu J, Melhem N, Abdulnaby A, Elzahed AA, Saleh T, Gewirtz DA. Is Autophagy Targeting a Valid Adjuvant Strategy in Conjunction with Tyrosine Kinase Inhibitors? Cancers (Basel) 2024; 16:2989. [PMID: 39272847 PMCID: PMC11394573 DOI: 10.3390/cancers16172989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) represent a relatively large class of small-molecule inhibitors that compete with ATP for the catalytic binding site of tyrosine kinase proteins. While TKIs have demonstrated effectiveness in the treatment of multiple malignancies, including chronic myelogenous leukemia, gastrointestinal tumors, non-small cell lung cancers, and HER2-overexpressing breast cancers, as is almost always the case with anti-neoplastic agents, the development of resistance often imposes a limit on drug efficacy. One common survival response utilized by tumor cells to ensure their survival in response to different stressors, including anti-neoplastic drugs, is that of autophagy. The autophagic machinery in response to TKIs in multiple tumor models has largely been shown to be cytoprotective in nature, although there are a number of cases where autophagy has demonstrated a cytotoxic function. In this review, we provide an overview of the literature examining the role that autophagy plays in response to TKIs in different preclinical tumor model systems in an effort to determine whether autophagy suppression or modulation could be an effective adjuvant strategy to increase efficiency and/or overcome resistance to TKIs.
Collapse
Affiliation(s)
- Ahmed M Elshazly
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Jingwen Xu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Nebras Melhem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, The Hashemite University, Zarqa 13133, Jordan
| | - Alsayed Abdulnaby
- Department of Pharmacognosy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Aya A Elzahed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Tareq Saleh
- Department of Pharmacology and Public Health, Faculty of Medicine, Hashemite University, Zarqa 13133, Jordan
| | - David A Gewirtz
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298, USA
| |
Collapse
|
3
|
Zhao X, Chen C, Han W, Liang M, Cheng Y, Chen Y, Pang D, Lei H, Feng X, Cao S, Li Z, Wang J, Zhang Y, Yang B. EEBR induces Caspase-1-dependent pyroptosis through the NF-κB/NLRP3 signalling cascade in non-small cell lung cancer. J Cell Mol Med 2024; 28:e18094. [PMID: 38214430 PMCID: PMC10844718 DOI: 10.1111/jcmm.18094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/13/2024] Open
Abstract
Lung cancer is a leading cause of cancer-related deaths worldwide. Recent studies have identified pyroptosis, a type of programmed cell death, as a critical process in the development and progression of lung cancer. In this study, we investigated the effect of EEBR, a new compound synthesized by our team, on pyroptosis in non-small cell lung cancer cells (NSCLC) and the underlying molecular mechanisms. Our results demonstrated that EEBR significantly reduced the proliferation and metastasis of NSCLC cells in vitro. Moreover, EEBR-induced pyroptosis in NSCLC cells, as evidenced by cell membrane rupture, the release of cytokines such as interleukin-18 and interleukin-1 beta and the promotion of Gasdermin D cleavage in a Caspase-1-dependent manner. Furthermore, EEBR promoted the nuclear translocation of NF-κB and upregulated the protein level of NLRP3. Subsequent studies revealed that EEBR-induced pyroptosis was suppressed by the inhibition of NF-κB. Finally, EEBR effectively suppressed the growth of lung cancer xenograft tumours by promoting NSCLC pyroptosis in animal models. Taken together, our findings suggest that EEBR induces Caspase-1-dependent pyroptosis through the NF-κB/NLRP3 signalling cascade in NSCLC, highlighting its potential as a candidate drug for NSCLC treatment.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD)HarbinChina
| | - Chao Chen
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Weina Han
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of PharmacyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Min Liang
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Yuanyuan Cheng
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Yingjie Chen
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Defeng Pang
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Haoqi Lei
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Xuefei Feng
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Shifeng Cao
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
| | - Zhixiong Li
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of PharmacyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Jinhui Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of PharmacyHarbin Medical UniversityHarbinHeilongjiangChina
| | - Yan Zhang
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD)HarbinChina
| | - Baofeng Yang
- Department of Pharmacology (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of PharmacyHarbin Medical UniversityHarbinChina
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD)HarbinChina
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone, 2019RU070Chinese Academy of Medical SciencesHarbinChina
| |
Collapse
|
4
|
Singh S, Ghosh P, Roy R, Behera A, Sahadevan R, Kar P, Sadhukhan S, Sonawane A. 4″-Alkyl EGCG Derivatives Induce Cytoprotective Autophagy Response by Inhibiting EGFR in Glioblastoma Cells. ACS OMEGA 2024; 9:2286-2301. [PMID: 38250397 PMCID: PMC10795032 DOI: 10.1021/acsomega.3c06110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/08/2023] [Accepted: 11/16/2023] [Indexed: 01/23/2024]
Abstract
Epidermal growth factor receptor (EGFR)-targeted therapy has been proven vital in the last two decades for the treatment of multiple cancer types, including nonsmall cell lung cancer, glioblastoma, breast cancer and head and neck squamous cell carcinoma. Unfortunately, the majority of approved EGFR inhibitors fall into the drug resistance category because of continuous mutations and acquired resistance. Recently, autophagy has surfaced as one of the emerging underlying mechanisms behind resistance to EGFR-tyrosine kinase inhibitors (TKIs). Previously, we developed a series of 4″-alkyl EGCG (4″-Cn EGCG, n = 6, 8, 10, 12, 14, 16, and 18) derivatives with enhanced anticancer effects and stability. Therefore, the current study hypothesized that 4″-alkyl EGCG might induce cytoprotective autophagy upon EGFR inhibition, and inhibition of autophagy may lead to improved cytotoxicity. In this study, we have observed growth inhibition and caspase-3-dependent apoptosis in 4″-alkyl EGCG derivative-treated glioblastoma cells (U87-MG). We also confirmed that 4″-alkyl EGCG could inhibit EGFR in the cells, as well as mutant L858R/T790M EGFR, through an in vitro kinase assay. Furthermore, we have found that EGFR inhibition with 4″-alkyl EGCG induces cytoprotective autophagic responses, accompanied by the blockage of the AKT/mTOR signaling pathway. In addition, cytotoxicity caused by 4″-C10 EGCG, 4″-C12 EGCG, and 4″-C14 EGCG was significantly increased after the inhibition of autophagy by the pharmacological inhibitor chloroquine. These findings enhance our understanding of the autophagic response toward EGFR inhibitors in glioblastoma cells and suggest a potent combinatorial strategy to increase the therapeutic effectiveness of EGFR-TKIs.
Collapse
Affiliation(s)
- Satyam Singh
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Priya Ghosh
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Rajarshi Roy
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Ananyaashree Behera
- School
of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Orissa 751 024, India
| | - Revathy Sahadevan
- Department
of Chemistry, Indian Institute of Technology
Palakkad, Palakkad, Kerala 678 623, India
| | - Parimal Kar
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| | - Sushabhan Sadhukhan
- Department
of Chemistry, Indian Institute of Technology
Palakkad, Palakkad, Kerala 678 623, India
| | - Avinash Sonawane
- Department
of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh 453 552, India
| |
Collapse
|
5
|
Mishra S, Sahu A, Kaur A, Kaur M, Kumar J, Wal P. Recent Development in the Search for Epidermal Growth Factor Receptor (EGFR) Inhibitors based on the Indole Pharmacophore. Curr Top Med Chem 2024; 24:581-613. [PMID: 37909440 DOI: 10.2174/0115680266264206231020111820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 11/03/2023]
Abstract
The signal transduction and cell proliferation are regulated by the epidermal growth factor receptor. The proliferation of tumor cells, apoptosis, invasion, and angiogenesis is inhibited by the epidermal growth factor receptor. Thus, breast cancer, non-small cell lung cancer, cervical cancer, glioma, and bladder cancer can be treated by targeting the epidermal growth factor receptor. Although third-generation epidermal growth factor receptor inhibitors are potent drugs, patients exhibit drug resistance after treatment. Thus, the search for new drugs is being continued. Among the different potent epidermal growth factor receptor inhibitors, we have reviewed the indole-based inhibitors. We have discussed the structure-activity relationship of the compounds with the active sites of the epidermal growth factor receptor receptors, their synthesis, and molecular docking studies.
Collapse
Affiliation(s)
- Shweta Mishra
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, 122505, India
| | - Adarsh Sahu
- Department of Pharmaceutical Sciences, Dr. Hari Singh Gour Vishwavidyalaya (A Central University), Sagar, 473003, Madhya Pradesh, India
- Amity Institute of Pharmacy, Amity University Rajasthan, NH11C Kant Kanwar Jaipur, 300202, India
| | - Avneet Kaur
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, 122505, India
| | | | - Jayendra Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology, Delhi-NCR Campus, Ghaziabad, UP, 201204, India
| | - Pranay Wal
- Pranveer Singh Institute of Technology, Pharmacy, Kanpur, UP, India
| |
Collapse
|
6
|
Shaban N, Kamashev D, Emelianova A, Buzdin A. Targeted Inhibitors of EGFR: Structure, Biology, Biomarkers, and Clinical Applications. Cells 2023; 13:47. [PMID: 38201251 PMCID: PMC10778338 DOI: 10.3390/cells13010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Members of the EGFR family of tyrosine kinase receptors are major regulators of cellular proliferation, differentiation, and survival. In humans, abnormal activation of EGFR is associated with the development and progression of many cancer types, which makes it an attractive target for molecular-guided therapy. Two classes of EGFR-targeted cancer therapeutics include monoclonal antibodies (mAbs), which bind to the extracellular domain of EGFR, and tyrosine kinase inhibitors (TKIs), which mostly target the intracellular part of EGFR and inhibit its activity in molecular signaling. While EGFR-specific mAbs and three generations of TKIs have demonstrated clinical efficacy in various settings, molecular evolution of tumors leads to apparent and sometimes inevitable resistance to current therapeutics, which highlights the need for deeper research in this field. Here, we tried to provide a comprehensive and systematic overview of the rationale, molecular mechanisms, and clinical significance of the current EGFR-targeting drugs, highlighting potential candidate molecules in development. We summarized the underlying mechanisms of resistance and available personalized predictive approaches that may lead to improved efficacy of EGFR-targeted therapies. We also discuss recent developments and the use of specific therapeutic strategies, such as multi-targeting agents and combination therapies, for overcoming cancer resistance to EGFR-specific drugs.
Collapse
Affiliation(s)
- Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Dmitri Kamashev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Aleksandra Emelianova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia;
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (D.K.); (A.B.)
- Laboratory for Translational Genomic Bioinformatics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
- Institute of Personalized Oncology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
7
|
Onken MD, Erdmann-Gilmore P, Zhang Q, Thapa K, King E, Kaltenbronn KM, Noda SE, Makepeace CM, Goldfarb D, Babur Ö, Townsend RR, Blumer KJ. Protein Kinase Signaling Networks Driven by Oncogenic Gq/11 in Uveal Melanoma Identified by Phosphoproteomic and Bioinformatic Analyses. Mol Cell Proteomics 2023; 22:100649. [PMID: 37730182 PMCID: PMC10616553 DOI: 10.1016/j.mcpro.2023.100649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/22/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023] Open
Abstract
Metastatic uveal melanoma (UM) patients typically survive only 2 to 3 years because effective therapy does not yet exist. Here, to facilitate the discovery of therapeutic targets in UM, we have identified protein kinase signaling mechanisms elicited by the drivers in 90% of UM tumors: mutant constitutively active G protein α-subunits encoded by GNAQ (Gq) or GNA11 (G11). We used the highly specific Gq/11 inhibitor FR900359 (FR) to elucidate signaling networks that drive proliferation, metabolic reprogramming, and dedifferentiation of UM cells. We determined the effects of FR on the proteome and phosphoproteome of UM cells as indicated by bioinformatic analyses with CausalPath and site-specific gene set enrichment analysis. We found that inhibition of oncogenic Gq/11 caused deactivation of PKC, Erk, and the cyclin-dependent kinases CDK1 and CDK2 that drive proliferation. Inhibition of oncogenic Gq/11 in UM cells with low metastatic risk relieved inhibitory phosphorylation of polycomb-repressive complex subunits that regulate melanocytic redifferentiation. Site-specific gene set enrichment analysis, unsupervised analysis, and functional studies indicated that mTORC1 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 drive metabolic reprogramming in UM cells. Together, these results identified protein kinase signaling networks driven by oncogenic Gq/11 that regulate critical aspects of UM cell biology and provide targets for therapeutic investigation.
Collapse
Affiliation(s)
- Michael D Onken
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, St Louis, Missouri, USA.
| | | | - Qiang Zhang
- Department of Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Kisan Thapa
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Emily King
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Kevin M Kaltenbronn
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Sarah E Noda
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Carol M Makepeace
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Dennis Goldfarb
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Özgün Babur
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - R Reid Townsend
- Department of Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Kendall J Blumer
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA.
| |
Collapse
|
8
|
Zhao W, Zheng XD, Tang PYZ, Li HM, Liu X, Zhong JJ, Tang YJ. Advances of antitumor drug discovery in traditional Chinese medicine and natural active products by using multi-active components combination. Med Res Rev 2023; 43:1778-1808. [PMID: 37183170 DOI: 10.1002/med.21963] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/03/2023] [Accepted: 04/12/2023] [Indexed: 05/16/2023]
Abstract
The antitumor efficacy of Chinese herbal medicines has been widely recognized. Leading compounds such as sterols, glycosides, flavonoids, alkaloids, terpenoids, phenylpropanoids, and polyketides constitute their complex active components. The antitumor monomers derived from Chinese medicine possess an attractive anticancer activity. However, their use was limited by low bioavailability, significant toxicity, and side effects, hindering their clinical applications. Recently, new chemical entities have been designed and synthesized by combining natural drugs with other small drug molecules or active moieties to improve the antitumor activity and selectivity, and reduce side effects. Such a novel conjugated drug that can interact with several vital biological targets in cells may have a more significant or synergistic anticancer activity than a single-molecule drug. In addition, antitumor conjugates could be obtained by combining pharmacophores containing two or more known drugs or leading compounds. Based on these studies, the new drug research and development could be greatly shortened. This study reviews the research progress of conjugates with antitumor activity based on Chinese herbal medicine. It is expected to serve as a valuable reference to antitumor drug research and clinical application of traditional Chinese medicine.
Collapse
Affiliation(s)
- Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xiao-Di Zheng
- Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | | | - Hong-Mei Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Xue Liu
- Jinan Intellectual Property Protection Center, Jinan, China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
9
|
Deng Z, Gao Y, Nguyen T, Chai J, Wu J, Li J, Abdel-Rahman MA, Xu X, Chen X. The Potent Antitumor Activity of Smp43 against Non-Small-Cell Lung Cancer A549 Cells via Inducing Membranolysis and Mitochondrial Dysfunction. Toxins (Basel) 2023; 15:toxins15050347. [PMID: 37235381 DOI: 10.3390/toxins15050347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/06/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Research has been conducted to investigate the potential application of scorpion venom-derived peptides in cancer therapy. Smp43, a cationic antimicrobial peptide from Scorpio maurus palmatus venom, has been found to exhibit suppressive activity against the proliferation of multiple cancer cell lines. However, its impact on non-small-cell lung cancer (NSCLC) cell lines has not been previously investigated. This study aimed to determine the cytotoxicity of Smp43 towards various NSCLC cell lines, particularly A549 cells with an IC50 value of 2.58 μM. The results indicated that Smp43 was internalized into A549 cells through membranolysis and endocytosis, which caused cytoskeleton disorganization, a loss of mitochondrial membrane potential, an accumulation of reactive oxygen species (ROS), and abnormal apoptosis, cell cycle distribution, and autophagy due to mitochondrial dysfunction. Additionally, the study explored the in vivo protective effect of Smp43 in xenograft mice. The findings suggest that Smp43 has potential anticarcinoma properties exerted via the inducement of cellular processes related to cell membrane disruption and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Ze Deng
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yahua Gao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tienthanh Nguyen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jinwei Chai
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiena Wu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiali Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | | | - Xueqing Xu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
10
|
Al-Huseini I, Sirasanagandla SR, Babu KS, Sofin RGS, Das S. Kinase Inhibitors Involved in the Regulation of Autophagy: Molecular Concepts and Clinical Implications. Curr Med Chem 2023; 30:1502-1528. [PMID: 35078392 DOI: 10.2174/0929867329666220117114306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 11/22/2022]
Abstract
All cells and intracellular components are remodeled and recycled in order to replace the old and damaged cells. Autophagy is a process by which damaged, and unwanted cells are degraded in the lysosomes. There are three different types of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. Autophagy has an effect on adaptive and innate immunity, suppression of any tumour, and the elimination of various microbial pathogens. The process of autophagy has both positive and negative effects, and this pertains to any specific disease or its stage of progression. Autophagy involves various processes which are controlled by various signaling pathways, such as Jun N-terminal kinase, GSK3, ERK1, Leucine-rich repeat kinase 2, and PTEN-induced putative kinase 1 and parkin RBR E3. Protein kinases are also important for the regulation of autophagy as they regulate the process of autophagy either by activation or inhibition. The present review discusses the kinase catalyzed phosphorylated reactions, the kinase inhibitors, types of protein kinase inhibitors and their binding properties to protein kinase domains, the structures of active and inactive kinases, and the hydrophobic spine structures in active and inactive protein kinase domains. The intervention of autophagy by targeting specific kinases may form the mainstay of treatment of many diseases and lead the road to future drug discovery.
Collapse
Affiliation(s)
- Isehaq Al-Huseini
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| | - Srinivasa Rao Sirasanagandla
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| | - Kondaveeti Suresh Babu
- Department of Biochemistry, Symbiosis Medical College for Women, Symbiosis International (Deemed) University, Pune, Maharashtra, India
| | | | - Srijit Das
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| |
Collapse
|
11
|
Lee JH, Lee SB, Kim H, Shin JM, Yoon M, An HS, Han JW. Anticancer Activity of Mannose-Specific Lectin, BPL2, from Marine Green Alga Bryopsis plumosa. Mar Drugs 2022; 20:md20120776. [PMID: 36547923 PMCID: PMC9788543 DOI: 10.3390/md20120776] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Lectin is a carbohydrate-binding protein that recognizes specific cells by binding to cell-surface polysaccharides. Tumor cells generally show various glycosylation patterns, making them distinguishable from non-cancerous cells. Consequently, lectin has been suggested as a good anticancer agent. Herein, the anticancer activity of Bryopsis plumosa lectins (BPL1, BPL2, and BPL3) was screened and tested against lung cancer cell lines (A549, H460, and H1299). BPL2 showed high anticancer activity compared to BPL1 and BPL3. Cell viability was dependent on BPL2 concentration and incubation time. The IC50 value for lung cancer cells was 50 μg/mL after 24 h of incubation in BPL2 containing medium; however, BPL2 (50 μg/mL) showed weak toxicity in non-cancerous cells (MRC5). BPL2 affected cancer cell growth while non-cancerous cells were less affected. Further, BPL2 (20 μg/mL) inhibited cancer cell invasion and migration (rates were ˂20%). BPL2 induced the downregulation of epithelial-to-mesenchymal transition-related genes (Zeb1, vimentin, and Twist). Co-treatment with BPL2 and gefitinib (10 μg/mL and 10 μM, respectively) showed a synergistic effect compared with monotherapy. BPL2 or gefitinib monotherapy resulted in approximately 90% and 70% cell viability, respectively, with concomitant treatment showing 40% cell viability. Overall, BPL2 can be considered a good candidate for development into an anticancer agent.
Collapse
|
12
|
Guo CH, Li WC, Peng CL, Chen PC, Lee SY, Hsia S. Targeting EGFR in Combination with Nutritional Supplements on Antitumor Efficacy in a Lung Cancer Mouse Model. Mar Drugs 2022; 20:md20120751. [PMID: 36547898 PMCID: PMC9783964 DOI: 10.3390/md20120751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Selenium (Se) and fish oil (FO) exert anti-epidermal growth factor receptor (EGFR) action on tumors. This study aimed to compare the anti-cancer efficacy of EGFR inhibitors (gefitinib and erlotinib) alone and in combination with nutritional supplements of Se/FO in treating lung cancer. Lewis LLC1 tumor-bearing mice were treated with a vehicle or Se/FO, gefitinib or gefitinib plus Se/FO, and erlotinib or erlotinib plus Se/FO. The tumors were assessed for mRNA and protein expressions of relevant signaling molecules. Untreated tumor-bearing mice had the lowest body weight and highest tumor weight and volume of all the mice. Mice receiving the combination treatment with Se/FO and gefitinib or erlotinib had a lower tumor volume and weight and fewer metastases than did those treated with gefitinib or erlotinib alone. The combination treatment exhibited greater alterations in receptor signaling molecules (lower EGFR/TGF-β/TβR/AXL/Wnt3a/Wnt5a/FZD7/β-catenin; higher GSK-3β) and immune checkpoint molecules (lower PD-1/PD-L1/CD80/CTLA-4/IL-6; higher NKp46/CD16/CD28/IL-2). These mouse tumors also had lower angiogenesis, cancer stemness, epithelial to mesenchymal transitions, metastases, and proliferation of Ki-67, as well as higher cell cycle arrest and apoptosis. These preliminary results showed the Se/FO treatment enhanced the therapeutic efficacies of gefitinib and erlotinib via modulating multiple signaling pathways in an LLC1-bearing mouse model.
Collapse
Affiliation(s)
- Chih-Hung Guo
- Micronutrition and Biomedical Nutrition Laboratories, Institute of Biomedical Nutrition, Hung-Kuang University, Taichung 433, Taiwan
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
| | - Wen-Chin Li
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
| | - Chia-Lin Peng
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
| | | | - Shih-Yu Lee
- Biotechnology, Health, and Innovation Research Center, Hung-Kuang University, Taichung 433, Taiwan
| | - Simon Hsia
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
- Correspondence: ; Tel.: +886-2-2546-8824; Fax: +886-2-2545-9225
| |
Collapse
|
13
|
Wu H, Qian D, Bai X, Sun S. Targeted Pyroptosis Is a Potential Therapeutic Strategy for Cancer. JOURNAL OF ONCOLOGY 2022; 2022:2515525. [PMID: 36467499 PMCID: PMC9715319 DOI: 10.1155/2022/2515525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/21/2022] [Accepted: 11/15/2022] [Indexed: 12/01/2023]
Abstract
As a type of regulated cell death (RCD) mode, pyroptosis plays an important role in several kinds of cancers. Pyroptosis is induced by different stimuli, whose pathways are divided into the canonical pathway and the noncanonical pathway depending on the formation of the inflammasomes. The canonical pathway is triggered by the assembly of inflammasomes, and the activation of caspase-1 and then the cleavage of effector protein gasdermin D (GSDMD) are promoted. While in the noncanonical pathway, the caspase-4/5/11 (caspase 4/5 in humans and caspase 11 in mice) directly cleave GSDMD without the assembly of inflammasomes. Pyroptosis is involved in various cancers, such as lung cancer, gastric cancer, hepatic carcinoma, breast cancer, and colorectal carcinoma. Pyroptosis in gastric cancer, hepatic carcinoma, breast cancer, and colorectal carcinoma is related to the canonical pathway, while both the canonical and noncanonical pathway participate in lung cancer. Moreover, simvastatin, metformin, and curcumin have effect on these cancers and simultaneously promote the pyroptosis of cancer cells. Accordingly, pyroptosis may be an important therapeutic target for cancer.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
- Clinical Medicine, Three Class, 2020 Grade, Kunming Medical University, Kunming, China
| | - Dianlun Qian
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiangfeng Bai
- Department of Cardiothoracic Surgery, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
14
|
Zahran SS, Ragab FA, El-Gazzar MG, Soliman AM, Mahmoud WR, Ghorab MM. Antiproliferative, antiangiogenic and apoptotic effect of new hybrids of quinazoline-4(3H)-ones and sulfachloropyridazine. Eur J Med Chem 2022; 245:114912. [DOI: 10.1016/j.ejmech.2022.114912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
|
15
|
Angiotensin II type 1 receptor blockade attenuates gefitinib-induced cardiac hypertrophy via adjusting angiotensin II-mediated oxidative stress and JNK/P38 MAPK pathway in a rat model. Saudi Pharm J 2022; 30:1159-1169. [PMID: 36164571 PMCID: PMC9508643 DOI: 10.1016/j.jsps.2022.06.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/17/2022] [Indexed: 11/10/2022] Open
Abstract
Gefitinib is a tyrosine kinase inhibitor (TKI) of the epidermal growth factor receptor (EGFR), used for the treatment of advanced or metastatic non-small cell lung cancer. Recently, studies proved that Gefitinib-induced cardiotoxicity through induction of oxidative stress leads to cardiac hypertrophy. The current study was conducted to understand the mechanisms underlying gefitinib-induced cardiac hypertrophy through studying the roles of angiotensin II (AngII), oxidative stress, and mitogen-activated protein kinase (MAPK) pathway. Male Wistar albino rats were treated with valsartan, gefitinib, or both for four weeks. Blood samples were collected for AngII and cardiac markers measurement, and hearts were harvested for histological study and biochemical analysis. Gefitinib caused histological changes in the cardiac tissues and increased levels of cardiac hypertrophy markers, AngII and its receptors. Blocking of AngII type 1 receptor (AT1R) via valsartan protected hearts and normalized cardiac markers, AngII levels, and the expression of its receptors during gefitinib treatment. valsartan attenuated gefitinib-induced NADPH oxidase and oxidative stress leading to down-regulation of JNK/p38-MAPK pathway. Collectively, AT1R blockade adjusted AngII-induced NADPH oxidase and JNK/p38-MAPK leading to attenuation of gefitinib-induced cardiac hypertrophy. This study found a pivotal role of AngII/AT1R signaling in gefitinib-induced cardiac hypertrophy, which may provide novel approaches in the management of EGFRIs-induced cardiotoxicity.
Collapse
|
16
|
Eshraghi M, Ahmadi M, Afshar S, Lorzadeh S, Adlimoghaddam A, Rezvani Jalal N, West R, Dastghaib S, Igder S, Torshizi SRN, Mahmoodzadeh A, Mokarram P, Madrakian T, Albensi BC, Łos MJ, Ghavami S, Pecic S. Enhancing autophagy in Alzheimer's disease through drug repositioning. Pharmacol Ther 2022; 237:108171. [PMID: 35304223 DOI: 10.1016/j.pharmthera.2022.108171] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is one of the biggest human health threats due to increases in aging of the global population. Unfortunately, drugs for treating AD have been largely ineffective. Interestingly, downregulation of macroautophagy (autophagy) plays an essential role in AD pathogenesis. Therefore, targeting autophagy has drawn considerable attention as a therapeutic approach for the treatment of AD. However, developing new therapeutics is time-consuming and requires huge investments. One of the strategies currently under consideration for many diseases is "drug repositioning" or "drug repurposing". In this comprehensive review, we have provided an overview of the impact of autophagy on AD pathophysiology, reviewed the therapeutics that upregulate autophagy and are currently used in the treatment of other diseases, including cancers, and evaluated their repurposing as a possible treatment option for AD. In addition, we discussed the potential of applying nano-drug delivery to neurodegenerative diseases, such as AD, to overcome the challenge of crossing the blood brain barrier and specifically target molecules/pathways of interest with minimal side effects.
Collapse
Affiliation(s)
- Mehdi Eshraghi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Aida Adlimoghaddam
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada
| | | | - Ryan West
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America
| | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benedict C Albensi
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; Nova Southeastern Univ. College of Pharmacy, Davie, FL, United States of America; University of Manitoba, College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America.
| |
Collapse
|
17
|
Moiseyenko FV, Kuligina ES, Zhabina AS, Belukhin SA, Laidus TA, Martianov AS, Zagorodnev KA, Sokolova TN, Chuinyshena SA, Kholmatov MM, Artemieva EV, Stepanova EO, Shuginova TN, Volkov NM, Yanus GA, Imyanitov EN. Changes in the concentration of EGFR-mutated plasma DNA in the first hours of targeted therapy allow the prediction of tumor response in patients with EGFR-driven lung cancer. Int J Clin Oncol 2022; 27:850-862. [PMID: 35171360 PMCID: PMC8853017 DOI: 10.1007/s10147-022-02128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/23/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE This study aimed to analyze changes in the plasma concentration of EGFR-mutated circulating tumor DNA (ctDNA) occurring immediately after the start of therapy with EGFR tyrosine kinase inhibitors (TKIs). METHODS Serial plasma samples were collected from 30 patients with EGFR-driven non-small cell lung cancer before intake of the first tablet and at 0.5, 1, 2, 3, 6, 12, 24, 36 and 48 h after the start of the therapy. The content of EGFR alleles (exon 19 deletions or L858R) in ctDNA was measured by ddPCR. RESULTS ctDNA was detected at base-line in 25/30 (83%) subjects. Twelve (50%) out of 24 informative patients showed > 25% reduction of the ctDNA content at 48 h time point; all these patients demonstrated disease control after 4 and 8-12 weeks of therapy. The remaining 12 individuals showed either stable content of EGFR-mutated ctDNA (n = 5) or the elevation of ctDNA concentration (n = 7). 10 of 12 patients with elevated or stable ctDNA level achieved an objective response at 4 weeks, but only 5 of 10 evaluable patients still demonstrated disease control at 8-12 weeks (p = 0.032, when compared to the group with ctDNA decrease). The decline of the amount of circulating EGFR mutant copies at 48 h also correlated with longer progression-free survival (14.7 months vs. 8.5 months, p = 0.013). CONCLUSION Comparison of concentration of EGFR-mutated ctDNA at base-line and at 48 h after the start of therapy is predictive for the duration of TKI efficacy.
Collapse
Affiliation(s)
- Fedor V. Moiseyenko
- City Cancer Center, 68A Leningradskaya street, Pesochny, Saint Petersburg, 197758 Russia
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
| | - Ekaterina S. Kuligina
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
- St.-Petersburg Pediatric Medical University, 2 Litovskaya street, Saint Petersburg, 194100 Russia
| | - Albina S. Zhabina
- City Cancer Center, 68A Leningradskaya street, Pesochny, Saint Petersburg, 197758 Russia
| | - Sergey A. Belukhin
- City Cancer Center, 68A Leningradskaya street, Pesochny, Saint Petersburg, 197758 Russia
| | - Tatiana A. Laidus
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
- St.-Petersburg Pediatric Medical University, 2 Litovskaya street, Saint Petersburg, 194100 Russia
| | - Aleksandr S. Martianov
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
- St.-Petersburg Pediatric Medical University, 2 Litovskaya street, Saint Petersburg, 194100 Russia
| | - Kirill A. Zagorodnev
- St.-Petersburg Pediatric Medical University, 2 Litovskaya street, Saint Petersburg, 194100 Russia
| | - Tatyana N. Sokolova
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
| | - Svetlana A. Chuinyshena
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
- St.-Petersburg Pediatric Medical University, 2 Litovskaya street, Saint Petersburg, 194100 Russia
| | - Maxim M. Kholmatov
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
| | - Elizaveta V. Artemieva
- City Cancer Center, 68A Leningradskaya street, Pesochny, Saint Petersburg, 197758 Russia
| | - Ekaterina O. Stepanova
- City Cancer Center, 68A Leningradskaya street, Pesochny, Saint Petersburg, 197758 Russia
| | - Tatiana N. Shuginova
- City Cancer Center, 68A Leningradskaya street, Pesochny, Saint Petersburg, 197758 Russia
| | - Nikita M. Volkov
- City Cancer Center, 68A Leningradskaya street, Pesochny, Saint Petersburg, 197758 Russia
| | - Grigoriy A. Yanus
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
- St.-Petersburg Pediatric Medical University, 2 Litovskaya street, Saint Petersburg, 194100 Russia
| | - Evgeny N. Imyanitov
- Laboratory of Molecular Oncology, Department of Tumor Biology, N.N. Petrov Institute of Oncology, 68 Leningradskaya street, Pesochny-2, St.-Petersburg, 197758 Russia
- St.-Petersburg Pediatric Medical University, 2 Litovskaya street, Saint Petersburg, 194100 Russia
- I.I. Mechnikov North-Western Medical University, 41 Kirochnaya street, Saint Petersburg, 191015 Russia
| |
Collapse
|
18
|
Sipos F, Bohusné Barta B, Simon Á, Nagy L, Dankó T, Raffay RE, Petővári G, Zsiros V, Wichmann B, Sebestyén A, Műzes G. Survival of HT29 Cancer Cells Is Affected by IGF1R Inhibition via Modulation of Self-DNA-Triggered TLR9 Signaling and the Autophagy Response. Pathol Oncol Res 2022; 28:1610322. [PMID: 35651701 PMCID: PMC9148969 DOI: 10.3389/pore.2022.1610322] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023]
Abstract
Purpose: In HT29 colon cancer cells, a close interplay between self-DNA-induced TLR9 signaling and autophagy response was found, with remarkable effects on cell survival and differentiation. IGF1R activation drives the development and malignant progression of colorectal cancer. IGF1R inhibition displays a controversial effect on autophagy. The interrelated roles of IGF1R inhibition and TLR9/autophagy signaling in HT29 cancer cells have not yet been clarified. In our study, we aimed to investigate the complex interplay of IGF1R inhibition and TLR9/autophagy signaling in HT29 cells. Methods: HT29 cells were incubated with tumor-originated self-DNA with or without inhibitors of IGF1R (picropodophyllin), autophagy (chloroquine), and TLR9 (ODN2088), respectively. Cell proliferation and metabolic activity measurements, direct cell counting, NanoString and Taqman gene expression analyses, immunocytochemistry, WES Simple Western blot, and transmission electron microscopy investigations were performed. Results: The concomitant use of tumor-derived self-DNA and IGF1R inhibitors displays anti-proliferative potential, which can be reversed by parallel TLR9 signaling inhibition. The distinct effects of picropodophyllin, ODN2088, and chloroquine per se or in combination on HT29 cell proliferation and autophagy suggest that either the IGF1R-associated or non-associated autophagy machinery is "Janus-faced" regarding its actions on cell proliferation. Autophagy, induced by different combinations of self-DNA and inhibitors is not sufficient to rescue HT29 cells from death but results in the survival of some CD133-positive stem-like HT29 cells. Conclusion: The creation of new types of combined IGF1R, autophagy, and/or TLR9 signaling inhibitors would play a significant role in the development of more personalized anti-tumor therapies for colorectal cancer.
Collapse
Affiliation(s)
- Ferenc Sipos
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
- *Correspondence: Ferenc Sipos,
| | - Bettina Bohusné Barta
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| | - Ágnes Simon
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| | - Lőrinc Nagy
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| | - Titanilla Dankó
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Regina Eszter Raffay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Petővári
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Viktória Zsiros
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | | | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Györgyi Műzes
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
19
|
Manogaran P, Somasundaram B, Viswanadha VP. Reversal of cisplatin resistance by neferine/isoliensinine and their combinatorial regimens with cisplatin-induced apoptosis in cisplatin-resistant colon cancer stem cells (CSCs). J Biochem Mol Toxicol 2021; 36:e22967. [PMID: 34921482 DOI: 10.1002/jbt.22967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 08/24/2021] [Accepted: 10/18/2021] [Indexed: 12/28/2022]
Abstract
Cisplatin chemotherapy to the colorectal cancer cells (CRCs) is accompanied by dose-limiting adverse effects along with the acquisition of drug resistance implicating low therapeutic outcomes. The present study is aimed to evaluate the chemosensitizing efficacy of neferine/isoliensinine or combinatorial regimen of neferine/isoliensinine with cisplatin against CSCs (cisplatin resistant colon stem cells). CSCs were developed using pulse exposure of cisplatin to parental HCT-15 cells. Neferine/isoliensinine or combinatorial regimens of Neferine/isoliensinine and cisplatin exhibited a stronger cytotoxic activity against CSCs compared to control. IC50 doses were found to be 6.5 μM for neferine, 12.5 μM for isoliensinine, and 120 μM for cisplatin respectively. Furthermore, the combinatorial regimen of a low dose of cisplatin (40 μM) with 4 μM neferine/8 μM isoliensinine induced cell death in a synergistic manner as described by isobologram. Neferine/isoliensinine could confer extensive intracellular reactive oxygen species generation in CSCs. Neferine/isoliensinine or combinatorial regimens dissipated mitochondrial membrane potential and enhanced intracellular [Ca2+ ]i, which were measured by spectroflurimetry. Furthermore, these combinatorial regimens induced a significant increase in the sub G0 phase of cell cycle arrest and PI uptake and alleviated the expression of ERCC1 in CSCs. Combinatorial regimens or neferine/isoliensinine treatments downregulated the cell survival protein expression (PI3K/pAkt/mTOR) and activated mitochondria-mediated apoptosis by upregulating Bax, cytochrome c, caspase-3, and PARP cleavage expression while downregulating the BCl-2 expression in CSCs. Our study confirms the chemosensitizing efficacy of neferine/isoliensinine or combinatorial regimens of neferine/isoliensinine with a low dose of cisplatin against CSCs.
Collapse
Affiliation(s)
- Prasath Manogaran
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Bharath Somasundaram
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Vijaya Padma Viswanadha
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
20
|
Alqahtani AS, Ghorab MM, Nasr FA, Ahmed MZ, Al-Mishari AA, Attia SM. Novel sulphonamide-bearing methoxyquinazolinone derivatives as anticancer and apoptosis inducers: synthesis, biological evaluation and in silico studies. J Enzyme Inhib Med Chem 2021; 37:86-99. [PMID: 34894963 PMCID: PMC8667930 DOI: 10.1080/14756366.2021.1983807] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
We synthesised a new series of sulphonamide-bearing quinazolinone derivatives 5-18 and evaluated their in vitro cytotoxicity in various cancer cell lines (A549, HepG-2, LoVo and MCF-7) and in normal human cells (HUVEC). Compounds 6 and 10 exhibited the higher activity against all the cancer cell lines compared with 5-flourourcil as positive control. The ability of the most promising compounds 6 and 10 to induce cell cycle arrest and apoptosis in breast cancer (MCF-7) cells was evaluated by flow cytometry. Reverse transcriptase-polymerase chain reaction and western blotting were used to evaluate the expression of apoptosis-related markers. We found that the 2-tolylthioacetamide derivative 6 and the 3-ethyl phenyl thioacetamide derivative 10 exhibited cytotoxic activity comparable to that of 5-fluorouracil as reference drug in MCF-7 and LoVo colon cancer cells. Cell cycle analysis showed a concentration-dependent accumulation of cells in the sub-G1 phase upon treatment with both compounds. The Annexin V-fluorescein isothiocyanate/propidium iodide assay showed that the compounds 6 and 10 increased the early and late apoptosis cell death modes in a dose-dependent manner. These compounds downregulated the expression of B-cell lymphoma-2 (Bcl-2), while increasing that of p53, Bcl-2-like protein 4, and caspase-7, at the mRNA and protein levels. Molecular docking of compounds 6 and 10 with Bcl-2 predicted them to show moderate − high binding affinity (6: −7.5 kcal/mol, 10: −7.9 kcal/mol) and interactions with key central substrate cavity residues. Overall, compounds 6 and 10 were found to be promising anticancer and apoptosis-inducing agents.
Collapse
Affiliation(s)
- Ali S Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mostafa M Ghorab
- Department of Drug Radiation Research, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Fahd A Nasr
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Z Ahmed
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.,Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah A Al-Mishari
- Medicinal, Aromatic and Poisonous Plants Research Center, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
21
|
Liu Y, Du Y. Influence of Autophagy Inhibition on Lung Adenocarcinoma Cell Migration and Invasion Ability, and Efficacy of Gefitinib. Technol Cancer Res Treat 2021; 20:15330338211049000. [PMID: 34657484 PMCID: PMC8521425 DOI: 10.1177/15330338211049000] [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] [Indexed: 01/10/2023] Open
Abstract
An increasing number of studies have emphasized the role of autophagy in cancer cell metastasis and treatment of malignant tumors. Autophagy inhibitors have been widely used in combination therapies to treat advanced malignancies. Several lung adenocarcinoma cells harbor epidermal growth factor receptor (EGFR) gene mutations, and EGFR tyrosine kinase inhibitors (TKIs) are routinely used in the treatment of lung adenocarcinoma. However, a number of lung adenocarcinoma tumors do not respond or develop resistance to EGFR TKIs. The aim of the present study was to explore the effect of autophagy inhibition on the biological behavior of lung adenocarcinoma cells. In addition, whether autophagy inhibition increases the efficacy of gefitinib in lung adenocarcinoma was investigated. The activation of autophagy was inhibited via the reduction of the expression of ATG5 in A549, H1975 and HCC827 cells. ATG5 knockdown using ATG5 siRNA partially suppressed the LC3B-II expression, decreased the LC3B-I/II conversion rate and enhanced the P62 expression. Cell scratch test and Transwell assay showed that the inhibition of autophagy could impair the migration and invasion ability of cells. These studies suggested that autophagy may play a pro-survival role in lung adenocarcinoma.
Collapse
Affiliation(s)
- Ying Liu
- 117878Department of Cytology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yun Du
- 117878Department of Cytology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
22
|
Kumar D, Kommalapati VK, Jerald MK, Tangutur AD. Fluorinated thiazolidinol drives autophagic cell death in pancreatic cancer cells via AMPK activation and perturbation of critical sentinels of oncogenic signaling. Chem Biol Interact 2021; 343:109433. [PMID: 33689707 DOI: 10.1016/j.cbi.2021.109433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/23/2021] [Accepted: 03/03/2021] [Indexed: 12/25/2022]
Abstract
Pancreatic cancer is one of the most malignant cancers around the world. The co-occurrence of mutation in KRAS and p53 makes it highly aggressive, proliferative, metastatic, and resistant to apoptotic cell death. Therefore, there is a need to trigger an alternate mechanism of cancer cell death in apoptosis-resistant pancreatic cancer. Autophagic cell death could be an alternate viable option for treatment in such cases. Thus, the identification of small molecules as autophagy modulators with potent anticancer efficacy would be of great importance in pancreatic cancer. The present study investigates fluorinated thiazolidionol (FTZ) driven autophagy modulation, underlying mechanism, and regulation of critical sentinels of oncogenic signaling in pancreatic cancer cells. We identified that FTZ triggered autophagic cell death in pancreatic cancer cells, independent of apoptosis evidenced by an increase in cytoplasmic vacuoles formation, autophagy flux, LC3-II expression, and p62 degradation. Further, the crucial events of apoptosis i.e., Caspase-3 activation and PARP cleavage, were not observed, indicating the non-occurrence of apoptotic cell death. Moreover, FTZ was able to activate AMPK and suppress PI3k/Akt/mTOR as well as MEK/ERK, the key oncogenic signaling pathways in cancer cells. Furthermore, treatment with FTZ suppressed migration, invasion, and angiogenesis in pancreatic cancer cells. Studies in vivo revealed significant regression of tumors by FTZ in nude mice model. Overall, our study demonstrates that FTZ induces autophagic cell death in pancreatic cancer cells independent of apoptosis, which is accompanied by AMPK activation and suppression of critical sentinels of oncogenic signaling in pancreatic cancer cells.
Collapse
Affiliation(s)
- Dinesh Kumar
- Department of Applied Biology, CSIR- Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201 002, India
| | - Vamsi Krishna Kommalapati
- Department of Applied Biology, CSIR- Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201 002, India
| | - Mahesh Kumar Jerald
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201 002, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR- Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Sector 19, Kamla Nehru Nagar, Ghaziabad, Uttar Pradesh, 201 002, India.
| |
Collapse
|
23
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
24
|
Li S, Yang H, Zhao M, Gong L, Wang Y, Lv Z, Quan Y, Wang Z. Demethylation of HACE1 gene promoter by propofol promotes autophagy of human A549 cells. Oncol Lett 2020; 20:280. [PMID: 33014158 PMCID: PMC7520799 DOI: 10.3892/ol.2020.12143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/28/2020] [Indexed: 12/15/2022] Open
Abstract
Propofol (2,6-diisopropylphenol) is one of the most commonly used intravenous anesthetics and possesses a number of non-anesthetic effects, including antitumor function. The aim of the present study was to elucidate the antitumor molecular mechanism of propofol on A549 and H1299 cells. A549 and H1299 cells were treated in the presence or absence of different concentrations (0, 60 or 120 µmol) of propofol for different durations (0, 24, 48 or 72 h), and proliferation was detected by MTT and colony formation assays; the protein levels of optineurin (OPTN) ubiquitination, HECT domain and ankyrin repeat containing E3 ubiquitin protein ligase 1 (HACE1), methyl-CpG binding domain protein 3 (MBD3) and Microtubule-associated protein 1A/1B-light chain 3 were detected by immunoblotting or quantitative (q)PCR; the methylation state of the HACE1 gene promoter was detected by bisulfite DNA sequencing; and binding of MBD3 on HACE1 gene promoter was detected by chromatin immunoprecipitation-qPCR. Propofol inhibited proliferation of A549 and H1299 cells and promoted HACE1-OPTN axis-mediated selective autophagy activity by increasing the protein expression levels of HACE1 via demethylating its promoter region. Furthermore, propofol promoted expression levels of MBD3 and binding to the -1,000 to -1 bp (transcription start site) region of HACE1 gene promoter. MBD3-knockdown experiments indicated that propofol inhibited proliferation of A549 cells in a MBD3-dependent manner. Thus, the findings of the present study provided a potential antitumor molecular mechanism mediated by propofol.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| | - Hui Yang
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| | - Min Zhao
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| | - Linli Gong
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| | - Yahong Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| | - Zhiyong Lv
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| | - Yuhang Quan
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| | - Zhonghui Wang
- Department of Anesthesiology, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan 650118, P.R. China
| |
Collapse
|
25
|
Tong CWS, Wu MMX, Yan VW, Cho WCS, To KKW. Repurposing loperamide to overcome gefitinib resistance by triggering apoptosis independent of autophagy induction in KRAS mutant NSCLC cells. Cancer Treat Res Commun 2020; 25:100229. [PMID: 33152554 DOI: 10.1016/j.ctarc.2020.100229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/17/2020] [Accepted: 10/23/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Gefitinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) approved for first-line treatment of non-small cell lung cancer (NSCLC) with sensitizing EGFR mutations. However, NSCLC patients bearing mutant KRAS are inherently unresponsive to gefitinib. Defective autophagy was proposed to mediate resistance to EGFR-TKIs. In this study, the reversal of primary resistance to gefitinib in NSCLC by putative autophagy inducers was investigated. MATERIALS AND METHODS A few putative autophagy inducers were investigated in NSCLC cells harboring KRAS or EGFR mutations. Quantitative real-time PCR and Western blot analysis were used to evaluate expression of autophagy-related genes and proteins. Sulforhodamine B assay was used to evaluate cytotoxicity of drug combinations. Flow cytometric asssays were used to study apoptotic and cell cycle effects. RESULTS The antidiarrheal agent loperamide was identified as an autophagy inducer. Loperamide promoted the formation of autophagosomes and it potentiated the cytotoxic effect of gefitinib specifically in NSCLC cells bearing mutant KRAS and wild-type EGFR. Gefitinib-loperamide combination enhanced apoptosis and G1 cell cycle arrest, both of which could not be reversed by pharmacological autophagy inhibitor (3-methyladenine). Moreover, synergistic anticancer effect of gefitinib-loperamide combination was observed in both autophagy-proficient (Atg5-wild type) and -deficient (Atg5-knockout) mouse embryonic fibroblasts. Loperamide overcome gefitinib resistance in NSCLC harboring mutant KRAS and wild-type EGFR through increased apoptosis but independent of autophagy induction. CONCLUSION Loperamide could be repurposed to overcome primary resistance to gefitinib in KRAS-mutation bearing NSCLC as it also helps relieve the common side effect of diarrhea caused by EGFR-TKIs.
Collapse
Affiliation(s)
- Christy W S Tong
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mia M X Wu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vivi W Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
26
|
Zhu ML, Zhang PM, Jiang M, Yu SW, Wang L. Myricetin induces apoptosis and autophagy by inhibiting PI3K/Akt/mTOR signalling in human colon cancer cells. BMC Complement Med Ther 2020; 20:209. [PMID: 32631392 PMCID: PMC7336643 DOI: 10.1186/s12906-020-02965-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [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: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The compound 3,3',4',5,5',7-hexahydroxyflavone (myricetin) is a natural flavonoid with antitumour activity. Most of the studies on myricetin have focused on the induction of tumour cell apoptosis, and little is known about the regulatory effects of myricetin on autophagy in colorectal cancer. METHODS Here, we studied the effects of myricetin on colon cancer cell proliferation, apoptosis and autophagy. We detected colon cancer cell apoptosis induced by myricetin via flow cytometry and Hoechst 33258 staining. Transmission electron microscopy was performed to observe the morphological changes associated with autophagy. The expression levels of apoptosis-, autophagy- and PI3K/Akt/mTOR signalling-related proteins were measured by Western blot analysis. RESULTS This study confirmed that myricetin inhibits the proliferation of 4 kinds of colon cancer cell lines. Myricetin induced cell apoptosis and autophagy by inhibiting PI3K/Akt/mTOR signalling pathway. In addition, the inhibition of autophagy with 3-methyladenine (3-MA) promoted the apoptosis of myricetin-treated colon cancer cells. CONCLUSIONS Considering that myricetin induces apoptosis and autophagy in colon cancer cells, myricetin may become a viable candidate for chemotherapy; it could be used to exert tumour inhibitory effects alone or as adjuvant chemotherapy to inhibit autophagy. These studies may provide further evidence for the potential use of myricetin in the treatment of colon cancer.
Collapse
Affiliation(s)
- Ming-Liang Zhu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Pei-Min Zhang
- Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, China
| | - Min Jiang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shu-Wen Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, China.
| | - Lu Wang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250013, China.
| |
Collapse
|
27
|
Ajoolabady A, Aghanejad A, Bi Y, Zhang Y, Aslkhodapasandhukmabad H, Abhari A, Ren J. Enzyme-based autophagy in anti-neoplastic management: From molecular mechanisms to clinical therapeutics. Biochim Biophys Acta Rev Cancer 2020; 1874:188366. [PMID: 32339608 DOI: 10.1016/j.bbcan.2020.188366] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 02/08/2023]
Abstract
Autophagy is an evolutionarily conserved self-cannibalization process commonly found in all eukaryotic cells. Through autophagy, long-lived or damaged organelles, superfluous proteins, and pathogens are sequestered and encapsulated into the double-membrane autophagosomes prior to fusion with lysosomes for ultimate degradation and recycling. Given that autophagy is deemed both protective and detrimental in malignancies, the clinical therapeutic utilization of autophagy modulators in cancer has attracted immense attentions over the past decades. Dependence of tumor cells on autophagy during amino acid insufficiency or deprivation has prompted us to explore the underlying autophagy regulatory mechanisms to inject amino acid degrading enzymes and enzyme-based strategies into therapeutic maneuvers of autophagy in cancer.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yaguang Bi
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yingmei Zhang
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | | | - Alireza Abhari
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA; Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
28
|
Sueangoen N, Tantiwetrueangdet A, Panvichian R. HCC-derived EGFR mutants are functioning, EGF-dependent, and erlotinib-resistant. Cell Biosci 2020; 10:41. [PMID: 32190291 PMCID: PMC7076995 DOI: 10.1186/s13578-020-00407-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Background Epidermal growth factor receptor (EGFR) has emerged as an important therapeutic target. Overexpression of EGFR is frequently observed in hepatocellular carcinoma (HCC) and EGFR activation has been proven to be a potential determinant of primary resistance of HCC cells to sorafenib. In our previous study, we found 13 missense mutations in EGFR exon 19-23 from hepatocellular carcinoma (HCC) tissues, but the functions of these mutations have not been determined. This study aims to determine the kinase activity and sensitivity to erlotinib, a 1st-generation EGFR-tyrosine kinase inhibitor (TKI), of seven HCC-derived mutants (K757E, N808S, R831C, V897A, P937L, T940A, and M947T). Results Using transduction of pBabe-puro retroviral vector with or without EGFR, we constructed and determined the function of EGFRs in NIH-3T3 cells stably harboring each of the seven mutants, as well as the erlotinib-sensitive L858R-mutant, the erlotinib-resistant T790M-mutant, and EGFR wild type (WT). Our results indicate that the seven mutants are functioning, EGF-dependent, EGFRs. Cells harboring six of the seven mutants could generate some level of EGFR phosphorylation in the absence of EGF, indicating some constitutive kinase activity, but all of the seven mutants remain primarily EGF-dependent. Our results demonstrate that erlotinib induces differential degree of apoptosis and autophagy among cells harboring different EGFRs: complete apoptosis and autophagy (cleavage of both caspase-3 and PARP, and marked LC3-II increment) in L858R-mutant; partial apoptosis and autophagy (only cleavage of caspase-3, and moderate LC3-II increment) in WT and HCC-derived mutants; and no apoptosis and minimal autophagy (no cleavage of caspase-3 and PARP, and minimal LC3-II increment) in T790M-mutant. The seven HCC-derived mutants are erlotinib-resistant, as treatment with erlotinib up to high concentration could only induce partial inhibition of EGFR phosphorylation, partial or no inhibition of AKT and ERK phosphorylation, and partial apoptosis and autophagy. Conclusion The seven HCC-derived EGFR mutants in this study are functioning, EGF-dependent, and erlotinib-resistant. Erlotinib induces differential degree of apoptosis and autophagy among cells harboring different EGFRs. The degree of inhibition of EGFR phosphorylation by erlotinib is the determining factor for the degree of apoptosis and autophagy amongst cells harboring EGFR mutants. This study paves the way for further investigation into the sensitivity of these HCC-derived mutants to the 3rd-generation irreversible EGFR-TKI, osimertinib.
Collapse
Affiliation(s)
- Natthaporn Sueangoen
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Ravat Panvichian
- Department of Internal Medicine, Division of Medical Oncology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Rama 6 Road, Ratchathewi, Bangkok, 10400 Thailand
| |
Collapse
|
29
|
Wu G, Yan Y, Zhou Y, Duan Y, Zeng S, Wang X, Lin W, Ou C, Zhou J, Xu Z. Sulforaphane: Expected to Become a Novel Antitumor Compound. Oncol Res 2020; 28:439-446. [PMID: 32111265 PMCID: PMC7851526 DOI: 10.3727/096504020x15828892654385] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Natural products are becoming increasingly popular in a variety of traditional, complementary, and alternative systems due to their potency and slight side effects. Natural compounds have been shown to be effective against many human diseases, especially cancers. Sulforaphane (SFE) is a traditional Chinese herbal medicine. In recent years, an increasing number of studies have been conducted to evaluate the antitumor effect of SFE. The roles of SFE in cancers are mainly through the regulation of potential biomarkers to activate or inhibit related signaling pathways. SFE has exhibited promising inhibitory effects on breast cancer, lung cancer, liver cancer, and other malignant tumors. In this review, we summarized the reports on the activity and functional mechanisms of SFE in cancer treatment and explored the efficacy and toxicity of SFE.
Collapse
Affiliation(s)
- Geting Wu
- Department of Pathology, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Yangying Zhou
- Department of Oncology, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Yumei Duan
- Department of Pathology, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Xiang Wang
- Department of Pharmacy, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Wei Lin
- Department of Pathology, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South UniversityChangshaP.R. China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South UniversityChangshaP.R. China
| |
Collapse
|
30
|
Polyphyllin VI Induces Caspase-1-Mediated Pyroptosis via the Induction of ROS/NF-κB/NLRP3/GSDMD Signal Axis in Non-Small Cell Lung Cancer. Cancers (Basel) 2020; 12:cancers12010193. [PMID: 31941010 PMCID: PMC7017302 DOI: 10.3390/cancers12010193] [Citation(s) in RCA: 211] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/08/2020] [Accepted: 01/10/2020] [Indexed: 12/24/2022] Open
Abstract
Trillium tschonoskii Maxim (TTM), a traditional Chinese medicine, has been demonstrated to have a potent anti-tumor effect. Recently, polyphyllin VI (PPVI), a main saponin isolated from TTM, was reported by us to significantly suppress the proliferation of non-small cell lung cancer (NSCLC) via the induction of apoptosis and autophagy in vitro and in vivo. In this study, we further found that the NLRP3 inflammasome was activated in PPVI administrated A549-bearing athymic nude mice. As is known to us, pyroptosis is an inflammatory form of caspase-1-dependent programmed cell death that plays an important role in cancer. By using A549 and H1299 cells, the in vitro effect and action mechanism by which PPVI induces activation of the NLRP3 inflammasome in NSCLC were investigated. The anti-proliferative effect of PPVI in A549 and H1299 cells was firstly measured and validated by MTT assay. The activation of the NLRP3 inflammasome was detected by using Hoechst33324/PI staining, flow cytometry analysis and real-time live cell imaging methods. We found that PPVI significantly increased the percentage of cells with PI signal in A549 and H1299, and the dynamic change in cell morphology and the process of cell death of A549 cells indicated that PPVI induced an apoptosis-to-pyroptosis switch, and, ultimately, lytic cell death. In addition, belnacasan (VX-765), an inhibitor of caspase-1, could remarkably decrease the pyroptotic cell death of PPVI-treated A549 and H1299 cells. Moreover, by detecting the expression of NLRP3, ASC, caspase-1, IL-1β, IL-18 and GSDMD in A549 and h1299 cells using Western blotting, immunofluorescence imaging and flow cytometric analysis, measuring the caspase-1 activity using colorimetric assay, and quantifying the cytokines level of IL-1β and IL-18 using ELISA, the NLRP3 inflammasome was found to be activated in a dose manner, while VX-765 and necrosulfonamide (NSA), an inhibitor of GSDMD, could inhibit PPVI-induced activation of the NLRP3 inflammasome. Furthermore, the mechanism study found that PPVI could activate the NF-κB signaling pathway via increasing reactive oxygen species (ROS) levels in A549 and H1299 cells, and N-acetyl-L-cysteine (NAC), a scavenger of ROS, remarkably inhibited the cell death, and the activation of NF-κB and the NLRP3 inflammasome in PPVI-treated A549 and H1299 cells. Taken together, these data suggested that PPVI-induced, caspase-1-mediated pyroptosis via the induction of the ROS/NF-κB/NLRP3/GSDMD signal axis in NSCLC, which further clarified the mechanism of PPVI in the inhibition of NSCLC, and thereby provided a possibility for PPVI to serve as a novel therapeutic agent for NSCLC in the future.
Collapse
|
31
|
Alkahtani HM, Abdalla AN, Obaidullah AJ, Alanazi MM, Almehizia AA, Alanazi MG, Ahmed AY, Alwassil OI, Darwish HW, Abdel-Aziz AAM, El-Azab AS. Synthesis, cytotoxic evaluation, and molecular docking studies of novel quinazoline derivatives with benzenesulfonamide and anilide tails: Dual inhibitors of EGFR/HER2. Bioorg Chem 2020; 95:103461. [PMID: 31838290 DOI: 10.1016/j.bioorg.2019.103461] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/11/2019] [Accepted: 11/20/2019] [Indexed: 12/19/2022]
Abstract
We synthesized a new series of 2-[(3-(4-sulfamoylphenethyl)-4(3H)-quinazolinon-2-yl)thio]anilide derivatives (2-16) and evaluated their cytotoxic activity against breast adenocarcinoma (MCF-7), colorectal adenocarcinoma (HT-29), and acute myeloid leukemia (HL-60 and K562) cells. To reveal their selectivity toward cancer cells, the compounds were also tested against the human fibroblast cell line, MRC-5. Compounds 1-5 exhibited potent cytotoxic activity against the tested cell lines with IC50 values of 0.65-3.86, 0.68-4.60, 0.41-1.45, 0.42-4.07, and 3.77-25.55 μM, respectively compared to sorafenib, the standard drug (IC50 2.50, 2.50, and 3.14 μM against MCF-7, HT-29, and HL60 cells, respectively). Interestingly, compounds 1-5 displayed selectivity toward the cancer cell lines over MRC-5 (IC50 3.77-25.55 μM). These compounds also displayed potent inhibitory activity against EGFR and HER2 kinases (IC50 0.09-0.43 and 0.15-0.33 μM, respectively) compared to the standard drug, sorafenib (IC50 0.11 and 0.13 μM, respectively). Likewise, compounds 1, 4, and 5 showed strong inhibitory activity against VEGFR2 (IC50 0.34, 0.28 and 0.39 μM, respectively) compared to sorafenib (IC50 0.17 μM). We also employed molecular docking to identify the structural features required for the EGFR/HER2 inhibitory activity of the new series. Ultimately, compounds 1, 4, and 5 were demonstrated to be candidates for further preclinical investigations.
Collapse
Affiliation(s)
- Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Ahmad J Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdulrahman A Almehizia
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mashael G Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ahmed Y Ahmed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Osama I Alwassil
- Department of Pharmaceutical Sciences, College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, 3163, P.O. Box 3660, Riyadh 11481, Saudi Arabia
| | - Hany W Darwish
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Alaa A-M Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Adel S El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
32
|
Shou J, Wang M, Cheng X, Wang X, Zhang L, Liu Y, Fei C, Wang C, Gu F, Xue F, Li J, Zhang K. Tizoxanide induces autophagy by inhibiting PI3K/Akt/mTOR pathway in RAW264.7 macrophage cells. Arch Pharm Res 2020; 43:257-270. [PMID: 31894502 DOI: 10.1007/s12272-019-01202-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/06/2019] [Indexed: 12/25/2022]
Abstract
As the main metabolite of nitazoxanide, tizoxanide (TIZ) has a broad-spectrum anti-infective effect against parasites, bacteria, and virus. In this study, we investigated the effects of TIZ on autophagy by regulating the PI3K/Akt/mTOR signaling pathway. RAW264.7 macrophage cells were treated with various TIZ concentrations. Cell viability assay, transmission electron microscope, and immunofluorescence staining were used to detect the biological function of the macrophage cells, and the expression levels of the autophagy pathway-related proteins were measured by Western blot. Results revealed that TIZ promoted the conversion of LC3-I to LC3-II, the formation of autophagy vacuoles, and the degradation of SQSTM1/p62 in a concentration- and time-dependent manner in RAW264.7 cells. Treatment with TIZ increased the Beclin-1 expression level and inhibited PI3K, Akt, mTOR, and ULK1 activation. These effects were enhanced by pretreatment with rapamycin but attenuated by pretreatment with LY294002. In addition, the conversion of LC3-I to LC3-II was observed in Vero, 293T, and HepG2 cells treated with TIZ. These data suggested that TIZ may induce autophagy by inhibiting the Akt/mTOR/ULK1 signaling pathway in macrophages and other cells.
Collapse
Affiliation(s)
- Jiaoqin Shou
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
- College of Chemistry, Xiangtan University, Yuhu District, Xiangtan, 411105, Hunan, China
| | - Mi Wang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Xiaolei Cheng
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Xiaoyang Wang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Lifang Zhang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Yingchun Liu
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Chenzhong Fei
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Chunmei Wang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Feng Gu
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Feiqun Xue
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China
| | - Juan Li
- College of Chemistry, Xiangtan University, Yuhu District, Xiangtan, 411105, Hunan, China.
| | - Keyu Zhang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 518 Ziyue RD, Minhang District, Shanghai, 200241, China.
| |
Collapse
|
33
|
TIPRL potentiates survival of lung cancer by inducing autophagy through the eIF2α-ATF4 pathway. Cell Death Dis 2019; 10:959. [PMID: 31862913 PMCID: PMC6925247 DOI: 10.1038/s41419-019-2190-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022]
Abstract
Autophagy, an intracellular system of degrading damaged organelles and misfolded proteins, is essential for cancer cell survival. Despite the progress made towards understanding the mechanism, identification of novel autophagy regulators presents a major obstacle in developing anticancer therapies. Here, we examine the association between the TOR signaling pathway regulator-like (TIPRL) protein and autophagy in malignant transformation of tumors. We show that TIPRL upregulation in non-small cell lung cancer (NSCLC) potentiated autophagy activity and enabled autophagic clearance of metabolic and cellular stress, conferring a survival advantage to cancer cells. Importantly, the interaction of TIPRL with eukaryotic initiation factor 2α (eIF2α) led to eIF2α phosphorylation and activation of the eIF2α-ATF4 pathway, thereby inducing autophagy. Conversely, TIPRL depletion increased apoptosis by reducing autophagic clearance, which was markedly enhanced in TIPRL-depleted A549 xenografts treated with 2-deoxy-D-glucose. Overall, the study indicated that TIPRL is a potential regulator of autophagy and an important drug target for lung cancer therapy.
Collapse
|
34
|
Kumar G, Gupta R, Sharan S, Roy P, Pandey DM. Anticancer activity of plant leaves extract collected from a tribal region of India. 3 Biotech 2019; 9:399. [PMID: 31656737 DOI: 10.1007/s13205-019-1927-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/28/2019] [Indexed: 12/21/2022] Open
Abstract
The goal of this research was to explore the preliminary anticancer properties of five plants namely Calotropis procera, Moringa oleifera, Millettia pinnata, Basela alba and Euphorbia neriifolia available in Jharkhand which is used for the medicinal purpose by local tribes. In the present study, plant leaves from five species were collected, dried and extracted with solvents of increasing polarity, followed by assessment of their cytotoxicity in A549 non-small-cell lung cancer cells. In the antimicrobial assay, the methanol extract of the M. pinnata leaves exhibited comparatively higher zone of inhibition of 0.7 ± 0.20 cm against a Salmonella typhi culture than the other extracts. M. pinnata leaves extract also displayed the maximum percentage inhibition in the DPPH, 83.97 ± 0.01 FRAP, 193.14 ± 3.01 mM assays. Furthermore, the cytotoxicity of the chloroform (37.45 ± 1.04) and ethyl acetate extracts (34.20 ± 0.81) of M. pinnata against A549 cells was found relatively higher with respect to another extract. In contrast, a study with the L132 normal epithelial lung cell line revealed less toxicity from the chloroform extract (0.33 ± 0.19) compared to the ethyl acetate extract (6.65 ± 0.59). Based on these findings, phytochemical investigation on chloroform and ethyl acetate extract of M. pinnata was performed using UPLC-ESI-MS/MS analysis revealing the presence of β-sitosterol, lanceolatin B, karanjin, and stigmasterol. Congruently, a complete phytochemical and cytotoxic investigation of the M. pinnata extract constituents might infer the potency of this extract/s as anticancer, antioxidant and antimicrobial agents.
Collapse
Affiliation(s)
- Gourav Kumar
- 1Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi India
| | - Rashmi Gupta
- 1Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi India
| | - Shruti Sharan
- 2Department of Bio-Technology, Indian Institute of Technology, Roorkee, India
| | - Partha Roy
- 2Department of Bio-Technology, Indian Institute of Technology, Roorkee, India
| | - Dev Mani Pandey
- 1Department of Bio-Engineering, Birla Institute of Technology, Mesra, Ranchi India
| |
Collapse
|
35
|
Tong Y, Wang M, Huang H, Zhang J, Huang Y, Chen Y, Pan H. Inhibitory effects of genistein in combination with gefitinib on the hepatocellular carcinoma Hep3B cell line. Exp Ther Med 2019; 18:3793-3800. [PMID: 31611933 PMCID: PMC6781792 DOI: 10.3892/etm.2019.8027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/08/2019] [Indexed: 01/27/2023] Open
Abstract
Combination therapy is an important method for treating advanced hepatocellular carcinoma (HCC). Gefitinib is an epidermal growth factor receptor (EGFR) inhibitor, which has profound effects on HCC. The purpose of the present study was to investigate the effects of genistein in combination with gefitinib on the proliferation and apoptosis of HCC cells and the associated mechanism. Cell counting kit-8 assay was performed to calculate the IC50 values and cytotoxicity, whilst flow cytometry was used to assess cell apoptosis. Protein expression was detected using western blot analysis. The IC50 of genistein and gefitinib on Hep3B cells were calculated to be 128.078 and 13.657 µM, respectively. Genistein in combination with gefitinib significantly inhibited cell viability, promoted apoptosis and reduced EGFR, vascular endothelial growth factor receptor and platelet-derived growth factor receptor phosphorylation. Genistein in combination with gefitinib promoted the expression of cleaved caspase-3 and cleaved poly ADP-ribose polymerase. In addition, combined treatment of genistein and gefitinib strongly inhibited the activation of the Akt/Erk/mTOR signaling pathway. In conclusion, findings from the present study suggest that genistein in combination with gefitinib inhibit HCC cell proliferation and promote apoptosis by inhibiting the Akt/Erk/mTOR pathway.
Collapse
Affiliation(s)
- Yongxi Tong
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Mingshan Wang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Haijun Huang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Jiajie Zhang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yicheng Huang
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| | - Yingjun Chen
- Department of Infectious Diseases, Tiantai People's Hospital of Zhejiang Province, Taizhou, Zhejiang 317200, P.R. China
| | - Hongying Pan
- Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
36
|
Hao Y, Yin H, Zhu C, Li F, Zhang Y, Li Y, Wang X, Li D. [Matrine inhibits proliferation and promotes autophagy and apoptosis in non-small cell lung cancer cells by deactivating PI3K/AKT/mTOR pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:760-765. [PMID: 31340906 DOI: 10.12122/j.issn.1673-4254.2019.07.02] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To investigate the inhibitory effect of matrine on the proliferation of human non-small cell lung cancer (NSCLC) and explore the possible molecular mechanism. METHODS Cultured human NSCLC A549 cells were treated with 0.4, 0.8, 1.2, 1.6, and 2.0 g/L matrine for 24, 48 or 72 h. CCK-8 assay was used for measuring the changes in A549 cell viability. The morphological changes of the cells were observed under a fluorescence microscope, and flow cytometry was employed for analyzing the cell apoptosis. The effects of matrine and the PI3K specific inhibitor LY294002 (10 nmol/L) on AKT pathway and autophagy-related proteins in A549 cells were investigated using Western blotting. RESULTS Matrine significantly inhibited the proliferation of A549 cells in a time- and dose-dependent manner (P < 0.05). At the concentration of 1.6 g/L or higher, matrine caused obvious cell shrinkage and fragmentation and significantly increased floating cells; autophagy vacuoles could be observed in the cells after acridine orange staining. Within the concentrations range of 0.8-1.6 g/L, matrine time- and dosedependently increased the cell apoptosis. Treatment of the cells with 1.6 g/L matrine and 10 nmol/L LY294002 resulted in significantly lowered expressions of p-AKT and p-mTOR proteins and increased the expression of light chain 3B (LC 3B), an autophagy-related protein, as compared with those in the control cells (P < 0.05). CONCLUSIONS We demonstrate that matrine inhibits the proliferation and induces autophagy and apoptosis of A549 cells by deactivating AKT pathway, suggesting the potential of matrine as an anti-cancer agent for lung cancer.
Collapse
Affiliation(s)
- Yanmei Hao
- Department of Clinical Laboratory, Bengbu Medical College, Bengbu 233030, China
| | - Hongmei Yin
- Department of Radiotherapy, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Chaomang Zhu
- Department of Radiotherapy, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Feng Li
- Department of Radiotherapy, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Yingjie Zhang
- Department of Clinical Laboratory, Bengbu Medical College, Bengbu 233030, China
| | - Yuyun Li
- Department of Clinical Laboratory, Bengbu Medical College, Bengbu 233030, China
| | - Xiaojing Wang
- Anhui Clinical and Preclinical Key laboratory of Respiratory Disease, Bengbu Medical College, Bengbu 233030, China
| | - Duojie Li
- Department of Radiotherapy, First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| |
Collapse
|
37
|
Xia T, Zhang J, Zhou C, Li Y, Duan W, Zhang B, Wang M, Fang J. 20(S)-Ginsenoside Rh2 displays efficacy against T-cell acute lymphoblastic leukemia through the PI3K/Akt/mTOR signal pathway. J Ginseng Res 2019; 44:725-737. [PMID: 32913402 PMCID: PMC7471214 DOI: 10.1016/j.jgr.2019.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/20/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background T-cell acute lymphoblastic leukemia (T-ALL) is a kind of aggressive hematological cancer, and the PI3K/Akt/mTOR signaling pathway is activated in most patients with T-ALL and responsible for poor prognosis. 20(S)-Ginsenoside Rh2 (20(S)-GRh2) is a major active compound extracted from ginseng, which exhibits anti-cancer effects. However, the underlying anticancer mechanisms of 20(S)-GRh2 targeting the PI3K/Akt/mTOR pathway in T-ALL have not been explored. Methods Cell growth and cell cycle were determined to investigate the effect of 20(S)-GRh2 on ALL cells. PI3K/Akt/mTOR pathway–related proteins were detected in 20(S)-GRh2–treated Jurkat cells by immunoblotting. Antitumor effect of 20(S)-GRh2 against T-ALL was investigated in xenograft mice. The mechanisms of 20(S)-GRh2 against T-ALL were examined by cell proliferation, apoptosis, and autophagy. Results In the present study, the results showed that 20(S)-GRh2 decreased cell growth and arrested cell cycle at the G1 phase in ALL cells. 20(S)-GRh2 induced apoptosis through enhancing reactive oxygen species generation and upregulating apoptosis-related proteins. 20(S)-GRh2 significantly elevated the levels of pEGFP-LC3 and autophagy-related proteins in Jurkat cells. Furthermore, the PI3K/Akt/mTOR signaling pathway was effectively blocked by 20(S)-GRh2. 20(S)-GRh2 suppressed cell proliferation and promoted apoptosis and autophagy by suppressing the PI3K/Akt/mTOR pathway in Jurkat cells. Finally, 20(S)-GRh2 alleviated symptoms of leukemia and reduced the number of white blood cells and CD3 staining in the spleen of xenograft mice, indicating antitumor effects against T-ALL invivo. Conclusion These findings indicate that 20(S)-GRh2 exhibits beneficial effects against T-ALL through the PI3K/Akt/mTOR pathway and could be a natural product of novel target for T-ALL therapy.
Collapse
Affiliation(s)
- Ting Xia
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jin Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Chuanxin Zhou
- Department of Pediatrics, The Fifth Hospital of Sun Yat Sen University, Sun Yat sen University, Zhuhai, Guangdong, China
| | - Yu Li
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Wenhui Duan
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Bo Zhang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Min Wang
- State Key Laboratory of Food Nutrition and Safety, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Jianpei Fang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guang Dong, China.,Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-sen University, Guangzhou, Guang Dong, China
| |
Collapse
|
38
|
Yang L, Ying S, Hu S, Zhao X, Li M, Chen M, Zhu Y, Song P, Zhu L, Jiang T, An H, Yousafzai NA, Xu W, Zhang Z, Wang X, Feng L, Jin H. EGFR TKIs impair lysosome-dependent degradation of SQSTM1 to compromise the effectiveness in lung cancer. Signal Transduct Target Ther 2019; 4:25. [PMID: 31637005 PMCID: PMC6799834 DOI: 10.1038/s41392-019-0059-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/30/2019] [Accepted: 06/02/2019] [Indexed: 01/07/2023] Open
Abstract
Tyrosine kinase inhibitors for epidermal growth factor receptor (EGFR TKIs) greatly improved clinical outcomes of patients with non-small cell lung cancer (NSCLC). Unfortunately, primary and acquired resistance limits their clinical benefits. To overcome such resistance, new generations of EGFR TKIs have been developed by targeting newly identified mutations in EGFR. However, much less effort has been put into alternative strategies, such as targeting the intrinsic protective responses to EGFR TKIs. In this study, we found that EGFR TKIs, including gefitinib and AZD9291, impaired lysosome-dependent degradation of SQSTM1, thus compromising their anti-cancer efficiency. By accumulating in the lysosome lumen, gefitinib and AZD9291 attenuated lysosomal acidification and impaired autolysosomal degradation of SQSTM1 owing to their intrinsic alkalinity. As a result, SQSTM1 protein was stabilized in response to gefitinib and AZD9291 treatment and conferred EGFR TKI resistance. Depleting SQSTM1 significantly increased the sensitivity of NSCLC cells to gefitinib and AZD9291 both in vitro and in vivo. Furthermore, a chemically modified gefitinib analog lacking alkalinity displayed stronger inhibitory effects on NSCLC cells. Therefore, targeting accumulated SQSTM1 or chemically modified EGFR TKIs may represent new strategies to increase the effectiveness of EGFR targeted therapy.
Collapse
Affiliation(s)
- Lixian Yang
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Shilong Ying
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Shiman Hu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Xiangtong Zhao
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Muchun Li
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Miaoqin Chen
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Yiran Zhu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Ping Song
- Department of Medical Oncology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Liyuan Zhu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Tingting Jiang
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Huimin An
- Department of Pathology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Neelum Aziz Yousafzai
- Department of Medical Oncology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Wenxia Xu
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Zhiguo Zhang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Xian Wang
- Department of Medical Oncology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Lifeng Feng
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Hongchuan Jin
- Laboratory of Cancer Biology, Key Lab of Biotherapy in Zhejiang, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, China
| |
Collapse
|
39
|
Wattanathamsan O, Hayakawa Y, Pongrakhananon V. Molecular mechanisms of natural compounds in cell death induction and sensitization to chemotherapeutic drugs in lung cancer. Phytother Res 2019; 33:2531-2547. [DOI: 10.1002/ptr.6422] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/06/2019] [Accepted: 05/26/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Onsurang Wattanathamsan
- Inter‐department program of Pharmacology, Graduate SchoolChulalongkorn University Bangkok Thailand
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research ClusterChulalongkorn University Bangkok Thailand
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural MedicineUniversity of Toyama Toyama Japan
| | - Varisa Pongrakhananon
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research ClusterChulalongkorn University Bangkok Thailand
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical SciencesChulalongkorn University Bangkok Thailand
| |
Collapse
|
40
|
Wang Q, Shen B, Qin X, Liu S, Feng J. Akt/mTOR and AMPK signaling pathways are responsible for liver X receptor agonist GW3965-enhanced gefitinib sensitivity in non-small cell lung cancer cell lines. Transl Cancer Res 2019; 8:66-76. [PMID: 35116735 PMCID: PMC8797756 DOI: 10.21037/tcr.2018.12.34] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/14/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND This study was to systemically analyze the mechanism of LXR ligand GW3965-induced sensitivity to EGFR-TKI in EGFR-TKI-resistant non-small cell lung cancer (NSCLC) cell lines. METHODS Gefitinib-resistant PC9 cell line (EGFR exon 19 deletion) was treated with single and combined treatment with GW3965 and gefitinib. Cell viability, apoptosis and autophagy were detected using MTT, flow cytometric analysis and immunofluorescent analysis, respectively. Autophagy-related signaling pathways were detected using Western blot analysis. RESULTS Inhibited cell viability by single and combined treatment with gefitinib and GW3965 were observed. Combined treatment with gefitinib and GW3965 increased LC3 II/I ratio and Beclin 1 expression. Synergistic effect of gefitinib and GW3965 on apoptosis and autophagosome accumulation as well as on the inhibition of Akt/mTOR signaling and activation of AMP-activated protein kinase (AMPK) was observed in gefitinib-resistant PC9 cells. AMPK expression showed similar profile with apoptosis and autophagy of PC9 cells. CONCLUSIONS We confirmed that GW3965 and gefitinib showed synergistic effect on Akt/mTOR inhibition, apoptosis and autophagy of lung cancer cells. Gefitinib sensitivity in PC9 cell line might be mediated by Akt/mTOR, AMPK and JNK signaling pathways.
Collapse
Affiliation(s)
- Qingbo Wang
- The Fourth Clinical School of Nanjing Medical University, Nanjing 210029, China
| | - Bo Shen
- Department of Chemotherapy, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing 210009, China
| | - Xiaobing Qin
- The Fourth Clinical School of Nanjing Medical University, Nanjing 210029, China
| | - Siwen Liu
- Department of Chemotherapy, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing 210009, China
| | - Jifeng Feng
- Department of Chemotherapy, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing 210009, China
| |
Collapse
|
41
|
Zhang Y, Wu Z, Yu H, Wang H, Liu G, Wang S, Ji X. Chinese Herbal Medicine Wenxia Changfu Formula Reverses Cell Adhesion-Mediated Drug Resistance via the Integrin β1-PI3K-AKT Pathway in Lung Cancer. J Cancer 2019; 10:293-304. [PMID: 30719123 PMCID: PMC6360309 DOI: 10.7150/jca.25163] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 10/04/2018] [Indexed: 12/24/2022] Open
Abstract
In the treatment of lung cancer, the multidrug resistance to chemotherapeutic drugs is one of the reasons of low rates for cure and treatment failure, the combination of chemotherapeutic drugs and traditional Chinese medicine can increase the sensitivity of chemotherapy and reduce its adverse effects. Our previous study has proved that Chinese herbal medicine (CHM) Wenxia Changfu Formula (WCF for short) effectively enhances chemotherapeutic efficacy in lung cancer treatment and reverses multidrug resistance in lung cancer cells in vitro. The present study aims to investigate the effect and mechanism of WCF in reversing cell adhesion-mediated drug resistance of lung cancer by using A549 three-dimensional cell culture and nude mouse model of the A549 cell line with Integrin β1 overexpression. We show that the combination of WCF with DDP can decrease proliferation of lung cancer cells by inducing cell cycle arrest and apoptosis. Moreover, we find that the combination of WCF with DDP suppresses the expression of certain molecules which regulate cell cycle and apoptosis. Mechanistically, we show that the Integrin β1, FAK, PI3K, and AKT protein expressions are suppressed by DDP and even more responses are observed when DDP and WCF are combined, showing WCF treatment enhances the effect of commonly used anticancer drugs. In line with the above findings, our results confirm that WCF reverses cell adhesion-mediated drug resistance of lung cancer via inactivating Integrin β1/PI3K/AKT and apoptosis induction.
Collapse
Affiliation(s)
- YaNan Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China.,Shandong Provincial Chinese Medicine Classical Prescription Demonstration Engineering Technology Research Center, Jinan, Shangdong Province 250355, China
| | - ZhiChun Wu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China.,Shandong Provincial Chinese Medicine Classical Prescription Demonstration Engineering Technology Research Center, Jinan, Shangdong Province 250355, China
| | - HuaYun Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China.,Shandong Provincial Chinese Medicine Classical Prescription Demonstration Engineering Technology Research Center, Jinan, Shangdong Province 250355, China
| | - HuaXin Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China.,Shandong Provincial Chinese Medicine Classical Prescription Demonstration Engineering Technology Research Center, Jinan, Shangdong Province 250355, China
| | - Guowei Liu
- Shandong Provincial Chinese Medicine Classical Prescription Demonstration Engineering Technology Research Center, Jinan, Shangdong Province 250355, China
| | - ShiJun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shangdong Province 250355, China.,Shandong Provincial Chinese Medicine Classical Prescription Demonstration Engineering Technology Research Center, Jinan, Shangdong Province 250355, China
| | - XuMing Ji
- College of Basic Medicine,Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China.,Shandong Provincial Chinese Medicine Classical Prescription Demonstration Engineering Technology Research Center, Jinan, Shangdong Province 250355, China
| |
Collapse
|
42
|
Yu JH, Chen L, Yu JY, Luo HQ, Wang L. PI3K-PKB-mTOR hyperactivation in relation to nasopharyngeal carcinoma progression and prognosis. J Cell Biochem 2018; 120:10186-10194. [PMID: 30582216 DOI: 10.1002/jcb.28303] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/28/2018] [Indexed: 12/27/2022]
Abstract
Nasopharyngeal carcinoma (NPC) has a unique and complex etiology, which is not completely understood. The aim of this study is to investigate the expression patterns of phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB), and mammalian target of rapamycin (mTOR) proteins in patients with NPC and their relationship with NPC progression and prognosis. Between January 2008 and March 2010, PI3K, PKB, and mTOR protein expressions were detected using immunohistochemistry among 119 patients with NPC and 30 healthy people. A 5-year follow-up was conducted for all patients. Correlations of PI3K, PKB, and mTOR proteins with the clinicopathological features and prognosis of NPC were evaluated using Spearman's rank correlation coefficient and Kaplan-Meier curve. Cox's regression analysis was performed to analyze the risk factors for the prognosis of NPC. First, PI3K, PKB, and mTOR were highly expressed in patients with NPC. The expressions of PI3K, PKB, and mTOR proteins were associated with T stage, N stage, clinical stage, relapse, and distant metastasis. Meanwhile, PI3K is positively correlated with PKB and PKB is positively correlated with mTOR in NPC. Higher PI3K, PKB, and mTOR protein expressions were related to a shorter survival time and a lower survival rate in NPC. Cox regression analysis revealed that age, T stage, N stage, PI3K, PKB, and mTOR were independent risk factors for NPC patient survival. Altogether, our data suggest that overexpression of PI3K, PKB, and mTOR proteins is an important indicator of poor survival in NPC. In addition, inhibition of PI3K-PKB-mTOR signaling may also contribute to the development of new therapeutic strategies for NPC.
Collapse
Affiliation(s)
- Jian-Hua Yu
- Department of Oncology, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Li Chen
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian-Yong Yu
- Department of Pharmacy, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Hong-Qiang Luo
- Department of ENT, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Ling Wang
- Department of Oncology, Jiangxi Provincial People's Hospital, Nanchang, China
| |
Collapse
|
43
|
Xiao X, Xu M, Yang C, Yao Y, Liang LN, Ed Chung P, Long Q, Zacksenhaus E, He Z, Liu S, Ben-David Y. Novel racemosin B derivatives as new therapeutic agents for aggressive breast cancer. Bioorg Med Chem 2018; 26:6096-6104. [PMID: 30471828 DOI: 10.1016/j.bmc.2018.11.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/09/2018] [Accepted: 11/11/2018] [Indexed: 02/01/2023]
Abstract
Carbazole derivatives show anti-cancer activity and are of great interest for drug development. In this study, we synthesized and analyzed several new alkylamide derivatives of racemocin B, a natural indolo[3,2-a]carbazole molecule originally isolated from the green alga Caulerpa racemose. Several alkylamide derivatives were found to exhibit moderate to strong growth inhibition against human breast cancer cell lines. They induced G2/M cell cycle arrest and apoptosis in the aggressive triple-negative breast cancer cell line MDA-MB-231. Among these derivatives, compound 25 with the lowest IC50 induced cell death by suppressing autophagy. This was accompanied by inhibition of autophagic flux and accumulation of autophagy protein 1 light chain 3, LC3II, and p62. The novel alkylamide derivative offers a potential new treatment for human breast cancer.
Collapse
Affiliation(s)
- Xiao Xiao
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China; Stem Cell and Tissue Engineering Research Center, Laboratory Animal Center, Department of Immunology, Guizhou Medical University, Guiyang 550004, PR China
| | - Mei Xu
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Chao Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Yao Yao
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Li-Na Liang
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China
| | - Philip Ed Chung
- Department of Medicine, University of Toronto, Toronto, Ontario M5G2M1, Canada; Division of Advanced Diagnostics, Toronto General Research Institute-University Health Network, Toronto, Ontario M5G2M1, Canada
| | - Qun Long
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; Stem Cell and Tissue Engineering Research Center, Laboratory Animal Center, Department of Immunology, Guizhou Medical University, Guiyang 550004, PR China
| | - Eldad Zacksenhaus
- Department of Medicine, University of Toronto, Toronto, Ontario M5G2M1, Canada; Division of Advanced Diagnostics, Toronto General Research Institute-University Health Network, Toronto, Ontario M5G2M1, Canada
| | - Zhixu He
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; Stem Cell and Tissue Engineering Research Center, Laboratory Animal Center, Department of Immunology, Guizhou Medical University, Guiyang 550004, PR China.
| | - Sheng Liu
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| | - Yaacov Ben-David
- State Key Laboratory for Functions and Applications of Medicinal Plants/College of Basic Medical Sciences, Guizhou Medical University, Guiyang 550014, PR China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academic of Sciences, Guiyang 550014, PR China.
| |
Collapse
|
44
|
Mphahlele MJ, Maluleka MM, Aro A, McGaw LJ, Choong YS. Benzofuran-appended 4-aminoquinazoline hybrids as epidermal growth factor receptor tyrosine kinase inhibitors: synthesis, biological evaluation and molecular docking studies. J Enzyme Inhib Med Chem 2018; 33:1516-1528. [PMID: 30274538 PMCID: PMC6171423 DOI: 10.1080/14756366.2018.1510919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A series of 2-arylbenzo[b]furan–appended 4-aminoquinazoline hybrids were prepared and evaluated for cytotoxicity in vitro against the human lung cancer (A549), colorectal adenocarcinoma (Caco-2), hepatocellular carcinoma (C3A) and cervical cancer (HeLa) cell lines. Compounds 10d and 10j exhibited significant cytotoxicity against the C3A and Caco-2 cell lines and induced apoptosis in these cell lines. Likewise, compounds 10d and 10e exhibited significant inhibitory activity towards epidermal growth factor receptor-tyrosine kinase phosphorylation (IC50 values of 29.3 nM and 31.1 nM, respectively) against Gefitinib (IC50 = 33.1 nM). Molecular docking of compounds 10 into EGFR-TK active site suggests that they bind to the region of EGFR like Gefitinib does.
Collapse
Affiliation(s)
- Malose J Mphahlele
- a Department of Chemistry, College of Science, Engineering and Technology , University of South Africa , Florida , South Africa
| | - Marole M Maluleka
- a Department of Chemistry, College of Science, Engineering and Technology , University of South Africa , Florida , South Africa
| | - Abimbola Aro
- b Phytomedicine Programme , Department of Paraclinical Sciences , University of Pretoria , Onderstepoort , South Africa
| | - Lyndy J McGaw
- b Phytomedicine Programme , Department of Paraclinical Sciences , University of Pretoria , Onderstepoort , South Africa
| | - Yee Siew Choong
- c Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia , Minden , Malaysia
| |
Collapse
|
45
|
Bednarczyk M, Zmarzły N, Grabarek B, Mazurek U, Muc-Wierzgoń M. Genes involved in the regulation of different types of autophagy and their participation in cancer pathogenesis. Oncotarget 2018; 9:34413-34428. [PMID: 30344951 PMCID: PMC6188136 DOI: 10.18632/oncotarget.26126] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 08/30/2018] [Indexed: 12/13/2022] Open
Abstract
Autophagy is a highly conserved mechanism of self-digestion that removes damaged organelles and proteins from cells. Depending on the way the protein is delivered to the lysosome, four basic types of autophagy can be distinguished: macroautophagy, selective autophagy, chaperone-mediated autophagy and microautophagy. Macroautophagy involves formation of autophagosomes and is controlled by specific autophagy-related genes. The steps in macroautophagy are initiation, phagophore elongation, autophagosome maturation, autophagosome fusion with the lysosome, and proteolytic degradation of the contents. Selective autophagy is macroautophagy of a specific cellular component. This work focuses on mitophagy (selective autophagy of abnormal and damaged mitochondria), in which the main participating protein is PINK1 (phosphatase and tensin homolog-induced putative kinase 1). In chaperone-mediated autophagy, the substrate is bound to a heat shock protein 70 chaperone before it is delivered to the lysosome. The least characterized type of autophagy is microautophagy, which is the degradation of very small molecules without participation of an autophagosome. Autophagy can promote or inhibit tumor development, depending on the severity of the disease, the type of cancer, and the age of the patient. This paper describes the molecular basis of the different types of autophagy and their importance in cancer pathogenesis.
Collapse
Affiliation(s)
- Martyna Bednarczyk
- Department of Internal Diseases, School of Public Health in Bytom, Medical University of Silesia in Katowice, 40–055 Katowice, Poland
| | - Nikola Zmarzły
- Department of Molecular Biology, School of Pharmacy with The Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, 40–055 Katowice, Poland
| | - Beniamin Grabarek
- Department of Molecular Biology, School of Pharmacy with The Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, 40–055 Katowice, Poland
| | - Urszula Mazurek
- Department of Molecular Biology, School of Pharmacy with The Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia in Katowice, 40–055 Katowice, Poland
| | - Małgorzata Muc-Wierzgoń
- Department of Internal Diseases, School of Public Health in Bytom, Medical University of Silesia in Katowice, 40–055 Katowice, Poland
| |
Collapse
|
46
|
Synthesis, Biological Evaluation and Molecular Docking of Novel Indole-Aminoquinazoline Hybrids for Anticancer Properties. Int J Mol Sci 2018; 19:ijms19082232. [PMID: 30065164 PMCID: PMC6121530 DOI: 10.3390/ijms19082232] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
A series of indole-aminoquinazolines was prepared via amination of the 2-aryl-4-chloroquinazolines with the 7-amino-2-aryl-5-bromoindoles. It was then evaluated for cytotoxicity in vitro against human lung cancer (A549), epithelial colorectal adenocarcinoma (Caco-2), hepatocellular carcinoma (C3A), breast adenocarcinoma (MCF-7), and cervical cancer (HeLa) cells. A combination on the quinazoline and indole moieties of a 2-phenyl and 2-(4-fluorophenyl) rings in compound 4b; 2-(4-fluorophenyl) and 3-chlorophenyl rings in compound 4f; or the two 2-(4-fluorophenyl) rings in compound 4g, resulted in significant and moderate activity against the Caco-2 and C3A cell lines. The indole-aminoquinazoline hybrids compounds 4f and 4g induced apoptosis in Caco-2 and C3A cells, and were also found to exhibit moderate (IC50 = 52.5 nM) and significant (IC50 = 40.7 nM) inhibitory activity towards epidermal growth factor receptor (EGFR) against gefitinib (IC50 = 38.9 nM). Molecular docking suggests that 4a–h could bind to the ATP region of EGFR like erlotinib.
Collapse
|
47
|
Zhu J, Cai Y, Xu K, Ren X, Sun J, Lu S, Chen J, Xu P. Beclin1 overexpression suppresses tumor cell proliferation and survival via an autophagy‑dependent pathway in human synovial sarcoma cells. Oncol Rep 2018; 40:1927-1936. [PMID: 30066884 PMCID: PMC6111547 DOI: 10.3892/or.2018.6599] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/17/2018] [Indexed: 12/19/2022] Open
Abstract
Beclin1 is an important autophagy‑related prot-ein, which is involved in both autophagy and apoptosis. In recent years, the antitumor effect of Beclin1 has received increased attention. In the present study, we established a stable Beclin1‑overexpressing cell line with SW982 human synovial sarcoma cells. We found that Beclin1 overexpression decreased the cell viability, inhibited proliferation and induced apoptosis in SW982 cells. The expression levels of Bcl‑2 and PCNA were decreased, while the levels of cleaved‑caspase‑3 and cleaved‑PARP were increased. Beclin1 is closely related with autophagy, thus the autophagy‑related markers LC3 and p62 were detected by western blot analysis, and transmission electron microscopy was used to observe autophagosomes. The results showed that the expression level of LC3II was increased and that of p62 was decreased. Moreover, many double membrane‑enclosed autophagosomes were found in cells with Beclin1 overexpression, which indicated that the autophagic activity was enhanced. To explore the effect of autophagy on the viability of SW982 cells, Atg5 was knocked down using siRNA to inhibit the autophagic activity. We found that autophagy contributed to the decrease in cell viability. Knockdown of Atg5 increased the viability and decreased the apoptotic rate of SW982 cells with Beclin1 overexpression. The expression level of Bcl‑2 was increased, while the expression levels of cleaved‑caspase‑3 and cleaved‑PARP were decreased. We also found that the Akt/Bcl‑2/caspase‑9 pathway was involved. The phosphorylation of AKT was positively correlated with cell viability. The cleavage of caspase‑9 was increased by Beclin1 overexpression and decreased by inhibition of autophagy. Altogether, our results suggested that both autophagy and apoptosis contributed to the antitumor effect of Beclin1 in SW982 cells.
Collapse
Affiliation(s)
- Jialin Zhu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Yongsong Cai
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Ke Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaoyu Ren
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| | - Jian Sun
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Shemin Lu
- Department of Genetics and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Jinghong Chen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Institute of Endemic Diseases, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Xu
- Department of Joint Surgery, Xi'an Hong Hui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
48
|
Feng FB, Qiu HY. RETRACTED: Effects of Artesunate on chondrocyte proliferation, apoptosis and autophagy through the PI3K/AKT/mTOR signaling pathway in rat models with rheumatoid arthritis. Biomed Pharmacother 2018; 102:1209-1220. [PMID: 29710540 DOI: 10.1016/j.biopha.2018.03.142] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 03/06/2018] [Accepted: 03/12/2018] [Indexed: 12/20/2022] Open
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figure 5D and 5F, which appear to have a similar phenotype as contained within another publication, as detailed here: https://pubpeer.com/publications/CD4DF7B6DCA28182EC6809846F3653; and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. Concerns were also raised about the provenance of the flow cytometry data in Figure 7A. In addition, suspected duplications were detected in Western blots contained within Figure 5E and 5F. The journal requested the corresponding author comment on these concerns and provide raw data. The corresponding author communicated that the raw data was not available and some of the results were not repeatable and therefore not solid enough to support the conclusions. The authors requested retraction of this article. The Editor-in-Chief assessed this case and decided to retract the article.
Collapse
Affiliation(s)
- Fa-Bo Feng
- Department of Orthopedics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, PR China
| | - Hai-Yan Qiu
- Department of Endocrinology, Hangzhou First People's Hospital, Nanjing Medical University, Hangzhou 310006, PR China.
| |
Collapse
|
49
|
Zhang XJ, Yu HY, Cai YJ, Ke M. Lycium barbarum polysaccharides inhibit proliferation and migration of bladder cancer cell lines BIU87 by suppressing Pi3K/AKT pathway. Oncotarget 2018; 8:5936-5942. [PMID: 27992374 PMCID: PMC5351602 DOI: 10.18632/oncotarget.13963] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 12/09/2016] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to verify whether Lycium barbarum polysaccharides inhibits proliferation and migration of BIU87 cells through Pi3K/AKT pathway. Different concentrations of Lycium barbarum polysaccharides were used to incubate with BIU87cells. LY-294002 and IGF-1 were used to inhibit and activate Pi3K/AKT pathway respectively. MTT were used to investigate the proliferation of BIU87cells. Transwell chambers and wound healing were used to test the migratory ability of BIU87cells. Western blotting were used to investigate the expressions of P21,P27,MMP-2, MMP-9, AKT and p-AKT in BIU87cells. Compared with the control group, the proliferation and migration of BIU87cells and the expression of p-AKT were significantly decreased in the study group; the inhibitory effect of the downregulation of p-AKT by LY-294002on the induction of BIU87cells proliferation and migration was identical to that of Lycium barbarum polysaccharides; upregulation of p-AKT by IGF-1 reversed the Lycium barbarum polysaccharides-induced inhibition of BIU87cells dedifferentiation. In conclusion, LBP inhibits the proliferation and migration of BIU87 cells by suppressing Pi3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Xian-Jun Zhang
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, Zhejiang Province, PR China
| | - Hong-Yuan Yu
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, Zhejiang Province, PR China
| | - Yong-Jian Cai
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, Zhejiang Province, PR China
| | - Mang Ke
- Department of Urology, Taizhou Hospital of Zhejiang Province, Wenzhou Medical University, Linhai, Zhejiang Province, PR China
| |
Collapse
|
50
|
Liu Z, Gao W. Leptomycin B reduces primary and acquired resistance of gefitinib in lung cancer cells. Toxicol Appl Pharmacol 2017; 335:16-27. [PMID: 28942004 PMCID: PMC5643250 DOI: 10.1016/j.taap.2017.09.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 09/07/2017] [Accepted: 09/19/2017] [Indexed: 12/22/2022]
Abstract
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) gefitinib has demonstrated dramatic clinical efficacy in non-small cell lung cancer (NSCLC) patients. However, its therapeutic efficacy is ultimately limited by the development of acquired drug resistance. The aim of this study was to explore the potential utility of chromosome region maintenance 1 (CRM1) inhibitor leptomycin B (LMB) in combination with gefitinib to overcome primary and acquired gefitinib resistance in NSCLC cells. The combinative effects of gefitinib and LMB were evaluated by MTT and its underlining mechanism was assessed by flow cytometry and Western blot. LMB displayed a synergistic effect on gefitinib-induced cytotoxicity in A549 (IC50: 25.0±2.1μM of gefitinib+LMB vs. 32.0±2.5μM of gefitinib alone, p<0.05). Gefitinib+LMB caused a significantly different cell cycle distribution and signaling pathways involved in EGFR/survivin/p21 compared with gefitinib. A549 cells then were treated with progressively increased concentrations of gefitinib (A549GR) or in combination with LMB (A549GLR) over 10months to generate gefitinib resistance. IC50 of gefitinib in A549GLR (37.0±2.8μM) was significantly lower than that in A549GR (53.0±3.0μM, p<0.05), which indicates that LMB could reverse gefitinib-induced resistance in A549. Further mechanism investigation revealed that the expression patterns of EGFR pathway and epithelial-mesenchymal transition (EMT) markers in A549, A549GR, and A549GLR were significantly different. In conclusion, LMB at a very low concentration (0.5nM) combined with gefitinib showed synergistic therapeutic effects and ameliorated the development of gefitinib-induced resistance in lung cancer cells.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, TX, United States
| | - Weimin Gao
- Department of Environmental Toxicology, The Institute of Environmental and Human Health, Texas Tech University, Lubbock, TX, United States.
| |
Collapse
|