1
|
Ge W, Gao M, Dai Y, Zheng G, Yang L, Zuo W, Tian X. Comprehensive network pharmacology and experimental study to investigate the effect and mechanism of solasonine on breast carcinoma treatment. Cancer Cell Int 2025; 25:49. [PMID: 39962568 PMCID: PMC11834262 DOI: 10.1186/s12935-025-03665-6] [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: 11/05/2024] [Accepted: 01/26/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Ferroptosis is a therapeutic strategy for breast carcinoma (BC). Solasonine (SS) was linked to ferroptosis as a tumor suppressor. However, whether SS could treat BC by activating ferroptosis and its underlying mechanisms has not been reported. METHODS We obtained the intersection of genes targeting SS and BC disease through network pharmacology. Bioinformatics analysis revealed that the intersection genes were primarily enriched in the extracellular signal-regulated kinase 2/mitogen-activated protein kinase (ERK2/MAPK) signaling pathway. The interaction modes of SS with ERK2 and epidermal growth factor receptor (EGFR) were simulated by molecular docking. We further detected the expressions of ERK2 and p-ERK2 in BC patients and the correlation between ERK2/p-ERK2 and ferroptosis. The effects and mechanism of SS on ferroptosis in BC were validated by mutation plasmids construction, immunohistology, wound healing, transwell assay, and western blotting using in vitro and in vivo models. RESULTS ERK2 and p-ERK2 were up-regulated in BC patients, and the ERK2/p-ERK2 ratio was negatively correlated with ferroptosis. Molecular docking indicated that SS could bind to ERK2 and EGFR to inhibit the activity of the ERK2/MAPK pathway. In vitro and in vivo experiments confirmed that SS induced ferroptosis by inhibiting the ERK2/MAPK pathway, inhibiting proliferation, migration, and invasion of BC cells. CONCLUSION SS could inactivate the ERK2/MAPK pathway, thereby inducing ferroptosis and further inhibiting BC cell proliferation, migration, and invasion. This study clarified the potential mechanism of SS in BC and provided a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Wenkai Ge
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Min Gao
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yingqi Dai
- Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Gang Zheng
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Li Yang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wenshu Zuo
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xingsong Tian
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
2
|
Lin TP, Chen PC, Lin CY, Wang BJ, Kuo YY, Yeh CC, Tseng JC, Huo C, Kao CL, Shih LJ, Chen JK, Li CY, Hour TC, Chuu CP. Prostate cancer cells elevate glycolysis and G6PD in response to caffeic acid phenethyl ester-induced growth inhibition. BMC Cancer 2025; 25:95. [PMID: 39819475 PMCID: PMC11737093 DOI: 10.1186/s12885-025-13477-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Caffeic acid phenethyl ester (CAPE) is the main bioactive component of poplar type propolis. We previously reported that treatment with caffeic acid phenethyl ester (CAPE) suppressed the cell proliferation, tumor growth, as well as migration and invasion of prostate cancer (PCa) cells via inhibition of signaling pathways of AKT, c-Myc, Wnt and EGFR. We also demonstrated that combined treatment of CAPE and docetaxel altered the genes involved in glycolysis and tricarboxylic acid (TCA) cycle. We therefore suspect that CAPE treatment may interfere glucose metabolism in PCa cells. METHODS Seahorse Bioenergetics platform was applied to analyzed the extra cellular acidification rate (ECAR) and oxygen consumption rate (OCR) of PCa cells under CAPE treatment. UPLC-MSMS with Multiple Reaction Monitoring (MRM), PCR, and western blot were used to analyze the effects of CAPE on metabolites, genes, and proteins involved in glycolysis, TCA cycle and pentose phosphate pathway in PCa cells. Flow cytometry and ELISA were used to determine the level of reactive oxygen species in PCa cells being treated with CAPE. RESULTS Seahorse Bioenergetics analysis revealed that ECAR, glycolysis, OCR, and ATP production were elevated in C4-2B cells under CAPE treatment. Protein levels of glucose-6-phosphate dehydrogenase (G6PD), phosphogluconate dehydrogenase (PGD), glutaminase (GLS), phospho-AMPK Thr172 as well as abundance of pyruvate, lactate, ribulose-5-phosphate, and sedoheptulose-7-phosphate were increased in CAPE-treated C4-2B cells. ROS level decreased 48 h after treatment with CAPE. Co-treatment of AMPK inhibitor with CAPE exhibited additive growth inhibition on PCa cells. CONCLUSIONS Our study indicated that PCa cells attempted to overcome the CAPE-induced stress by upregulation of glycolysis and G6PD but failed to impede the growth inhibition caused by CAPE. Concurrent treatment of CAPE and inhibitors targeting glycolysis may be effective therapy for advanced PCa.
Collapse
Grants
- TYAFGH-A-111008, TYAFGH-A-112006, TYAFGH-A-113007 Taoyuan Armed Forces General Hospital
- TYAFGH-A-111008, TYAFGH-A-112006, TYAFGH-A-113007 Taoyuan Armed Forces General Hospital
- TYAFGH-A-111008, TYAFGH-A-112006, TYAFGH-A-113007 Taoyuan Armed Forces General Hospital
- NSTC 111-2811-B-400-029; NSTC 112-2320-B-400-008; NSTC 112-2314-B-400-031; NSTC 112-2314-B-037-128 Ministry of Science and Technology, Taiwan
- NSTC 111-2811-B-400-029; NSTC 112-2320-B-400-008; NSTC 112-2314-B-400-031; NSTC 112-2314-B-037-128 Ministry of Science and Technology, Taiwan
- NSTC 111-2811-B-400-029; NSTC 112-2320-B-400-008; NSTC 112-2314-B-400-031; NSTC 112-2314-B-037-128 Ministry of Science and Technology, Taiwan
- NSTC 111-2811-B-400-029; NSTC 112-2320-B-400-008; NSTC 112-2314-B-400-031; NSTC 112-2314-B-037-128 Ministry of Science and Technology, Taiwan
- CS-110-PP-03; CS-111-PP-03; CS-113-SP-04 National Health Research Institutes
Collapse
Affiliation(s)
- Tzu-Ping Lin
- Faculty of Medicine, National Yang Ming Chiao Tung University, Hsinchu City, 30010, Taiwan
- Department of Urology, Taipei Veterans General Hospital, Taipei City, 11217, Taiwan
| | - Pei-Chun Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Ching-Yu Lin
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, Taipei Medical University, Taipei City, 11031, Taiwan
| | - Bi-Juan Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chien-Chih Yeh
- Department of Education and Medical Research, Taoyuan Armed Forces General Hospital, Taoyuan City, 325208, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114202, Taiwan
| | - Jen-Chih Tseng
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chieh Huo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Cheng-Li Kao
- Division of Urology, Departments of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114202, Taiwan
- Division of Urology, Department of Surgery, Taoyuan Armed Forces General Hospital, Taoyuan City, 325208, Taiwan
| | - Li-Jane Shih
- Division of Colon and Rectal Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114202, Taiwan
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei City, 114202, Taiwan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
| | - Tzyh-Chyuan Hour
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung City, 80708, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City, 80708, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan.
- PhD Program for Aging, China Medical University, Taichung City, 40402, Taiwan.
- Biotechnology Center, National Chung Hsing University, Taichung City, 40227, Taiwan.
- Department of Life Sciences, National Central University, Taoyuan City, 32031, Taiwan.
| |
Collapse
|
3
|
Zhang F, Cui X, Yang K, Guo R, Zhu L, Zhao W, Liu Z, Liu B. Activin A inhibits the migration of human lung adenocarcinoma A549 cells induced by EGF. Int Immunopharmacol 2024; 142:113170. [PMID: 39288626 DOI: 10.1016/j.intimp.2024.113170] [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: 03/11/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Activin A, a member of the transforming growth factor β (TGF-β) superfamily, is involved in tumorigenesis and tumor progression. However, it remains unclear whether activin A can affect the migration of lung adenocarcinoma (LUAD) cells. In this study, the results of differentially expressed genes (DEGs) identification revealed that lung adenocarcinoma tissues exhibited lower expression of activin βA mRNA, but higher expression of epidermal growth factor (EGF) and MMP9 mRNA compared to nontumor tissues. Moreover, we found that activin A inhibited human LUAD A549 cell proliferation promoted by EGF. Additionally, EGF induced A549 cell migration in microfluidic device, while activin A attenuated EGF actions. Simultaneously, EGF increased the levels of migration-related proteins, but activin A played the opposite role. Furthermore, the study revealed that EGF upregulated the ratio of p-ERK/ERK in A549 cells, which was weakened by activin A, and A549 cell migration regulated by activin A was not related to calcium signaling. In addition, the inhibitory effect of activin A on EGF-induced A549 cell migration was attenuated by the ERK inhibitor FR180204. These findings demonstrate that activin A effectively hinders the migration of A549 cells induced by EGF through ERK1/2 signaling, suggesting that targeting activin A may hold promise in the treatment of EGF-dependent LUAD growth and metastasis.
Collapse
Affiliation(s)
- Fenglin Zhang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China
| | - Xueling Cui
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China; Key Laboratory of Neuroimmunology and Clinical Immunology in Jilin Province, Jilin Province 130021, China
| | - Ke Yang
- Institute of Applied Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui Province 230031, China
| | - Rui Guo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China
| | - Linjing Zhu
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China
| | - Wei Zhao
- Key Laboratory of Neuroimmunology and Clinical Immunology in Jilin Province, Jilin Province 130021, China; Department of Internal Medicine, The First Hospital of Jilin University, Changchun, Jilin Province 130021, China
| | - Zhonghui Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China; Key Laboratory of Neuroimmunology and Clinical Immunology in Jilin Province, Jilin Province 130021, China
| | - Boyang Liu
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province 130021, China; Department of Scientific Research, Jilin Jianzhu University, Changchun, Jilin Province 130118, China.
| |
Collapse
|
4
|
de Carvalho Braga G, Coiado JV, de Melo VC, Loureiro BB, Bagatini MD. Cutaneous melanoma and purinergic modulation by phenolic compounds. Purinergic Signal 2024; 20:581-593. [PMID: 38498100 PMCID: PMC11555167 DOI: 10.1007/s11302-024-10002-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Cutaneous melanoma is a complex pathology that still has only treatments that lack efficiency and offer many adverse effects. Due to this scenario emerges the need to analyze other possible treatments against this disease, such as the effect of phenolic compounds. These substances have proven antitumor effects, but still have not been fully explored as a form of therapy to combat melanoma. Also, the purinergic receptors, along with its system molecules, take part in the formation of tumors from many pathways, such as the actions of ectoenzymes and receptors activity, especially P2Rs family, and are formed by structures that can be modulated by the phenolic compounds. Therefore, more studies have to be made with the aim of explaining the purinergic system activity in carcinogenesis of cutaneous melanoma and the effects of its modulation by phenolic compound, in order to enable the development of new therapies to combat this aggressive and feared cancer.
Collapse
Affiliation(s)
| | - João Victor Coiado
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | | | | | | |
Collapse
|
5
|
Elagawany M, Abdel Ghany LMA, Ibrahim TS, Alharbi AS, Abdel-Aziz MS, El-labbad EM, Ryad N. Development of certain benzylidene coumarin derivatives as anti-prostate cancer agents targeting EGFR and PI3Kβ kinases. J Enzyme Inhib Med Chem 2024; 39:2311157. [PMID: 38348846 PMCID: PMC10866054 DOI: 10.1080/14756366.2024.2311157] [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: 12/04/2023] [Accepted: 01/22/2024] [Indexed: 02/15/2024] Open
Abstract
Novel coumarin derivatives were synthesised and tested for their cytotoxicity against human cancer cells (PC-3 and MDA-MB-231). Compounds 5, 4b, and 4a possessed potent cytotoxic activity against PC-3 cells with IC50 3.56, 8.99, and 10.22 µM, respectively. Compound 4c displayed cytotoxicity more than erlotinib in the MDA-MB-231 cells with IC50 8.5 µM. Moreover, compound 5 exhibited potent inhibitory activity on EFGR with IC50 0.1812 µM, as well as PI3Kβ inhibitory activity that was twofold higher than LY294002, suggesting that this compound has a dual EGFR and PI3Kβ inhibiting activity. Docking aligns with the in vitro results and sheds light on the molecular mechanisms underlying dual targeting. Furthermore, compound 5 decreased AKT and m-TOR expression in PC-3 cells, showing that it specifically targets these cells via the EGFR/PI3K/Akt/m-TOR signalling pathway. Simultaneously, compound 5 caused cell cycle arrest at S phase and induced activation of both intrinsic and extrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Mohamed Elagawany
- Department of Pharmaceutical Chemistry, Damanhour University, Damanhour, Buhaira, Egypt
| | - Lina M. A. Abdel Ghany
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza, Egypt
| | - Tarek S. Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrhman S. Alharbi
- Department of Chemistry, College of Science and Arts, Shaqra University, Sajir, Shaqra, Saudi Arabia
| | - Mohamed S. Abdel-Aziz
- Microbial Chemistry Department, Biotechnology Research Institute, National Research Centre, Cairo, Egypt
| | - Eman M. El-labbad
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, United Arab Emirates
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, Egypt
| | - Noha Ryad
- Pharmaceutical Organic Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th of October City, Giza, Egypt
| |
Collapse
|
6
|
Ali E, Helmy MW, Radwan EH, Abdul Aziz KK, El-Wahed AAA, El-Samad LM, El Wakil A. Evaluation of the cytotoxic activity of chemically characterized propolis originating from different geographic regions and vitamin D co-supplementation against human ovarian cancer cells. J Ovarian Res 2024; 17:181. [PMID: 39244585 PMCID: PMC11380329 DOI: 10.1186/s13048-024-01500-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 08/14/2024] [Indexed: 09/09/2024] Open
Abstract
Ovarian cancer is the second most common and lethal gynecologic malignancy. Among natural product-based therapy, the honeybee products, particularly propolis, serve a valuable source contributing directly to human nutrition and health.In the present study, we determined the chemical composition of different types of propolis originating from Egypt, Germany and France using liquid chromatography-tandem mass spectrometry. The compounds identified belong to different metabolite classes, including flavonoids, cinnamic acid, chalcones, terpenoids, phenolic lipids, stilbenes, phenolic compounds, carbohydrates, vitamins, coumarins, polyprenylated benzophenone, benzoic acids, fatty acid methyl ester, and coumaric acid, and their derivatives. The most active extract is from France then Egypt and Germany.Afterwards, we treated the human ovarian cancer cells, OVCAR4, with different concentrations (1-400 μg/mL) of variable propolis types supplemented or not with vitamin D (0.0015-0.15 μg/mL) in order to evaluate the efficacy and the cytotoxic activities of our local P as compared to other types collected from different geographic regions. Importantly, the combinatorial treatment of OVCAR4 cancer cells with propolis and vitamin D in the same concentration ranges resulted in enhanced cell viability inhibition. Furthermore, such co-supplementation with vitamin D inhibits predominately the proliferative activity of cell population with the French propolis type as manifested by Ki67 expression, while it reduces considerably its expression, particularly with the German type, followed by the Egyptian one.Nowadays, scientists are interested by natural products which have risen to the forefront of drug discovery. Chemically characterized propolis showing cell viability inhibition and antiproliferative potential seems a valuable extract for further consideration as anti-carcinogenic agent.
Collapse
Affiliation(s)
- Eman Ali
- Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Maged W Helmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
- Department of Pharmacology and Toxicology, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria, Egypt
| | - Eman H Radwan
- Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | | | - Aida A Abd El-Wahed
- Department of Bee Research, Plant Protection Research Institute, Agricultural Research Centre, Giza, 12627, Egypt
| | - Lamia M El-Samad
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Abeer El Wakil
- Department of Biological and Geological Sciences, Faculty of Education, Alexandria University, Alexandria, 21526, Egypt.
| |
Collapse
|
7
|
Kuo YY, Huo C, Li CY, Chuu CP. Caffeic acid phenethyl ester suppresses the expression of androgen receptor variant 7 via inhibition of CDK1 and AKT. Cancer Gene Ther 2024; 31:807-815. [PMID: 38480977 DOI: 10.1038/s41417-024-00753-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 06/23/2024]
Abstract
Androgen receptor (AR) splice variant 7 (AR-V7) is capable to enter nucleus and activate downstream signaling without ligand. AR-V7 assists the tumor growth, cancer metastasis, cancer stemness, and the evolvement of therapy-resistant prostate cancer (PCa). We discovered that caffeic acid phenethyl ester (CAPE) can repress the expression and downstream signaling of AR-V7 in PCa cells. CAPE blocked the gene transcription, nuclear localization, and protein abundance of AR-V7. CAPE inhibited the expression of U2AF65, SF2 and hnRNPF, which were splicing factors for AR-V7 intron. Additionally, CAPE decreased protein stability of AR-V7 and enhanced the proteosome-degradation of AR-V7. We observed that CDK1 and AKT regulated the expression and stability of AR-V7 via phosphorylation of Ser81 and Ser213, respectively. CAPE decreased the expression of CDK1 and AKT. Overexpression of CDK1 restored the abundance of AR-V7 in CAPE-treated PCa cells. Overexpression of AR-V7, AKT or CDK1 rescued the proliferation of PCa cells under CAPE treatment. Intraperitoneal injection of 10 mg/kg CAPE retarded the growth of 22Rv1 xenografts in nude mice and suppressed the protein levels of AR-V7, CDK1 and AKT in 22Rv1 xenografts. Our study provided the rationale of applying CAPE for inhibition of AR-V7 in prostate tumors.
Collapse
Affiliation(s)
- Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Chieh Huo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli County, Taiwan.
- PhD Program for Aging and Graduate Institute of Basic Medical Science, China Medical University, Taichung City, Taiwan.
- Biotechnology Center, National Chung Hsing University, Taichung City, Taiwan.
- Department of Life Sciences, National Central University, Taoyuan City, Taiwan.
| |
Collapse
|
8
|
Zhang Y, Deng Q, Hong H, Qian Z, Wan B, Xia M. Caffeic acid phenethyl ester inhibits neuro-inflammation and oxidative stress following spinal cord injury by mitigating mitochondrial dysfunction via the SIRT1/PGC1α/DRP1 signaling pathway. J Transl Med 2024; 22:304. [PMID: 38528569 PMCID: PMC10962082 DOI: 10.1186/s12967-024-05089-8] [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: 11/13/2023] [Accepted: 03/13/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND The treatment of spinal cord injury (SCI) has always been a significant research focus of clinical neuroscience, with inhibition of microglia-mediated neuro-inflammation as well as oxidative stress key to successful SCI patient treatment. Caffeic acid phenethyl ester (CAPE), a compound extracted from propolis, has both anti-inflammatory and anti-oxidative effects, but its SCI therapeutic effects have rarely been reported. METHODS We constructed a mouse spinal cord contusion model and administered CAPE intraperitoneally for 7 consecutive days after injury, and methylprednisolone (MP) was used as a positive control. Hematoxylin-eosin, Nissl, and Luxol Fast Blue staining were used to assess the effect of CAPE on the structures of nervous tissue after SCI. Basso Mouse Scale scores and footprint analysis were used to explore the effect of CAPE on the recovery of motor function by SCI mice. Western blot analysis and immunofluorescence staining assessed levels of inflammatory mediators and oxidative stress-related proteins both in vivo and in vitro after CAPE treatment. Further, reactive oxygen species (ROS) within the cytoplasm were detected using an ROS kit. Changes in mitochondrial membrane potential after CAPE treatment were detected with 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-imidacarbocyanine iodide. Mechanistically, western blot analysis and immunofluorescence staining were used to examine the effect of CAPE on the SIRT1/PGC1α/DRP1 signaling pathway. RESULTS CAPE-treated SCI mice showed less neuronal tissue loss, more neuronal survival, and reduced demyelination. Interestingly, SCI mice treated with CAPE showed better recovery of motor function. CAPE treatment reduced the expression of inflammatory and oxidative mediators, including iNOS, COX-2, TNF-α, IL-1β, 1L-6, NOX-2, and NOX-4, as well as the positive control MP both in vitro and in vivo. In addition, molecular docking experiments showed that CAPE had a high affinity for SIRT1, and that CAPE treatment significantly activated SIRT1 and PGC1α, with down-regulation of DRP1. Further, CAPE treatment significantly reduced the level of ROS in cellular cytoplasm and increased the mitochondrial membrane potential, which improved normal mitochondrial function. After administering the SIRT1 inhibitor nicotinamide, the effect of CAPE on neuro-inflammation and oxidative stress was reversed.On the contrary, SIRT1 agonist SRT2183 further enhanced the anti-inflammatory and antioxidant effects of CAPE, indicating that the anti-inflammatory and anti-oxidative stress effects of CAPE after SCI were dependent on SIRT1. CONCLUSION CAPE inhibits microglia-mediated neuro-inflammation and oxidative stress and supports mitochondrial function by regulating the SIRT1/PGC1α/DRP1 signaling pathway after SCI. These effects demonstrate that CAPE reduces nerve tissue damage. Therefore, CAPE is a potential drug for the treatment of SCI through production of anti-inflammatory and anti-oxidative stress effects.
Collapse
Affiliation(s)
- Yanan Zhang
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Qian Deng
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
- Postgraduate School, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongxiang Hong
- Department of Spine Surgery, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, Research Institute for Spine and Spinal Cord Disease of Nantong University, No. 666, ShengLi Road, Chongchuan District, Nantong, Jiangsu, China
| | - Zhanyang Qian
- Department of Orthopedics, Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China.
| | - Bowen Wan
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, China.
| | - Mingjie Xia
- Department of Spine Surgery, Nantong First People's Hospital, The Second Affiliated Hospital of Nantong University, Research Institute for Spine and Spinal Cord Disease of Nantong University, No. 666, ShengLi Road, Chongchuan District, Nantong, Jiangsu, China.
| |
Collapse
|
9
|
Abdulrahman FG, Abulkhair HS, Saeed HSE, El-Dydamony NM, Husseiny EM. Design, synthesis, and mechanistic insight of novel imidazolones as potential EGFR inhibitors and apoptosis inducers. Bioorg Chem 2024; 144:107105. [PMID: 38219482 DOI: 10.1016/j.bioorg.2024.107105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
As regards to the structural analysis and optimization of diverse potential EGFR inhibitors, two series of imidazolyl-2-cyanoprop-2-enimidothioates and ethyl imidazolylthiomethylacrylates were designed and constructed as potential EGFR suppressors. The cytotoxic effect of the prepared derivatives was assessed toward hepatic, breast, and prostate cancerous cells (Hep-G2, MCF-7, and PC-3). Three derivatives 3d, 3e, and 3f presented potent antiproliferative activity and selectivity against the examined tumor cells showing IC50 values at low micromolar levels. Hence, successive biological assays were applied to determine the probable mechanism of action of the new compounds. They exhibited significant EGFR suppression with an IC50 range of 0.137-0.507 µM. The most effective EGFR inhibitor 3f arrested the MCF-7 cell cycle at the S phase by inducing the apoptotic pathway that was confirmed via increasing the expression of Caspases 8, 9, and Bax, which are associated with Bcl-2 decline. Additionally, molecular docking displayed a distinctive interaction between 3f and EGFR binding pocket. Overall, this work introduces some novel imidazolyl-2-cyanoprop-2-enimidothioates and ethyl imidazolylthiomethylacrylates as potential cytotoxic and EGFR inhibitors that deserve further research in tumor therapy.
Collapse
Affiliation(s)
- Fatma G Abdulrahman
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Hamada S Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11884, Cairo, Egypt; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Horus University-Egypt, International Coastal Road, New Damietta 34518, Egypt.
| | - Hoda S El Saeed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt
| | - Nehad M El-Dydamony
- Pharmaceutical Chemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6(th) of October City, Egypt
| | - Ebtehal M Husseiny
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls), Al-Azhar University, Nasr City 11754, Cairo, Egypt.
| |
Collapse
|
10
|
Bahrami A, Farasat A, Zolghadr L, Sabaghi Y, PourFarzad F, Gheibi N. The anticancer impacts of free and liposomal caffeic acid phenethyl ester (CAPE) on melanoma cell line (A375). Cell Biochem Funct 2024; 42:e3900. [PMID: 38111127 DOI: 10.1002/cbf.3900] [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: 06/20/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023]
Abstract
The deadliest type of skin cancer, malignant melanoma, is also the reason for the majority of skin cancer-related deaths. The objective of this article was to investigate the efficiency of free caffeic acid phenethyl ester (CAPE) and liposomal CAPE in inducing apoptosis in melanoma cells (A375) in in vitro. CAPE was loaded into liposomes made up of hydrogenated soybean phosphatidylcholine, cholesterol, and 1,2-distearoyl-sn-glycero-3 phosphoethanolamine-N-[methoxy (polyethylene glycol)-2000], and their physicochemical properties were assessed. (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) test was performed for comparing the cytotoxicity of free CAPE and liposomal CAPE at dosages of 10, 15, 25, 50, 75 and the highest dose of 100 μg/mL for period of 24 and 48 h on A375 cell line to calculate IC50. Apoptosis and necrosis were evaluated in A375 melanoma cancer cells using flow cytometry. Atomic force microscopy was utilized to determine the nanomechanical attributes of the membrane structure of A375 cells. To determine whether there were any effects on apoptosis, the expression of PI3K/AKT1 and BAX/BCL2 genes was analyzed using the real-time polymerase chain reaction technique. According to our results, the maximum amount of drug release from nanoliposomes was determined to be 91% and the encapsulation efficiency of CAPE in liposomes was 85.24%. Also, the release of free CAPE was assessed to be 97%. Compared with liposomal CAPE, free CAPE showed a greater effect on reducing the cancer cell survival after 24 and 48 h. Therefore, IC50 values of A375 cells treated with free and liposomal CAPE were calculated as 47.34 and 63.39 μg/mL for 24 h. After 48 h of incubation of A375 cells with free and liposomal CAPE, IC50 values were determined as 30.55 and 44.83 μg/mL, respectively. The flow cytometry analysis revealed that the apoptosis induced in A375 cancer cells was greater when treated with free CAPE than when treated with liposomal CAPE. The highest nanomechanical changes in the amount of cell adhesion forces, and elastic modulus value were seen in free CAPE. Subsequently, the greatest decrease in PI3K/AKT1 gene expression ratio occurred in free CAPE.
Collapse
Affiliation(s)
- Azita Bahrami
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Alireza Farasat
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Leila Zolghadr
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin, Iran
| | - Yalda Sabaghi
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Farnaz PourFarzad
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Nematollah Gheibi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
11
|
Hasan N, Nadaf A, Imran M, Jiba U, Sheikh A, Almalki WH, Almujri SS, Mohammed YH, Kesharwani P, Ahmad FJ. Skin cancer: understanding the journey of transformation from conventional to advanced treatment approaches. Mol Cancer 2023; 22:168. [PMID: 37803407 PMCID: PMC10559482 DOI: 10.1186/s12943-023-01854-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/30/2023] [Indexed: 10/08/2023] Open
Abstract
Skin cancer is a global threat to the healthcare system and is estimated to incline tremendously in the next 20 years, if not diagnosed at an early stage. Even though it is curable at an early stage, novel drug identification, clinical success, and drug resistance is another major challenge. To bridge the gap and bring effective treatment, it is important to understand the etiology of skin carcinoma, the mechanism of cell proliferation, factors affecting cell growth, and the mechanism of drug resistance. The current article focusses on understanding the structural diversity of skin cancers, treatments available till date including phytocompounds, chemotherapy, radiotherapy, photothermal therapy, surgery, combination therapy, molecular targets associated with cancer growth and metastasis, and special emphasis on nanotechnology-based approaches for downregulating the deleterious disease. A detailed analysis with respect to types of nanoparticles and their scope in overcoming multidrug resistance as well as associated clinical trials has been discussed.
Collapse
Affiliation(s)
- Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Arif Nadaf
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Imran
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, 4102, Australia
| | - Umme Jiba
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, 24381, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, 61421, Asir-Abha, Saudi Arabia
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Kuthambakkam, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
12
|
Jameel M, Fatma H, Nadtochii LA, Siddique HR. Molecular Insight into Prostate Cancer: Preventive Role of Selective Bioactive Molecules. Life (Basel) 2023; 13:1976. [PMID: 37895357 PMCID: PMC10608662 DOI: 10.3390/life13101976] [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: 08/21/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Prostate cancer (CaP) is one of the most prevalent male malignancies, accounting for a considerable number of annual mortalities. However, the prompt identification of early-stage CaP often faces delays due to diverse factors, including socioeconomic inequalities. The androgen receptor (AR), in conjunction with various other signaling pathways, exerts a central influence on the genesis, progression, and metastasis of CaP, with androgen deprivation therapy (ADT) serving as the primary therapeutic strategy. Therapeutic modalities encompassing surgery, chemotherapy, hormonal intervention, and radiotherapy have been formulated for addressing early and metastatic CaP. Nonetheless, the heterogeneous tumor microenvironment frequently triggers the activation of signaling pathways, culminating in the emergence of chemoresistance, an aspect to which cancer stem cells (CSCs) notably contribute. Phytochemicals emerge as reservoirs of bioactive agents conferring manifold advantages against human morbidity. Several of these phytochemicals demonstrate potential chemoprotective and chemosensitizing properties against CaP, with selectivity exhibited towards malignant cells while sparing their normal counterparts. In this context, the present review aims to elucidate the intricate molecular underpinnings associated with metastatic CaP development and the acquisition of chemoresistance. Moreover, the contributions of phytochemicals to ameliorating CaP initiation, progression, and chemoresistance are also discussed.
Collapse
Affiliation(s)
- Mohd Jameel
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| | - Homa Fatma
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| | - Liudmila A. Nadtochii
- Department of Microbiology, Saint Petersburg State Chemical & Pharmaceutical University, 197022 Saint Petersburg, Russia
| | - Hifzur R. Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh 202002, India (H.F.)
| |
Collapse
|