1
|
Valivand N, Aravand S, Lotfi H, Esfahani AJ, Ahmadpour-Yazdi H, Gheibi N. Propolis: a natural compound with potential as an adjuvant in cancer therapy - a review of signaling pathways. Mol Biol Rep 2024; 51:931. [PMID: 39177837 DOI: 10.1007/s11033-024-09807-9] [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: 12/10/2023] [Accepted: 07/18/2024] [Indexed: 08/24/2024]
Abstract
Propolis is a natural product used in cancer treatment, which is produced by bees via different sources. The chemical composition of Propolis is determined based on the climatic and geographical conditions, as well as harvesting time and method. This compound has been the subject of numerous investigational endeavors due to its expansive therapeutic capacity which includes antibacterial, anti-fungal, anti-inflammatory, anti-oxidant, anti-viral, and anti-cancer effects. The growing incidence rate of different cancers necessitates the need for developing novel preventive and therapeutic strategies. Chemotherapy, radiotherapy, and stem cell therapy have proved effective in cancer treatment, regardless of the adverse events associated with these modalities. Clinical application of natural compounds such as Propolis may confer promise as an adjuvant therapeutic intervention, particularly in certain subpopulations of patients that develop adverse events associated with anticancer regimens. The diverse biologically active compounds of propolis are believed to confer anti-cancer potential by modulation of critical signaling cascades such as caffeic acid phenethyl ester, Galangin, Artepillin C, Chrysin, Quercetin, Caffeic acid, Nymphaeols A and C, Frondoside A, Genistein, p-coumaric acid, and Propolin C. This review article aims to deliver a mechanistic account of anti-cancer effects of propolis and its components. Propolis can prevent angiogenesis by downregulating pathways involving Jun-N terminal kinase, ERK1/2, Akt and NF-ƘB, while counteracting metastatic progression of cancer by inhibiting Wtn2 and FAK, and MAPK and PI3K/AKT signaling pathways. Moreover, propolis or its main components show regulatory effects on cyclin D, CDK2/4/6, and their inhibitors. Additionally, propolis-induced up-regulation of p21 and p27 may result in cell cycle arrest at G2/M or G0/G1. The broad anti-apoptotic effects of propolis are mediated through upregulation of TRAIL, Bax, p53, and downregulation of the ERK1/2 signaling pathway. Considering the growing body of evidence regarding different anti-cancers effects of propolis and its active components, this natural compound could be considered an effective adjuvant therapy aimed at reducing related side effects associated with chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Nassim Valivand
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sara Aravand
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hajie Lotfi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Azam Janati Esfahani
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Hossein Ahmadpour-Yazdi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran.
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
| |
Collapse
|
2
|
Prades-Sagarra E, Laarakker F, Dissy J, Lieuwes NG, Biemans R, Dubail M, Fouillade C, Yaromina A, Dubois LJ. Caffeic Acid Phenethyl Ester (CAPE), a natural polyphenol to increase the therapeutic window for lung adenocarcinomas. Radiother Oncol 2024; 190:110021. [PMID: 38000688 DOI: 10.1016/j.radonc.2023.110021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/16/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND AND PURPOSE Lung cancers are highly resistant to radiotherapy, necessitating the use of high doses, which leads to radiation toxicities such as radiation pneumonitis and fibrosis. Caffeic Acid Phenethyl Ester (CAPE) has been suggested to have anti-proliferative and pro-apoptotic effects in tumour cells, while radioprotective anti-inflammatory and anti-oxidant effects in the normal tissue. We investigated the radiosensitizing and radioprotective effects of CAPE in lung cancer cell lines and normal tissue in vitro and ex vivo, respectively. MATERIALS AND METHODS The cytotoxic and radiosensitizing effects of CAPE in lung cancer were investigated using viability and clonogenic survival assays. The radioprotective effects of CAPE were assessed in vitro and ex vivo using precision cut lung slices (PCLS). Potential underlying molecular mechanisms of CAPE focusing on cell cycle, cell metabolism, mitochondrial function and pro-inflammatory markers were investigated. RESULTS Treatment with CAPE decreased cell viability in a dose-dependent manner (IC50 57.6 ± 16.6 μM). Clonogenic survival assays showed significant radiosensitization by CAPE in lung adenocarcinoma lines (p < 0.05), while no differences were found in non-adenocarcinoma lines (p ≥ 0.13). Cell cycle analysis showed an increased S-phase (p < 0.05) after incubation with CAPE in the majority of cell lines. Metabolic profiling showed that CAPE shifted cellular respiration towards glycolysis (p < 0.01), together with mitochondrial membrane depolarization (p < 0.01). CAPE induced a decrease in NF-κB activity in adenocarcinomas and decreased pro-inflammatory gene expression in PCLS. CONCLUSION The combination of CAPE and radiotherapy may be a potentially effective approach to increase the therapeutic window in lung cancer patients.
Collapse
Affiliation(s)
- E Prades-Sagarra
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - F Laarakker
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - J Dissy
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - N G Lieuwes
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - R Biemans
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - M Dubail
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405 Orsay Cedex, France
| | - C Fouillade
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL University, Centre Universitaire, 91405 Orsay Cedex, France
| | - A Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - L J Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
3
|
Bjørklund G, Storchylo O, Peana M, Hangan T, Lysiuk R, Lenchyk L, Koshovyi O, Antonyak H, Hudz N, Chirumbolo S. Caffeic Acid Phenethyl Ester: A Potential Therapeutic Cancer Agent? Curr Med Chem 2024; 31:6760-6774. [PMID: 37933215 DOI: 10.2174/0109298673252993230921073502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/20/2023] [Accepted: 07/24/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Propolis and its major phenolic compound, caffeic acid phenethyl ester (CAPE), have garnered considerable scientific interest due to their anti- inflammatory properties and potential therapeutic applications. OBJECTIVES This narrative review explores the potential utility of CAPE in cancer treatment. METHODS We comprehensively reviewed relevant studies from scientific databases (PubMed and Web of Science) from 2000 to 2022. Our search focused on keywords such as cancer, natural drugs, caffeic acid phenethyl ester, CAPE, cancer cell lines, antitumor effects, and propolis. RESULTS CAPE exhibits diverse biological benefits, including antimicrobial, antioxidant, antiviral, anti-inflammatory, cytotoxic, and potentially anti-carcinogenic properties. Numerous studies have demonstrated its wide-ranging antitumor effects on various cancer cell lines, including growth inhibition, apoptosis induction, tumor invasiveness prevention, malignancy suppression, and anti-angiogenic activity. CONCLUSION Following comprehensive preclinical toxicity assessments, further evaluation of CAPE's efficacy and safety through clinical trials is highly recommended to elucidate its potential health benefits in diverse forms of human cancer.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine (CONEM), Mo i Rana, Norway
| | - Olha Storchylo
- Medical Chemistry Department, Odessa National Medical University, Odessa, Ukraine
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Sassari, Italy
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, Constanta, Romania
| | - Roman Lysiuk
- Department of Pharmacognosy and Botany, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- Department of Pharmacognosy and Botany, CONEM Ukraine Life Science Research Group, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Larysa Lenchyk
- Department of Chemistry of Natural Compounds, National University of Pharmacy, Kharkiv, Ukraine
- CONEM Ukraine Pharmacognosy and Natural Product Chemistry Research Group, National University of Pharmacy, Kharkiv, Ukraine
| | - Oleh Koshovyi
- CONEM Ukraine Pharmacognosy and Natural Product Chemistry Research Group, National University of Pharmacy, Kharkiv, Ukraine
- Department of Pharmacognosy, National University of Pharmacy, Kharkiv, Ukraine
| | - Halyna Antonyak
- Department of Ecology, Ivan Franko National University of Lviv, Lviv, Ukraine
| | - Nataliia Hudz
- Department of Drug Technology and Biopharmaceutics, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
- Faculty of Chemistry, University of Opole, Opole, Poland
| | - Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- CONEM Scientific Secretary, Verona, Italy
| |
Collapse
|
4
|
Wu Q, Li J, Hao S, Guo Y, Li Z, Liu Z, Xuan H. Caffeic acid phenethyl ester inhibits MDA-MB-231 cell proliferation in inflammatory microenvironment by suppressing glycolysis and lipid metabolism. Biomed Pharmacother 2023; 168:115766. [PMID: 37864895 DOI: 10.1016/j.biopha.2023.115766] [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: 06/26/2023] [Revised: 10/08/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023] Open
Abstract
Caffeic acid phenethyl ester (CAPE) is one of the main active ingredients of propolis with good antitumor activities. However, the potential effects of CAPE on the glycolysis and lipid metabolism of tumor cells are unclear. Here, the anti-tumor effects of CAPE on MDA-MB-231 cells in an inflammatory microenvironment stimulated with lipopolysaccharide (LPS) were studied by estimating the inflammatory mediators and the key factors of glycolysis and lipid metabolism. The CAPE treatment obviously inhibited proliferation, migration, invasion, and angiogenesis, and the mitochondrial membrane potential was decreased in the LPS-stimulated MDA-MB-231 cells. Compared with the LPS group, pro-inflammatory mediators, including toll-like receptor 4 (TLR4), tumor necrosis factor alpha (TNF-α), NF-kappa-B inhibitor alpha (IκBα), interleukin (IL)-1β, and IL-6, as well as interleukin-1 receptor-associated kinase 4 (IRAK4), declined after the CAPE treatment. Additionally, CAPE significantly down-regulated the levels of glucose transporter 1 (GLUT1), glucose transporter 3 (GLUT3), and the key enzymes of glycolysis-hexokinase 2 (HK2), phosphofructokinase (PFK), pyruvate kinase muscle isozyme M2 (PKM2), and lactate dehydrogenase A (LDHA). Moreover, CAPE treatment decreased the levels of key lipid metabolism proteins, including acetyl coenzyme A carboxylase (ACC), fatty acid synthase (FASN), and free fatty acid (FFA)-transported-related protein CD36. After adding the glycolysis inhibitor 2-deoxy-D-glucose (2-DG), the inhibitory effects of CAPE on cell viability and migration were not significant when compared with the LPS group. In summary, the antitumor activity of CAPE in vitro was mainly via the modulation of the inflammatory mediators and the inhibition of key proteins and enzymes in glucose and lipid metabolism.
Collapse
Affiliation(s)
- Qian Wu
- School of Life Science, Liaocheng University, Liaocheng 252059, China
| | - Junya Li
- School of Life Science, Liaocheng University, Liaocheng 252059, China
| | - Shengyu Hao
- School of Physical Science and Information Technology, Liaocheng University, Liaocheng 252059, China
| | - Yuyang Guo
- School of Life Science, Liaocheng University, Liaocheng 252059, China
| | - Zongze Li
- School of Life Science, Liaocheng University, Liaocheng 252059, China
| | - Zhengxin Liu
- School of Life Science, Liaocheng University, Liaocheng 252059, China
| | - Hongzhuan Xuan
- School of Life Science, Liaocheng University, Liaocheng 252059, China.
| |
Collapse
|
5
|
Kleczka A, Dzik R, Kabała-Dzik A. Caffeic Acid Phenethyl Ester (CAPE) Synergistically Enhances Paclitaxel Activity in Ovarian Cancer Cells. Molecules 2023; 28:5813. [PMID: 37570782 PMCID: PMC10420888 DOI: 10.3390/molecules28155813] [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: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Caffeic acid phenethyl ester (CAPE) belongs to the phenols found in propolis. It has already shown strong antiproliferative, cytotoxic and pro-apoptotic activities against head and neck cancers and against breast, colorectal, lung and leukemia cancer cells. Ovarian cancer is one of the most dangerous gynecological cancers. Its treatment involves intensive chemotherapy with platinum salts and paclitaxel (PTX). The purpose of this study was to evaluate whether the combined use of CAPE and paclitaxel increases the effectiveness of chemotherapeutic agents. The experiment was performed on three ovarian cancer lines: OV7, HTB78, and CRL1572. The effect of the tested compounds was assessed using H-E staining, a wound-healing test, MTT and the cell death detection ELISAPLUS test. The experiment proved that very low doses of PTX (10 nM) showed a cytotoxic effect against all the cell lines tested. Also, the selected doses of CAPE had a cytotoxic effect on the tested ovarian cancer cells. An increase in the cytotoxic effect was observed in the OV7 line after the simultaneous administration of 10 nM PTX and 100 µM CAPE. The increase in the cytotoxicity was dependent on the CAPE dosage (50 vs. 100 µM) and on the duration of the experiment. In the other cell lines tested, the cytotoxic effect of PTX did not increase after the CAPE administration. The administration of PTX together with CAPE increased the percentage of apoptotic cells in the tested ovarian cancer cell lines. Moreover, the simultaneous administration of PTX and CAPE enhanced the anti-migration activity of the chemotherapeutic used in this study.
Collapse
Affiliation(s)
- Anna Kleczka
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Ostrogórska 30, 41-200 Sosnowiec, Poland;
| | - Radosław Dzik
- Department of Biosensors and Processing of Biomedical Signals, Faculty of Biomedical Engineering, Silesian University of Technology, Roosevelta 40, 41-800 Zabrze, Poland;
| | - Agata Kabała-Dzik
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Ostrogórska 30, 41-200 Sosnowiec, Poland;
| |
Collapse
|
6
|
He YX, Shen H, Ji YZ, Hua HR, Zhu Y, Zeng XF, Wang F, Wang KX. N-myc downstream regulated gene 1 inhibition of tumor progression in Caco2 cells. World J Gastrointest Oncol 2022; 14:2313-2328. [PMID: 36568939 PMCID: PMC9782617 DOI: 10.4251/wjgo.v14.i12.2313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/17/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Invasion and migration are the irreversible stages of colorectal cancer (CRC). The key is to find a sensitive, reliable molecular marker that can predict the migration of CRC at an early stage. N-myc downstream regulated gene 1 (NDRG1) is a multifunctional gene that has been tentatively reported to have a strong relationship with tumor invasion and migration, however the current molecular role of NDRG1 in CRC remains unknown.
AIM To explore the role of NDRG1 in the development of CRC.
METHODS NDRG1 stably over-expressed Caco2 cell line was established by lentiviral infection and NDRG1 knock-out Caco2 cell line was established by CRISPR/Cas9. Furthermore, the mRNA and protein levels of NDRG1 in Caco2 cells after NDRG1 over-expression and knockout were detected by real-time polymerase chain reaction and western blot. The cell proliferation rate was measured by the cell counting kit-8 method; cell cycle and apoptosis were detected by flow cytometry; invasion and migration ability were detected by the 24-transwell method.
RESULTS NDRG1 over-expression inhibited Caco2 proliferation and the cell cycle could be arrested at the G1/S phase when NDRG1 was over-expressed, while the number of cells in the G2 phase was significantly increased when NDRG1 was knocked out. This suggests that NDRG1 inhibited the proliferation of Caco2 cells by arresting the cell cycle in the G1/S phase. Our data also demonstrated that NDRG1 promotes early cell apoptosis. Invasion and migration of cells were extensively inhibited when NDRG1 was over-expressed.
CONCLUSION NDRG1 inhibits tumor progression in Caco2 cells which may represent a potential novel therapeutic strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Yi-Xiao He
- Department of Pathology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, Sichuan Province, China
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Hong Shen
- Department of Pathology, Zhaotong First People’s Hospital, Zhaotong 657000, Yunnan Province, China
| | - Yu-Zhu Ji
- Department of Pathology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang 621000, Sichuan Province, China
| | - Hai-Rong Hua
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Yu Zhu
- School of Nursing, Henan Vocational College of Applied Technology, Kaifeng 450000, Henan Province, China
| | - Xiang-Fei Zeng
- Department of Clinical Pathology, West China Hospital, Sichuan University, Chengdu 610000, Sichuan Province, China
| | - Fang Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, Yunnan Province, China
| | - Kai-Xin Wang
- Department of Pathology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen 518052, Guangdong Province, China
| |
Collapse
|
7
|
Si Y, Xu J, Meng L, Wu Y, Qi J. Role of STAT3 in the pathogenesis of nasopharyngeal carcinoma and its significance in anticancer therapy. Front Oncol 2022; 12:1021179. [PMID: 36313702 PMCID: PMC9615247 DOI: 10.3389/fonc.2022.1021179] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a type of head and neck tumor with noticeable regional and ethnic differences. It is associated with Epstein-Barr virus infection and has a tendency for local and distant metastasis. NPC is also highly sensitive to radiotherapy and chemotherapy. Over 70% of patients present with locoregionally advanced disease, and distant metastasis is the primary reason for treatment failure. A signal transducer and activator of transcription 3 (STAT3) promotes NPC oncogenesis through mechanisms within cancerous cells and their interactions with the tumor microenvironment, which is critical in the initiation, progression, and metastasis of NPC. Further, p-STAT3 is strongly associated with advanced NPC. Recent research on STAT3 has focused on its expression at the center of various oncogenic pathways. Here, we discuss the role of STAT3 in NPC and its potential therapeutic inhibitors and analogs for the treatment and control of NPC.
Collapse
|
8
|
Hou CP, Tsui KH, Chen ST, Chang KS, Sung HC, Hsu SY, Lin YH, Feng TH, Juang HH. The Upregulation of Caffeic Acid Phenethyl Ester on Growth Differentiation Factor 15 Inhibits Transforming Growth Factor β/Smad Signaling in Bladder Carcinoma Cells. Biomedicines 2022; 10:biomedicines10071625. [PMID: 35884930 PMCID: PMC9312961 DOI: 10.3390/biomedicines10071625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is known as a TGFβ-like cytokine acting on the TGFβ receptor to modulate target genes. GDF15 is regarded as a tumor suppressor gene in the human bladder and the caffeic acid phenethyl ester (CAPE) induces GDF15 expression to inhibit the tumor growth in vitro and in vivo. However, the interactions among GDF15, CAPE, and TGFβ/Smads signaling in the human bladder carcinoma cells remain unexplored. Results revealed that TGFβ downregulated the expression of GDF15 via the activation of Smad 2/3 and Smad 1/5. Induction of GDF15 on its downstream genes, NDRG1 and maspin, is dependent on the TGFβ/Smad pathways. Moreover, TGFβ blocked the CAPE-inducing expressions of GDF15, maspin, and NDRG1. Pretreatment of TGF receptor kinase inhibitor not only blocked the activation of TGFβ but also attenuated the activation of GDF15 on the expressions of maspin and NDRG1. The CAPE treatment attenuated the activation of TGFβ on cell proliferation and invasion. Our findings indicate that TGFβ downregulated the expressions of GDF15, maspin, and NDRG1 via TGFβ/Smad signaling. Whereas, CAPE acts as an antagonist on TGFβ/Smad signaling to block the effect of TGFβ on the GDF15 expression and cell proliferation and invasion in bladder carcinoma cells.
Collapse
Affiliation(s)
- Chen-Pang Hou
- Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan;
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
- Department of Healthcare Management, Yuanpei University of Medical Technology, Hsinchu 300, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City 235041, Taiwan;
- TMU Research Center of Urology and Kindey, Department of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Syue-Ting Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Kang-Shuo Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
| | - Hsin-Ching Sung
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan;
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Taoyuan 33302, Taiwan;
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan; (S.-T.C.); (K.-S.C.); (H.-C.S.); (S.-Y.H.)
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 33302, Taiwan
- Correspondence: ; Tel.: +886-3-2118800; Fax: +886-3-2118112
| |
Collapse
|
9
|
Pua LJW, Mai CW, Chung FFL, Khoo ASB, Leong CO, Lim WM, Hii LW. Functional Roles of JNK and p38 MAPK Signaling in Nasopharyngeal Carcinoma. Int J Mol Sci 2022; 23:ijms23031108. [PMID: 35163030 PMCID: PMC8834850 DOI: 10.3390/ijms23031108] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 02/05/2023] Open
Abstract
c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) family members integrate signals that affect proliferation, differentiation, survival, and migration in a cell context- and cell type-specific way. JNK and p38 MAPK activities are found upregulated in nasopharyngeal carcinoma (NPC). Studies have shown that activation of JNK and p38 MAPK signaling can promote NPC oncogenesis by mechanisms within the cancer cells and interactions with the tumor microenvironment. They regulate multiple transcription activities and contribute to tumor-promoting processes, ranging from cell proliferation to apoptosis, inflammation, metastasis, and angiogenesis. Current literature suggests that JNK and p38 MAPK activation may exert pro-tumorigenic functions in NPC, though the underlying mechanisms are not well documented and have yet to be fully explored. Here, we aim to provide a narrative review of JNK and p38 MAPK pathways in human cancers with a primary focus on NPC. We also discuss the potential therapeutic agents that could be used to target JNK and p38 MAPK signaling in NPC, along with perspectives for future works. We aim to inspire future studies further delineating JNK and p38 MAPK signaling in NPC oncogenesis which might offer important insights for better strategies in diagnosis, prognosis, and treatment decision-making in NPC patients.
Collapse
Affiliation(s)
- Lesley Jia Wei Pua
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (L.J.W.P.); (C.-O.L.)
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-W.M.); (A.S.-B.K.)
| | - Chun-Wai Mai
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-W.M.); (A.S.-B.K.)
| | - Felicia Fei-Lei Chung
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway 47500, Malaysia;
| | - Alan Soo-Beng Khoo
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-W.M.); (A.S.-B.K.)
| | - Chee-Onn Leong
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (L.J.W.P.); (C.-O.L.)
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-W.M.); (A.S.-B.K.)
- AGTC Genomics, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Wei-Meng Lim
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-W.M.); (A.S.-B.K.)
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
- Correspondence: (W.-M.L.); (L.-W.H.)
| | - Ling-Wei Hii
- Center for Cancer and Stem Cell Research, Development and Innovation (IRDI), Institute for Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (C.-W.M.); (A.S.-B.K.)
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
- Correspondence: (W.-M.L.); (L.-W.H.)
| |
Collapse
|
10
|
The Antitumor Effect of Caffeic Acid Phenethyl Ester by Downregulating Mucosa-Associated Lymphoid Tissue 1 via AR/p53/NF-κB Signaling in Prostate Carcinoma Cells. Cancers (Basel) 2022; 14:cancers14020274. [PMID: 35053438 PMCID: PMC8773797 DOI: 10.3390/cancers14020274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022] Open
Abstract
Caffeic acid phenethyl ester (CAPE), a honeybee propolis-derived bioactive ingredient, has not been extensively elucidated regarding its effect on prostate cancer and associated mechanisms. The mucosa-associated lymphoid tissue 1 gene (MALT1) modulates NF-κB signal transduction in lymphoma and non-lymphoma cells. We investigated the functions and regulatory mechanisms of CAPE in relation to MALT1 in prostate carcinoma cells. In p53- and androgen receptor (AR)-positive prostate carcinoma cells, CAPE downregulated AR and MALT1 expression but enhanced that of p53, thus decreasing androgen-induced activation of MALT1 and prostate-specific antigen expressions. p53 downregulated the expression of MALT in prostate carcinoma cells through the putative consensus and nonconsensus p53 response elements. CAPE downregulated MALT1 expression and thus inhibited NF-κB activity in p53- and AR-negative prostate carcinoma PC-3 cells, eventually reducing cell proliferation, invasion, and tumor growth in vitro and in vivo. CAPE induced the ERK/JNK/p38/AMPKα1/2 signaling pathways; however, pretreatment with the corresponding inhibitors of MAPK or AMPK1/2 did not inhibit the CAPE effect on MALT1 blocking in PC-3 cells. Our findings verify that CAPE is an effective antitumor agent for human androgen-dependent and -independent prostate carcinoma cells in vitro and in vivo through the inhibition of MALT1 expression via the AR/p53/NF-κB signaling pathways.
Collapse
|
11
|
Hou CP, Tsui KH, Chang KS, Sung HC, Hsu SY, Lin YH, Yang PS, Chen CL, Feng TH, Juang HH. Caffeic acid phenethyl ester inhibits the growth of bladder carcinoma cells by upregulating growth differentiation factor 15. Biomed J 2021; 45:763-775. [PMID: 34662721 DOI: 10.1016/j.bj.2021.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Caffeic acid phenethyl ester (CAPE), a bioactive component of propolis, has beneficial effects on cancer prevention. Growth differentiation factor 15 (GDF15) is an antitumor gene of bladder cancer. Therefore, this study investigated the anti-cancer effect of CAPE on bladder carcinoma cells and related mechanisms. METHODS The expressions of GDF15, N-myc downstream-regulated gene 1 (NDRG1), and maspin, and the activations of ERK, JNK, p38, and AMPKα1/2 in human bladder cells after gene transfection or knockdown were determined by immunoblot, RT-qPCR, and reporter assays. The assays of 5-ethynyl-2'-deoxyuridine (EdU), CyQUANT cell proliferation, and Matrigel invasion, and the xenograft animal study were used to assess the cell proliferation, invasion, and tumorigenesis. RESULTS GDF15 expression in epithelial cells was negatively correlated with neoplasia in vitro. Also, GDF15 exhibits in bladder fibroblasts and smooth muscle cells. CAPE-induced expressions of NDRG1 and maspin decreased cell proliferation and invasion of bladder carcinoma cells in a GDF15-dependent manner in vitro. The xenograft animal study suggesting CAPE attenuated tumor growth in vivo. CAPE increased phosphorylation of ERK, JNK, p38, and AMPKα1/2 to modulate the GDF15 expressions. Pretreatments with ERK, JNK, or p38 inhibitors partially inhibited the CAPE effects on the inductions of GDF15, NDRG1, or maspin. Knockdown of AMPKα1/2 attenuated the CAPE-induced GDF15 expression and cell proliferation in bladder carcinoma cells. CONCLUSIONS Our findings indicate that CAPE is a promising agent for anti-tumor growth in human bladder carcinoma cells via the upregulation of GDF15.
Collapse
Affiliation(s)
- Chen-Pang Hou
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ke-Hung Tsui
- Department of Urology, Shuang Ho Hospital, New Taipei City, Taiwan; Department of Medicine; TMU Research Center of Urology and Kindey, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Hsiang Lin
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Pei-Shan Yang
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chien-Lun Chen
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Tsui-Hsia Feng
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; School of Nursing, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Horng-Heng Juang
- Department of Urology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
12
|
Anticancer Activity of Propolis and Its Compounds. Nutrients 2021; 13:nu13082594. [PMID: 34444754 PMCID: PMC8399583 DOI: 10.3390/nu13082594] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
Propolis is a natural material that honey bees (Apis mellifera) produce from various botanical sources. The therapeutic activity of propolis, including antibacterial, antifungal, and anti-inflammatory effects, have been known since antiquity. Cancer is one of the major burdens of disease worldwide, therefore, numerous studies are being conducted to develop new chemotherapeutic agents and treatments for cancer. Propolis is a rich source of biologically active compounds, which affect numerous signaling pathways regulating crucial cellular processes. The results of the latest research show that propolis can inhibit proliferation, angiogenesis, and metastasis of cancer cells and stimulate apoptosis. Moreover, it may influence the tumor microenvironment and multidrug resistance of cancers. This review briefly summarizes the molecular mechanisms of anticancer activity of propolis and its compounds and highlights the potential benefits of propolis to reduce the side effects of chemotherapy and radiotherapy.
Collapse
|
13
|
Mirzaei S, Gholami MH, Zabolian A, Saleki H, Farahani MV, Hamzehlou S, Far FB, Sharifzadeh SO, Samarghandian S, Khan H, Aref AR, Ashrafizadeh M, Zarrabi A, Sethi G. Caffeic acid and its derivatives as potential modulators of oncogenic molecular pathways: New hope in the fight against cancer. Pharmacol Res 2021; 171:105759. [PMID: 34245864 DOI: 10.1016/j.phrs.2021.105759] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/18/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023]
Abstract
As a phenolic acid compound, caffeic acid (CA) can be isolated from different sources such as tea, wine and coffee. Caffeic acid phenethyl ester (CAPE) is naturally occurring derivative of CA isolated from propolis. This medicinal plant is well-known due to its significant therapeutic impact including its effectiveness as hepatoprotective, neuroprotective and anti-diabetic agent. Among them, anti-tumor activity of CA has attracted much attention, and this potential has been confirmed both in vitro and in vivo. CA can induce apoptosis in cancer cells via enhancing ROS levels and impairing mitochondrial function. Molecular pathways such as PI3K/Akt and AMPK with role in cancer progression, are affected by CA and its derivatives in cancer therapy. CA is advantageous in reducing aggressive behavior of tumors via suppressing metastasis by inhibiting epithelial-to-mesenchymal transition mechanism. Noteworthy, CA and CAPE can promote response of cancer cells to chemotherapy, and sensitize them to chemotherapy-mediated cell death. In order to improve capacity of CA and CAPE in cancer suppression, it has been co-administered with other anti-tumor compounds such as gallic acid and p-coumaric acid. Due to its poor bioavailability, nanocarriers have been developed for enhancing its ability in cancer suppression. These issues have been discussed in the present review with a focus on molecular pathways to pave the way for rapid translation of CA for clinical use.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | | | - Fatemeh Bakhtiari Far
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyed Omid Sharifzadeh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Vice President at Translational Sciences, Xsphera Biosciences Inc. 6 Tide Street, Boston, MA, 02210, USA
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey.
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
14
|
Lv L, Cui H, Ma Z, Liu X, Yang L. Recent progresses in the pharmacological activities of caffeic acid phenethyl ester. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:1327-1339. [PMID: 33492405 DOI: 10.1007/s00210-021-02054-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/14/2021] [Indexed: 12/16/2022]
Abstract
The past decades have seen a growing interest in natural products. Caffeic acid phenethyl ester (CAPE), a flavonoid isolated from honeybee propolis, has shown multiple pharmacological potentials, including anti-cancer, anti-inflammatory, antioxidant, antibacterial, antifungal, and protective effects on nervous systems and multiple organs, since it was found as a potent nuclear factor κB (NF-κB) inhibitor. This review summarizes the advances in these beneficial effects of CAPE, as well as the underlying mechanisms, and proposes that CAPE offers an opportunity for developing therapeutics in multiple diseases. However, clinical trials on CAPE are necessary and encouraged to obtain certain clinically relevant conclusions.
Collapse
Affiliation(s)
- Lili Lv
- Jilin University, Changchun, 130021, China
| | | | - Zhiming Ma
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Xin Liu
- Eye Center, The Second Hospital of Jilin University, Changchun, 130041, China.
| | - Longfei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
15
|
Šuran J, Cepanec I, Mašek T, Radić B, Radić S, Tlak Gajger I, Vlainić J. Propolis Extract and Its Bioactive Compounds-From Traditional to Modern Extraction Technologies. Molecules 2021; 26:molecules26102930. [PMID: 34069165 PMCID: PMC8156449 DOI: 10.3390/molecules26102930] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Propolis is a honeybee product known for its antioxidant, anti-inflammatory, anticancer, and antimicrobial effects. It is rich in bioactive molecules whose content varies depending on the botanical and geographical origin of propolis. These bioactive molecules have been studied individually and as a part of propolis extracts, as they can be used as representative markers for propolis standardization. Here, we compare the pharmacological effects of representative polyphenols and whole propolis extracts. Based on the literature data, polyphenols and extracts act by suppressing similar targets, from pro-inflammatory TNF/NF-κB to the pro-proliferative MAPK/ERK pathway. In addition, they activate similar antioxidant mechanisms of action, like Nrf2-ARE intracellular antioxidant pathway, and they all have antimicrobial activity. These similarities do not imply that we should attribute the action of propolis solely to the most representative compounds. Moreover, its pharmacological effects will depend on the efficacy of these compounds’ extraction. Thus, we also give an overview of different propolis extraction technologies, from traditional to modern ones, which are environmentally friendlier. These technologies belong to an open research area that needs further effective solutions in terms of well-standardized liquid and solid extracts, which would be reliable in their pharmacological effects, environmentally friendly, and sustainable for production.
Collapse
Affiliation(s)
- Jelena Šuran
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia;
| | - Ivica Cepanec
- Director of Research & Development and CTO, Amelia Ltd., Zagorska 28, Bunjani, 10314 Kriz, Croatia;
| | - Tomislav Mašek
- Department of Animal Nutrition and Dietetics, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia;
| | - Božo Radić
- Hedera Ltd., 4. Gardijske Brigade 35, 21311 Split, Croatia; (B.R.); (S.R.)
| | - Saša Radić
- Hedera Ltd., 4. Gardijske Brigade 35, 21311 Split, Croatia; (B.R.); (S.R.)
| | - Ivana Tlak Gajger
- Department for Biology and Pathology of Fish and Bees, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia;
| | - Josipa Vlainić
- Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
- Correspondence:
| |
Collapse
|
16
|
Radiosensitization potential of caffeic acid phenethyl ester and the long non-coding RNAs in response to 60Coγ radiation in mouse hepatoma cells. Radiat Phys Chem Oxf Engl 1993 2021. [DOI: 10.1016/j.radphyschem.2020.109326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
17
|
Liu X, Du Q, Tian C, Tang M, Jiang Y, Wang Y, Cao Y, Wang Z, Wang Z, Yang J, Li Y, Jiao X, Xie P. Discovery of CAPE derivatives as dual EGFR and CSK inhibitors with anticancer activity in a murine model of hepatocellular carcinoma. Bioorg Chem 2020; 107:104536. [PMID: 33342565 DOI: 10.1016/j.bioorg.2020.104536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022]
Abstract
Caffeic acid phenethyl ester (CAPE), a bioactive component extracted from propolis of honeybee hives, can inhibit hepatocellular carcinoma (HCC). In order to explore more stable CAPE derivatives, 25 compounds were designed, synthesized, and pharmacologically assessed in vitro and in vivo as anti-tumor agents in HCC. Compounds 8d, 8f, 8l, 8j, and 8k showed favorable antiproliferative activity than other compounds including CAPE in the HCC cell lines. Based on the result of QTRP (Quantitative Thiol Reactivity Profiling), epidermal growth factor receptor (EGFR) and C-terminal Src kinase (CSK) were supposed to the targets of 8f, which was confirmed by binding mode analysis. Furthermore, compounds 8f, 8l, 8j, 8k, 8g, and 8h showed potent inhibitory effects against both CSK and EGFR than other derivatives in an ADP-Glo™ kinase assay. The representative compound, 8f, potently inhibited various tumor growth in murine model including murine hepatocellular carcinoma H22, meanwhile downregulating the EGFR/AKT pathway and enhancing T cell proliferation through inhibition of CSK. Metabolic stability in vitro suggested 8f and 8k were more stable in mouse plasma than CAPE and susceptible to metabolism in liver microsomes. The overall excellent profile of compound 8f makes it a potential candidate for further preclinical investigation.
Collapse
Affiliation(s)
- Xiaoyu Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qianqian Du
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Caiping Tian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Beijing Institute of Lifeomics, Beijing 102206, China; School of Medicine, Tsinghua University, Beijing, China
| | - Mei Tang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yingjun Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yong Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yang Cao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhenwei Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yan Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Xiaozhen Jiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ping Xie
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
18
|
Chiang KC, Chang KS, Hsu SY, Sung HC, Feng TH, Chao M, Juang HH. Human Heme Oxygenase-1 Induced by Interleukin-6 via JAK/STAT3 Pathways Is a Tumor Suppressor Gene in Hepatoma Cells. Antioxidants (Basel) 2020; 9:antiox9030251. [PMID: 32204510 PMCID: PMC7139670 DOI: 10.3390/antiox9030251] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
Heme oxygenase-1 (HO-1) has several important roles in hepatocytes in terms of anti-inflammation, anti-apoptosis, and antioxidant properties. Interleukin-6 (IL-6) is a pleiotropic cytokine associated with liver regeneration and protection against injury. The aim of this study was to determine the potential crosstalk between HO-1 and IL-6, and to elucidate the signaling pathways involved in the induction of HO-1 by IL-6 in human hepatoma cells. Ectopic overexpression of HO-1 not only attenuated cell proliferation in vitro and in vivo, but also blocked the reactive oxygen species (ROS) induced by H2O2 and the pyocyanin in HepG2 or Hep3B cells. IL-6 expression was negatively regulated by HO-1, while IL-6 induced signal transducer and activator of transcription 3 (STAT3) phosphorylation and HO-1 gene expression in HepG2 cells. The co-transfected HO-1 reporter vector and a protein inhibitor of the activated STAT3 (PIAS3) expression vector blocked the IL-6-induced HO-1 reporter activity. Both interferon γ and interleukin-1β treatments induced STAT1 but not STAT3 phosphorylation, which had no effects on the HO-1 expression. Treatments of AG490 and luteolin blocked the JAK/STAT3 signaling pathways which attenuated IL-6 activation on the HO-1 expression. Our results indicated that HO-1 is the antitumor gene induced by IL-6 through the IL-6/JAK/STAT3 pathways; moreover, a feedback circuit may exist between IL-6 and HO-1 in hepatoma cells.
Collapse
Affiliation(s)
- Kun-Chun Chiang
- Department of General Surgery, Min-Sheng General Hospital, Tao-Yuan 33302, Taiwan;
| | - Kang-Shuo Chang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
- Institute of Medicine Science, College of Medicine, ChSang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
| | - Shu-Yuan Hsu
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
| | - Hsin-Ching Sung
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
| | - Tsui-Hsia Feng
- School of Nursing, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan;
| | - Mei Chao
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Department of Hepato-Gastroenterology, Liver Research Center, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Correspondence: (M.C.); (H.-H.J.); Tel.:+886-3-2118800 (M.C. & H.-H.J.); Fax: +886-3-2118112 (M.C. & H.-H.J.)
| | - Horng-Heng Juang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan; (K.-S.C.); (S.-Y.H.); (H.-C.S.)
- Institute of Medicine Science, College of Medicine, ChSang Gung University, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Department of Urology, Chang Gung Memorial Hospital-Linkou, Kwei-Shan, Tao-Yuan 33302, Taiwan
- Correspondence: (M.C.); (H.-H.J.); Tel.:+886-3-2118800 (M.C. & H.-H.J.); Fax: +886-3-2118112 (M.C. & H.-H.J.)
| |
Collapse
|
19
|
Therapeutic Potential of Plant Phenolic Acids in the Treatment of Cancer. Biomolecules 2020; 10:biom10020221. [PMID: 32028623 PMCID: PMC7072661 DOI: 10.3390/biom10020221] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/26/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
Globally, cancer is the second leading cause of death. Different conventional approaches to treat cancer include chemotherapy or radiotherapy. However, these are usually associated with various deleterious effects and numerous disadvantages in clinical practice. In addition, there are increasing concerns about drug resistance. In the continuous search for safer and more effective treatments, plant-derived natural compounds are of major interest. Plant phenolics are secondary metabolites that have gained importance as potential anti-cancer compounds. Phenolics display a great prospective as cytotoxic anti-cancer agents promoting apoptosis, reducing proliferation, and targeting various aspects of cancer (angiogenesis, growth and differentiation, and metastasis). Phenolic acids are a subclass of plant phenolics, furtherly divided into benzoic and cinnamic acids, that are associated with potent anticancer abilities in various in vitro and in vivo studies. Moreover, the therapeutic activities of phenolic acids are reinforced by their role as epigenetic regulators as well as supporters of adverse events or resistance associated with conventional anticancer therapy. Encapsulation of phyto-substances into nanocarrier systems is a challenging aspect concerning the efficiency of natural substances used in cancer treatment. A summary of phenolic acids and their effectiveness as well as phenolic-associated advances in cancer treatment will be discussed in this review.
Collapse
|
20
|
Liang Y, Feng G, Wu L, Zhong S, Gao X, Tong Y, Cui W, Qin Y, Xu W, Xiao X, Zhang Z, Huang G, Zhou X. Caffeic acid phenethyl ester suppressed growth and metastasis of nasopharyngeal carcinoma cells by inactivating the NF-κB pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1335-1345. [PMID: 31118570 PMCID: PMC6499142 DOI: 10.2147/dddt.s199182] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/23/2019] [Indexed: 12/12/2022]
Abstract
Purpose: Caffeic acid phenethyl ester (CAPE) is the main polyphenol extracted from honeybee propolis, which inhibits the growth of several kinds of tumor. This study aimed to assess the inhibitory effect of CAPE in nasopharyngeal carcinoma (NPC), evaluate the synergistic action of CAPE in radiotherapy sensitivity of NPC cell lines and further elucidate the possible molecular mechanism involved. Materials and methods: CCK-8 assay was used to analyze cell proliferation ability. Colony formation assay was used to evaluate the clonogenic ability and radio-sensitiveness of NPC cells by CAPE treatment. Wound-healing and transwell assay were used to assess the motility of cells. The expression of key molecules of the epithelial–mesenchymal transition (EMT) was determined by western blot analysis and changes in radiation sensitivity were measured by colony-formation assay. cDNA microarray analysis was used to determine differentially expressed genes with and without CAPE treatment, with Gene Ontology enrichment of gene function and KEGG pathways determined. Cell cycle and apoptosis were detected by flow cytometry and western blot analysis. Results: CAPE suppressed the viability of NPC cell lines time- and dose-dependently. It induced apoptosis in NPC cells along with decreased expression of Bcl-XL and increased cleavage of PARP and expression of Bax. G1 phase arrest was induced by CAPE with ower expression of CDK4, CDK6, Rb and p-Rb. The migratory and invasive ability of NPC cells was decreased by the EMT pathway. The irradiation sensitivity of NPC cells was enhanced with CAPE treatment. CAPE specifically inhibited nuclear factor κB (NF-κB) signaling pathway by suppressing p65 subunit translocation from cytoplasm to nucleus. CAPE treatment was synergistic with chemotherapy and radiotherapy. Conclusion: CAPE may inhibit the proliferation and metastasis of NPC cells but enhance radiosensitivity in NPC therapy by inhibiting the NF-κB pathway. CAPE could be a potential therapeutic compound for NPC therapy.
Collapse
Affiliation(s)
- Yushan Liang
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guofei Feng
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Liang Wu
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing, People's Republic of China
| | - Suhua Zhong
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Xiaoyu Gao
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Yan Tong
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Wanmeng Cui
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Yongying Qin
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - WenQing Xu
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Xue Xiao
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Zhe Zhang
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Guangwu Huang
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Department of Otolaryngology Head & Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Xiaoying Zhou
- Key laboratory of High-Incidence-Tumor Prevention & Treatment, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China.,Life Science Institute, Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
21
|
Crowe SM, Kintzios S, Kaltsas G, Palmer CS. A Bioelectronic System to Measure the Glycolytic Metabolism of Activated CD4+ T Cells. BIOSENSORS-BASEL 2019; 9:bios9010010. [PMID: 30634392 PMCID: PMC6468583 DOI: 10.3390/bios9010010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/30/2018] [Accepted: 01/01/2019] [Indexed: 01/02/2023]
Abstract
The evaluation of glucose metabolic activity in immune cells is becoming an increasingly standard task in immunological research. In this study, we described a sensitive, inexpensive, and non-radioactive assay for the direct and rapid measurement of the metabolic activity of CD4+ T cells in culture. A portable, custom-built Cell Culture Metabolite Biosensor device was designed to measure the levels of acidification (a proxy for glycolysis) in cell-free CD4+ T cell culture media. In this assay, ex vivo activated CD4+ T cells were incubated in culture medium and mini electrodes were placed inside the cell free culture filtrates in 96-well plates. Using this technique, the inhibitors of glycolysis were shown to suppress acidification of the cell culture media, a response similar to that observed using a gold standard lactate assay kit. Our findings show that this innovative biosensor technology has potential for applications in metabolic research, where acquisition of sufficient cellular material for ex vivo analyses presents a substantial challenge.
Collapse
Affiliation(s)
- Suzanne M Crowe
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC 3001, Australia.
- Department of Infectious Diseases, Monash University, Melbourne, VIC 3004, Australia.
- Infectious Diseases Department, The Alfred hospital, Melbourne, VIC 3004, Australia.
| | - Spyridon Kintzios
- Laboratory of Cell Technology, School of Food Science, Biotechnology and Development, Agricultural University of Athens, 11855 Athens, Greece.
| | - Grigoris Kaltsas
- Department of Electrical and Electronics Engineering, microSENSES lab, University of West Attika, 12244 Athens, Greece.
| | - Clovis S Palmer
- Life Sciences Discipline, Burnet Institute, Melbourne, VIC 3001, Australia.
- Department of Infectious Diseases, Monash University, Melbourne, VIC 3004, Australia.
| |
Collapse
|