1
|
Zaidi AK, Kumar A, Kumar R, Singh J, Yadav S, Sonkar AB, Kumar D, Shrivastava NK, Ansari MN, Saeedan AS, Kaithwas G. Repurposing brucine as a chemopreventive agent in mammary gland carcinoma: Regulating lactate transport through MCT-4. Toxicol Rep 2025; 14:101902. [PMID: 39897398 PMCID: PMC11787605 DOI: 10.1016/j.toxrep.2025.101902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/17/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
In the present study, we aim to identify a potential drug candidate that targets the Monocarboxylate Transporter-4 (MCT-4) protein. Syrosingopine (SRY) is a well-established inhibitor of lactate transport through MCT-4. We screened 2,11,192 potential leads through ZINC database, which were atleast 50 % structurally similar with SYR. After in-depth analysis, 900 molecules were shortlisted based on Lipinski's rule, optimal molecular weight, binding energy, hydrogen bonding, and ADMET (absorption, distribution, metabolism, excretion, and toxicity) properties that render them viable MCT-4 inhibitors. The outcome underscored Brucine (BRU) as the most promising lead molecule within a cohort of ten potential compounds. BRU is a monoterpenoid indole alkaloid and is used in the regulation of high blood pressure and other comparatively benign cardiac ailments. As such, no reports is available emphasizing the efficacy of BRU on lactate transport or mammary gland carcinoma. BRU demonstrated strong affinity for the MCT-4 transporter's catalytic domain, forming significant hydrophobic and polar interactions with essential amino acids at the binding site. BRU demonstrated significant cytotoxicity and increased the extracellular lactate levels in MCF-7 cells. The findings strongly encouraged BRU's effectiveness, offering promising paths for subsequent investigations.
Collapse
Affiliation(s)
- Asma Khatoon Zaidi
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Anurag Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Rohit Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Jyoti Singh
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Sneha Yadav
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Archana Bharti Sonkar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Dharmendra Kumar
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Neeraj Kumar Shrivastava
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Mohd Nazam Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Abdulaziz S. Saeedan
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Gaurav Kaithwas
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| |
Collapse
|
2
|
Lalmuansangi C, Lalfakawmi, Nghakliana F, Sailo H, Tochhawng L, Trivedi AK, Kharat KR, Vellingiri B, Kumar NS, Siama Z. Anticancer activity of Stemona tuberosa (wild asparagus) against type-II human lung adenocarcinoma (A549) cells and identification of SRC inhibitor using integrated network pharmacology and molecular dynamic simulation. Discov Oncol 2025; 16:429. [PMID: 40159570 PMCID: PMC11955439 DOI: 10.1007/s12672-025-02138-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Stemona tuberosa is widely recognized for its traditional applications as an anti-cancer agent. This study aimed to assess the anti-cancer properties of S. tuberosa in human lung adenocarcinoma A549 cells. Among the various solvent extracts of S. tuberosa, the methanolic extract showed the highest toxicity against A549 cells. The S. tuberosa extract elicited cytotoxic effects and suppressed colony formation in A549 cells in a dose-dependent manner. S. tuberosa activity was further supported by AO/EtBr staining, increased caspase 3/6 activity, upregulation of pro-apoptotic genes, DNA damage, and elevated lipid peroxidation, with decreasing antioxidant levels. LC-MS analysis identified 80 predominant secondary metabolites in the methanolic extracts of S. tuberosa. A network pharmacology study identified SRC as the primary target of compounds identified from S. tuberosa. SRC protein is crucial for advancing lung cancer because of its function in cell proliferation, survival, and metastasis. Among the various compounds identified from S. tuberosa extract, 4-Azatricyclo [4.3.1.13,8] undecan-5-one (ADE) (- 10.88 kcal/mol) and Dihydro-normorphine, 3-desoxy- (DNY) (- 10.83 kcal/mol) exhibited notable binding affinities for SRC. Further analysis using molecular dynamics simulations (100 ns) validated the stability of SRC-ligand complexes, with RMSD of 1.8 and 2.2 Å for ADE and DNY, respectively, alongside the establishment of essential hydrogen bonds with pivotal residues, including ASP408, ALA403, and THR438. Finally, gmx._MMPBSA showed favourable ΔGbind values for ADE (- 15.06 ± 0.11 kcal/mol) and DNY (- 15.66 ± 0.25 kcal/mol), which highlights the significant potential of ADE and DNY as effective SRC inhibitors, suggesting S. tuberosa as a novel candidate for cancer therapy.
Collapse
Affiliation(s)
- C Lalmuansangi
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Lalfakawmi
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Fanai Nghakliana
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Hmingremhlua Sailo
- Department of Botany, Mizoram University (a Central University), Aizawl, 796004, India
| | - Lalchhandami Tochhawng
- Mizoram Science, Technology and Innovation Council (MISTIC), Aizawl, 796001, Mizoram, India
| | - Amit Kumar Trivedi
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Kiran R Kharat
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India
| | - Balachandar Vellingiri
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Ghudda, India
| | | | - Zothan Siama
- Department of Zoology, Mizoram University (a Central University), Aizawl, 796004, India.
| |
Collapse
|
3
|
Gunathilaka TL, Kumarasinghe HS, Bandaranayake UE, Athapaththu M, Samarakoon KW, Ranasinghe P, Peiris LDC. Integration of In Vitro and In-Silico Analysis of Gracilaria edulis on Anti-Cancer Potential and Apoptotic Signaling Pathway Activity. Cell Biochem Biophys 2025:10.1007/s12013-025-01685-7. [PMID: 39939528 DOI: 10.1007/s12013-025-01685-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2025] [Indexed: 02/14/2025]
Abstract
Breast cancer, the most common malignancy in females, and rhabdomyosarcoma (RMS), the most prevalent soft tissue sarcoma in children, remain significant clinical challenges. This study evaluated the anticancer potential and apoptotic signaling pathways of Gracilaria edulis extracts and identified their mechanisms of action against RMS and breast adenocarcinoma (MCF-7) cell lines. Cytotoxicity was assessed using MTT assays, while apoptotic potential was evaluated through phase contrast and fluorescence microscopy, caspase 3/7 activity, DNA fragmentation, and gene expression analysis of apoptosis regulatory genes. In silico analysis was also performed to examine the molecular interactions of bioactive compounds present in Gracilaria edulis with cancer-related proteins involved in apoptotic signaling. The methanol extract was fractionated into hexane, chloroform, and ethyl acetate, with the hexane fraction demonstrating the strongest cytotoxicity (IC50RMS: 32.52 ± 2.15 μg/mL; IC50MCF-7:29.84 ± 0.65 μg/mL) in MTT assays. Apoptotic features, including chromatin condensation, membrane blebbing, cellular shrinkage, and DNA fragmentation, were observed, particularly in RMS cells. The hexane fraction significantly activated caspase 3/7 in RMS cells, while lower activation was noted in MCF-7 cells, possibly due to the partial deletion of the CASP-3 gene. Real-time PCR analysis revealed differential gene expression, with p21 showing dominant upregulation in RMS cells and p53 being more prominently expressed in MCF-7 cells. These findings reflect their distinct roles in apoptotic signaling pathways. A significant increase in the Bax/Bcl-2 ratio in RMS cells (8.45) and MCF-7 cells (29.69) indicated a pro-apoptotic shift. GC-MS analysis identified key bioactive compounds, including 9-octadecenoic acid methyl ester, hexadecenoic acid methyl ester, and 1,2-benzenedicarboxylic acid mono(2-ethylhexyl) ester. In silico docking revealed that 1,2-benzenedicarboxylic acid mono(2-ethylhexyl) ester demonstrated the most promising binding interactions, particularly with BCL-2, while 9-octadecenoic acid methyl ester exhibited weaker binding affinities across all targets (p53, p21, and BCL-2), suggesting limited therapeutic relevance without structural optimization. However, the hexane fraction of G. edulis and its bioactive compounds remain promising as potential anticancer agents, warranting further in vitro and in vivo validation and molecular optimization.
Collapse
Affiliation(s)
- Thilina Lakmini Gunathilaka
- Department of Zoology, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka.
- Department of Basic Science and Social Science for Nursing, Faculty of Nursing, University of Colombo, Colombo, Sri Lanka.
| | - Hiruni S Kumarasinghe
- Department of Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si, South Korea
| | - U E Bandaranayake
- ECU Sri Lanka campus, Sri Jayewardenepura Mawatha, Rajagiriya, Sri Lanka
| | | | - Kalpa W Samarakoon
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defence University, Ratmalana, 10390, Sri Lanka
| | | | - L Dinithi C Peiris
- Department of Zoology/Genetics & Molecular Biology Unit, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka.
| |
Collapse
|
4
|
Siddique TA, Rafi KJ, Akter S, Bithy FY, Aktar R, Khan AN, Majid M, Sultana F, Saha S, Ahmed AMA, Rahman A. Nutritional, Antibacterial and Thrombolytic Prospects of Freshwater Paludomas conica Protein Hydrolysate and Its Anti-Inflammatory Potential in LPS-Induced RAW 264.7 Macrophage Cells. Food Sci Nutr 2025; 13:e4711. [PMID: 39803236 PMCID: PMC11717026 DOI: 10.1002/fsn3.4711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Chronic inflammation and heme-iron overload can result from bacterial hemolysis. Along with the synthetic drugs, numerous traditional and functional food approaches are equally trialed to eradicate the problem. As a prospective new source of dietary protein hydrolysates, freshwater mollusks (Paludomas conica) have recently drawn huge interest from researchers. In this research, protein hydrolysate (PhPC) of Paludomas conica, prepared by the enzyme digestion method, was analyzed for proximate nutritional and minerals contents and deciphered its suppressive effects on inflammatory gene expression in LPS-stimulated RAW264.7 macrophage cells. The inhibitory action of protein denaturation is also unfolded with established in vitro and in vivo models. Anti-hemolytic, antibacterial, and thrombolytic effects of PhPC were respectively assessed by H2O2-induced hemolysis of RBCs, the disc diffusion method, and the clot lysis method. The proximate nutritional and mineral contents of PhPC revealed it to be an enriched source of nutrients, crude protein, carbohydrates, Calcium, and Magnesium. Heavy metals were found to be within the prescribed limit. The PhPC suppressed the expression of inflammatory genes, including COX-2, iNOS, IL-6, TNF-α, and IL-1, multifold in LPS-stimulated RAW264.7 macrophages. The inhibition concentrations (IC50) of PhPC in the bovine serum albumin denaturation inhibition test and membrane stabilization tests were 431.39 and 285.25 μg/mL, respectively. The PhPC was discerned to be active against Shigella flexneri, Pseudomonas aeruginosa, and Shigella dysenteriae; its maximum thrombolytic effect was displayed to be 23.72% ± 2.71%. The findings demonstrate that the nutritionally enriched PhPC could be affirmed as an exciting invertebrate anti-inflammatory agent Extending other biological functions needs to be further characterized with its pure protein or protein products.
Collapse
Affiliation(s)
- Tanvir Ahmed Siddique
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Khalid Juhani Rafi
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Sumaiya Akter
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Farhana Yesmin Bithy
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Rasheda Aktar
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Asif Nadim Khan
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Mumtahina Majid
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Farjana Sultana
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - Srabonti Saha
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| | - A. M. Abu Ahmed
- Department of Genetic Engineering and BiotechnologyUniversity of ChittagongChittagongBangladesh
| | - Atiar Rahman
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChittagongBangladesh
| |
Collapse
|
5
|
Gautam P, Ciuta I, Teif VB, Sinha SK. Predicting p53-dependent cell transitions from thermodynamic models. J Chem Phys 2024; 161:135101. [PMID: 39356070 DOI: 10.1063/5.0225166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
A cell's fate involves transitions among its various states, each defined by a distinct gene expression profile governed by the topology of gene regulatory networks, which are affected by 3D genome organization. Here, we develop thermodynamic models to determine the fate of a malignant cell as governed by the tumor suppressor p53 signaling network, taking into account long-range chromatin interactions in the mean-field approximation. The tumor suppressor p53 responds to stress by selectively triggering one of the potential transcription programs that influence many layers of cell signaling. These range from p53 phosphorylation to modulation of its DNA binding affinity, phase separation phenomena, and internal connectivity among cell fate genes. We use the minimum free energy of the system as a fundamental property of biological networks that influences the connection between the gene network topology and the state of the cell. We constructed models based on network topology and equilibrium thermodynamics. Our modeling shows that the binding of phosphorylated p53 to promoters of target genes can have properties of a first order phase transition. We apply our model to cancer cell lines ranging from breast cancer (MCF-7), colon cancer (HCT116), and leukemia (K562), with each one characterized by a specific network topology that determines the cell fate. Our results clarify the biological relevance of these mechanisms and suggest that they represent flexible network designs for switching between developmental decisions.
Collapse
Affiliation(s)
- Pankaj Gautam
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| | - Isabella Ciuta
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Vladimir B Teif
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, United Kingdom
| | - Sudipta Kumar Sinha
- Theoretical and Computational Biophysical Chemistry Group, Department of Chemistry, Indian Institute of Technology Ropar, Rupnagar, Punjab 140001, India
| |
Collapse
|
6
|
Kaur S, Mendonca P, Soliman KFA. The Anticancer Effects and Therapeutic Potential of Kaempferol in Triple-Negative Breast Cancer. Nutrients 2024; 16:2392. [PMID: 39125273 PMCID: PMC11314279 DOI: 10.3390/nu16152392] [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: 06/15/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Breast cancer is the second-leading cause of cancer death among women in the United States. Triple-negative breast cancer (TNBC), a subtype of breast cancer, is an aggressive phenotype that lacks estrogen (ER), progesterone (PR), and human epidermal growth (HER-2) receptors, which is challenging to treat with standardized hormonal therapy. Kaempferol is a natural flavonoid with antioxidant, anti-inflammatory, neuroprotective, and anticancer effects. Besides anti-tumorigenic, antiproliferative, and apoptotic effects, kaempferol protects non-cancerous cells. Kaempferol showed anti-breast cancer effects by inducing DNA damage and increasing caspase 3, caspase 9, and pAMT expression, modifying ROS production by Nrf2 modulation, inducing apoptosis by increasing cleaved PARP and Bax and downregulating Bcl-2 expression, inducing cell cycle arrest at the G2/M phase; inhibiting immune evasion by modulating the JAK-STAT3 pathway; and inhibiting the angiogenic and metastatic potential of tumors by downregulating MMP-3 and MMP-9 levels. Kaempferol holds promise for boosting the efficacy of anticancer agents, complementing their effects, or reversing developed chemoresistance. Exploring novel TNBC molecular targets with kaempferol could elucidate its mechanisms and identify strategies to overcome limitations for clinical application. This review summarizes the latest research on kaempferol's potential as an anti-TNBC agent, highlighting promising but underexplored molecular pathways and delivery challenges that warrant further investigation to achieve successful clinical translation.
Collapse
Affiliation(s)
- Sukhmandeep Kaur
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA;
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA;
| |
Collapse
|
7
|
Bel’skaya LV, Dyachenko EI. Oxidative Stress in Breast Cancer: A Biochemical Map of Reactive Oxygen Species Production. Curr Issues Mol Biol 2024; 46:4646-4687. [PMID: 38785550 PMCID: PMC11120394 DOI: 10.3390/cimb46050282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024] Open
Abstract
This review systematizes information about the metabolic features of breast cancer directly related to oxidative stress. It has been shown those redox changes occur at all levels and affect many regulatory systems in the human body. The features of the biochemical processes occurring in breast cancer are described, ranging from nonspecific, at first glance, and strictly biochemical to hormone-induced reactions, genetic and epigenetic regulation, which allows for a broader and deeper understanding of the principles of oncogenesis, as well as maintaining the viability of cancer cells in the mammary gland. Specific pathways of the activation of oxidative stress have been studied as a response to the overproduction of stress hormones and estrogens, and specific ways to reduce its negative impact have been described. The diversity of participants that trigger redox reactions from different sides is considered more fully: glycolytic activity in breast cancer, and the nature of consumption of amino acids and metals. The role of metals in oxidative stress is discussed in detail. They can act as both co-factors and direct participants in oxidative stress, since they are either a trigger mechanism for lipid peroxidation or capable of activating signaling pathways that affect tumorigenesis. Special attention has been paid to the genetic and epigenetic regulation of breast tumors. A complex cascade of mechanisms of epigenetic regulation is explained, which made it possible to reconsider the existing opinion about the triggers and pathways for launching the oncological process, the survival of cancer cells and their ability to localize.
Collapse
Affiliation(s)
- Lyudmila V. Bel’skaya
- Biochemistry Research Laboratory, Omsk State Pedagogical University, 644099 Omsk, Russia;
| | | |
Collapse
|
8
|
Xie J, Wang H, Xie W, Liu Y, Chen Y. Gallic acid promotes ferroptosis in hepatocellular carcinoma via inactivating Wnt/β-catenin signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2437-2445. [PMID: 37847411 DOI: 10.1007/s00210-023-02770-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/03/2023] [Indexed: 10/18/2023]
Abstract
Hepatocellular carcinoma (HCC) has high morbidity and mortality, and effective therapies are lacking. Gallic acid (GA), a natural phenolic compound derived from plants, has been reported to prevent the onset and progression of various cancers. However, there is limited elaboration on the potential mechanisms and anticancer effects of GA on hepatocellular carcinoma. Inducing ferroptosis of tumor cells has become one of the most promising ways to eradicate tumor cells. However, the effect of GA on HCC ferroptosis remains unknown. We evaluated the impact of GA on cell viability, migration, and mitochondrial morphology in HepG2 cells. Our study identified a critical role of GA in inducing ferroptosis in HepG2 cells. Mechanistically, we found that GA could inhibit the expression of a ferroptosis-related protein SLC7A11 and GPX4 in HepG2, by blocking β-catenin transport from nuclear to the cytoplasm, thus inducing the inactivation of the Wnt/β-catenin pathway. Our study has confirmed that GA is a novel ferroptosis inducer of HC, suggesting GA could be a promising candidate for the clinical treatment of HCC.
Collapse
Affiliation(s)
- Jingyi Xie
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Haijiao Wang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Wuxing Xie
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yongping Liu
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| | - Yi Chen
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
9
|
El-Hawary SS, Albalawi MA, Montasser AOS, Ahmed SR, Qasim S, Shati AA, Alfaifi MY, Elbehairi SEI, Hassan OF, Sadakah AA, Mokhtar FA. Network pharmacology and molecular docking study for biological pathway detection of cytotoxicity of the yellow jasmine flowers. BMC Complement Med Ther 2023; 23:164. [PMID: 37210476 DOI: 10.1186/s12906-023-03987-w] [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: 10/17/2022] [Accepted: 05/04/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND The yellow jasmine flower (Jasminum humile L.) is a fragrant plant belonging to the Oleaceae family with promising phytoconstituents and interesting medicinal uses. The purpose of this study was to characterize the plant metabolome to identify the potential bioactive agents with cytotoxic effects and the underlying mechanism of cytotoxic activity. METHODS First, HPLC-PDA-MS/MS was used to identify the potential bioactive compounds in the flowers. Furthermore, we assessed the cytotoxic activity of the flower extract against breast cancer (MCF-7) cell line using MTT assay followed by the cell cycle, DNA-flow cytometry, and Annexin V-FITC analyses alongside the effect on reactive oxygen species (ROS). Finally, Network pharmacology followed by a molecular docking study was performed to predict the pathways involved in anti-breast cancer activity. RESULTS HPLC-PDA-MS/MS tentatively identified 33 compounds, mainly secoiridoids. J. humile extract showed a cytotoxic effect on MCF-7 breast cancer cell line with IC50 value of 9.3 ± 1.2 µg/mL. Studying the apoptotic effect of J. humile extract revealed that it disrupts G2/M phase in the cell cycle, increases the percentage of early and late apoptosis in Annexin V-FTIC, and affects the oxidative stress markers (CAT, SOD, and GSH-R). Network analysis revealed that out of 33 compounds, 24 displayed interaction with 52 human target genes. Relationship between compounds, target genes, and pathways revealed that J. humile exerts its effect on breast cancer by altering, Estrogen signaling pathway, HER2, and EGFR overexpression. To further verify the results of network pharmacology, molecular docking was performed with the five key compounds and the topmost target, EGFR. The results of molecular docking were consistent with those of network pharmacology. CONCLUSION Our findings suggest that J. humile suppresses breast cancer proliferation and induces cell cycle arrest and apoptosis partly by EGFR signaling pathway, highlighting J. humile as a potential therapeutic candidate against breast cancer.
Collapse
Affiliation(s)
- Seham S El-Hawary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| | - Marzough A Albalawi
- Department of Chemistry, Alwajh College, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | | | - Shaimaa R Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
- Department of Pharmacognosy, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Sumera Qasim
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72341, Saudi Arabia
| | - Ali A Shati
- King Khalid University, Faculty of Science, Biology Department, Abha, 9004, Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha, 9004, Saudi Arabia
| | - Serag Eldin I Elbehairi
- King Khalid University, Faculty of Science, Biology Department, Abha, 9004, Saudi Arabia
- Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company), Giza, Egypt
| | - Omnia F Hassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, MSA University, 6th of October City, Egypt
| | - Abdelfattah A Sadakah
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, ALsalam University, Kafr Alzayat, Al Gharbia, Egypt
| | - Fatma A Mokhtar
- Department of Pharmacognosy, Faculty of Pharmacy, Al Salam University, Kafr Alzayat, Al Gharbia, Egypt.
- Department of pharmacognosy, Faculty of Pharmacy, El Saleheya El Gadida University, El Saleheya El Gadida 44813, Sharkia, Egypt.
| |
Collapse
|
10
|
Wani AK, Akhtar N, Mir TUG, Singh R, Jha PK, Mallik SK, Sinha S, Tripathi SK, Jain A, Jha A, Devkota HP, Prakash A. Targeting Apoptotic Pathway of Cancer Cells with Phytochemicals and Plant-Based Nanomaterials. Biomolecules 2023; 13:biom13020194. [PMID: 36830564 PMCID: PMC9953589 DOI: 10.3390/biom13020194] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/13/2023] [Accepted: 01/15/2023] [Indexed: 01/20/2023] Open
Abstract
Apoptosis is the elimination of functionally non-essential, neoplastic, and infected cells via the mitochondrial pathway or death receptor pathway. The process of apoptosis is highly regulated through membrane channels and apoptogenic proteins. Apoptosis maintains cellular balance within the human body through cell cycle progression. Loss of apoptosis control prolongs cancer cell survival and allows the accumulation of mutations that can promote angiogenesis, promote cell proliferation, disrupt differentiation, and increase invasiveness during tumor progression. The apoptotic pathway has been extensively studied as a potential drug target in cancer treatment. However, the off-target activities of drugs and negative implications have been a matter of concern over the years. Phytochemicals (PCs) have been studied for their efficacy in various cancer cell lines individually and synergistically. The development of nanoparticles (NPs) through green synthesis has added a new dimension to the advancement of plant-based nanomaterials for effective cancer treatment. This review provides a detailed insight into the fundamental molecular pathways of programmed cell death and highlights the role of PCs along with the existing drugs and plant-based NPs in treating cancer by targeting its programmed cell death (PCD) network.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Tahir ul Gani Mir
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Rattandeep Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Prakash Kumar Jha
- Feed the Future Innovation Lab for Collaborative Research on Sustainable Intensification, Kansas State University, Manhattan, KS 66506, USA
| | - Shyam Kumar Mallik
- College of Medical and Allied Sciences, Purbanchal University, Morang 56600, Nepal
| | - Shruti Sinha
- UNC Blood Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Surya Kant Tripathi
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Abha Jain
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Aprajita Jha
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar 751024, India
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
- Headquarters for Admissions and Education, Kumamoto University, Kurokami, 2-39-1, Chuo-ku, Kumamoto 860-8555, Japan
- Pharmacy Program, Gandaki University, Pokhara 33700, Nepal
- Correspondence: (H.P.D.); (A.P.)
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
- Correspondence: (H.P.D.); (A.P.)
| |
Collapse
|
11
|
Cancer-Associated Fibroblasts Hinder Lung Squamous Cell Carcinoma Oxidative Stress-Induced Apoptosis via METTL3 Mediated m6A Methylation of COL10A1. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4320809. [PMID: 36246404 PMCID: PMC9560815 DOI: 10.1155/2022/4320809] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/08/2022] [Accepted: 08/28/2022] [Indexed: 11/18/2022]
Abstract
Background. Cancer-associated fibroblasts (CAFs) within the tumor microenvironment are key players in tumorigenesis and tumor development. Nevertheless, the regulatory mechanisms of CAFs on lung squamous cell carcinoma- (LUSC-) associated remain poorly elucidated. Methods. The microarray dataset GSE22874, containing 30 specimens of primary culture of normal fibroblasts (NFs) and 8 specimens of cancer-associated fibroblasts (CAFs) samples derived from LUSC, was retrieved from the Gene Expression Omnibus (GEO) database and then calculated by using the R language (limma package) to identify differentially expressed genes (DEGs). CAF-conditioned medium (CAF-CM) was collected and used to culture LUSC cells, followed by assessment of cell proliferation, apoptosis, and oxidative stress levels by using CCK-8, annexin V-FITC/PI double staining and ELISA assays. Subsequently, COL10A1 was knocked down in CAFs to assess the role of COL10A1 in CAF regulation of LUSC behavior. Bioinformatics online analysis and MeRIP were applied to predict and test the m6A modification of COL10A1 mRNA and the regulatory relationship with METTL3. Rescue experiments were next performed to explore the effects of METTL3 and COL10A1 in CAFs on LUSC cell proliferation, apoptosis, and oxidative stress. LUSC tumor cells with or without (COL10A1-silenced) CAFs were subcutaneously inoculated in nude mice to evaluate the effect of COL10A1 in CAFs on LUSC tumor growth. Results. Elevated expression of COL10A1 was found in LUSC-derived CAFs by GSE22874 dataset analysis. We discovered that COL10A1 and METTL3 was expressed in both LUSC cells and matched CAFs, while COL10A1 expression was prominently higher in CAFs than in LUSC cells. CAF-CM memorably encouraged LUSC cell proliferation and suppressed apoptosis-induced oxidative stress, which was reversed by interfering with COL10A1 expression in CAFs, suggesting that COL10A1 might be secreted by CAFs into the culture medium to exert its effects inside LUSC cells. Global m6A modification was decreased in METTL3 knocked down CAFs. M6A modification, expression levels, and stability of COL10A1 mRNA were impaired upon METTL3 knockdown in CAFs. Overexpression of COL10A1 in CAFs partially reversed the effect of METTL3 knockdown on the malignant behavior of LUSC cells. In vivo studies confirmed that CAFs accelerated LUSC tumor growth, and this effect was counteracted by COL10A1 silencing. Conclusions. COL10A1 secreted by CAFs could facilitate LUSC cell proliferation and repress apoptosis-induced oxidative stress, and the mechanism was due to elevated expression mediated by METTL3 promoting its mRNA m6A modification, thereby accelerating tumor growth.
Collapse
|
12
|
Franco C, Viana AR, Ourique AF, Vizzotto BS, Krause LMF. Protective Effect of Indomethacin-loaded Polymeric Nanoparticles Against Oxidative Stress-Induced Cytotoxicity in Human Breast Adenocarcinoma Cell Model. REVISTA BRASILEIRA DE CANCEROLOGIA 2022. [DOI: 10.32635/2176-9745.rbc.2022v68n4.2545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Introduction: Anti-inflammatory drugs are being utilized to treat cancer because of its inflammatory microenvironment. Objective: The objective of this study is to investigate the antioxidant potential of indomethacin and its genotoxicity, since free or loaded in polymeric nanocapsules using MCF-7 (human breast cancer) cells as an in vitro model. Method: Development of indomethacin-loaded polyepsiloncaprolactone (PCL) nanocapsules by interfacial deposition method. It is characterized by pH determination by potentiometer, mean diameter and polydispersity index by dynamic light scattering; zeta potential by electrophoretic mobility; encapsulation efficacy by high performance liquid chromatography method; corona effect formation; 2ʹ,7ʹ-dichlorofluorescin diacetate (DCFH-DA) method by spectrofluorimetric assay; nitric oxide (NO) determination by spectrophotometric and genotoxicity assay by plasmid DNA cleavage method. Results: The results showed a mild acidic pH (4.78 ± 0.10), sizes around 200 nm and PDI<0.2 with a zeta potential around -20 mV and encapsulation efficiency of 99% (1 mg mL-1), showing a dose-dependent corona formation profile in 24h incubation. Conclusion: DCFH-DA assay showed no production of reactive oxygen species (ROS) while NO determination showed that Ind-OH-NC from 26.7 to 100 μM increased reactive nitrogen species (RNS), demonstrating antioxidant potential against MCF-7 cells. No sample at the concentrations evaluated induced DNA cleavage, being considered a safe treatment.
Collapse
|
13
|
Chaudhry GES, Md Akim A, Sung YY, Sifzizul TMT. Cancer and apoptosis: The apoptotic activity of plant and marine natural products and their potential as targeted cancer therapeutics. Front Pharmacol 2022; 13:842376. [PMID: 36034846 PMCID: PMC9399632 DOI: 10.3389/fphar.2022.842376] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/13/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is a multifactorial, multi-stage disease, including complex cascades of signaling pathways—the cell growth governed by dysregulated and abrupt cell division. Due to the complexity and multi-regulatory cancer progression, cancer is still a challenging disease to treat and survive. The screening of extracts and fractions from plants and marine species might lead to the discovery of more effective compounds for cancer therapeutics. The isolated compounds and reformed analogs were known as future prospective contenders for anti-cancer chemotherapy. For example, Taxol, a potent mitotic inhibitor discovered from Taxus brevifolia, suppresses cell growth and arrest, induces apoptosis, and inhibits proliferation. Similarly, marine sponges show remarkable tumor chemo preventive and chemotherapeutic potential. However, there is limited research to date. Several plants and marine-derived anti-cancer compounds having the property to induce apoptosis have been approved for clinical trials. The anti-cancer activity kills the cell and slows the growth of cancer cells. Among cell death mechanisms, apoptosis induction is a more profound mechanism of cell death triggered by naturally isolated anti-cancer agents. Evading apoptosis is the major hurdle in killing cancer cells, a mechanism mainly regulated as intrinsic and extrinsic. However, it is possible to modify the apoptosis-resistant phenotype of the cell by altering many of these mechanisms. Various extracts and fractions successfully induce apoptosis, cell-cycle modulation, apoptosis, and anti-proliferative activity. Therefore, there is a pressing need to develop new anti-cancer drugs of natural origins to reduce the effects on normal cells. Here, we’ve emphasized the most critical elements: i) A better understanding of cancer progression and development and its origins, ii) Molecular strategies to inhibit the cell proliferation/Carcino-genesis, iii) Critical regulators of cancer cell proliferation and development, iv) Signaling Pathways in Apoptosis: Potential Targets for targeted therapeutics, v) Why Apoptosis induction is mandatory for effective chemotherapy, vi) Plants extracts/fractions as potential apoptotic inducers, vii) Marine extracts as Apoptotic inducers, viii) Marine isolated Targeted compounds as Apoptotic inducers (FDA Approved/treatment Phase). This study provides a potential therapeutic option for cancer, although more clinical studies are needed to verify its efficacy in cancer chemotherapy.
Collapse
Affiliation(s)
- Gul-e-Saba Chaudhry
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu, Malaysia
- *Correspondence: Gul-e-Saba Chaudhry, ,
| | - Abdah Md Akim
- Department of Biomedical Sciences, Faculty of Medicine and Health sciences, University of Putra Malaysia, Seri Kembangan, Malaysia
| | - Yeong Yik Sung
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu, Malaysia
| | | |
Collapse
|
14
|
Bhaduri R, Mandal S, Kumar Tarai S, Pan A, Mukherjee S, Bagchi A, Biswas A, Ch. Moi S. Cytotoxic activity of nitrogen, sulfur, and oxygen chelated Pt(II) complexes; their DNA/BSA binding by in vitro and in silico approaches. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.119529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Kamarudin AA, Sayuti NH, Saad N, Razak NAA, Esa NM. Induction of apoptosis by Eleutherine bulbosa (Mill.) Urb. bulb extracted under optimised extraction condition on human retinoblastoma cancer cells (WERI-Rb-1). JOURNAL OF ETHNOPHARMACOLOGY 2022; 284:114770. [PMID: 34688803 DOI: 10.1016/j.jep.2021.114770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The bulb of Eleutherine bulbosa (Mill.) Urb. is an indigenous medicinal plant traditionally used among Dayak people for the management of diabetes, breast cancer, hypertension, stroke, and fertility problems in women. The bulb has been reported with a potent cytotoxic potential but with limited underlying mechanisms. AIM OF THE STUDY This study aimed to investigate the cytotoxic properties of E. bulbosa ethanolic bulb extracted under optimised extraction condition on retinoblastoma cancer cells (WERI-Rb-1) through in vitro cell culture bioassays. The optimised extraction condition has been determined in the previous reports. MATERIALS AND METHODS Cytotoxic assay was analysed through MTT assay. Comparison between non-optimised and optimised extraction condition from E. bulbosa ethanolic bulb extract was evaluated. Morphological assessment of apoptotic cells was conducted through acridine orange propidium iodide (AOPI) staining using fluorescence microscopy. Apoptosis assay was carried out through Annexin V-FITC and cell cycle analysis through PI staining. The effect of varying concentrations (IC25, IC50, IC75) of the optimised E. bulbosa ethanolic bulb extract was observed. The mRNA expression was also conducted to confirm the underlying mechanism. RESULTS The optimised E. bulbosa ethanolic bulb extract markedly suppressed the proliferation of retinoblastoma cancer cells significantly with an IC50 value of 15.7 μg/mL as compared to non-optimised extract (p < 0.01). Fluorescence microscopy revealed that retinoblastoma cancer cells manifested early features of apoptosis-like membrane blebbing, chromatin condensation and formation of apoptotic bodies in a dose-dependent manner. The number of apoptotic cells were greatly observed in early and late apoptosis through Annexin V-FITC and the extract also induced cell arrestment as compared to the untreated group. The apoptosis was confirmed with the upregulation of Bax, Bad, p53, Caspase 3, Caspase 8, and Caspase 9 genes meanwhile, Bcl-2, BcL-xL, Nrf-2, and HO-1 genes were downregulated. CONCLUSION The optimised E. bulbosa ethanolic bulb extract induced a significant cell death and cell cycle arrestment on retinoblastoma cancer cells. It could be suggested that the induction of apoptosis in retinoblastoma cancer cells may be due to the synergistic effect of the bioactive compounds extracted under optimised extraction condition. Our findings indicated that E. bulbosa bulb could be promising chemotherapeutic potential to treat retinoblastoma cancer cells.
Collapse
Affiliation(s)
- Ammar Akram Kamarudin
- Natural Medicines and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Nor Hafiza Sayuti
- Natural Medicines and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Norazalina Saad
- UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Nor Asma Ab Razak
- Natural Medicines and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Norhaizan Mohd Esa
- Natural Medicines and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
16
|
Jabbari N, Feghhi M, Esnaashari O, Soraya H, Rezaie J. Inhibitory effects of gallic acid on the activity of exosomal secretory pathway in breast cancer cell lines: A possible anticancer impact. BIOIMPACTS : BI 2022; 12:549-559. [PMID: 36644548 PMCID: PMC9809134 DOI: 10.34172/bi.2022.23489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 01/18/2023]
Abstract
Introduction: Breast cancer cells produce exosomes that promote tumorigenesis. The anticancer properties of gallic acid have been reported. However, the mechanism underlying its anticancer effect on the exosomal secretory pathway is still unclear. We investigated the effect of gallic acid on exosome biogenesis in breast cancer cell lines. Methods: The cytotoxic effect of gallic acid on MCF-10a, MCF-7, and MDA-MD-231 cells was measured by MTT assay after 48 hours treatment. Expression of miRNAs including miRNA-21, -155, and 182 as well as exosomal genes such as Rab27a, b, Rab11, Alix, and CD63; along with HSP-70 (autophagy gene), was determined using Q-PCR. The subcellular distribution of it was monitored by flow cytometry analysis. Isolated exosomes were characterized by transmission and scanning electron microscopes and flow cytometry. Acetylcholinesterase activity is used to measure the number of exosomes in supernatants. In addition, autophagy markers including LC3 and P62 were measured by ELISA. Results: Data showed that gallic acid was cytotoxic to cells (P < 0.05). Gallic acid modulated expression of miRNAs and down-regulated transcript levels of exosomal genes and up-regulated the HSP-70 gene in three cell lines (P < 0.05). The surface CD63/total CD63 ratio as well as acetylcholinesterase activity decreased in treated cells (P < 0.05). The protein level of LC3 was increased in three cell lines, while the expression of P62 increased in MCF-7 and MDA-MB-231 cancer cell lines. Conclusion: Together, gallic acid decreased the activity of the exosomal secretory pathway in breast cancer cell lines, providing evidence for its anti-cancer effects.
Collapse
Affiliation(s)
- Nasrollah Jabbari
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Feghhi
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | | | - Hamid Soraya
- Department of Pharmacology Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
,Corresponding author: Jafar Rezaie,
| |
Collapse
|
17
|
Chaudhry GES, Jan R, Akim A, Zafar MN, Sung YY, Muhammad TST. Breast Cancer: A Global Concern, Diagnostic and Therapeutic Perspectives, Mechanistic Targets in Drug Development. Adv Pharm Bull 2021; 11:580-594. [PMID: 34888205 PMCID: PMC8642807 DOI: 10.34172/apb.2021.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 08/10/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is a complex multifactorial process, unchecked and abrupt division, and cell growth—conventional chemotherapy, along with radiotherapy, is used to treat breast cancer. Due to reduce efficacy and less survival rate, there is a particular need for the discovery of new active anticancer agents. Natural resources such as terrestrial/marine plants or organisms are a promising source for the generation of new therapeutics with improving efficacy. The screening of natural plant extracts and fractions, isolations of phytochemicals, and mechanistic study of those potential compounds play a remarkable role in the development of new therapeutic drugs with increased efficacy. Cancer is a multistage disease with complex signaling cascades. The initial study of screening whole extracts or fractions and later the isolation of secondary compounds and their mechanism of action study gives a clue of potential therapeutic agents for future drug development. The phytochemicals present in extracts/fractions produce remarkable effects due to synergistically targeting multiple signals. In this review, the molecular targets of extracts/ fractions and isolated compounds highlighted. The therapeutic agent's mechanistic targets in drug development focused involves; i) Induction of Apoptosis, ii) modulating cell cycle arrest, iii) Inhibition or suppression of invasion and metastasis and iv) various other pro-survival signaling pathways. The phytochemicals and their modified analogs identified as future potential candidates for anticancer chemotherapy.
Collapse
Affiliation(s)
- Gul-E-Saba Chaudhry
- Institute of Marine Biotechnology, University Malaysia Terengganu, 21030 Kuala Terengganu, Malaysia
| | - Rehmat Jan
- Department of Environmental Sciences, Fatima Jinnah University, Rawalpindi, Pakistan
| | - Abdah Akim
- Department of Biomedical Sciences, Universiti Putra Malaysia, Seri Kembangan, Selangor, Malaysia
| | | | - Yeong Yik Sung
- Institute of Marine Biotechnology, University Malaysia Terengganu, 21030 Kuala Terengganu, Malaysia
| | | |
Collapse
|
18
|
Gordon JL, Hinsen KJ, Reynolds MM, Brown MA. Anticancer Impact of Nitric Oxide (NO) and NO Combination with SMYD-3 Inhibitor on Breast Carcinomas. Diseases 2021; 9:82. [PMID: 34842655 PMCID: PMC8628812 DOI: 10.3390/diseases9040082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Despite enormous advances in the detection and treatment of breast cancer, it still remains the leading cancer diagnosis and has the second highest mortality rate. Thus, breast cancer research is a high priority for academics and clinicians alike. Based on previous research indicating the potential of nitric oxide (NO) and SMYD-3 inhibition, this work sought to expand upon these concepts and combine the two approaches. Both NO (from S-Nitrosoglutathione (GSNO)), termed Group 1, and a combination therapeutic, inhibitor-4 (SMYD-3 inhibitor) plus NO (from GSNO), termed Group 2, were evaluated for their efficacy on breast carcinoma cell lines MCF7 and MDA-MB-231, and the normal MCF10A breast cell line, using cellular viability, colony formation capacity, cytotoxicity, and cellular apoptosis analysis. These results indicated that, in Group 1, breast carcinoma lines MCF7 and MDA-MB-231, cells experienced a moderate reduction in cellular viability (~20-25%), a large reduction in colony formation capacity (~80-90%), a moderate increase in the relative number of dead cells, and a moderate increase in cellular apoptosis. Group 2 was significantly more impactful, with a ~50% knockdown in cellular viability, a 100% reduction in colony formation capacity, a large increase in the relative number of dead cells, and a large increase in cellular apoptosis. Additionally, Group 2 induced a very small impact on the normal MCF10A cell line. Cumulatively, this work revealed the exciting impact of this combination therapeutic, indicating its potential for clinical application and further research.
Collapse
Affiliation(s)
- Jenna L. Gordon
- Department of Chemistry, Colorado State University, Fort Collins, CO 80521, USA;
| | - Kristin J. Hinsen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA;
| | - Melissa M. Reynolds
- Department of Chemistry, Department of Chemical and Biological Engineering, School of Biomedical Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Mark A. Brown
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80521, USA;
| |
Collapse
|
19
|
Gunathilaka TL, Dilrangi KH, Ranasinghe P, Samarakoon KW, Peiris LDC. Mechanistic Insight into Apoptotic Induction in Human Rhabdomyosarcoma and Breast Adenocarcinoma Cells by Chnoospora minima: A Sri Lankan Brown Seaweed. Pharmaceuticals (Basel) 2021; 14:1154. [PMID: 34832937 PMCID: PMC8622854 DOI: 10.3390/ph14111154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022] Open
Abstract
The current study determined the cytotoxic and apoptotic potential of the polyphenol-rich methanol extract of Chnoospora minima (C. minima) and its fractions against human breast adenocarcinoma (MCF-7) and rhabdomyosarcoma (RMS) cells. MTT and neutral red assays were used to determine cytotoxicity. The clonogenic assay evaluated the antineoplastic activity, while the apoptotic activity was determined by cellular morphological changes, caspase 3/7 activity, and DNA fragmentation. Morphological alterations in apoptosis were observed by an inverted phase-contrast microscope and Hoechst 33342 staining methods. The total phenolic, flavonoids, alkaloids, and antioxidant activity in the hexane and chloroform fractions were determined, based on their cytotoxic activity. The hexane fraction of C. minima effectively reduced the cell growth that is concentration-dependent in human RMS and MCF-7 cell lines. It also exhibited low cytotoxicity on Vero cells. The characteristic cellular and nuclear apoptotic morphological features were observed. A noticeable caspase 3/7 activation and the fragmented DNA were detected only in the hexane fraction treated RMS cells, whereas MCF-7 cells showed low caspase 3/7 activation due to a lack of caspase 3 and no evidence of having a typical ladder pattern of apoptosis. Further analysis revealed that the hexane fraction-treated RMS cells upregulated the p53 gene twofold (2.72) compared to the p21 (0.77) gene, whereas in the MCF-7 cells, a 2.21-fold upregulation of p53 was observed compared to the p21 (0.64) gene. The hexane fraction exhibited moderate total phenolics, flavonoids, alkaloids content, and antioxidant activity. According to the different antioxidant mechanisms, hexane and chloroform fractions showed the highest antioxidant activities by FRAP and ORAC assays, respectively. GC-MS analysis of hexane fraction revealed the presence of methyl tetradecanoate (38.314%) as the most abundant compound. The study's findings highlighted that the non-polar compounds present in the hexane fraction of C. minima suppressed cell proliferation and induced apoptosis-mediated cell death in RMS and MCF-7 cells, mainly via the activation of the p53 gene. Hence, the isolation of compounds is warranted. However, more studies are required to understand the mechanistic insights of these observations.
Collapse
Affiliation(s)
- Thilina Lakmini Gunathilaka
- Department of Zoology, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka; (T.L.G.); (K.H.D.)
| | - Kulathungage Hiranthi Dilrangi
- Department of Zoology, Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka; (T.L.G.); (K.H.D.)
| | | | - Kalpa W. Samarakoon
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defence University, Ratmalana 10390, Sri Lanka
| | - L. Dinithi C. Peiris
- Department of Zoology/Genetics & Molecular Biology Unit (Center for Biotechnology), Faculty of Applied Sciences, University of Sri Jayewardenepura, Nugegoda 10250, Sri Lanka
| |
Collapse
|
20
|
Joshi BC, Juyal V, Sah AN, Verma P, Mukhija M. Review On Documented Medicinal Plants Used For The Treatment Of Cancer. CURRENT TRADITIONAL MEDICINE 2021. [DOI: 10.2174/2215083807666211011125110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background:
Cancer is a frightful disease and it is the second leading cause of death worldwide. Naturally derived compounds are gaining interest of research workers as they have less toxic side effects as compared to currently used treatments such as chemotherapy. Plants are the pool of chemical compounds which provides a promising future for research on cancer.
Objective:
This review paper provides updated information gathered on medicinal plants and isolated phytoconstituents used as anticancer agents and summarises the plant extracts and their isolated chemical constituents exhibiting anticancer potential on clinical trials.
Methods:
An extensive bibliographic investigation was carried out by analysing worldwide established scientific databases like SCOPUS, PUBMED, SCIELO, ScienceDirect, Springerlink, Web of Science, Wiley, SciFinder and Google Scholar etc. In next few decades, herbal medicine may become a new epoch of medical system.
Results:
Many researches are going on medicinal plants for the treatment of cancer but it is a time to increase further experimental studies on plant extracts and their chemical constituents to find out their mechanism of action at molecular level.
Conclusion:
The article may help many researchers to start off further experimentation that might lead to the drugs for the cancer treatment.
Collapse
Affiliation(s)
- Bhuwan Chandra Joshi
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Bhimtal Campus, Nainital-263136, India
| | - Vijay Juyal
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Bhimtal Campus, Nainital-263136, India
| | - Archana N. Sah
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Bhimtal Campus, Nainital-263136, India
| | - Piyush Verma
- Department of Pharmacology, School of Pharmaceutical science and Technology, Sardar Bhagwan Singh University, Dehradun-248001, India
| | - Minky Mukhija
- Department of Pharmaceutical Sciences, Ch. Devi Lal College of Pharmacy, Buria Road, Bhagwangarh, Jagadhri-135003, India
| |
Collapse
|
21
|
Troschel FM, Palenta H, Borrmann K, Heshe K, Hua SH, Yip GW, Kiesel L, Eich HT, Götte M, Greve B. Knockdown of the prognostic cancer stem cell marker Musashi-1 decreases radio-resistance while enhancing apoptosis in hormone receptor-positive breast cancer cells via p21 WAF1/CIP1. J Cancer Res Clin Oncol 2021; 147:3299-3312. [PMID: 34291358 PMCID: PMC8484224 DOI: 10.1007/s00432-021-03743-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/13/2021] [Indexed: 02/02/2023]
Abstract
Purpose While the stem cell marker Musashi-1 (MSI-1) has been identified as a key player in a wide array of malignancies, few findings exist on its prognostic relevance and relevance for cancer cell death and therapy resistance in breast cancer. Methods First, we determined prognostic relevance of MSI-1 in database analyses regarding multiple survival outcomes. To substantiate findings, MSI-1 was artificially downregulated in MCF-7 breast cancer cells and implications for cancer stem cell markers, cell apoptosis and apoptosis regulator p21, proliferation and radiation response were analyzed via flow cytometry and colony formation. Radiation-induced p21 expression changes were investigated using a dataset containing patient samples obtained before and after irradiation and own in vitro experiments. Results MSI-1 is a negative prognostic marker for disease-free and distant metastasis-free survival in breast cancer and tends to negatively influence overall survival. MSI-1 knockdown downregulated stem cell gene expression and proliferation, but increased p21 levels and apoptosis. Similar to the MSI-1 knockdown effect, p21 expression was strongly increased after irradiation and was expressed at even higher levels in MSI-1 knockdown cells after irradiation. Finally, combined use of MSI-1 silencing and irradiation reduced cancer cell survival. Conclusion MSI-1 is a prognostic marker in breast cancer. MSI-1 silencing downregulates proliferation while increasing apoptosis. The anti-proliferation mediator p21 was upregulated independently after both MSI-1 knockdown and irradiation and even more after both treatments combined, suggesting synergistic potential. Radio-sensitization effects after combining radiation and MSI-1 knockdown underline the potential of MSI-1 as a therapeutic target. Supplementary Information The online version contains supplementary material available at 10.1007/s00432-021-03743-y.
Collapse
Affiliation(s)
- Fabian M Troschel
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany.
| | - Heike Palenta
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Katrin Borrmann
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - Kristin Heshe
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - San Hue Hua
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - George W Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117594, Singapore
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Hans Theodor Eich
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital Münster, 48149, Münster, Germany
| | - Burkhard Greve
- Department of Radiation Oncology, University Hospital Münster, 48149, Münster, Germany
| |
Collapse
|
22
|
Palanivel S, Yli-Harja O, Kandhavelu M. Alkylamino Phenol Derivative Induces Apoptosis by Inhibiting EGFR Signaling Pathway in Breast Cancer Cells. Anticancer Agents Med Chem 2021; 20:809-819. [PMID: 32053080 DOI: 10.2174/1871520620666200213101407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 10/08/2019] [Accepted: 12/30/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE The present study was carried out to evaluate the anticancer property of an alkylamino phenol derivative -2-((3,4-Dihydroquinolin-1(2H)-yl)(p-tolyl)methyl)phenol) (THTMP) against human breast cancer cells. The cytotoxicity of the THTMP was assessed to know its specificity towards breast cancer cells without affecting the normal cells. METHODS The THTMP was synthesized and the cytotoxicity was assessed by MTT assay, Caspases enzyme activity, DNA fragmentation and FITC/Annexin V, AO/EtBr staining, RT-PCR and QSAR. In addition, ADME analysis was executed to understand the mode of action of THTMP. RESULTS THTMP showed potential cytotoxic activity against the growth of MCF7 and SK-BR3 cells with the IC50 values of 87.92μM and 172.51μM, respectively. Interestingly, THTMP found to activate caspase 3 and caspase 9 enzymes in cancer cells, which are the key enzymes implicated in apoptosis. THTMP induced apoptosis in which 33% of the cells entered the late apoptotic stage after 24h of treatment. The results also revealed that the apoptotic response could be influenced by the association of THTMP with the Epidermal Growth Factor Receptor (EGFR) mediated inhibition of the Phosphatidylinositol 3-Kinase (PI3K)/S6K1 signaling pathway. In addition, docking was performed to study the binding mode of the THTMP, which shows better interaction with EGFR. The structural elucidation of THTMP by Quantitative Structure-Activity Relationship model (QSAR) and ADMET screening suggested, THTMP as an effective anticancer compound. CONCLUSION This work strengthens the potential of a promising drug-like compound, THTMP, for the discovery of anticancer drug against breast cancer.
Collapse
Affiliation(s)
- Suresh Palanivel
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland,Institute of Biosciences and Medical Technology, Tampere, Finland
| | - Olli Yli-Harja
- Institute of Biosciences and Medical Technology, Tampere, Finland,Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland,Institute for Systems Biology, 1441N 34th Street, Seattle, WA 98103-8904, USA
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University and BioMediTech, P.O. Box 553, 33101 Tampere, Finland,Institute of Biosciences and Medical Technology, Tampere, Finland
| |
Collapse
|
23
|
Özdemi̇r F, Sever A, Keçeci̇ YÖ, Incesu Z. Resveratrol increases the sensitivity of breast cancer MDA-MB-231 cell line to cisplatin by regulating intrinsic apoptosis. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:66-72. [PMID: 33643572 PMCID: PMC7894626 DOI: 10.22038/ijbms.2020.50485.11501] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/13/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Breast cancer is one of the most common types of cancer. Chemotherapeutic agents used during treatment induce cytotoxic effects also on normal cells in the tissues. Anti-oxidants used in combination with chemotherapeutic agents have been shown to reduce toxicity on normal cells to a minimum, and some anti-oxidant substances have chemotherapeutic effects. Cisplatin (CDDP) is a platinum class drug that is used clinically in the treatment of many cancers. Resveratrol (RSV) is a natural polyphenol with potent anti-oxidant and anticancer properties. In this study, we aimed to investigate apoptotic effects of using cisplatin and RSV alone or in combined treatment of MDA-MB-231 cells. MATERIALS AND METHODS The cytotoxic effects of the drugs on MDA-MB-231 cells were determined by MTT method. Subsequently, the change in CDDP-induced apoptotic effect after RSV addition was examined using the AnnexinV FITC labeling, and TUNEL staining method. Activation of caspase-9, -3 in MDA-MB-231 cells was measured by flow cytometer. The mitochondrial membrane potential (MMP), the major factor on the intrinsic pathway, was measured using flowcytometry. RESULTS The combined dose (23 μM CDDP + 72 μM RSV) produced more cytotoxicity than the agents used alone, leading to early apoptosis (8.2%), 31% depolarization, and 23% DNA fragmentation. Caspase-9 was found to be 30.5% in this combined group and caspase-3 was 26.3%. CONCLUSION RSV, an effective anti-oxidant, and CDDP as an effective drug in cancer treatment, were found to increase apoptosis when given in the MDA-MB-231 cell.
Collapse
Affiliation(s)
- Filiz Özdemi̇r
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Arda Sever
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Yüksel Öğünç Keçeci̇
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Zerrin Incesu
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
24
|
Tanaka Y, Obinata H, Konishi A, Yamagiwa N, Tsuneoka M. Production of ROS by Gallic Acid Activates KDM2A to Reduce rRNA Transcription. Cells 2020; 9:E2266. [PMID: 33050392 PMCID: PMC7601038 DOI: 10.3390/cells9102266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/04/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Metformin, which is suggested to have anti-cancer effects, activates KDM2A to reduce rRNA transcription and proliferation of cancer cells. Thus, the specific activation of KDM2A may be applicable to the treatment of cancers. In this study, we screened a food-additive compound library to identify compounds that control cell proliferation. We found that gallic acid activated KDM2A to reduce rRNA transcription and cell proliferation in breast cancer MCF-7 cells. Gallic acid accelerated ROS production and activated AMPK. When ROS production or AMPK activity was inhibited, gallic acid did not activate KDM2A. These results suggest that both ROS production and AMPK activation are required for activation of KDM2A by gallic acid. Gallic acid did not reduce the succinate level, which was required for KDM2A activation by metformin. Metformin did not elevate ROS production. These results suggest that the activation of KDM2A by gallic acid includes mechanisms distinct from those by metformin. Therefore, signals from multiple intracellular conditions converge in KDM2A to control rRNA transcription. Gallic acid did not induce KDM2A-dependent anti-proliferation activity in non-tumorigenic MCF10A cells. These results suggest that the mechanism of KDM2A activation by gallic acid may be applicable to the treatment of breast cancers.
Collapse
Affiliation(s)
- Yuji Tanaka
- Laboratory of Molecular and Cellular Biology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan;
| | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan;
| | - Akimitsu Konishi
- Department of Biochemistry, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan;
| | - Noriyuki Yamagiwa
- Laboratory of Molecular Design Chemistry, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan;
| | - Makoto Tsuneoka
- Laboratory of Molecular and Cellular Biology, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan;
| |
Collapse
|
25
|
Tran Trung H, Truong Thi Huynh H, Nguyen Thi Thuy L, Nguyen Van Minh H, Thi Nguyen MN, Luong Thi MN. Growth-Inhibiting, Bactericidal, Antibiofilm, and Urease Inhibitory Activities of Hibiscus rosa sinensis L. Flower Constituents toward Antibiotic Sensitive- and Resistant-Strains of Helicobacter pylori. ACS OMEGA 2020; 5:20080-20089. [PMID: 32832762 PMCID: PMC7439261 DOI: 10.1021/acsomega.0c01640] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/22/2020] [Indexed: 05/18/2023]
Abstract
The aim of the present study was to assess antimicrobial effects of naringenin (NRG), luteolin (LUT), myricetin (MCT), and protocatechuic acid (PCA) identified in a Hibiscus rosa sinensis flower against two reference strains and five clinical isolates of Helicobacter pylori. NRG displayed the most growth inhibitory and bactericidal activities to seven bacterial strains including six strains resistant to one or several antibiotics, azithromycin (MIC, 16-32 mg/L), erythromycin (MIC, 32 mg/L), levofloxacin (MIC, 32 mg/L), and/or metronidazole (24-64 mg/L), followed by LUT and MCT, while PCA showed weak activities toward the strains. These constituents had similar antibacterial activities toward the seven tested strains suggesting that these constituents and the antibiotics do not have a common mechanism of anti-H. pylori activity. NRG, LUT, and MCT resulted in a high percentage of coccoid forms of H. pylori. NRG exhibited the highest anti-biofilm formation activity. MCT produced the strongest inhibition of urease activity followed by LUT and PCA, whereas the activity of NRG was similar to standard inhibitor thiourea. The four constituents had no significant toxicity to human cell lines. A global attempt to decrease utilization of antibiotics justifies the need for further research on H. rosa sinensis derived materials containing NRG, LUT, MCT, and PCA as potential products or lead compounds for the prevention or treatment of diseases caused by H. pylori infection.
Collapse
Affiliation(s)
- Hieu Tran Trung
- Faculty
of Biology and Biotechnology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, District 5, Ho Chi Minh City 700000, Vietnam
- . Tel: (+84) 2838304093
| | - Hoa Truong Thi Huynh
- Central
Laboratory for Analysis, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, District 5, Ho Chi Minh City 700000, Vietnam
| | - Linh Nguyen Thi Thuy
- Faculty
of Chemistry, University of Science, Vietnam
National University, 227 Nguyen Van Cu Street, District 5, Ho Chi
Minh City 700000, Vietnam
| | - Hoang Nguyen Van Minh
- Oxford
University Clinical Research Unit, Hospital for Tropical Diseases, 764 Vo Van Kiet Street, District
5, Ho Chi Minh City 700000, Vietnam
| | - My-Nuong Thi Nguyen
- Faculty
of Biology and Biotechnology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, District 5, Ho Chi Minh City 700000, Vietnam
| | - My Ngan Luong Thi
- Faculty
of Biology and Biotechnology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, District 5, Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
26
|
Song J, He Y, Luo C, Feng B, Ran F, Xu H, Ci Z, Xu R, Han L, Zhang D. New progress in the pharmacology of protocatechuic acid: A compound ingested in daily foods and herbs frequently and heavily. Pharmacol Res 2020; 161:105109. [PMID: 32738494 DOI: 10.1016/j.phrs.2020.105109] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 12/13/2022]
Abstract
Protocatechuic acid is a natural phenolic acid, which widely exists in our daily diet and herbs. It is also one of the main metabolites of complex polyphenols, such as anthocyanins and proanthocyanins. In recent years, a large number of studies on the pharmacological activities of protocatechuic acid have emerged. Protocatechuic acid has a wide range of pharmacological activities including antioxidant, anti-inflammatory, neuroprotective, antibacterial, antiviral, anticancer, antiosteoporotic, analgesia, antiaging activties; protection from metabolic syndrome; and preservation of liver, kidneys, and reproductive functions. Pharmacokinetic studies showed that the absorption and elimination rate of protocatechuic acid are faster, with glucuronidation and sulfation being the major metabolic pathways. However, protocatechuic acid displays a dual-directional regulatory effect on some pharmacological activities. When the concentration is very high, it can inhibit cell proliferation and reduce survival rate. This review aims to comprehensively summarize the pharmacology, pharmacokinetics, and toxicity of protocatechuic acid with emphasis on its pharmacological activities discovered in recent 5 years, so as to provide more up-to-date and thorough information for the preclinical and clinical research of protocatechuic acid in the future. Moreover, it is hoped that the clinical application of protocatechuic acid can be broadened, giving full play to its characteristics of rich sources, low toxicity and wide pharmacological activites.
Collapse
Affiliation(s)
- Jiao Song
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China
| | - Yanan He
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China
| | - Chuanhong Luo
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China
| | - Bi Feng
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China
| | - Fei Ran
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China
| | - Hong Xu
- Chengdu Yongkang Pharmaceutical Co., Ltd., Chengdu 610041, PR China
| | - Zhimin Ci
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China
| | - Runchun Xu
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China
| | - Li Han
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China.
| | - Dingkun Zhang
- Pharmacy College, Chengdu University of Traditional Chinese Medicine, Key Laboratory Breeding Base of Systematic Research and Utilization on Chinese Meterial Medical Resources Co-founded by Sichuan Province and Ministry of Science and Technology, Chengdu 611137, PR China.
| |
Collapse
|
27
|
Kantapan J, Paksee S, Chawapun P, Sangthong P, Dechsupa N. Pentagalloyl Glucose- and Ethyl Gallate-Rich Extract from Maprang Seeds Induce Apoptosis in MCF-7 Breast Cancer Cells through Mitochondria-Mediated Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:5686029. [PMID: 32382295 PMCID: PMC7193289 DOI: 10.1155/2020/5686029] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/26/2020] [Accepted: 03/25/2020] [Indexed: 01/26/2023]
Abstract
Bouea macrophylla Griffith, locally known as maprang, has important economic value as a Thai fruit tree. The maprang seed extract (MPSE) has been shown to exhibit antibacterial and anticancer activities. However, the bioactive constituents in MPSE and the molecular mechanisms underlying these anticancer activities remain poorly understood. This study aims to identify the active compounds in MPSE and to investigate the mechanisms involved in MPSE-induced apoptosis in MCF-7 treated cancer cells. The cytotoxic effect was determined by MTT assay. The apoptosis induction of MPSE was evaluated in terms of ROS production, mitochondrial membrane potential depolarization, and apoptosis-related gene expression. The compounds identified by HPLC and LC/MS analysis were pentagalloyl glucose, ethyl gallate, and gallic acid. MPSE treatment decreased cell proliferation in MCF-7 cells, and MPSE was postulated to induce G2/M phase cell cycle arrest. MPSE was found to promote intracellular ROS production in MCF-7 treated cells and to also influence the depolarization of mitochondrial membrane potential. In addition, MPSE treatment can lead to increase in the Bax/Bcl-2 gene expression ratio, suggesting that MPSE-induced apoptosis is mitochondria-dependent pathway. Our results suggest that natural products obtained from maprang seeds have the potential to target the apoptosis pathway in breast cancer treatments.
Collapse
Affiliation(s)
- Jiraporn Kantapan
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siwaphon Paksee
- Department of Radiological Technology, Kanchanabhishek Institute of Medical and Public Health Technology, Nonthaburi 11150, Thailand
| | - Pornthip Chawapun
- Interdisciplinary Program in Biotechnology, Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Padchanee Sangthong
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
- Research Center on Chemistry for Development of Health Promoting Products from Northern Resources, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nathupakorn Dechsupa
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
28
|
SUMOylation Protects FASN Against Proteasomal Degradation in Breast Cancer Cells Treated with Grape Leaf Extract. Biomolecules 2020; 10:biom10040529. [PMID: 32244364 PMCID: PMC7226518 DOI: 10.3390/biom10040529] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Existing therapeutic strategies for breast cancer are limited by tumor recurrence and drug-resistance. Antioxidant plant-derived compounds such as flavonoids reduce adverse outcomes and have been identified as a potential source of antineoplastic agent with less undesirable side effects. Here, we describe the novel regulation of fatty-acid synthase (FASN), the key enzyme in de novo fatty-acid synthesis, whereby Vitis vinifera L. cv Vermentino leaf hydroalcoholic extract lowers its protein stability that is regulated by small ubiquitin-like modifier (SUMO)ylation. The phenolic compounds characterization was performed by liquid chromatography–mass spectrometry (LC–MS), whereas mass spectrometry (LC–MS/MS), Western blotting/co-immunoprecipitation (Co-IP) and RT-PCR, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), clonogenicity assays, and FACS analysis were used to measure the expression of targets and tumorigenicity. Vermentino extract exhibits antitumorigenic effects, and we went on to determine that FASN and ubiquitin-conjugating enzyme 9 (UBC9), the sole E2 enzyme required for SUMOylation, were significantly reduced. Moreover, FASN was found SUMOylated in human breast cancer tissues and cell lines, and lack of SUMOylation caused by SUMO2 silencing reduced FASN protein stability. These results suggest that SUMOylation protects FASN against proteasomal degradation and may exert oncogenic activity through alteration of lipid metabolism, whereas Vermentino extract inhibits these effects which supports the additional validation of the therapeutic value of this compound in breast cancer.
Collapse
|
29
|
Roy S, Singh M, Rawat A, Kumar D, Kaithwas G. Mitochondrial apoptosis and curtailment of hypoxia-inducible factor-1α/fatty acid synthase: A dual edge perspective of gamma linolenic acid in ER+ mammary gland cancer. Cell Biochem Funct 2020; 38:591-603. [PMID: 32207176 DOI: 10.1002/cbf.3513] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/22/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
Gamma linolenic acid is a polyunsaturated fatty acid having selective anti-tumour properties with negligible systemic toxicity. In the present study, the anti-cancer potential of gamma linolenic acid and its effects on mitochondrial as well as hypoxia-associated marker was evaluated. The effect of gamma linolenic acid was scrutinised against ER + MCF-7 cells by using fluorescence microscopy, JC-1 staining, dot plot assay and cell cycle analysis. The in vitro results were also confirmed using carcinogen (n-methyl-n-nitrosourea) induced in vivo model. The early and late apoptotic signals in the conjugation with mitochondrial depolarisation were found once scrutinised through mitochondrial membrane potential and life death staining after gamma linolenic acid treatment. Gamma linolenic acid arrested the cell cycle in G0/G1 phase with the majority of cell populations in the early apoptotic stage. The translocation of phosphatidylserine was studied through annexin-V FITC dot plot assay. The markers of cellular proliferation (decreased alveolar bud count, histopathological architecture restoration and loss of tumour micro-vessels) were diminished after gamma linolenic acid treatment. Gamma linolenic acid ameliorates the biological effects of n-methyl-n-nitrosourea persuading the mitochondrial mediated death pathway and impeding the hypoxic microenvironment to make a halt in palmitic acid synthesis. SIGNIFICANCE: The present study elaborates the effect of gamma linolenic acid on mammary gland cancer by following mitochondrial-mediated death apoptosis pathway. Gamma linolenic acid also inhibits cell-wall synthesis by the curtailment of HIF-1α and FASN level in mammary gland cancer.
Collapse
Affiliation(s)
- Subhadeep Roy
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, Uttar Pradesh, India
| | - Manjari Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, Uttar Pradesh, India
| | - Atul Rawat
- Centre for Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus, Lucknow, Uttar Pradesh, India
| | - Dinesh Kumar
- Centre for Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus, Lucknow, Uttar Pradesh, India
| | - Gaurav Kaithwas
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, Uttar Pradesh, India
| |
Collapse
|
30
|
Kim S, Li Y, Lin L, Sayasith PR, Tarr AT, Wright EB, Yasmin S, Lannigan DA, O'Doherty GA. Synthesis and Biological Evaluation of 4'-Substituted Kaempfer-3-ols. J Org Chem 2020; 85:4279-4288. [PMID: 32056430 DOI: 10.1021/acs.joc.9b03461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The synthesis of two series of five kaempfer-3-ols was described. The first set all have a C-3 hydroxyl group and the second has a carboxymethoxy ether at the C-3 position. Both series have variable substitution at the C-4' position (i.e., OH, Cl, F, H, OMe). Both kaempferols and carboxymethoxy ethers were evaluated for their ability to inhibit ribosomal s6 kinase (RSK) activity and cancer cell proliferation.
Collapse
Affiliation(s)
- Sugyeom Kim
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Yu Li
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Lin Lin
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Peyton R Sayasith
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Ariel T Tarr
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | | | | | | | - George A O'Doherty
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
31
|
Effect of S-Allyl -L-Cysteine on MCF-7 Cell Line 3-Mercaptopyruvate Sulfurtransferase/Sulfane Sulfur System, Viability and Apoptosis. Int J Mol Sci 2020; 21:ijms21031090. [PMID: 32041330 PMCID: PMC7037078 DOI: 10.3390/ijms21031090] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/24/2020] [Accepted: 02/04/2020] [Indexed: 12/24/2022] Open
Abstract
The S-Allyl-L-cysteine (SAC) component of aged garlic extract (AGE) is proven to have anticancer, antihepatotoxic, neuroprotective and neurotrophic properties. γ-Cystathionase (CTH), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (MPST) are involved in H2S/sulfane sulfur endogenous formation from L-cysteine. The aim of the study was to determine the effect of SAC on MCF-7 cells survival and apoptosis, which is a widely known approach to reduce the number of cancer cells. An additional goal of this paper was to investigate the effect of SAC on the activity and expression of enzymes involved in H2S production. The experiments were carried out in the human breast adenocarcinoma cell line MCF-7. Changes in the cell viability were determined by MTT assay. Cell survival was determined by flow cytometry (FC). Changes in enzymes expression were analyzed using Western blot. After 24 h and 48 h incubation with 2245 µM SAC, induction of late apoptosis was observed. A decrease in cell viability was observed with increasing SAC concentration and incubation time. SAC had no significant cytotoxic effect on the MCF-7 cells upon all analyzed concentrations. CTH, MPST and CBS expression were confirmed in non-treated MCF-7 cells. Significant decrease in MPST activity at 2245 µM SAC after 24 h and 48 h incubation vs. 1000 µM SAC was associated with decrease in sulfane sulfur levels. The presented results show promising SAC effects regarding the deterioration of the MCF-7 cells’ condition in reducing their viability through the downregulation of MPST expression and sulfate sulfur level reduction.
Collapse
|
32
|
A Natural Quinazoline Derivative from Marine Sponge Hyrtios erectus Induces Apoptosis of Breast Cancer Cells via ROS Production and Intrinsic or Extrinsic Apoptosis Pathways. Mar Drugs 2019; 17:md17120658. [PMID: 31771152 PMCID: PMC6950652 DOI: 10.3390/md17120658] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/23/2019] [Accepted: 10/23/2019] [Indexed: 12/20/2022] Open
Abstract
Here, we report the therapeutic potential of a natural quinazoline derivative (2-chloro-6-phenyl-8H-quinazolino[4,3-b]quinazolin-8-one) isolated from marine sponge Hyrtios erectus against human breast cancer. The cytotoxicity of the compound was investigated on a human breast carcinoma cell line (MCF-7). Antiproliferative activity of the compound was estimated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. MTT assay showed significant inhibition of MCF-7 cells viability with the IC50 value of 13.04 ± 1.03 µg/mL after 48 h. The compound induced down-regulation of anti-apoptotic Bcl-2 protein and increase in the pro-apoptotic Bax/Bcl-2 ratio in MCF-7 cells. The compound activated the expression of Caspases-9 and stimulated downstream signal transducer Caspase-7. In addition, Caspase-8 showed remarkable up-regulation in MCF-7 cells treated with the compound. Moreover, the compound was found to promote oxidative stress in MCF-7 cells that led to cell death. In conclusion, the compound could induce apoptosis of breast carcinoma cells via a mechanism that involves ROS production and either extrinsic or intrinsic apoptosis pathways. The systemic toxic potential of the compound was evaluated in an in vivo mouse model, and it was found non-toxic to the major organs.
Collapse
|
33
|
Novel Antrodia cinnamomea Extract Reduced Cancer Stem-Like Phenotype Changes and Resensitized KRAS-Mutant Colorectal Cancer via a MicroRNA-27a Pathway. Cancers (Basel) 2019; 11:cancers11111657. [PMID: 31717759 PMCID: PMC6896121 DOI: 10.3390/cancers11111657] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/14/2019] [Accepted: 10/23/2019] [Indexed: 01/20/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common causes of death in Taiwan. Previous studies showed that Antrodia cinnamomea (AC) can treat poisoning, diarrhea, and various types of cancer. Therefore, we purified a novel ubiquinone derivative, AC009, and investigated its antitumor effects. Cell viability assays revealed that AC009 reduced the viability of several human CRC cell lines. AC009 treatment resulted in cell-cycle arrest/apoptosis, and these effects may occur via caspase and Bcl-2 signaling pathways. We demonstrated that AC009 could significantly inhibit in vivo tumor growth in xenograft mouse models. Using messenger RNA (mRNA) and microRNA (miRNA) microarrays, we found that KRAS gene expression was also regulated by AC009, possibly through specific miRNAs. AC009 also reduced cancer stem-cell marker CD44+/CD24+ expression and restored the tumor inhibition effect of cetuximab in KRAS-mutant CRC. Moreover, we found that miRNA-27a could restore the tumor inhibition effect of cetuximab in KRAS-mutant CRC cells. Taken together, our results suggest that AC009 has therapeutic potential against human wild-type and KRAS-mutant CRC.
Collapse
|
34
|
Gimenes L, Batista Junior JM, Martins Dos Santos Junior F, da Silva Souza M, Luna-Dulcey L, Ellena JE, Cominetti MR, Fatima das Graças Fernandes da Silva M, Vieira PC, Fernandes JB, Staerk D. Picraviane A and B: Nortriterpenes with limonoid-like skeletons containing a heptanolide E-ring system from Picramnia glazioviana. PHYTOCHEMISTRY 2019; 163:38-45. [PMID: 31003042 DOI: 10.1016/j.phytochem.2019.03.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 06/09/2023]
Abstract
Two highly oxygenated nortriterpenes, picraviane A and B, were isolated from the ethanolic extract of Picramnia glazioviana Engl. The structures were determined by analysis of HRMS and 2D NMR spectroscopic data. Single-crystal X-ray diffraction data was also obtained for picraviane B. The absolute configuration of both compounds were assigned by comparison of experimental and calculated vibrational and electronic circular dichroism spectroscopy, respectively. Picraviane A showed a moderate cytotoxic activity against the triple negative MDA-MB-231 breast cancer cell line. These compounds represent an undescribed class of natural products with limonoid-like skeletons containing a heptanolide as the E-ring.
Collapse
Affiliation(s)
- Leila Gimenes
- Department of Chemistry, Federal University of Sao Carlos (UFSCar), 13565-905, Sao Carlos, SP, Brazil; Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100, Copenhagen, Denmark
| | - Joao Marcos Batista Junior
- Department of Chemistry, Federal University of Sao Carlos (UFSCar), 13565-905, Sao Carlos, SP, Brazil; Institute of Science and Technology, Federal University of Sao Paulo, 12231-280, Sao Jose dos Campos, SP, Brazil
| | - Fernando Martins Dos Santos Junior
- Department of Chemistry, Federal University of Sao Carlos (UFSCar), 13565-905, Sao Carlos, SP, Brazil; Department of Organic Chemistry, Chemistry Institute, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Matheus da Silva Souza
- São Carlos Institute of Physics, University of São Paulo, 13566-590, São Carlos, SP, Brazil
| | - Liany Luna-Dulcey
- Department of Gerontology, Federal University of Sao Carlos, 13565-905, Sao Carlos, SP, Brazil
| | - Javier Esteves Ellena
- São Carlos Institute of Physics, University of São Paulo, 13566-590, São Carlos, SP, Brazil
| | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of Sao Carlos, 13565-905, Sao Carlos, SP, Brazil
| | | | - Paulo Cezar Vieira
- Department of Chemistry, Federal University of Sao Carlos (UFSCar), 13565-905, Sao Carlos, SP, Brazil; School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, 14040-903, Ribeirao Preto, SP, Brazil
| | - Joao Batista Fernandes
- Department of Chemistry, Federal University of Sao Carlos (UFSCar), 13565-905, Sao Carlos, SP, Brazil
| | - Dan Staerk
- Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
35
|
Naz H, Tarique M, Ahamad S, Alajmi MF, Hussain A, Rehman MT, Luqman S, Hassan MI. Hesperidin-CAMKIV interaction and its impact on cell proliferation and apoptosis in the human hepatic carcinoma and neuroblastoma cells. J Cell Biochem 2019; 120:15119-15130. [PMID: 31021496 DOI: 10.1002/jcb.28774] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/08/2019] [Accepted: 01/14/2019] [Indexed: 01/11/2023]
Abstract
Calcium/calmodulin-dependent protein kinase IV (CAMKIV) is a key regulatory molecule of cell signaling, and thereby controls its growth and proliferation, including expression of certain genes. The overexpression of CAMKIV is directly associated with the development of different types of cancers. Hesperidin is abundantly found in citrus fruits and exhibits wide range of pharmacological activities including anti-inflammatory, antibacterial and anticancerous effects. We have investigated binding mechanism of hesperidin with the CAMKIV using molecular docking methods followed by fluorescence quenching and isothermal titration calorimetric assays. An appreciable binding affinity of hesperidin was observed with CAMKIV during fluorescence quenching and isothermal titration calorimetric studies. Efficacy of hesperidin to inhibit the growth of human hepatic carcinoma (HepG2) and neuroblastoma (SH-SY5Y) cancer cell lines were investigated. Hesperidin has significantly reduced the proliferation of HepG2 and SH-SY5Y cells and induces apoptosis by activating the caspase-3-dependent intrinsic pathway through the upregulation of proapoptotic Bax protein. Hesperidin treatment reduces the mitochondrial membrane potential of HepG2 and SH-SY5Y cells. All these observations clearly anticipated hesperidin a potent inhibitor of CAMKIV which may be further exploited a newer therapeutic approach for the management of different cancer types.
Collapse
Affiliation(s)
- Huma Naz
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Mohd Tarique
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Shahzaib Ahamad
- Department of Biotechnology, College of Engineering & Technology, IFTM University, Delhi Road, Moradabad, India
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Suaib Luqman
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
36
|
Thangarasu R, Pachaiappan P, Subbaiyan T. Anti-Estrogenic and Anti-Cell Proliferative Effect of Allyl Isothiocyanate in Chemoprevention of Chemically Induced Mammary Carcinogenesis in Rats. Pathol Oncol Res 2019; 26:913-925. [PMID: 30895454 DOI: 10.1007/s12253-019-00638-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/11/2019] [Indexed: 10/27/2022]
Abstract
The anti-estrogenic and anti-cell proliferative effect of allyl isothiocyanate (AITC) was carried out by analyzing the status of sex hormones and its receptors and cell proliferative markers in chemically induced mammary carcinogenesis in rats. Mammary tumor was induced by a single dose of DMBA (25 mg/rat) and MNU (50 mg/kg bw) injected subcutaneously near mammary gland. RT-PCR, western blotting and immunohistochemical analysis of mammary tissues show an upregulation of ER-α, PR, aromatase, PCNA, cyclin D1 and AgNORs staining and down regulation of p53 expression as well as plasma estradiol, prolactin and testosterone levels increased in DMBA and MNU-induced tumor bearing rats. Oral administration of AITC at a dose of 20 mg/kg bw restored the levels of sex hormones and its receptors, aromatase, cell proliferative markers and AgNORs staining near to normal levels. Molecular docking studies also supported these findings. The results suggest that anti-estrogenic and anti-proliferative effect of AITC prevent the development of DMBA and MNU-induced mammary carcinogenesis in rat.
Collapse
Affiliation(s)
- Rajakumar Thangarasu
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, 608 002, India
| | - Pugalendhi Pachaiappan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, 608 002, India.
| | - Thilagavathi Subbaiyan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, 608 002, India
| |
Collapse
|
37
|
Maruszewska A, Tarasiuk J. Antitumour effects of selected plant polyphenols, gallic acid and ellagic acid, on sensitive and multidrug-resistant leukaemia HL60 cells. Phytother Res 2019; 33:1208-1221. [PMID: 30838722 DOI: 10.1002/ptr.6317] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 01/18/2019] [Accepted: 01/24/2019] [Indexed: 12/30/2022]
Abstract
The aim of this study was to examine the antitumour effects of plant phenolic acids, gallic acid (GA) and ellagic acid (EA), on human promyelocytic leukaemia sensitive HL60 cell line and its resistant sublines exhibiting two MDR phenotypes: HL60/VINC (overexpressing P-glycoprotein) and HL60/MX2 (characterized by the presence of mutated α isoform of topoisomerase II). Both studied compounds exerted comparable cytotoxic activities towards sensitive HL60 cells and their MDR counterparts. It was also found that GA and EA modulated the cellular level of reactive oxygen species in a dose-dependent and time-dependent manner. Furthermore, it was demonstrated that GA (IC90 ) and EA (IC50 and IC90 ) significantly increased the percentage of sub-G1 subpopulation of all studied leukaemia cells causing oligonucleosomal DNA fragmentation. Both compounds used at IC90 triggered mainly the apoptotic death of these cells. However, GA had no effect on the activity of caspase-3 as well as caspase-8 in sensitive HL60 cells and their MDR counterparts. In contrast, EA provoked a significant activation of these caspases in all studied leukaemia cells. It was also found that lysosomes were not involved in triggering programmed death of sensitive HL60 and MDR cells by GA and EA.
Collapse
Affiliation(s)
- Agnieszka Maruszewska
- Department of Biochemistry, Faculty of Biology, University of Szczecin, 3c Felczaka St, Szczecin, 71-412, Poland.,Molecular Biology and Biotechnology Center, Faculty of Biology, University of Szczecin, 13 Wąska St, Szczecin, 71-415, Poland
| | - Jolanta Tarasiuk
- Department of Biochemistry, Faculty of Biology, University of Szczecin, 3c Felczaka St, Szczecin, 71-412, Poland.,Molecular Biology and Biotechnology Center, Faculty of Biology, University of Szczecin, 13 Wąska St, Szczecin, 71-415, Poland
| |
Collapse
|
38
|
Pan X, Lin Z, Jiang D, Yu Y, Yang D, Zhou H, Zhan D, Liu S, Peng G, Chen Z, Yu Z. Erastin decreases radioresistance of NSCLC cells partially by inducing GPX4-mediated ferroptosis. Oncol Lett 2019; 17:3001-3008. [PMID: 30854078 PMCID: PMC6365906 DOI: 10.3892/ol.2019.9888] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 11/16/2018] [Indexed: 12/23/2022] Open
Abstract
The aim of the present study was to examine whether erastin influences radioresistance in non-small cell lung cancer (NSCLC) cells and produce a preliminary investigation into its mechanism of action. The radioresistant subtype of NSCLC cells, A549-R and H460-R, were induced by high-dose hypofractionated irradiation. Erastin was used to treat the radioresistant cells and radiosensitivity was examined by colony formation assays. Cell death was determined after the cells were treated with erastin, irradiation (IR) or erastin together with IR. The expression of glutathione peroxidase 4 (GPX4) expression in the parental cells and radioresistance cells was detected by western blotting. GPX4 expression in the radioresistance cells was subsequently inhibited, radiosensitivity and cell death was measured, and erastin enhanced radiosensitivity in A549-R and H460-R cells. Erastin and IR exhibited a combined effect on killing cells, as co-treatment with erastin and IR demonstrated a higher effect on killing cells compared with erastin or IR alone. GPX4 expression was inhibited by erastin in the radioresistant cells. Inhibiting GPX4 expression also radiosensitized NSCLC cells to radiation in the radioresistant cell lines. Erastin-induced and GPX4-inhibition-induced cell death could partially be rescued by deferoxamine, but not Z-VAD-FMK and olaparib, which indicated that erastin and GPX4-inhibition induced ferroptosis in the radioresistant cells. Erastin decreased radioresistance of NSCLC cells partially by inducing GPX4-mediated ferroptosis.
Collapse
Affiliation(s)
- Xiaofen Pan
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518000, P.R. China
| | - Zhixiu Lin
- Department of Pharmacy, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| | - Danxian Jiang
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| | - Ying Yu
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| | - Donghong Yang
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| | - Hechao Zhou
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| | - Dechao Zhan
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| | - Sha Liu
- Department of Oncology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| | - Gang Peng
- Department of Head and Neck Cancer, Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zihong Chen
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| | - Zhonghua Yu
- Department of Head and Neck Cancer, Oncology Center, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524000, P.R. China
| |
Collapse
|
39
|
Ray A, Jena S, Dash B, Sahoo A, Kar B, Patnaik J, Panda PC, Nayak S, Mahapatra N. Hedychium coronarium extract arrests cell cycle progression, induces apoptosis, and impairs migration and invasion in HeLa cervical cancer cells. Cancer Manag Res 2019; 11:483-500. [PMID: 30655700 PMCID: PMC6322495 DOI: 10.2147/cmar.s190004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Hedychium coronarium Koen. (Zingiberaceae) is traditionally used as medicine in countries such as India, China, and Vietnam for treatment of various ailments including cancer. However, in spite of its implied significance in cancer treatment regimes, there are no reports so far involving the anticancerous attributes of H, coronarium ethanol extract (HCEE) on cancer cells and a more comprehensive study on its mechanism is still lacking. Materials and methods The cytotoxicity of HCEE was evaluated by MTT and clonogenic survival assay. Annexin V/propidium iodide (PI), Hoechst 33342 staining, and TUNEL assay were performed to detect apoptosis. Cell cycle analysis was performed using PI staining. JC-1 and 2′,7′-dichlorodihydrofluorescein diacetate assay were used to check the levels of MMP and ROS, respectively. Western blot analysis was carried out to measure the expression levels of proteins. Migration and invasion activity were assessed by wound healing and Transwell membrane assay, respectively. Results Antiproliferative effect of HCEE was investigated in various cancerous and normal cell lines. Among these, HCEE significantly inhibited the survival of HeLa cells without affecting the viability of normal human umbilical vein endothelial cells. Annexin V/PI, Hoechst staining, and TUNEL assay showed HCEE induced apoptosis in HeLa cells in a dose-dependent manner. HCEE promoted cell cycle arrest at G1 phase in HeLa cells by upregulating the levels of p53 and p21 and downregulating the levels of cyclin D1, CDK-4, and CDK-6. Moreover, HCEE treatment upregulated the expression of Bax and downregulated the expression of Bcl-2. Additionally, HCEE activated the caspase cascade by increasing the activities of caspase-9, caspase-8, and caspase-3. The expression levels of Fas ligand and Fas were also upregulated. Further, HCEE inhibited the migratory potential of HeLa cells by downregulating MMP-2 and MMP-9 expression levels. Conclusion Our results indicate H. coronarium exerts antiproliferative and apoptotic effects against HeLa cells, and therefore may be used for treatment against cervical cancer.
Collapse
Affiliation(s)
- Asit Ray
- Regional Medical Research Centre (Indian Council of Medical Research), Chandrasekharpur, Bhubaneswar 751023, Odisha, India.,Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Sudipta Jena
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Biswabhusan Dash
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Ambika Sahoo
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Basudeba Kar
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Jeetendranath Patnaik
- Department of Botany, Sri Krushna Chandra Gajapati College, Paralakhemundi 761200, Odisha, India
| | - Pratap Chandra Panda
- Taxonomy and Conservation Division, Regional Plant Resource Centre, Nayapalli, Bhubaneswar 751015, Odisha, India
| | - Sanghamitra Nayak
- Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha O Anusandhan University, Kalinganagar, Ghatikia, Bhubaneswar 751003, Odisha, India,
| | - Namita Mahapatra
- Regional Medical Research Centre (Indian Council of Medical Research), Chandrasekharpur, Bhubaneswar 751023, Odisha, India
| |
Collapse
|
40
|
Foo JB, Ng LS, Lim JH, Tan PX, Lor YZ, Loo JSE, Low ML, Chan LC, Beh CY, Leong SW, Saiful Yazan L, Tor YS, How CW. Induction of cell cycle arrest and apoptosis by copper complex Cu(SBCM)2 towards oestrogen-receptor positive MCF-7 breast cancer cells. RSC Adv 2019; 9:18359-18370. [PMID: 35515266 PMCID: PMC9064738 DOI: 10.1039/c9ra03130h] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/03/2019] [Indexed: 12/29/2022] Open
Abstract
Copper complexes have the potential to be developed as targeted therapy for cancer because cancer cells take up larger amounts of copper than normal cells. Copper complex Cu(SBCM)2 has been reported to induce cell cycle arrest and apoptosis towards triple-negative breast cancer cells. Nevertheless, its effect towards other breast cancer subtypes has not been explored. Therefore, the present study was conducted to investigate the effect of Cu(SBCM)2 towards oestrogen-receptor positive MCF-7 breast cancer cells. Growth inhibition of Cu(SBCM)2 towards MCF-7 and human non-cancerous MCF-10A breast cells was determined by MTT assay. Morphological changes of Cu(SBCM)2-treated-MCF-7 cells were observed under an inverted microscope. Annexin V/PI apoptosis assay and cell cycle analysis were evaluated by flow cytometry. The expression of wild-type p53 protein was evaluated by Western blot analysis. The intracellular ROS levels of MCF-7 treated with Cu(SBCM)2 were detected using DCFH-DA under a fluorescence microscope. The cells were then co-treated with Cu(SBCM)2 and antioxidants to evaluate the involvement of ROS in the cytotoxicity of Cu(SBCM)2. Docking studies of Cu(SBCM)2 with DNA, DNA topoisomerase I, and human ribonucleotide reductase were also performed. The growth of MCF-7 cells was inhibited by Cu(SBCM)2 in a dose-dependent manner with less toxicity towards MCF-10A cells. It was found that Cu(SBCM)2 induced G2/M cell cycle arrest and apoptosis in MCF-7 cells, possibly via a p53 pathway. Induction of intracellular ROS was not detected in MCF-7 cells. Interestingly, antioxidants enhance the cytotoxicity of Cu(SBCM)2 towards MCF-7 cells. DNA topoisomerase I may be the most likely target that accounts for the cytotoxicity of Cu(SBCM)2. Cu(SBCM)2 binds to DNA topoisomerase I, which, in turn, induces cell cycle arrest and apoptosis in MCF-7 breast cancer cells, possibly via p53 signalling pathway.![]()
Collapse
Affiliation(s)
- Jhi Biau Foo
- Faculty of Pharmacy
- MAHSA University
- Malaysia
- School of Pharmacy
- Faculty of Health & Medical Sciences
| | - Li Shan Ng
- Faculty of Pharmacy
- MAHSA University
- Malaysia
| | - Ji Hui Lim
- Faculty of Pharmacy
- MAHSA University
- Malaysia
| | | | | | - Jason Siau Ee Loo
- School of Pharmacy
- Faculty of Health & Medical Sciences
- Taylor's University
- Malaysia
| | - May Lee Low
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- International Medical University
- Kuala Lumpur
- Malaysia
| | - Lee Chin Chan
- Virology Lab 1
- Faculty of Biotechnology and Biomolecular Sciences
- Universiti Putra Malaysia (UPM)
- Malaysia
| | - Chaw Yee Beh
- Laboratory of Vaccines and Immunotherapeutics
- Institute of Bioscience
- Universiti Putra Malaysia (UPM)
- Malaysia
| | - Sze Wei Leong
- Virology Lab 1
- Faculty of Biotechnology and Biomolecular Sciences
- Universiti Putra Malaysia (UPM)
- Malaysia
| | - Latifah Saiful Yazan
- Laboratory of Molecular Biomedicine
- Institute of Bioscience
- Universiti Putra Malaysia
- Malaysia
- Department of Biomedical Sciences
| | - Yin Sim Tor
- School of Biosciences
- Faculty of Health & Medical Sciences
- Taylor's University
- Malaysia
| | | |
Collapse
|
41
|
Chiou CS, Wu CM, Dubey NK, Lo WC, Tsai FC, Tung TDX, Hung WC, Hsu WC, Chen WH, Deng WP. Mechanistic insight into hyaluronic acid and platelet-rich plasma-mediated anti-inflammatory and anti-apoptotic activities in osteoarthritic mice. Aging (Albany NY) 2018; 10:4152-4165. [PMID: 30582743 PMCID: PMC6326674 DOI: 10.18632/aging.101713] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/06/2018] [Indexed: 12/17/2022]
Abstract
Osteoarthritis (OA) poses a major clinical challenges owing to limited regenerative ability of diseased or traumatized chondrocytes in articular cartilage. Previous studies have determined the individual therapeutic efficacies of hyaluronic acid (HA) and platelet-rich plasma (PRP) on OA; however, the underlying mechanism is still lacking. Therefore, we investigated mechanistic approach of HA+PRP therapy on chondrocyte apoptosis in IL-1β+TNF-α (I+T) treated in vitro OA model, in addition to in vivo anterior cruciate ligament transection-OA mice model. MTT assay showed an enhanced chondrocyte proliferation and viability in HA+PRP-treated group, compared to I+T, I+T/HA, I+T/PRP, I+T/HA+PRP groups. Further, HA+PRP also significantly suppressed ROS, apoptotic cleaved caspase-3 and PARP, p53 and p21 and MMP-1; whereas, cell cycle modulatory proteins including p-ERK, cyclin B1, D1, and E2 were upregulated. The sub-G1 population and TUNEL assay confirmed the higher abundance of healthy chondrocytes in HA+PRP group. A significantly decreased ARS staining in HA+PRP group was also noted, indicating reduced cartilaginous matrix mineralization compared to other groups. Conclusively, compared to HA or PRP, the combined HA+PRP might be a promising therapy for articular cartilage regeneration in osteoarthritic pathology, possibly via augmented anti-inflammatory, anti-oxidative chondrocyte proliferation and inhibited MMP-1 activity and matrix calcification.
Collapse
Affiliation(s)
- Chi-Sheng Chiou
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chi-Ming Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Wen-Cheng Lo
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Feng-Chou Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tran Dang Xuan Tung
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Stem Cells Center, Van Hanh General Hospital, Ho Chi Minh City, Vietnam
| | - Wei-Ching Hung
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taiwan
| | - Wei-Che Hsu
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taiwan
| | - Wei-Hong Chen
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taiwan
| | - Win-Ping Deng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taiwan
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, Taipei, Taiwan
| |
Collapse
|
42
|
Manilkara zapota (L.) P. Royen leaf water extract triggered apoptosis and activated caspase-dependent pathway in HT-29 human colorectal cancer cell line. Biomed Pharmacother 2018; 110:748-757. [PMID: 30554113 DOI: 10.1016/j.biopha.2018.12.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 01/08/2023] Open
Abstract
Manilkara zapota (L.) P. Royen (Family: Sapotaceae), commonly called as sapodilla, has been applied as traditional folk medicine for diarrhea and pulmonary infections. Conventional therapy in colorectal cancer is not likely effective due to undesirable outcomes. The anti-colon cancer properties of Manilkara zapota leaf water extract have yet to be investigated thus far. Therefore, our present study aimed to evaluate the ability to induce apoptosis and the underlying mechanisms of Manilkara zapota leaf water extract against human colorectal cancer (HT-29) cells. The cytotoxicity of Manilkara zapota leaf water extract was screened in different cancer cell lines using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) analyses. The morphological changes in HT-29 cell lines after exposure to Manilkara zapota leaf water extract were viewed under fluorescence and inverted light microscope. The apoptotic cell was measured by Annexin V-propidium iodide staining. The caspase-3 and -8 activities were assessed by colorimetric assay. Overall analyses revealed that treatment with Manilkara zapota leaf water extract for 72 h can inhibit the viability of HT-29 cells. Incubation with Manilkara zapota leaf water extract for 24, 48, and 72 h significantly increased (p < 0.05) the total apoptotic cells compared to the control. Treatment with 21, 42, and 84 μg/mL of Manilkara zapota leaf water extract for 72 h triggered both caspase-3 and -8 activities in a concentration-dependent pattern. We also found that the catalase level in the two treatment groups (21 and 42 μg/mL) was significantly elevated after 24 h incubation. Incubation with Manilkara zapota leaf water extract for 72 h triggered the transcriptional elevation of the adenomatous polyposis coli (APC), glycogen synthase kinase 3β (GSK3β), AXIN1, and casein kinase 1 (CK1). The β-catenin mRNA levels were reduced accordingly when the concentration of the Manilkara zapota leaf water extract was increased. Our results suggested that Manilkara zapota leaf water extract offer great potential against colorectal cancer through modulation of Wnt/β-catenin signaling pathway, caspase-dependent pathway, and antioxidant enzyme.
Collapse
|
43
|
de Carvalho Melo-Cavalcante AA, da Rocha Sousa L, Alencar MVOB, de Oliveira Santos JV, da Mata AMO, Paz MFCJ, de Carvalho RM, Nunes NMF, Islam MT, Mendes AN, Gonçalves JCR, da Silva FCC, Ferreira PMP, de Castro E Sousaa JM. Retinol palmitate and ascorbic acid: Role in oncological prevention and therapy. Biomed Pharmacother 2018; 109:1394-1405. [PMID: 30551390 DOI: 10.1016/j.biopha.2018.10.115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/14/2022] Open
Abstract
Cancer development has been directly related to oxidative stress. During chemotherapy, some cancer patients use dietary antioxidants to avoid nutritional deficiencies due to cancer treatment. Among the antioxidants consumed, there are vitamins, including retinyl palmitate (PR) and ascorbic acid (AA), which have the capacity to reduce free radicals formation, protect cellular structures and maintain the cellular homeostasis. This systematic review evaluated the antioxidant and antitumor mechanisms of retinol palmitate (a derivative of vitamin A) and/or ascorbic acid (vitamin C) in cancer-related studies. Ninety-seven (97) indexed articles in the databases PubMed and Science Direct, published between 2013 and 2017, including 23 clinical studies (5 for every single compound while 13 in interaction) and 74 non-clinical studies (37 for retinol palmitate, 36 for ascorbic acid and 1 in interaction) were considered. Antioxidant and antitumor effects, with controversies over dosage and route of administration, were observed for the test compounds in their isolated form or associated in clinical studies. Prevention of cancer risks against oxidative damage was seen in lower doses of retinol palmitate and/or vitamin C. However, at high doses, they can generate reactive oxygen species, cytotoxicity and apoptosis in test systems. Non-clinical studies using cell lines have allowed understanding the mechanisms related to antioxidants and antitumor effects of the isolated compounds, however, studies on vitamin interactions, acting as antioxidants and/or antitumor are still rare and controversial. More studies, mainly related to modulation of antineoplastic drugs are needed for understanding the risks and benefits of their use during treatment in order to achieve effectiveness in cancer therapy and patient's quality of life.
Collapse
Affiliation(s)
- Ana Amélia de Carvalho Melo-Cavalcante
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Leonardo da Rocha Sousa
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Marcus Vinícius Oliveira Barros Alencar
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - José Victor de Oliveira Santos
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Ana Maria Oliveira da Mata
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Márcia Fernanda Correia Jardim Paz
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Ricardo Melo de Carvalho
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Nárcia Mariana Fonseca Nunes
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Muhammad Torequl Islam
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Anderson Nogueira Mendes
- Department of Biophysics and Physiology of Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Juan Carlos Ramos Gonçalves
- Department of Biochemistry and Pharmacology, Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - Felipe Cavalcanti Carneiro da Silva
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam; Department of Biological Sciences, Federal University of Piauí, Picos, Piauí, 64.067-670, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Postgraduate Program in Pharmaceutical Sciences. Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Department of Biophysics and Physiology of Federal University of Piauí, Teresina, Piauí, 64.049-550, Brazil; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam
| | - João Marcelo de Castro E Sousaa
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City-700000, Vietnam; Department of Biological Sciences, Federal University of Piauí, Picos, Piauí, 64.067-670, Brazil.
| |
Collapse
|
44
|
DEPDC1, negatively regulated by miR-26b, facilitates cell proliferation via the up-regulation of FOXM1 expression in TNBC. Cancer Lett 2018; 442:242-251. [PMID: 30419349 DOI: 10.1016/j.canlet.2018.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/03/2018] [Accepted: 11/03/2018] [Indexed: 01/18/2023]
Abstract
Triple negative breast cancer (TNBC), characterized by lack of estrogen receptors, progesterone hormone receptors, and HER2 overexpression, is a more aggressive high grade tumor and not sensitive to current targeted drugs. The clinical prognosis of TNBC is poorer than other types of breast cancer, and there is no effective therapy strategy until now. Thus, it is necessary to determine important factors involved in regulating the progression of TNBC. In this study, we found DEPDC1 was up-regulated in the tissues of TNBC compared with their paired peritumoral tissues. DEPDC1 over-expression facilitated cell proliferation and tumor growth through increasing the expression of FOXM1 in TNBC cells. Conversely, knockdown of DEPDC1 had the opposite effects. Moreover, miR-26b, acting as a tumor suppressor in TNBC, directly repressed the expression of DEPDC1 and mitigated its promotive effects on cell growth and colony formation. These results indicate that DEPDC1, negatively regulated by miR-26b, promotes cell proliferation and tumor growth via up-regulating FOXM1 expression, implying an important underlying mechanism of regulating the progression of TNBC.
Collapse
|
45
|
Manilkara zapota (L.) P. Royen Leaf Water Extract Induces Apoptosis in Human Hepatocellular Carcinoma (HepG2) Cells via ERK1/2/Akt1/JNK1 Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:7826576. [PMID: 30519270 PMCID: PMC6241369 DOI: 10.1155/2018/7826576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/05/2018] [Accepted: 10/22/2018] [Indexed: 01/27/2023]
Abstract
Manilkara zapota (L.) P. Royen, called sapodilla, or locally known as ciku, belongs to the family Sapotaceae. We found that Manilkara zapota leaf water extract has cytotoxic effect against human hepatocellular carcinoma (HepG2) cell line in our earlier study. Therefore, this study aimed to explore the anticancer properties of Manilkara zapota leaf water extract in HepG2 cells. We also aimed to unravel yet undiscovered mechanisms and identified several expressed genes whose functions in cytotoxicity activity of Manilkara zapota leaf water extract in HepG2 cells have not been well-studied. The apoptosis and intracellular reactive oxygen species (ROS) activities were analyzed using Annexin V-propidium iodide staining and dichlorodihydrofluorescein diacetate, respectively, by NovoCyte Flow Cytometer. Bax and Bcl-2 expression were assessed using Enzyme-Linked Immunosorbent Assay. The associated molecular pathways were evaluated by quantitative real-time PCR. Overall analyses revealed that Manilkara zapota leaf water extract can increase percentage of early apoptotic cells, induce the formation of ROS, upregulate c-Jun N-terminal kinase 1 (JNK1) and inducible nitric oxide synthase (iNOS), and reduce Akt1 and vascular endothelial growth factor A (VEGFA) transcriptional activities. Our data suggest that Manilkara zapota leaf water extract can suppress the growth of HepG2 cells via modulation of ERK1/2/Akt1/JNK1 transcriptional expression.
Collapse
|
46
|
Tan BL, Norhaizan ME, Chan LC. An Intrinsic Mitochondrial Pathway Is Required for Phytic Acid-Chitosan-Iron Oxide Nanocomposite (Phy-CS-MNP) to Induce G₀/G₁ Cell Cycle Arrest and Apoptosis in the Human Colorectal Cancer (HT-29) Cell Line. Pharmaceutics 2018; 10:pharmaceutics10040198. [PMID: 30360519 PMCID: PMC6321496 DOI: 10.3390/pharmaceutics10040198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 01/10/2023] Open
Abstract
Magnetic iron oxide nanoparticles are among the most useful metal nanoparticles in biomedical applications. A previous study had confirmed that phytic acid-chitosan-iron oxide nanocomposite (Phy-CS-MNP) exhibited antiproliferative activity towards human colorectal cancer (HT-29) cells. Hence, in this work, we explored the in vitro cytotoxicity activity and mechanistic action of Phy-CS-MNP nanocomposite in modulating gene and protein expression profiles in HT-29 cell lines. Cell cycle arrest and apoptosis were evaluated by NovoCyte Flow Cytometer. The mRNA changes (cyclin-dependent kinase 4 (Cdk4), vascular endothelial growth factor A (VEGFA), c-Jun N-terminal kinase 1 (JNK1), inducible nitric oxide synthase (iNOS), and matrix metallopeptidase 9 (MMP9)) and protein expression (nuclear factor-kappa B (NF-κB) and cytochrome c) were assessed by quantitative real-time polymerase chain reaction (PCR) and western blotting, respectively. The data from our study demonstrated that treatment with Phy-CS-MNP nanocomposite triggered apoptosis and G0/G1 cell cycle arrest. The transcriptional activity of JNK1 and iNOS was upregulated after treatment with 90 μg/mL Phy-CS-MNP nanocomposite. Our results suggested that Phy-CS-MNP nanocomposite induced apoptosis and cell cycle arrest via an intrinsic mitochondrial pathway through modulation of Bax and Bcl-2 and the release of cytochrome c from the mitochondria into the cytosol.
Collapse
Affiliation(s)
- Bee Ling Tan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Mohd Esa Norhaizan
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Research Centre of Excellent, Nutrition and Non-Communicable Diseases (NNCD), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Lee Chin Chan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
47
|
Hsueh KC, Lin CL, Tung JN, Yang SF, Hsieh YH. Nimbolide induced apoptosis by activating ERK-mediated inhibition of c-IAP1 expression in human hepatocellular carcinoma cells. ENVIRONMENTAL TOXICOLOGY 2018; 33:913-922. [PMID: 29962003 DOI: 10.1002/tox.22576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/04/2018] [Accepted: 05/06/2018] [Indexed: 06/08/2023]
Abstract
Nimbolide is one of the major compounds from the leaves and flowers of the neem tree and exhibits antitumor properties on various cancer cells. However, no report has shown that nimbolide induces apoptosis in vitro and in vivo in human hepatocellular carcinoma cells. Our results indicated that it inhibited cell growth in Huh-7 and PLC/PRF/5 cells. We also found that nimbolide induced cell death through the induction of G2/M phase arrest and mitochondrial dysfunction, accompanied by the increased expression of cleaved caspase-7, caspase-9, caspase-3, caspase-PARP, and Bax and decreased expression of Mcl-1 and Bcl-2. A human apoptosis antibody array analysis demonstrated that inhibition of the apoptosis family proteins (XIAP, c-IAP1, and c-IAP2) was one of the major targets of nimbolide. Additionally, nimbolide sustained activation of ERK expression. Moreover, pretreatment with U0126 (MEK inhibitor) markedly abolished nimbolide-inhibited cell viability, induced cell apoptosis, ERK phosphorylation, cleaved caspase-9, caspase-3, cleaved-PARP activation, and increased c-IAP1 expression in Huh-7 cells. An in vivo study showed that nimbolide significantly reduced Huh-7 tumor growth and weight in a xenograft mouse model. This study indicated the antitumor potential of nimbolide in human hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Kuan-Chun Hsueh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of General Surgery, Department of Surgery, Tungs' Taichung MetroHarbour Hospital, Taichung, Taiwan
| | - Chia-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Jai-Nien Tung
- Department of Neurosurgery, Tungs'Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
48
|
Tanajak P, Pongkan W, Chattipakorn SC, Chattipakorn N. Increased plasma FGF21 level as an early biomarker for insulin resistance and metabolic disturbance in obese insulin-resistant rats. Diab Vasc Dis Res 2018; 15:263-269. [PMID: 29424246 DOI: 10.1177/1479164118757152] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED Propose: To investigate the temporal relationship between plasma fibroblast growth factor 21 levels, insulin resistance, metabolic dysfunction and cardiac fibroblast growth factor 21 resistance in long-term high-fat diet-induced obese rats. METHODS In total, 36 male Wistar rats were fed with either a normal diet or high-fat diet for 12 weeks. Blood was collected from the tail tip, and plasma was used to determine metabolic profiles and fibroblast growth factor 21 levels. Rats were sacrificed at weeks 4, 8 and 12, and the hearts were rapidly removed for the determination of cardiac fibroblast growth factor 21 signalling pathways. RESULTS Body weight and plasma fibroblast growth factor 21 levels were increased after 4 weeks of consumption of a high-fat diet. At weeks 8 and 12, high-fat diet rats had significantly increased body weight and plasma fibroblast growth factor 21 levels, together with increased plasma insulin, HOMA index, area under the curve of glucose, plasma total cholesterol, plasma low-density lipoprotein cholesterol, serum malondialdehyde and cardiac malondialdehyde levels. However, plasma high-density lipoprotein cholesterol levels and cardiac fibroblast growth factor 21 signalling proteins (p-FGFR1 Tyr154, p-ERK1/2 Thr202/Tyr204 and p-Akt Ser473) were decreased, compared with normal diet rats. CONCLUSION These findings suggest that plasma fibroblast growth factor 21 levels could be an early predictive biomarker prior to the development of insulin resistance, metabolic disturbance and cardiac fibroblast growth factor 21 resistance.
Collapse
Affiliation(s)
- Pongpan Tanajak
- 1 Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- 2 Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- 3 Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Wanpitak Pongkan
- 1 Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- 4 Division of Veterinary Pre-clinic, Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- 1 Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- 3 Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- 5 Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- 1 Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- 2 Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- 3 Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
49
|
2-aryl benzimidazole conjugate induced apoptosis in human breast cancer MCF-7 cells through caspase independent pathway. Apoptosis 2018; 22:118-134. [PMID: 27770267 DOI: 10.1007/s10495-016-1290-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Apoptosis is a representative form of programmed cell death, which has been assumed to be critical for cancer prevention. Thus, any agent that can induce apoptosis may be useful for cancer treatment and apoptosis induction is arguably the most potent defense against cancer promotion. In our previous studies, 2-aryl benzimidazole conjugates were synthesized and evaluated for their antiproliferative activity and one of the new molecule (2f) was considered as a potential lead. This lead molecule showed significant antiproliferative activity against human breast cancer cell line, MCF-7. The results of the present study revealed that this compound arrested the cell cycle at G2/M phase. Topoisomerase II inhibition assay and Western blot analysis suggested that this compound effectively inhibits topoisomerase II activity which leads to apoptotic cell death. Apoptosis induction in MCF-7 cells was further confirmed by loss of mitochondrial membrane potential (∆Ψm), release of cytochrome c from mitochondria, an increase in the level of apoptosis inducing factor (AIF), generation of reactive oxygen species (ROS), up regulation of proapoptotic protein Bax and down regulation of anti apoptotic protein Bcl-2. Apoptosis assay using Annexin V-FITC assay also suggested that this compound induced cell death by apoptosis. However, compound 2f induced apoptosis could not be reversed by Z-VAD-FMK (a pan-caspase inhibitor) demonstrated that the 2f induced apoptosis was caspase independent. Further, 2f treatment did not activate caspase-7 and caspase-9 activity, suggesting that this compound induced apoptosis in breast cancer cells via a caspase independent pathway. Most importantly, this compound was less toxic towards non-tumorigenic breast epithelial cells, MCF-10A. Furthermore, docking studies also support the potentiality of this molecule to bind to the DNA topoisomerase II.
Collapse
|
50
|
Abstract
Substitutions in thiophene structure give rise to new derivatives with different biological and pharmacological activities. The present study investigated the cytotoxicity activity of some thiophene derivatives in breast cancer cells maintained in two-dimensional (2D) or in three-dimensional (3D) culture and evaluated the anticancer mechanism of these compounds. Cytotoxicity assays were performed against untransformed cells and against breast cancer cell MCF-7. Apoptosis analysis and in-vitro migration assay were also performed to evaluate the mechanism of induction of cell death. All thiophene derivatives reduced the cell viability in breast cancer cells, showing cytotoxic activity (IC50<30 µmol/l), and SB-200 compound showed the best selectivity index in MCF-7 cells compared with doxorubicin in 2D culture. All thiophene derivatives significantly induced G0/G1 phase cell cycle arrest. However, only SB-83 treatment was effective against motility of MCF-7 cells in 2D culture (P=0.0059). The SB-200 derivative treatment induced an increased proportion of acridine orange/Hoechst double-stained cells (35.35 vs. 3.14%, P=0.0002) compared with nontreated cells, with apoptosis morphological alterations independent of caspase 7 activation (P>0.05). MCF-7 cells became less responsive to SB-200 and to doxorubicin in 3D culture compared with cells in 2D culture (higher IC50 values); however, SB-200 showed a better cytotoxic effect compared with doxorubicin in 3D culture. Therefore, the current study provides an insight into anticancer potential of thiophene derivatives, and further studies should be conducted to understand the mechanism by which thiophene derivatives act on cancer cells.
Collapse
|