1
|
Atanassova M, Martorell M, Sharopov F, Atanassov L, Kumar P, Sharifi-Rad J, Tejada-Gavela S, Iriti M, Pezzani R, Varoni EM. Cocoa as immunomodulatory agent: an update. Nat Prod Res 2024; 38:4196-4207. [PMID: 37909175 DOI: 10.1080/14786419.2023.2272025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/22/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
Cocoa is rich in polyphenols, mainly flavonoids, which correlate with several health benefits mediated by their antioxidant, anti-inflammatory and immunomodulatory properties. Cocoa and chocolate consumption have been reported to impact the regulation of the immune system, both in preclinical studies and in human trials. The mechanisms for immunomodulation can involve different effects of cocoa polyphenols on the immune system, acting as anti-inflammatory, antioxidant and anti-allergic agents, as well as the direct influence of cocoa on innate and acquired immunity, with cytokines production and activation of both lymphocyte-dependent and -independent pathways. Cocoa intake has been also correlated to changes in gut microbiota ecology and composition, also affecting the intestinal immune system. This review summarises the updates of the last two decades on cocoa as immunomodulatory agent and explores the health-related benefits of cocoa and chocolate intake.
Collapse
Affiliation(s)
- Maria Atanassova
- Scientific Consulting, Chemical Engineering, UCTM, Sofia, Bulgaria
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, Unidad de Desarrollo Tecnológico, UDT, University of Concepción, Concepción, Chile
| | - Farukh Sharopov
- Research Institution "Chinese-Tajik Innovation Center for Natural Products", Academy of Sciences of the Republic of Tajikistan, Dushanbe, Tajikistan
| | - Lyubomir Atanassov
- Faculty of International Relations, St. Petersburg State University, St. Petersburg, Russia
| | - Pradeep Kumar
- Department of Botany, University of Lucknow, Lucknow, India
| | | | - Silvia Tejada-Gavela
- Laboratory of Neurophysiology, Department of Biology, University of the Balearic Islands (UIB), and Health Research Institute of the Balearic Islands (IdISBa), IdISBa, Palma, España
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Marcello Iriti
- Department of Biomedical, Surgical and Dental Sciences, Milan State University, Milan, Italy
| | - Raffaele Pezzani
- AIROB, Associazione Italiana per la Ricerca Oncologica di Base, Padova, Italy
- Phytotherapy Lab, Endocrinology Unit, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Elena Maria Varoni
- Department of Biomedical, Surgical and Dental Sciences, Milan State University, Milan, Italy
| |
Collapse
|
2
|
Laurindo LF, Rodrigues VD, Minniti G, de Carvalho ACA, Zutin TLM, DeLiberto LK, Bishayee A, Barbalho SM. Pomegranate (Punica granatum L.) phytochemicals target the components of metabolic syndrome. J Nutr Biochem 2024; 131:109670. [PMID: 38768871 DOI: 10.1016/j.jnutbio.2024.109670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 04/08/2024] [Accepted: 05/14/2024] [Indexed: 05/22/2024]
Abstract
Pomegranate (Punica granatum L.) is a multipurpose dietary and medicinal plant known for its ability to promote various health benefits. Metabolic syndrome (MetS) is a complex metabolic disorder driving health and socioeconomic challenges worldwide. It may be characterized by insulin resistance, abdominal obesity, hypertension, and dyslipidemia. This study aims to conduct a review of pomegranate's effects on MetS parameters using a mechanistic approach relying on pre-clinical studies. The peel, juice, roots, bark, seeds, flowers, and leaves of the fruit present several bioactive compounds that are related mainly to anti-inflammatory and antioxidant activities as well as cardioprotective, antidiabetic, and antiobesity effects. The use of the juice extract can work as a potent inhibitor of angiotensin-converting enzyme activities, consequently regulating blood pressure. The major bioactive compounds found within the fruit are phenolic compounds (hydrolysable tannins and flavonoids) and fatty acids. Alkaloids, punicalagin, ellagitannins, ellagic acid, anthocyanins, tannins, flavonoids, luteolin, and punicic acid are also present. The antihyperglycemia, antihyperlipidemia, and weight loss promoting effects are likely related to the anti-inflammatory and antioxidant effects. When considering clinical application, pomegranate extracts are found to be frequently well-tolerated, further supporting its efficacy as a treatment modality. We suggest that pomegranate fruit, extract, or processed products can be used to counteract MetS-related risk factors. This review represents an important step towards exploring potential avenues for further research in this area.
Collapse
Affiliation(s)
- Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), São Paulo, São Paulo, Brazil; Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Victória Dogani Rodrigues
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), São Paulo, São Paulo, Brazil
| | - Giulia Minniti
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Antonelly Cassio Alves de Carvalho
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Tereza Laís Menegucci Zutin
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil
| | - Lindsay K DeLiberto
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL USA
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL USA.
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil; Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Universidade de Marília (UNIMAR), São Paulo, São Paulo, Brazil; Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), São Paulo, São Paulo, Brazil.
| |
Collapse
|
3
|
Sun J, Cao Y, Liu Q, Zhou Z, Xu Y, Liu C. Chemical Constituents, Anti-Tumor Mechanisms, and Clinical Application: A Comprehensive Review on Scutellaria barbata. Molecules 2024; 29:4134. [PMID: 39274982 PMCID: PMC11397148 DOI: 10.3390/molecules29174134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/22/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
With the increasing global incidence and mortality rates of cancer, the development of novel anti-tumor drugs has become particularly urgent. Scutellaria barbata D. Don, a perennial herb belonging to the genus Scutellaria in the family Lamiaceae, has aroused extensive attention for its medicinal value in recent years. This article presents an exhaustive review of the flavonoid, diterpene, and other chemical constituents harbored within Scutellaria barbata, delving into the intricate mechanisms by which these compounds orchestrate their anti-tumor effects via diverse biological pathways. Remarkably, these compounds distinguish themselves through their capability to regulate cellular signaling, inhibit cancer cell proliferation, trigger apoptosis, disrupt angiogenesis, and bolster immune responses. These anti-tumor effects are achieved through strategic modulation of pivotal signaling cascades, particularly the PI3K/Akt/mTOR, MAPK, and NFκB pathways. In addition, this article also summarizes the clinical applications of Scutellaria barbata in tumor treatment, especially its potential in alleviating the side effects of radiotherapy and chemotherapy and improving patients' quality of life. In conclusion, this review comprehensively summarizes and analyzes the chemical constituents, anti-tumor mechanisms, and clinical applications of Scutellaria barbata, with the aim of systematically reviewing the existing research results and exploring potential future research directions.
Collapse
Affiliation(s)
- Jiagui Sun
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin 150040, China
| | - Yuqi Cao
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin 150040, China
| | - Qiqi Liu
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin 150040, China
| | - Zhengshu Zhou
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin 150040, China
| | - Yanan Xu
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin 150040, China
| | - Chenggang Liu
- School Basic Medical Sciences, Heilongjiang University of Chinese Medicine, 24 Heping Road, Harbin 150040, China
| |
Collapse
|
4
|
Yang H, Zhao Y, Ren B, Wu Y, Qiu Z, Cheng Y, Qiu B. Poria acid inhibit the growth and metastasis of renal cell carcinoma by inhibiting the PI3K/akt/NF-κb signaling pathway. Heliyon 2024; 10:e31106. [PMID: 38779018 PMCID: PMC11109894 DOI: 10.1016/j.heliyon.2024.e31106] [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: 01/11/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Background Poria acid (PAC) is a triterpene compound found in Poria cocos, a traditional Chinese medicine (TCM). The current study aims to explore the therapeutic effects and potential mechanisms of PAC on the migration and proliferation of human renal cell carcinoma (RCC) cells as well as tumor growth in animal model. Methods Cell viability and proliferative capacity of normal renal cells and RCC cells were investigated by MTT assay. In addition, 786-O cells were divided into four groups and treated with different concentrations of PAC (0, 20, 40, and 60 μM) for 48 h. Cell scratch test and cell invasion assay were performed to evaluate the effects of PAC on the invasion and migration of RCC cells, respectively. The effects of PAC on apoptosis of RCC cells and expression levels of PI3K/Akt/NF-kB signaling pathway-related biomarkers were investigated using TUNEL staining and Western blotting methods, respectively. Effects of PAC on the inhibitory activity of RCC tumor in mice were evaluated in a 786-O CDX model. Results The study found that PAC inhibited the viability of RCC cells in a dose-dependent manner, as demonstrated by in vitro cell assays (p < 0.05). However, PAC showed no significant inhibitory effect on normal renal cells (p > 0.05). PAC also significantly inhibited the migration and invasion of RCC via EMT/MMP signaling pathways (p < 0.05). Immunofluorescence and immunoblotting results showed that PAC induced the apoptosis of RCC, which was accompanied by changes in the expression levels of apoptosis-related proteins (p < 0.05). Moreover, PAC significantly downregulated the PI3K/Akt/NF-kB signaling pathway in a concentration-dependent manner (p < 0.05). The effect of PAC on RCC apoptosis was dramatically reversed by 740Y-P (PI3K agonist) (p < 0.05) but significantly enhanced in the presence of LY294002 (PI3K inhibitor) (p < 0.05). The results of in vivo experiment also demonstrated that the antitumor activity of PAC was achieved by affecting the PI3K/Akt/NF-kB signaling pathway. Conclusions PAC can effectively suppress the proliferation, invasion and migration of RCC cells, and exhibit anti-tumor effects in RCC model by inhibiting the PI3K/Akt/NF-kB signaling pathway.
Collapse
Affiliation(s)
- Haotian Yang
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang 050051, China
| | - Yue Zhao
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang 050051, China
| | - Bingnan Ren
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang 050051, China
| | - Yin Wu
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang 050051, China
| | - Zhihong Qiu
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang 050051, China
| | - Yan Cheng
- Department of Medical Oncology, Hebei General Hospital, Shijiazhuang 050051, China
| | - Bo Qiu
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang 050051, China
| |
Collapse
|
5
|
Islam MT, Jang NH, Lee HJ. Natural Products as Regulators against Matrix Metalloproteinases for the Treatment of Cancer. Biomedicines 2024; 12:794. [PMID: 38672151 PMCID: PMC11048580 DOI: 10.3390/biomedicines12040794] [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: 02/29/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Cancers are currently the major cause of mortality in the world. According to previous studies, matrix metalloproteinases (MMPs) have an impact on tumor cell proliferation, which could lead to the onset and progression of cancers. Therefore, regulating the expression and activity of MMPs, especially MMP-2 and MMP-9, could be a promising strategy to reduce the risk of cancers. Various studies have tried to investigate and understand the pathophysiology of cancers to suggest potent treatments. In this review, we summarize how natural products from marine organisms and plants, as regulators of MMP-2 and MMP-9 expression and enzymatic activity, can operate as potent anticancer agents.
Collapse
Affiliation(s)
- Md. Towhedul Islam
- Department of Chemistry, Faculty of Science, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
| | - Nak Han Jang
- Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
| | - Hyuck Jin Lee
- Department of Chemistry Education, Kongju National University, Gongju 32588, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
6
|
Zhou T, Zhou H, Tian L, Tang M, Wang L, Kang Y, Chen T, Li X, Wu S, Xia R, Huang X, Peng L, Yin W. Pomegranate juice-containing serum inhibits migration of hepatocellular carcinoma cells and promotes apoptosis by induction of mitochondrial dysfunction. J Nutr Biochem 2024; 125:109557. [PMID: 38151194 DOI: 10.1016/j.jnutbio.2023.109557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, with an insidious onset and poor prognosis. Pomegranate is a fruit rich in many natural products with anti-cancer potential; however, its direct biological effects are difficult to evaluate in vitro because of changes in its active components after absorption and metabolism. This study was conducted to prepare pomegranate juice-containing serum (PJ serum) by gavage of pomegranate juice (PJ) in rats and to collect serum. The aim was to investigate the components and the effects of PJ serum on HCC cells by serum pharmacology. 56 compounds were identified in the PJ serum, including 6 prototype components. PJ serum selectively inhibited HCC cells proliferation and migration, and it promoted apoptosis of HCC cells without affecting LO2 cells activity. Furthermore, PJ serum reduced the mitochondrial membrane potential and increased the calcium ion concentration in HCC cells. Mechanistically, PJ serum up-regulated the expression of the Bax family, Caspases and TIMP2/MMP2, and down-regulated the expression of MMP9. This study revealed that PJ serum inhibited HCC cell migration by regulating the TIMP2/MMP2 balance and MMP9 expression and promoted HCC cell apoptosis by inducing mitochondrial dysfunction and causing a Caspase cascade. The polyphenols and flavonoids in PJ may be important components responsible for its anti-HCC activity after metabolism.
Collapse
Affiliation(s)
- Ting Zhou
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Heting Zhou
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Li Tian
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Minghai Tang
- Collaborative Innovation Center for Biotherapy and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Liqun Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Yuhong Kang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Tao Chen
- Chengdu Institute of Product Quality Inspection Co., Ltd, Chengdu, Sichuan, China
| | - Xingjie Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Shouxun Wu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Rui Xia
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Xiaoyi Huang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China
| | - Lijun Peng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China.
| | - Wenya Yin
- West China School of Public Health and West China Fourth Hospital, Sichuan University, West China Occupational Pneumoconiosis Cohort Study (WCOPCS) Workgroup, West China-PUMCC.C. Chen Institute of Health, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Shen L, Wang J, Li Y, Sun C, Teng M, Ye X, Feng X. Transcription Factor STAT3-Activated LDHB Promotes Tumor Properties of Endometrial Cancer Cells by Inducing MDH2 Expression. Mol Biotechnol 2024:10.1007/s12033-024-01067-z. [PMID: 38381377 DOI: 10.1007/s12033-024-01067-z] [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: 05/08/2023] [Accepted: 01/08/2024] [Indexed: 02/22/2024]
Abstract
The pathogenesis of endometrial cancer (EC) involves the regulation of lactate dehydrogenases. However, the role and mechanism of lactate dehydrogenase-B (LDHB) in EC progression have not been studied. The mRNA levels of LDHB and malate dehydrogenase 2 (MDH2) were detected by quantitative real-time polymerase chain reaction. Protein expression was checked by western blotting and immunohistochemistry assays. Cell proliferation, apoptosis, and invasion were analyzed by 5-Ethynyl-2'-deoxyuridine, transwell, and flow cytometry assay, respectively. Glycolysis was investigated using Glucose Assay Kit, CheKine™ Micro Lactate Assay Kit, and ADP/ATP ratio assay kit. An in vivo tumor formation assay was conducted to disclose the effect of LDHB on tumor growth in vivo. The associations among signal transducer and activator of transcription 3 (STAT3), LDHB, and MDH2 were predicted through JASPAR or GeneMANIA online database and identified by chromatin immunoprecipitation assay, dual-luciferase reporter assay, and co-immunoprecipitation assay. LDHB expression was increased in EC tissues and cells in comparison with normal endometrial tissues and human endometrial stromal cells. LDHB had the potential as a biomarker to predict the prognosis of EC patients. In addition, LDHB knockdown inhibited the proliferation, invasion, and glycolysis and promoted apoptosis of RL95-2 and Ishikawa cells. LDHB knockdown inhibited tumor property of Ishikawa cells in vivo. STAT3 bound to the promoter region of LDHB, and STAT3 silencing-induced effects were relieved after LDHB upregulation. LDHB interacted with and regulated MDH2 expression. Moreover, MDH2 overexpression rescued LDHB knockdown-induced effects on EC cell phenotypes. STAT3-activated LDHB promoted endometrial cancer cell malignancy by inducing MDH2 production.
Collapse
Affiliation(s)
- Li Shen
- Department of Obstetrics and Gynecology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, 430070, Hubei, China
| | - Juan Wang
- Department of Obstetrics and Gynecology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, 430070, Hubei, China
| | - Yanxia Li
- Department of Rehabilitation, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, 430070, Hubei, China
| | - Cuizhen Sun
- Department of Rehabilitation, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, 430070, Hubei, China
| | - Minjie Teng
- Department of Rehabilitation, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, 430070, Hubei, China
| | - Xiaohe Ye
- Department of Rehabilitation, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan City, 430070, Hubei, China
| | - Xiaomin Feng
- Department of Oncology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 39 Yanhu Road, East Lake Eco-Tourism Scenic Spot, Wuhan City, 430070, Hubei, China.
| |
Collapse
|
8
|
Bai Y, Chen R, Sun J, Guo Y. Evaluation of Therapeutic Mechanism of Hedyotis Diffusa Willd (HDW)‒ Scutellaria Barbata (SB) in Clear Cell Renal Cell Carcinoma via Singlecell RNA Sequencing and Network Pharmacology. Comb Chem High Throughput Screen 2024; 27:910-921. [PMID: 37526191 DOI: 10.2174/1386207326666230731155309] [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: 04/03/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 08/02/2023]
Abstract
OBJECTIVE The present study aimed to investigate the therapeutic mechanism of Hedyotis diffusa Willd (HDW) and Scutellaria barbata (SB) in ccRCC using a combination of single-cell RNA sequencing (scRNA-seq) and network pharmacology. METHODS The active ingredients and potential molecular targets of HDW-SB were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform. Gene expression data (GSE53757) were obtained from the Gene Expression Omnibus database. The hub genes of HDW-SB against ccRCC were identified via the protein-protein interaction network, and further analyzed by molecular complex detection. The roles of these genes in the diagnosis and immune infiltration of ccRCC were analyzed. The clinical significance of hub genes was verified using scRNA-seq data (GSE121638) and molecular docking. RESULTS Following the PPI network analysis, 29 hub genes of HDW-SB against ccRCC were identified. All hub genes, except for CENPE, had significantly different expressions in tumor tissue and a more accurate diagnosis of ccRCC. Fifteen cell clusters were defined based on the scRNA-seq dataset, and the clusters were annotated as six cell types using marker genes. TYMS and KIAA0101 from hub genes were highly expressed in NK cells. Three active compounds, quercetin, luteolin, and baicalein, were found to target TYMS and KIAA0101 from the compound-target interaction network. CONCLUSION 29 hub genes of HDW-SB against ccRCC were identified and showed good performance in terms of diagnosis and prognosis. Moreover, among these hub genes docking with the main ingredients of HDW-SB, TYMS and KIAA0101 exerted anti-ccRCC effects through NK cells.
Collapse
Affiliation(s)
- Yangyang Bai
- Department of Urology, Henan Province Hospital of TCM, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Ruiting Chen
- Department of Urology, Henan Province Hospital of TCM, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Jijian Sun
- Department of Urology, Henan Province Hospital of TCM, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Yilin Guo
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Salih R, Bajou K, Shaker B, Elgamouz A. Antitumor effect of algae silver nanoparticles on human triple negative breast cancer cells. Biomed Pharmacother 2023; 168:115532. [PMID: 37832405 DOI: 10.1016/j.biopha.2023.115532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/10/2023] [Accepted: 09/17/2023] [Indexed: 10/15/2023] Open
Abstract
In recent years, metallic nanoparticles have gained increasing attention due to their prospective applications in the field of nanomedicine, with increasing research into their use in cancer therapy. In this current research, we investigated the effect of green synthesized Silver Nanoparticles (AgNPs) capped with Noctiluca scintillans algae extract. The phytochemicals present in the shell of AgNPs were identified using GC-MS. Different compounds with anticancer activity such as n-hexadecanoic acid, beta-sitosterol, stigmasterol and palmitic acid were detected among others. The effects of Algae-AgNPs synthesized were tested on MDA-MB-231 human breast cancer cells and HaCat human keratinocyte normal cells. Cell viability assay revealed a time and dose-dependent effect against breast cancer cells with a less potent effect against normal cells. The cell viability reduction is not attributed to a cytotoxic nor an antiproliferative effect of the Algae-AgNPs as attested by LDH release and BrdU incorporation. Algae-AgNPs exhibited an exceptional ability to specifically induce apoptosis in cancer cells and not normal cells. The observed effects are not attributed to the AgNPs, as demonstrated by the lack of impact of the Starch-AgNPs (used as a negative control) on cell survival and apoptosis. In addition to that, we show that Algae-AgNPs significantly reduced tumor cell migration by downregulation of matrix metalloprotease-9 levels. In vivo, the breast cancer xenograft model showed a significant reduction of tumor growth in mice treated with Algae-AgNPs. These findings highlight the promising potential of the green synthesized AgNPs as a safe targeted therapy for cancer treatment.
Collapse
Affiliation(s)
- Rawan Salih
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Human Genetics and Stem Cells Research Group, Research Institute of Science and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Khalid Bajou
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Human Genetics and Stem Cells Research Group, Research Institute of Science and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Baraah Shaker
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Human Genetics and Stem Cells Research Group, Research Institute of Science and Engineering, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Abdelaziz Elgamouz
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
10
|
Liu Q, Wang L, He D, Wu Y, Liu X, Yang Y, Chen Z, Dong Z, Luo Y, Song Y. Application Value of Antimicrobial Peptides in Gastrointestinal Tumors. Int J Mol Sci 2023; 24:16718. [PMID: 38069041 PMCID: PMC10706433 DOI: 10.3390/ijms242316718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Gastrointestinal cancer is a common clinical malignant tumor disease that seriously endangers human health and lacks effective treatment methods. As part of the innate immune defense of many organisms, antimicrobial peptides not only have broad-spectrum antibacterial activity but also can specifically kill tumor cells. The positive charge of antimicrobial peptides under neutral conditions determines their high selectivity to tumor cells. In addition, antimicrobial peptides also have unique anticancer mechanisms, such as inducing apoptosis, autophagy, cell cycle arrest, membrane destruction, and inhibition of metastasis, which highlights the low drug resistance and high specificity of antimicrobial peptides. In this review, we summarize the related studies on antimicrobial peptides in the treatment of digestive tract tumors, mainly oral cancer, esophageal cancer, gastric cancer, liver cancer, pancreatic cancer, and colorectal cancer. This paper describes the therapeutic advantages of antimicrobial peptides due to their unique anticancer mechanisms. The length, net charge, and secondary structure of antimicrobial peptides can be modified by design or modification to further enhance their anticancer effects. In summary, as an emerging cancer treatment drug, antimicrobial peptides need to be further studied to realize their application in gastrointestinal cancer diseases.
Collapse
Affiliation(s)
- Qi Liu
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Lei Wang
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Dongxia He
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Yuewei Wu
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Xian Liu
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Yahan Yang
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Zhizhi Chen
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Zhan Dong
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Ying Luo
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Yuzhu Song
- College of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
- Medical College, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
11
|
Čižmáriková M, Michalková R, Mirossay L, Mojžišová G, Zigová M, Bardelčíková A, Mojžiš J. Ellagic Acid and Cancer Hallmarks: Insights from Experimental Evidence. Biomolecules 2023; 13:1653. [PMID: 38002335 PMCID: PMC10669545 DOI: 10.3390/biom13111653] [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: 10/13/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a complex and multifaceted disease with a high global incidence and mortality rate. Although cancer therapy has evolved significantly over the years, numerous challenges persist on the path to effectively combating this multifaceted disease. Natural compounds derived from plants, fungi, or marine organisms have garnered considerable attention as potential therapeutic agents in the field of cancer research. Ellagic acid (EA), a natural polyphenolic compound found in various fruits and nuts, has emerged as a potential cancer prevention and treatment agent. This review summarizes the experimental evidence supporting the role of EA in targeting key hallmarks of cancer, including proliferation, angiogenesis, apoptosis evasion, immune evasion, inflammation, genomic instability, and more. We discuss the molecular mechanisms by which EA modulates signaling pathways and molecular targets involved in these cancer hallmarks, based on in vitro and in vivo studies. The multifaceted actions of EA make it a promising candidate for cancer prevention and therapy. Understanding its impact on cancer biology can pave the way for developing novel strategies to combat this complex disease.
Collapse
Affiliation(s)
- Martina Čižmáriková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Gabriela Mojžišová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia;
| | - Martina Zigová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Annamária Bardelčíková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (M.Č.); (R.M.); (M.Z.); (A.B.)
| |
Collapse
|
12
|
Kolesarova A, Baldovska S, Kohut L, Vasicek J, Ivanisova E, Arvay J, Duracka M, Roychoudhury S. Modulatory effect of pomegranate peel extract on key regulators of ovarian cellular processes in vitro. Front Endocrinol (Lausanne) 2023; 14:1277155. [PMID: 38027211 PMCID: PMC10663288 DOI: 10.3389/fendo.2023.1277155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
In this study, response of ovarian cells (human granulosa cell line HGL5, and human adenocarcinoma cell line OVCAR-3) to short-term pomegranate peel extract (PPE) treatment (for 24 hours in cell culture) was evaluated in vitro. Quantitative and qualitative screening of polyphenols revealed punicalagins α and β as major polyphenolic components. Total phenolic content (TPC) was 93.76 mg GAE/g d.w. with a high antioxidant activity of 95.30 mg TEAC/g d.w. In OVCAR-3, PPE treatment inhibited the metabolic activity, and increased cyclin-dependent kinase 1 (CDKN1A, p21) level at the highest dose, but not in HGL5. Flow cytometry analysis could not detect any significant difference between proportions of live, dead, and apoptotic cells in both cell lines. Reactive oxygen species (ROS) revealed an antioxidant effect on HGL5, and a prooxidant effect by stimulating ROS generation in OVCAR-3 cells at the higher doses of PPE. However, in contrast to HGL5, PPE treatment decreased release of growth factors - TGF-β2 and EGF at the highest dose, as well as their receptors TGFBR2 and EGFR in OVCAR-3 cells. PPE also influenced steroidogenesis in granulosa cells HGL5 by stimulating 17β-estradiol secretion at higher doses. In conclusion, the present study highlighted the bioactive compounds in pomegranate peels and the possible mechanisms of action of PPE, shedding light on its promising role in ovarian cancer (chemo)prevention and/or management.
Collapse
Affiliation(s)
- Adriana Kolesarova
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
- AgroBioTech Research Centre, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Simona Baldovska
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Ladislav Kohut
- Institute of Applied Biology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Jaromir Vasicek
- Institute of Farm Animal Genetics and Reproduction, NPPC - Research Institute for Animal Production Nitra, Lužianky, Slovakia
- Institute of Biotechnology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Eva Ivanisova
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Julius Arvay
- Institute of Food Sciences, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Michal Duracka
- AgroBioTech Research Centre, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | | |
Collapse
|
13
|
Movahhed M, pazhouhi M, Ghaleh HEG, Kondori BJ. Anti-metastatic effect of taraxasterol on prostate cancer cell lines. Res Pharm Sci 2023; 18:439-448. [PMID: 37614618 PMCID: PMC10443670 DOI: 10.4103/1735-5362.378090] [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: 11/26/2022] [Revised: 01/04/2023] [Accepted: 05/30/2023] [Indexed: 08/25/2023] Open
Abstract
Background and purpose Prostate cancer is the second cause of death among men. Nowadays, treating various cancers with medicinal plants is more common than other therapeutic agents due to their minor side effects. This study aimed to evaluate the effect of taraxasterol on the prostate cancer cell line. Experimental approach The prostate cancer cell line (PC3) was cultured in a nutrient medium. MTT method and trypan blue staining were used to evaluate the viability of cells in the presence of different concentrations of taraxasterol, and IC50 was calculated. Real-time PCR was used to measure the expression of MMP-9, MMP-2, uPA, uPAR, TIMP-2, and TIMP-1 genes. Gelatin zymography was used to determine MMP-9 and MMP-2 enzyme activity levels. Finally, the effect of taraxasterol on cell invasion, migration, and adhesion was investigated. Findings/Results Taraxasterol decreased the survival rate of PC3 cells at IC50 time-dependently (24, 48, and 72 h). Taraxasterol reduced the percentage of PC3 cell adhesion, invasion, and migration by 74, 56, and 76 percent, respectively. Real-time PCR results revealed that uPA, uPAR, MMP-9, and MMP-2 gene expressions decreased in the taraxasterol-treated groups, but TIMP-2 and TIMP-1 gene expressions increased significantly. Also, a significant decrease in the level of MMP-9 and MMP-2 enzymes was observed in the PC3 cell line treated with taraxasterol. Conclusion and implications The present study confirmed the therapeutic role of taraxasterol in preventing prostate cancer cell metastasis in the in-vitro study.
Collapse
Affiliation(s)
- Morteza Movahhed
- Department of Pathology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mona pazhouhi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, I.R. Iran
| | | | - Bahman Jalali Kondori
- Department of Anatomical Sciences, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Yin Y, Martínez R, Zhang W, Estévez M. Crosstalk between dietary pomegranate and gut microbiota: evidence of health benefits. Crit Rev Food Sci Nutr 2023; 64:10009-10035. [PMID: 37335106 DOI: 10.1080/10408398.2023.2219763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Gut microbiota (GM) is an invisible organ that plays an important role in human health. Increasing evidence suggests that polyphenols in pomegranate (punicalagin, PU) could serve as prebiotics to modulate the composition and function of GM. In turn, GM transform PU into bioactive metabolites such as ellagic acid (EA) and urolithin (Uro). In this review, the interplay between pomegranate and GM is thoroughly described by unveiling a dialog in which both actors seem to affect each other's roles. In a first dialog, the influence of bioactive compounds from pomegranate on GM is described. The second act shows how the GM biotransform pomegranate phenolics into Uro. Finally, the health benefits of Uro and that related molecular mechanism are summarized and discussed. Intake of pomegranate promotes beneficial bacteria in GM (e.g. Lactobacillus spp., Bifidobacterium spp.) while reducing the growth of harmful bacteria (e.g. Bacteroides fragilis group, Clostridia). Akkermansia muciniphila, and Gordonibacter spp., among others, biotransform PU and EA into Uro. Uro contributes to strengthening intestinal barrier and reducing inflammatory processes. Yet, Uro production varies greatly among individuals and depend on GM composition. Uro-producing bacteria and precise metabolic pathways need to be further elucidated therefore contributing to personalized and precision nutrition.
Collapse
Affiliation(s)
- Yantao Yin
- Key Laboratory of Meat Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
- TECAL Research Group, IPROCAR Research Institute, Universidad de Extremadura, Caceres, Spain
| | - Remigio Martínez
- TECAL Research Group, IPROCAR Research Institute, Universidad de Extremadura, Caceres, Spain
- Infectious Diseases Unit. Animal Health Department, University of Extremadura, Caceres, Spain
- Departamento de Sanidad Animal, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), UIC Zoonosis y Enfermedades Emergentes ENZOEM, University of Córdoba, Córdoba, Spain
| | - Wangang Zhang
- Key Laboratory of Meat Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Mario Estévez
- TECAL Research Group, IPROCAR Research Institute, Universidad de Extremadura, Caceres, Spain
| |
Collapse
|
15
|
Hosseinzadeh A, Poursoleiman F, Biregani AN, Esmailzadeh A. Flavonoids target different molecules of autophagic and metastatic pathways in cancer cells. Cancer Cell Int 2023; 23:114. [PMID: 37308913 DOI: 10.1186/s12935-023-02960-4] [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: 03/12/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
Despite the success of cancer therapy, it has encountered a major obstacle due to the complicated nature of cancer, namely resistance. The recurrence and metastasis of cancer occur when anti-cancer therapeutic agents fail to eradicate all cancer cells. Cancer therapy aims to find the best agent that targets all cancer cells, including those sensitive or resistant to treatment. Flavonoids, natural products from our diet, show anti-cancer effects in different studies. They can inhibit metastasis and the recurrence of cancers. This review discusses metastasis, autophagy, anoikis in cancer cells, and their dynamic relationship. We present evidence that flavonoids can block metastasis and induce cell death in cancer cells. Our research suggests that flavonoids can serve as potential therapeutic agents in cancer therapy.
Collapse
Affiliation(s)
- Aysooda Hosseinzadeh
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Faezeh Poursoleiman
- Department of Cellular and Molecular Nutrition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Naghdipour Biregani
- Department of Nutrition, School of Health, Shahid Sadoughi University of Medical Scinences, Yazd, Iran
| | - Ahmad Esmailzadeh
- Students' Scientific Center, Tehran University of Medical Sciences, Tehran, Iran.
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
16
|
Nazam N, Jabir NR, Ahmad I, Alharthy SA, Khan MS, Ayub R, Tabrez S. Phenolic Acids-Mediated Regulation of Molecular Targets in Ovarian Cancer: Current Understanding and Future Perspectives. Pharmaceuticals (Basel) 2023; 16:274. [PMID: 37259418 PMCID: PMC9962268 DOI: 10.3390/ph16020274] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer is a global health concern with a dynamic rise in occurrence and one of the leading causes of mortality worldwide. Among different types of cancer, ovarian cancer (OC) is the seventh most diagnosed malignant tumor, while among the gynecological malignancies, it ranks third after cervical and uterine cancer and sadly bears the highest mortality and worst prognosis. First-line treatments have included a variety of cytotoxic and synthetic chemotherapeutic medicines, but they have not been particularly effective in extending OC patients' lives and are associated with side effects, recurrence risk, and drug resistance. Hence, a shift from synthetic to phytochemical-based agents is gaining popularity, and researchers are looking into alternative, cost-effective, and safer chemotherapeutic strategies. Lately, studies on the effectiveness of phenolic acids in ovarian cancer have sparked the scientific community's interest because of their high bioavailability, safety profile, lesser side effects, and cost-effectiveness. Yet this is a road less explored and critically analyzed and lacks the credibility of the novel findings. Phenolic acids are a significant class of phytochemicals usually considered in the nonflavonoid category. The current review focused on the anticancer potential of phenolic acids with a special emphasis on chemoprevention and treatment of OC. We tried to summarize results from experimental, epidemiological, and clinical studies unraveling the benefits of various phenolic acids (hydroxybenzoic acid and hydroxycinnamic acid) in chemoprevention and as anticancer agents of clinical significance.
Collapse
Affiliation(s)
- Nazia Nazam
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Noida 201301, Uttar Pradesh, India
| | - Nasimudeen R. Jabir
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur 613403, Tamil Nadu, India
| | - Iftikhar Ahmad
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
| | - Saif A. Alharthy
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Rashid Ayub
- Technology and Innovation Unit, Department of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21362, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
17
|
Ovejero-Sánchez M, González-Sarmiento R, Herrero AB. DNA Damage Response Alterations in Ovarian Cancer: From Molecular Mechanisms to Therapeutic Opportunities. Cancers (Basel) 2023; 15:448. [PMID: 36672401 PMCID: PMC9856346 DOI: 10.3390/cancers15020448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
The DNA damage response (DDR), a set of signaling pathways for DNA damage detection and repair, maintains genomic stability when cells are exposed to endogenous or exogenous DNA-damaging agents. Alterations in these pathways are strongly associated with cancer development, including ovarian cancer (OC), the most lethal gynecologic malignancy. In OC, failures in the DDR have been related not only to the onset but also to progression and chemoresistance. It is known that approximately half of the most frequent subtype, high-grade serous carcinoma (HGSC), exhibit defects in DNA double-strand break (DSB) repair by homologous recombination (HR), and current evidence indicates that probably all HGSCs harbor a defect in at least one DDR pathway. These defects are not restricted to HGSCs; mutations in ARID1A, which are present in 30% of endometrioid OCs and 50% of clear cell (CC) carcinomas, have also been found to confer deficiencies in DNA repair. Moreover, DDR alterations have been described in a variable percentage of the different OC subtypes. Here, we overview the main DNA repair pathways involved in the maintenance of genome stability and their deregulation in OC. We also recapitulate the preclinical and clinical data supporting the potential of targeting the DDR to fight the disease.
Collapse
Affiliation(s)
- María Ovejero-Sánchez
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-Spanish National Research Council, 37007 Salamanca, Spain
| | - Rogelio González-Sarmiento
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-Spanish National Research Council, 37007 Salamanca, Spain
| | - Ana Belén Herrero
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain
- Molecular Medicine Unit, Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-Spanish National Research Council, 37007 Salamanca, Spain
| |
Collapse
|
18
|
Sabry OM, Sabry MO, El-Sonbaty SM, Meselhy KM. In-vivo and in-silico studies of Eucalyptus kino polyphenolics: outstanding activity in quenching solid liver tumors through inhibition of MMP-9 and TGF-β gene expression. Nat Prod Res 2023; 37:343-347. [PMID: 34494929 DOI: 10.1080/14786419.2021.1961254] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
As a result of our continuous research efforts to investigate the molecular mechanisms of Eucalyptus kino polyphenolics for healing liver solid tumors, in-vivo histopathological studies of the solid tumor tissues and in-silico molecular docking were carried out. Histopathology of female mice treated with a dose of 200 mg/kg Eucalyptus kino methanolic extract in combination with low level ionising γ-radiation (0.25 Gy) recovered a near-to-normal histological structure revealing a substantial number of cells with hyper-chromachia in nuclei, fibroblast lobules and necrotic cells. Analysis for matrix metalloproteinase-9 (MMP-9) by Western blot and also molecular docking study were conducted to evaluate and rank the potential of the individual components dominated in the extract to inhibit MMP-9 and TGF-β. The current study presents new lead compounds for the design of novel MMP-9 and TGF-β strong inhibitors of natural origin.
Collapse
Affiliation(s)
- Omar M Sabry
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Miral O Sabry
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Sawsan M El-Sonbaty
- Department of Radiation Microbiology, the National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Nasr City, Egypt
| | - Khaled M Meselhy
- Department of Pharmacognosy, College of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
19
|
Liu C, Shen Y, Tan Q. Diagnostic and prognostic values of MMP-9 expression in ovarian cancer: A study based on bioinformatics analysis and meta-analysis. Int J Biol Markers 2022; 38:15-24. [PMID: 36448239 DOI: 10.1177/03936155221140421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background This study aims to explore the expression of matrix metalloproteinase-9 (MMP-9) associated with both diagnostic and prognostic value in ovarian cancer by meta-analysis and bioinformatics analyses. Methods We investigated the prognostic value of MMP-9 expression in ovarian cancer based on The Cancer Genome Atlas. Five databases were used to collect records about MMP-9 expression related to diagnostic and prognostic values in ovarian cancer from inception to June 2022. Using Stata 15.0 software, hazard ratio (HR) and odds ratio (OR) were calculated as the effect index of prognosis. We chose the pooled sensitivity, specificity, and area under the curve (AUC) to judge the diagnostic utility of MMP-9 for ovarian cancer. Results A total of 23 studies on prognosis, and five studies on diagnosis were entered into the meta-analysis. These suggest that high MMP-9 expression was detrimental to the overall survival of patients with ovarian cancer (HR = 1.34; 95% confidence interval (CI) 1.08∼1.66; P<0.01). High MMP-9 expression increased the risk of tumor stage (OR = 3.66; 95% CI 1.89∼7.07), but was not related to the tumor grade of ovarian cancer ( P>0.05). The pooled analysis of serum MMP-9 diagnosing for ovarian cancer gave the pooled sensitivity, specificity, and AUC the values of 0.72 (95% CI 0.61∼0.81), 0.81 (95% CI 0.77∼0.85), and 0.84 (95% CI 0.81∼0.87), respectively. Conclusion High MMP-9 expression can increase the tumor stage, and a correlation exists between high MMP-9 expression and poor prognosis in patients with ovarian cancer. Also, serum MMP-9 has a good diagnostic value for ovarian cancer.
Collapse
Affiliation(s)
- Changyu Liu
- Department of Clinical laboratory, Hainan general Hospital, Haikou, China
| | - Ying Shen
- Department of Clinical laboratory, Hainan general Hospital, Haikou, China
| | - Qiyan Tan
- Department of Clinical laboratory, Hainan general Hospital, Haikou, China
| |
Collapse
|
20
|
Exploring the Antiovarian Cancer Mechanisms of Salvia Miltiorrhiza Bunge by Network Pharmacological Analysis and Molecular Docking. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7895246. [PMID: 36483919 PMCID: PMC9726254 DOI: 10.1155/2022/7895246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 08/13/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022]
Abstract
Background Ovarian cancer was one of the gynecological malignant tumors. Salvia miltiorrhiza Bunge (SMB) was a kind of herbal medicine with an antitumor effect. However, the inhibitory effect of SMB on ovarian cancer and its potential mechanism were still unclear. Objective The antitumor effect of SMB on ovarian cancer was studied by network pharmacology and molecular docking techniques, and its possible molecular mechanisms were analyzed. Method The active ingredients of SMB and the target data of ovarian cancer were obtained from the Traditional Chinese Medicines for Systems Pharmacology Database (TCMSP) and the GeneCards database. The relationship between active ingredients of SMB and ovarian cancer targets was analyzed by String database, David 6.8 online database, and Cytoscape 3.7.2 software, and then potential pathways were screened out. In addition, molecular docking technology was used to verify further the binding effect of antiovarian cancer pathway targets with active ingredients of SMB. Finally, survival analysis was performed for all potential targets. Results We analyzed 71 SMB-ovarian cancer common targets, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that the PI3K-Akt signaling pathway might be an essential pathway for SMB to inhibit ovarian cancer. Luteolin, Tanshinone IIA, and Cryptotanshinone in SMB might play an important role. HSP90AA1, CDK2, and PIK3CG might be potential targets of SMB in inhibiting ovarian cancer. Conclusion Through network pharmacology and molecular docking analysis, we found that SMB might partially inhibit ovarian cancer by the PI3K-Akt signaling pathway. We believe that SMB might be a potential therapeutic agent for ovarian cancer patients.
Collapse
|
21
|
Xue P, Zhang G, Zhang J, Ren L. Synergism of ellagic acid in combination with radiotherapy and chemotherapy for cancer treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:153998. [PMID: 35217437 DOI: 10.1016/j.phymed.2022.153998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Ellagic acid (EA) is a polyphenol compound abundant in berries, walnuts, pecans, pomegranate, cranberries, and other plant foods and exerts a wide array of biological properties. In particular, EA has received considerable research attention in anti-cancer therapy. EA administered alone has been shown to exert effects against human cancers through multiple pathways. In addition, EA may increase tumor sensitivity to chemotherapy and radiotherapy. Namely, EA combination with a relatively low dosage of therapeutic drugs or optimized radiation dose could improve the treatment outcome. More importantly, EA could counteract chemotherapy-related adverse reactions. PURPOSE This review aims to summarize the in vitro and in vivo experimental evidence of synergism of EA in radiotherapy/chemotherapy for the treatment of cancers. In addition, the preventive effect of EA to counteract chemotherapy-induced toxicity is also discussed. METHODS The searches were performed in the PubMed, Web of Science and Google scholar and introduced the information about the role of EA in cancer treatment. RESULTS EA exhibits synergistic effects in radiotherapy/chemotherapy for the treatment of cancers and exerts a great potential in reducing the side effects of chemotherapy and radiotherapy due to its biological activities, such as antioxidant and anti-inflammatory activities. CONCLUSION EA could be a promising drug adjuvant for cancer treatment. In the near future, novel strategies for EA delivery systems that overcome the low EA solubility and bioavailability should be studied further to fully exploit the therapeutic potential of EA.
Collapse
Affiliation(s)
- Peiyu Xue
- College of Food Science and Engineering, Jilin University, Changchun 130062, China; School of Biology and Food Engineering, Anyang Institute of Technology, Anyang 455000, China
| | - Guangjie Zhang
- School of Biology and Food Engineering, Anyang Institute of Technology, Anyang 455000, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
22
|
Possible Beneficial Effects of Fresh Pomegranate Juice in SARS-CoV-2 Infection Conditions. J Nutr Metab 2022; 2022:5134560. [PMID: 35287379 PMCID: PMC8917946 DOI: 10.1155/2022/5134560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Rather than the prophylactic vaccination, any effective synthetic, natural, or nutritional therapy or regimen that may cure or remedy, albeit partially, the complications of SARS-CoV-2 should be highly acknowledged. Here, we reviewed and discussed possible beneficial biological effects of pomegranate juice in such diseased condition of viral infection based on the current published evidence (direct and indirect) and owing to the robust evidence that fresh pomegranate juice is highly rich with unique bioactive compounds that are approved in various occasions to be effective in several chronic diseased conditions. All related references that serve our aim are accessed through available electronic databases, particularly PubMed and Scopus. In summary, there is accepted evidence that pomegranate juice may be beneficial in SARS-CoV-2 infection conditions, especially for patients with the clinical history of chronic diseases such as hypertension, cardiovascular disease, diabetes, and cancer. However, the interventional studies that directly probe and confirm the effectiveness of fresh pomegranate juice in the management of SARS-CoV-2 infection are mandatory.
Collapse
|
23
|
Cheshomi H, Bahrami AR, Rafatpanah H, Matin MM. The effects of ellagic acid and other pomegranate ( Punica granatum L.) derivatives on human gastric cancer AGS cells. Hum Exp Toxicol 2022; 41:9603271211064534. [PMID: 35179410 DOI: 10.1177/09603271211064534] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Although surgery with or without (neo)adjuvant chemo/radiotherapy, as the standard treatments, can be suitable therapeutic strategies for gastric cancer, side effects and drug resistance are two main treatment obstacles. It has been discovered that pomegranate and its natural derivatives, especially ellagic acid (EA), offer significant anti-cancer effects while causing trivial side effects. In this study, we aimed to explore the anti-cancer effects of EA on a human gastric adenocarcinoma cell line (AGS) as well as in immunocompromised mice bearing human gastric tumors, for the first time. HPLC was used for determining EA in samples. MTT assay, apoptosis and scratch assay, gelatin zymography, and quantitative RT-PCR were used to determine the anti-cancer properties of different concentrations of pomegranate fruit juice, pomegranate peel extract, and EA. Furthermore, the effects of these compounds were investigated on immunosuppressed C57BL/6 mice carrying human gastric cancer tumors. EA could inhibit the proliferation and migration of gastric cancer cells. It also had significant effects on reducing both expression and activity of MMP-2 and MMP-9. Further, it was demonstrated that with alterations in the expression of genes involved in apoptosis and inflammation including P53, BAX, APAF1, BCL2, iNOS, NF-κB, IL-8, and TNF-α, EA treatment led to increased cancer cell death and reduced inflammation. Furthermore, its use in mice bearing gastric tumors resulted in a significant reduction in tumor volume without any obvious side effects. Ellagic acid exhibited anti-cancer effects on gastric adenocarcinoma, and can be considered as a safe anti-cancer agent for further preclinical studies on this cancer.
Collapse
Affiliation(s)
- Hamid Cheshomi
- Department of Biology, Faculty of Science, 48440Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, 48440Ferdowsi University of Mashhad, Mashhad, Iran.,Industrial Biotechnology Research Group, Institute of Biotechnology, 48440Ferdowsi University of Mashhad, Mashhad, Iran
| | - Houshang Rafatpanah
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, 48440Ferdowsi University of Mashhad, Mashhad, Iran.,Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
24
|
Jayatunga DPW, Hone E, Fernando WMADB, Garg ML, Verdile G, Martins RN. A Synergistic Combination of DHA, Luteolin, and Urolithin A Against Alzheimer's Disease. Front Aging Neurosci 2022; 14:780602. [PMID: 35250535 PMCID: PMC8890506 DOI: 10.3389/fnagi.2022.780602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common form of dementia worldwide. The classical AD brain is characterized by extracellular deposition of amyloid-β (Aβ) protein aggregates as senile plaques and intracellular neurofibrillary tangles (NFTs), composed of hyper-phosphorylated forms of the microtubule-associated protein Tau. There has been limited success in clinical trials for some proposed therapies for AD, so attention has been drawn toward using alternative approaches, including prevention strategies. As a result, nutraceuticals have become attractive compounds for their potential neuroprotective capabilities. The objective of the present study was to derive a synergistic nutraceutical combination in vitro that may act as a potential preventative therapy for AD. The compounds of interest were docosahexaenoic acid (DHA), luteolin (LUT), and urolithin A (UA). The cell viability and cytotoxicity assays MTS and LDH were used to evaluate the compounds individually and in two-compound combinations, for their ability to inhibit Aβ1-42-induced toxicity in human neuroblastoma BE(2)-M17 cells. The LDH-derived% protection values were used in the program CompuSyn v.1.0 to calculate the combination index (CI) of the two-compound combinations. The software-predicted potentially synergistic (CI < 1) two-compound combinations were validated using CellTiter Glo assay. Finally, a three-compound combination was predicted (D5L5U5) and shown to be the most effective at inhibiting Aβ1-42-induced toxicity. The synergistic combination, D5L5U5 warrants further research for its mechanism of action; however, it can serve as a basis to develop an advanced functional food for the prevention or co-treatment of AD.
Collapse
Affiliation(s)
- Dona P. W. Jayatunga
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Eugene Hone
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Cooperative Research Centre for Mental Health, Carlton, VIC, Australia
| | - W. M. A. D. Binosha Fernando
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Manohar L. Garg
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, Australia
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Giuseppe Verdile
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- Australian Alzheimer’s Research Foundation, Ralph and Patricia Sarich Neuroscience Research Institute, Nedlands, WA, Australia
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
25
|
Lee HS, Lee IH, Kang K, Park SI, Jung M, Yang SG, Kwon TW, Lee DY. A Network Pharmacology Study to Uncover the Mechanism of FDY003 for Ovarian Cancer Treatment. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221075432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological tumors responsible for 0.21 million deaths per year worldwide. Despite the increasing interest in the use of herbal drugs for cancer treatment, their pharmacological effects in OC treatment are not understood from a systems perspective. Using network pharmacology, we determined the anti-OC potential of FDY003 from a comprehensive systems view. We observed that FDY003 suppressed the viability of human OC cells and further chemosensitized them to cytotoxic chemotherapy. Through network pharmacological and pharmacokinetic approaches, we identified 16 active ingredients in FDY003 and their 108 targets associated with OC mechanisms. Functional enrichment investigation revealed that the targets may coordinate diverse cellular behaviors of OC cells, including their growth, proliferation, survival, death, and cell cycle regulation. Furthermore, the FDY003 targets are important constituents of diverse signaling pathways implicated in OC mechanisms (eg, phosphoinositide 3-kinase [PI3K]-Akt, mitogen-activated protein kinase [MAPK], focal adhesion, hypoxia-inducible factor [HIF]-1, estrogen, tumor necrosis factor [TNF], erythroblastic leukemia viral oncogene homolog [ErbB], Janus kinase [JAK]-signal transducer and activator of transcription [STAT], and p53 signaling). In summary, our data present a comprehensive understanding of the anti-OC effects and mechanisms of action of FDY003.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| | - In-Hee Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
| | - Kyungrae Kang
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| | - Sang-In Park
- Forestheal Hospitalo, Songpa-gu, Seoul, Republic of Korea
| | - Minho Jung
- Forest Hospital, Songpa-gu, Seoul, Republic of Korea
| | - Seung Gu Yang
- Kyunghee Naro Hospital, Bundang-gu, Seongnam, Republic of Korea
| | - Tae-Wook Kwon
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| | - Dae-Yeon Lee
- The Fore, Songpa-gu, Seoul, Republic of Korea
- Forest Hospital, Jongno-gu, Seoul, Republic of Korea
| |
Collapse
|
26
|
Chen Y, Luo Z, Lin J, Qi B, Sun Y, Li F, Guo C, Lin W, Kang X, He X, Wang Q, Chen S, Chen J. Exploring the Potential Mechanisms of Melilotus officinalis (L.) Pall. in Chronic Muscle Repair Patterns Using Single Cell Receptor-Ligand Marker Analysis and Molecular Dynamics Simulations. DISEASE MARKERS 2022; 2022:9082576. [PMID: 35692879 PMCID: PMC9177293 DOI: 10.1155/2022/9082576] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/28/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023]
Abstract
Information regarding the function of Melilotus officinalis (L.) Pall. in skeletal muscles is still unknown. In this study, we explored the possible regulatory targets of M. (L.) Pall. that affects the repair patterns in chronic muscle injury. We analyzed the potential target genes and chemical composition of M. (L.) Pall. and constructed a "drug-component-disease target genes" network analysis. Five active ingredients and 87 corresponding targets were obtained. Muscle-tendon junction (MTJ) cells were used to perform receptor-ligand marker analysis using the CellphoneDB algorithm. Targets of M. (L.) Pall. were screened further for the cellular ligand-receptor protein action on MTJs. Enrichment analysis suggests that those protein-associated ligand receptors may be associated with a range of intercellular signaling pathways. Molecular docking validation was then performed. Five proteins (CCL2, VEGFA, MMP2, MET, and EGFR) may be regulated by the active ingredient luteolin and scoparone. Finally, molecular dynamics simulations revealed that luteolin can stably target binding to MMP2. M. (L.) Pall. influences skeletal muscle repair patterns by affecting the fibroblast interactions in the muscle-tendon junctions through the active ingredients luteolin and scoparone.
Collapse
Affiliation(s)
- Yisheng Chen
- 1Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiwen Luo
- 1Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinrong Lin
- 1Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Beijie Qi
- 1Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaying Sun
- 1Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangqi Li
- 1Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Chenyang Guo
- 2Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| | - Weiwei Lin
- 3Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009 Zhejiang, China
| | - Xueran Kang
- 4Shanghai Jiao Tong University, Shanghai 200080, China
| | - Xinyi He
- 5State Key Laboratory of Genetics Engineering, Collaborative Innovation Center for Genetics and Development, School Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Qian Wang
- 6Postdoctoral Workstation, Department of Central Laboratory, The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, China
| | - Shiyi Chen
- 1Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiwu Chen
- 2Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China
| |
Collapse
|
27
|
Liu Y, Cao Y, Kai H, Han Y, Huang M, Gao L, Qiao H. Polyphyllin E inhibits proliferation, migration and invasion of ovarian cancer cells by down-regulating the AKT/NF-κB pathway. Biol Pharm Bull 2022; 45:561-568. [DOI: 10.1248/bpb.b21-00691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yinglei Liu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nantong University
| | - Yang Cao
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nantong University
| | - Haili Kai
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nantong University
| | - Yuwen Han
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nantong University
| | - Menghui Huang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nantong University
| | - Liusijie Gao
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nantong University
| | - Haifeng Qiao
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Nantong University
| |
Collapse
|
28
|
Xu X, Chen F, Zhang L, Liu L, Zhang C, Zhang Z, Li W. Exploring the mechanisms of anti-ovarian cancer of Hedyotis diffusa Willd and Scutellaria barbata D. Don through focal adhesion pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114343. [PMID: 34147618 DOI: 10.1016/j.jep.2021.114343] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/28/2021] [Accepted: 06/15/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hedyotis diffusa Willd and Scutellaria barbata D.Don (HD-SB) pairing were widely used as traditional medicine known for their anti-tumor effects. However, the inhibitory effect of HD-SB on ovarian cancer and its potential mechanisms were still not clear. AIM OF THE STUDY Our study identified the anti-tumor effect of HD-SB on ovarian cancer and analyzed the potential mechanisms by the network pharmacology and molecular docking method. MATERIALS AND METHODS The inhibitory effect of HD-SB combination on the growth and migration of ovarian cancer was detected by MTT and transwell assays. The effective ingredients of HD-SB and their potential targets were obtained from the Traditional Chinese Medicines for Systems Pharmacology Database (TCMSP), the GeneCards database, and the UniProt database. The relationships between active ingredients of HD-SB and potential targets or pathways of ovarian cancer were analyzed by String database, Cytoscape 3.7.2 software, and David 6.7 online database. The anti-ovarian cancer targets of HD-SB in the focal adhesion pathway were identified by RT-qPCR and molecular docking. RESULTS HD-SB combination significantly inhibited the proliferation and migration of ovarian cancer cells. We observed that the 1:2 ratio of HD-SB had the lowest IC50 value. 60 gene targets of 33 active ingredients in HD-SB were selected by pharmacokinetic parameters. The network pharmacological analysis showed that quercetin, luteolin, and baicalein might be the important anti-ovarian cancer ingredients in HD-SB, and the inhibitory effects of these three ingredients on the proliferation of ovarian cancer cells were verified respectively. Functional enrichment results suggested that HD-SB inhibited ovarian cancer growth and migration mainly through the focal adhesion pathway and the potential targets were EGFR, MAPK1, VEGFA, and PIK3CG. CONCLUSIONS HD-SB pairing significantly inhibited the proliferation and migration of ovarian cancer. Using network pharmacological methods and validation experiments, we found that HD-SB might, at least partially, inhibit ovarian cancer through the focal adhesion pathway. We believed that the HD-SB combination could be a potential therapeutic drug for the treatment of ovarian cancer patients.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Fenglin Chen
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Lin Zhang
- Academy of Integrative Medicine, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Cuili Zhang
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| | - Zhiwei Zhang
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Dalian, 116044, Liaoning, China.
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| |
Collapse
|
29
|
Maiuolo J, Gliozzi M, Carresi C, Musolino V, Oppedisano F, Scarano F, Nucera S, Scicchitano M, Bosco F, Macri R, Ruga S, Cardamone A, Coppoletta A, Mollace A, Cognetti F, Mollace V. Nutraceuticals and Cancer: Potential for Natural Polyphenols. Nutrients 2021; 13:nu13113834. [PMID: 34836091 PMCID: PMC8619660 DOI: 10.3390/nu13113834] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of death globally, associated with multifactorial pathophysiological components. In particular, genetic mutations, infection or inflammation, unhealthy eating habits, exposition to radiation, work stress, and/or intake of toxins have been found to contribute to the development and progression of cancer disease states. Early detection of cancer and proper treatment have been found to enhance the chances of survival and healing, but the side effects of anticancer drugs still produce detrimental responses that counteract the benefits of treatment in terms of hospitalization and survival. Recently, several natural bioactive compounds were found to possess anticancer properties, capable of killing transformed or cancerous cells without being toxic to their normal counterparts. This effect occurs when natural products are associated with conventional treatments, thereby suggesting that nutraceutical supplementation may contribute to successful anticancer therapy. This review aims to discuss the current literature on four natural bioactive extracts mostly characterized by a specific polyphenolic profile. In particular, several activities have been reported to contribute to nutraceutical support in anticancer treatment: (1) inhibition of cell proliferation, (2) antioxidant activity, and (3) anti-inflammatory activity. On the other hand, owing to their attenuation of the toxic effect of current anticancer therapies, natural antioxidants may contribute to improving the compliance of patients undergoing anticancer treatment. Thus, nutraceutical supplementation, along with current anticancer drug treatment, may be considered for better responses and compliance in patients with cancer. It should be noted, however, that when data from studies with bioactive plant preparations are discussed, it is appropriate to ensure that experiments have been conducted in accordance with accepted pharmacological research practices so as not to disclose information that is only partially correct.
Collapse
Affiliation(s)
- Jessica Maiuolo
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Vincenzo Musolino
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Oppedisano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Roberta Macri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Annarita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
| | - Annachiara Mollace
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Francesco Cognetti
- Medical Oncology 1, Regina Elena National Cancer Institute, IRCCS, 00144 Rome, Italy; (A.M.); (F.C.)
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (J.M.); (M.G.); (C.C.); (V.M.); (F.O.); (F.S.); (S.N.); (M.S.); (F.B.); (R.M.); (S.R.); (A.C.); (A.C.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
- Correspondence:
| |
Collapse
|
30
|
Paving Luteolin Therapeutic Potentialities and Agro-Food-Pharma Applications: Emphasis on In Vivo Pharmacological Effects and Bioavailability Traits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1987588. [PMID: 34594472 PMCID: PMC8478534 DOI: 10.1155/2021/1987588] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/30/2021] [Indexed: 11/25/2022]
Abstract
Luteolin is a naturally occurring secondary metabolite belonging to the class of flavones. As many other natural flavonoids, it is often found in combination with glycosides in many fruits, vegetables, and plants, contributing to their biological and pharmacological value. Many preclinical studies report that luteolin present excellent antioxidant, anticancer, antimicrobial, neuroprotective, cardioprotective, antiviral, and anti-inflammatory effects, and as a consequence, various clinical trials have been designed to investigate the therapeutic potential of luteolin in humans. However, luteolin has a very limited bioavailability, which consequently affects its biological properties and efficacy. Several drug delivery strategies have been developed to raise its bioavailability, with nanoformulations and lipid carriers, such as liposomes, being the most intensively explored. Pharmacological potential of luteolin in various disorders has also been underlined, but to some of them, the exact mechanism is still poorly understood. Given the great potential of this natural antioxidant in health, this review is aimed at providing an extensive overview on the in vivo pharmacological action of luteolin and at stressing the main features related to its bioavailability, absorption, and metabolism, while essential steps determine its absolute health benefits and safety profiles. In addition, despite the scarcity of studies on luteolin bioavailability, the different drug delivery formulations developed to increase its bioavailability are also listed here.
Collapse
|
31
|
Sha T, Li J, Sun S, Li J, Zhao X, Li Z, Cui Z. YEATS domain-containing 2 (YEATS2), targeted by microRNA miR-378a-5p, regulates growth and metastasis in head and neck squamous cell carcinoma. Bioengineered 2021; 12:7286-7296. [PMID: 34587874 PMCID: PMC8806651 DOI: 10.1080/21655979.2021.1977553] [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] [Indexed: 11/19/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide with poor prognosis and the development of HNSCC is a complex process. Some research have found that YEATS domain-containing 2 (YEATS2) is highly expressed in non-small cell lung cancer and pancreatic cancer, whereas its function in HNSCC is left to be studied. The primary aim was to investigate the role of YEATS2 in proliferation, apoptosis, invasion and migration in HNSCC cells and explore the possible mechanisms. We found YEATS2 expression was elevated in HNSCC clinical samples. Our work also indicated YEATS2 knockdown inhibited cell proliferation, induced apoptosis, and diminished the migration and invasion capability in HNSCC cell lines, including Detroit562 and FaDu cells. Besides, these inhibiting effects of YEATS2 knockdown could be crippled by microRNA-378a-5p (miR-378a-5p) inhibitor. In conclusion, our data suggested that YEATS2 expression was regulated by miR-378a-5p and YEATS2 knockdown inhibited proliferation and metastasis while induced apoptosis in HNSCC cells.
Collapse
Affiliation(s)
- Tong Sha
- The Third Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Jia Li
- Department of Oral and Maxillofacial Surgery Clinic, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Shiqun Sun
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Jianing Li
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Xuetao Zhao
- Department of Periodontics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Zehua Li
- Department of Pedodontics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Zhi Cui
- The Third Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
32
|
Wong TL, Strandberg KR, Croley CR, Fraser SE, Nagulapalli Venkata KC, Fimognari C, Sethi G, Bishayee A. Pomegranate bioactive constituents target multiple oncogenic and oncosuppressive signaling for cancer prevention and intervention. Semin Cancer Biol 2021; 73:265-293. [DOI: 10.1016/j.semcancer.2021.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/01/2020] [Accepted: 01/14/2021] [Indexed: 02/07/2023]
|
33
|
Farooqi AA. Regulation of deregulated cell signaling pathways by pomegranate in different cancers: Re-interpretation of knowledge gaps. Semin Cancer Biol 2021; 73:294-301. [DOI: 10.1016/j.semcancer.2021.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/05/2021] [Accepted: 01/21/2021] [Indexed: 12/27/2022]
|
34
|
Ziyuglycoside II exerts antiproliferative and antimetastasis effects on hepatocellular carcinoma cells. Anticancer Drugs 2021; 31:819-827. [PMID: 32097137 DOI: 10.1097/cad.0000000000000918] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide. Phytochemicals are important candidates for developing anticancer agents. Ziyuglycoside II is a major active compound of Sanguisorba officinalis, which exhibits antiproliferation activity in several cancers; however, its action in HCC remains unknown. In this study, we investigated the antitumor activity of ziyuglycoside II against HCC and explored the potential mechanisms. We found that ziyuglycoside II exerts significant inhibitory effects on the viability and clonogenic activity of HCC cells. The proliferation repression mediated by ziyuglycoside II was mainly due to increased apoptosis and reactive oxygen species accumulation, as well as a G0/G1 phase cell-cycle arrest. Additionally, ziyuglycoside II markedly impaired HCC cell migration and invasion, two important steps during metastasis, and these suppressive effects may be attributed to the downregulation of matrix metalloproteinases MMP2 and MMP9 expression. Moreover, ziyuglycoside II blocked the epidermal growth factor receptor/nuclear factor kappa-B (EGFR/NF-kB) signaling, which may contribute to its anticancer activity. Taken together, our findings reveal antiproliferative and antimetastasis activities of ziyuglycoside II in HCC cells, implying that ziyuglycoside II might be a promising candidate for the development of novel anti-HCC drugs.
Collapse
|
35
|
Shi ML, Chen YF, Wu WQ, Lai Y, Jin Q, Qiu WL, Yu DL, Li YZ, Liao HF. Luteolin inhibits the proliferation, adhesion, migration and invasion of choroidal melanoma cells in vitro. Exp Eye Res 2021; 210:108643. [PMID: 34058231 DOI: 10.1016/j.exer.2021.108643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/24/2021] [Accepted: 05/24/2021] [Indexed: 12/27/2022]
Abstract
Choroidal melanoma is a devastating disease that causes visual loss and a high mortality rate due to metastasis. Luteolin, a potential anticancer compound, is widely found in natural plants. The aim of this study was to evaluate the antiproliferative, antiadhesive, antimigratory and anti-invasive effects of luteolin on choroidal melanoma cells in vitro and to explore its potential mechanism. Cell counting kit-8 (CCK-8) assays, 5-ethynyl-2'-deoxyuridine (EdU) assays, Cell adhesion, migration, and invasion assays were performed to examine the inhibitory effects of luteolin on cell cell viability, proliferation, adhesion, migration and invasion capacities, respectively. Considering the correlation between Matrix metalloenzymes and tumor metastasis, Enzyme-linked immunosorbent assays (ELISAs) were used to assess matrix metalloproteases MMP-2 and MMP-9 secretion. Western blotting was performed to detect p-PI3K P85, Akt, and p-Akt protein expression. The cytoskeletal proteins vimentin were observed with cell immunofluorescence staining. Luteolin can inhibit OCM-1 cell proliferation, migration, invasion and adhesion and C918 cell proliferation, migration, and invasion through the PI3K/Akt signaling pathway. Therefore, Luteolin may have potential as a therapeutic medication for Choroidal melanoma.
Collapse
Affiliation(s)
- Meng-Lin Shi
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Province Blood Center, Nanchang, 330052, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China
| | - Yu-Fen Chen
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Wei-Qi Wu
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Yao Lai
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Qi Jin
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Wan-Lu Qiu
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Dong-Lian Yu
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Yi-Zhong Li
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Hong-Fei Liao
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China.
| |
Collapse
|
36
|
Mc Cormack B, Maenhoudt N, Fincke V, Stejskalova A, Greve B, Kiesel L, Meresman GF, Vankelecom H, Götte M, Barañao RI. The ellagic acid metabolites urolithin A and B differentially affect growth, adhesion, motility, and invasion of endometriotic cells in vitro. Hum Reprod 2021; 36:1501-1519. [PMID: 33748857 DOI: 10.1093/humrep/deab053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 02/08/2021] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION What are the effects of plant-derived antioxidant compounds urolithin A (UA) and B (UB) on the growth and pathogenetic properties of an in vitro endometriosis model? SUMMARY ANSWER Both urolithins showed inhibitory effects on cell behavior related to the development of endometriosis by differentially affecting growth, adhesion, motility, and invasion of endometriotic cells in vitro. WHAT IS KNOWN ALREADY Endometriosis is one of the most common benign gynecological diseases in women of reproductive age and is defined by the presence of endometrial tissue outside the uterine cavity. As current pharmacological therapies are associated with side effects interfering with fertility, we aimed at finding alternative therapeutics using natural compounds that can be administered for prolonged periods with a favorable side effects profile. STUDY DESIGN, SIZE, DURATION In vitro cultures of primary endometriotic stromal cells from 6 patients subjected to laparoscopy for benign pathologies with histologically confirmed endometriosis; and immortalized endometrial stromal (St-T1b) and endometriotic epithelial cells (12Z) were utilized to assess the effects of UA and UB on endometriotic cell properties. Results were validated in three-dimensional (3D) in vitro co-culture spheroids of 12Z and primary endometriotic stroma cells of one patient, and organoids from 3 independent donors with endometriosis. PARTICIPANTS/MATERIALS, SETTING, METHODS The effects on cell growth were measured by non-radioactive colorimetric assay to measure cellular metabolic activity as an indicator of cell viability (MTT assay) and flow cytometric cell cycle assay on primary cultures, St-T1b, and 12Z. Apoptosis analyses, the impact on in vitro adhesion, migration, and invasion were evaluated in the cell lines. Moreover, Real-Time Quantitative Reverse Transcription polymerase chain reaction (RT-qPCR) assays were performed on primary cultures, St- T1b and 12Z to evaluate a plausible mechanistic contribution by factors related to proteolysis (matrix metalloproteinase 2, 3 and 9 -MMP2, MMP3, MMP9-, and tissue inhibitor of metalloproteinases -TIMP-1-), cytoskeletal regulators (Ras-related C3 botulinum toxin substrate 1 -RAC1-, Rho-associated coiled-coil containing protein kinase 2 -ROCK2-), and cell adhesion molecules (Syndecan 1 -SDC1-, Integrin alpha V-ITGAV-). Finally, the urolithins effects were evaluated on spheroids and organoids by formation, viability, and drug screen assays. MAIN RESULTS AND THE ROLE OF CHANCE 40 µM UA and 20 µM UB produced a significant decrease in cell proliferation in the primary endometriotic cell cultures (P < 0.001 and P < 0.01, respectively) and in the St-T1b cell line (P < 0.001 and P < 0.05, respectively). In St-T1b, UA exhibited a mean half-maximum inhibitory concentration (IC50) of 39.88 µM, while UB exhibited a mean IC50 of 79.92 µM. Both 40 µM UA and 20 µM UB produced an increase in cells in the S phase of the cell cycle (P < 0.01 and P < 0.05, respectively). The same concentration of UA also increased the percentage of apoptotic ST-t1b cells (P < 0.05), while both urolithins decreased cell migration after 24 h (P < 0.001 both). Only the addition of 5 µM UB decreased the number of St-T1b adherent cells. TIMP-1 expression was upregulated in response to treating the cells with 40 µM UA (P < 0.05). Regarding the 12Z endometriotic cell line, only 40 µM UA decreased proliferation (P < 0.01); while both 40 µM UA and 20 µM UB produced an increase in cells in the G2/M phase (P < 0.05 and P < 0.01, respectively). In this cell line, UA exhibited a mean IC50 of 40.46 µM, while UB exhibited a mean IC50 of 54.79 µM. UB decreased cell migration (P < 0.05), and decreased the number of adherent cells (P < 0.05). Both 40 µM UA and 20 µM UB significantly decreased the cellular invasion of these cells; and several genes were altered when treating the cells with 40 µM UA and 10 µM UB. The expression of MMP2 was downregulated by UA (P < 0.001), and expression of MMP3 (UA P < 0.001 and UB P < 0.05) and MMP9 (P < 0.05, both) were downregulated by both urolithins. Moreover, UA significantly downregulated ROCK2 (P < 0.05), whereas UB treatment was associated with RAC1 downregulation (P < 0.05). Finally, the matrix adhesion receptors and signaling (co)receptors SDC1 and ITGAV were downregulated upon treatment with either UA or UB (P < 0.01 and P < 0.05, respectively in both cases). Regarding the effects of urolithins on 3D models, we have seen that they significantly decrease the viability of endometriosis spheroids (80 µM UA and UB: P < 0.05 both) as well as affecting their area (40 µM UA: P < 0.05, and 80 µM UA: P < 0.01) and integrity (40 µM UA and UB: P < 0.05, 80 µM UA and UB: P < 0.01). On the other hand, UA and UB significantly inhibited organoid development/outgrowth (40 and 80 µM UA: P < 0.0001 both; 40 µM UB: P < ns-0.05-0.001, and 80 µM UB: P < 0.01-0.001-0.001), and all organoid lines show urolithins sensitivity resulting in decreasing viability (UA exhibited a mean IC50 of 33.93 µM, while UB exhibited a mean IC50 of 52.60 µM). LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This study was performed on in vitro endometriosis models. WIDER IMPLICATIONS OF THE FINDINGS These in vitro results provide new insights into the pathogenetic pathways affected by these compounds and mark their use as a potential new therapeutic strategy for the treatment of endometriosis. STUDY FUNDING/COMPETING INTEREST(S) This study was funded EU MSCA-RISE-2015 project MOMENDO (691058). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- Barbara Mc Cormack
- Instituto de Biología y Medicina Experimental (IBYME)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - N Maenhoudt
- Laboratory of Tissue Plasticity in Health and Disease, Stem Cell and Developmental Biology Cluster, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - V Fincke
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - A Stejskalova
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - B Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Münster, Germany
| | - L Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - G F Meresman
- Instituto de Biología y Medicina Experimental (IBYME)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - H Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Stem Cell and Developmental Biology Cluster, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - M Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany
| | - R I Barañao
- Instituto de Biología y Medicina Experimental (IBYME)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
37
|
Ge S, Duo L, Wang J, Yang J, Li Z, Tu Y. A unique understanding of traditional medicine of pomegranate, Punica granatum L. and its current research status. JOURNAL OF ETHNOPHARMACOLOGY 2021; 271:113877. [PMID: 33515685 DOI: 10.1016/j.jep.2021.113877] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pomegranate, Punica granatum L., has been used in traditional medicine in China and several regions of the world including Ayurveda, Islamic, and Persian for the treatment of atherosclerosis, diabetes, hypertension, hyperlipidemia, and several types of cancer, as well as for peptic ulcer and oral diseases for hundreds of years. Presently, pomegranate is treated as both a "medicine food homology" herbal medicine and a healthy food supplemental product. AIM OF THE STUDY The aim of this work is to develop an overview of pomegranate in the context of the status of its traditional medicine theories, the spread along the Silk Road, ethnopharmacological uses, chemical compositions, pharmacological activities, toxicology, and the involved pathways. MATERIALS AND METHODS Information on P. granatum L. was acquired from published materials, including monographs on medicinal plants, ancient and modern recorded classical texts; and pharmacopoeias and electronic databases (PubMed, Science Direct, Web of Science, Google Scholar, CNKI, and Wanfang Data). RESULTS Pomegranate has been used in many traditional medical systems throughout history. It is widely cultivated in Central Asia and spread throughout China along the Silk Road. Many phytochemicals, such as tannins, organic acids, flavonoids, alkaloids, and volatile oils have been identified from different parts of pomegranate, these compounds have a wide range of activities, including antioxidant, antimicrobial, and anti-oncogenic properties, as well as conferring resistance to cerebrovascular disease. Furthermore, A summary of the four promising pharmacological pathways is provided. CONCLUSIONS The traditional uses, chemical compositions, pharmacological activities, and signaling pathways of pomegranate are summarized comprehensively in the review. It can be treated as a guidance for the future clinical and basic research. The information provided in this review will be very useful for further studies to develop novel therapeutic directions for application of pomegranate.
Collapse
Affiliation(s)
- Shasha Ge
- Medical Research Center, China Academy of Chinese Medical Science, Beijing, China; Development Research Center of TCM, China Academy of Chinese Medical Science, Beijing, China
| | - Lan Duo
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Junqi Wang
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Jingfan Yang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zhiyong Li
- School of Pharmacy, Minzu University of China, Beijing, China.
| | - Ya Tu
- Medical Research Center, China Academy of Chinese Medical Science, Beijing, China; Development Research Center of TCM, China Academy of Chinese Medical Science, Beijing, China.
| |
Collapse
|
38
|
Liu P, Guo Y, He Y, Tang Y. Radix Tetrastigma Hemsleyani Flavone Inhibits the Occurrence and Development of Ovarian Cancer Cells by Regulating miRNA-4458 Expression. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Ovarian cancer (OC) has been identified to have the highest mortality rate among gynecological tumors. Most patients are diagnosed at an advanced stage because of its asymptomatic nature and a lack of effective early diagnostic methods. Advanced-stage cancer cells are prone to metastasis
which reduces the efficacy of standard therapies. Thus, we evaluated the effect of different concentrations of radix tetrastigma hemsleyani flavone (RTHF) on SKOV3 OC cells. Our findings indicated a significant inhibition in cell proliferation, migration, and invasion. RTHF treatment resulted
in a significant increase in p21 protein expression, whereas the expression of cyclin D1, MMP-2, and MMP-9 has reportedly decreased. In addition, the expression of miRNA-4458 expression increased significantly in a dose-dependent manner. Co-transfection of miRNA-4458 mimics into SKOV3 cells
revealed that overexpressed miRNA-4458 can increase SKOV3 cell proliferation and p21 protein expression. Reduced cell migration and invasion were also observed along with decreased expression of cyclin D1, MMP-2, and MMP-9. Furthermore, inhibition of miRNA-4458 expression reversed the RTHF
effect on SKOV3 cell proliferation, migration, invasion, and cyclin D1, MMP-2, and MMP-9 expression. These results indicate that RTHF reduces the proliferation, migration, and invasion of OC cells, and the underlying mechanism is associated with the upregulation of miRNA-4458 expression. These
findings provide a new treatment strategy for advanced OC.
Collapse
Affiliation(s)
- Ping Liu
- Department of Obstetrics and Gynecology, The Affiliated Hospital North China University of Science and Technology, Tangshan 063000, Hebei, PR China
| | - Yanjuan Guo
- Department of Obstetrics and Gynecology, The Affiliated Hospital North China University of Science and Technology, Tangshan 063000, Hebei, PR China
| | - Yanfang He
- Department of Obstetrics and Gynecology, The Affiliated Hospital North China University of Science and Technology, Tangshan 063000, Hebei, PR China
| | - Yajuan Tang
- Department of Obstetrics and Gynecology, The Affiliated Hospital North China University of Science and Technology, Tangshan 063000, Hebei, PR China
| |
Collapse
|
39
|
Ciccone L, Vandooren J, Nencetti S, Orlandini E. Natural Marine and Terrestrial Compounds as Modulators of Matrix Metalloproteinases-2 (MMP-2) and MMP-9 in Alzheimer's Disease. Pharmaceuticals (Basel) 2021; 14:86. [PMID: 33498927 PMCID: PMC7911533 DOI: 10.3390/ph14020086] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/16/2022] Open
Abstract
Several studies have reported neuroprotective effects by natural products. A wide range of natural compounds have been investigated, and some of these may play a beneficial role in Alzheimer's disease (AD) progression. Matrix metalloproteinases (MMPs), a family of zinc-dependent endopeptidases, have been implicated in AD. In particular, MMP-2 and MMP-9 are able to trigger several neuroinflammatory and neurodegenerative pathways. In this review, we summarize and discuss existing literature on natural marine and terrestrial compounds, as well as their ability to modulate MMP-2 and MMP-9, and we evaluate their potential as therapeutic compounds for neurodegenerative and neuroinflammatory diseases, with a focus on Alzheimer's disease.
Collapse
Affiliation(s)
- Lidia Ciccone
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy; (L.C.); (S.N.)
| | - Jennifer Vandooren
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, KU Leuven—Herestraat 49—Box 1044, 3000 Leuven, Belgium;
| | - Susanna Nencetti
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy; (L.C.); (S.N.)
- Interdepartmental Research Centre “Nutraceuticals and Food for Health (NUTRAFOOD), University of Pisa, 56126 Pisa, Italy
| | - Elisabetta Orlandini
- Department of Earth Sciences, University of Pisa, via Santa Maria 53, 56126 Pisa, Italy
- Research Center “E. Piaggio”, University of Pisa, 56122 Pisa, Italy
| |
Collapse
|
40
|
Talib WH, AL-ataby IA, Mahmod AI, Jawarneh S, Al Kury LT, AL-Yasari IH. The Impact of Herbal Infusion Consumption on Oxidative Stress and Cancer: The Good, the Bad, the Misunderstood. Molecules 2020; 25:E4207. [PMID: 32937891 PMCID: PMC7570648 DOI: 10.3390/molecules25184207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 12/23/2022] Open
Abstract
The release of reactive oxygen species (ROS) and oxidative stress is associated with the development of many ailments, including cardiovascular diseases, diabetes and cancer. The causal link between oxidative stress and cancer is well established and antioxidants are suggested as a protective mechanism against cancer development. Recently, an increase in the consumption of antioxidant supplements was observed globally. The main sources of these antioxidants include fruits, vegetables, and beverage. Herbal infusions are highly popular beverages consumed daily for different reasons. Studies showed the potent antioxidant effects of plants used in the preparation of some herbal infusions. Such herbal infusions represent an important source of antioxidants and can be used as a dietary protection against cancer. However, uncontrolled consumption of herbal infusions may cause toxicity and reduced antioxidant activity. In this review, eleven widely consumed herbal infusions were evaluated for their antioxidant capacities, anticancer potential and possible toxicity. These herbal infusions are highly popular and consumed as daily drinks in different countries. Studies discussed in this review will provide a solid ground for researchers to have better understanding of the use of herbal infusions to reduce oxidative stress and as protective supplements against cancer development.
Collapse
Affiliation(s)
- Wamidh H. Talib
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (I.A.A.); (A.I.M.); (S.J.)
| | - Israa A. AL-ataby
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (I.A.A.); (A.I.M.); (S.J.)
| | - Asma Ismail Mahmod
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (I.A.A.); (A.I.M.); (S.J.)
| | - Sajidah Jawarneh
- Department of Clinical Pharmacy and Therapeutic, Applied Science Private University, Amman 11931-166, Jordan; (I.A.A.); (A.I.M.); (S.J.)
| | - Lina T. Al Kury
- Department of Health Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi 144534, UAE;
| | - Intisar Hadi AL-Yasari
- Department of Genetic Engineering, College of Biotechnology, Al-Qasim Green University, Babylon 00964, Iraq;
| |
Collapse
|
41
|
Sudha T, Mousa DS, El-Far AH, Mousa SA. Pomegranate ( Punica granatum) Fruit Extract Suppresses Cancer Progression and Tumor Angiogenesis of Pancreatic and Colon Cancer in Chick Chorioallantoic Membrane Model. Nutr Cancer 2020; 73:1350-1356. [PMID: 32757677 DOI: 10.1080/01635581.2020.1800768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pomegranate fruit extract contains many polyphenols and flavonoids of diverse biological importance including anticancer potential. In cancer, the angiogenesis process facilitates solid cancer growth and metastasis. Here, the antiangiogenic effect of pomegranate fruit extract against human pancreatic cancer (Suit-2) and colon (colo205) cell lines in the chick chorioallantoic membrane (CAM) model was studied along with the effect of pomegranate fruit extract on fibroblast growth factor (FGF2). Pomegranate fruit extract significantly reduced the tumor weight and hemoglobin content in CAM models of pancreatic Suit-2 and colon colo205.
Collapse
Affiliation(s)
- Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| | | | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, New York, USA
| |
Collapse
|
42
|
Başyiğit B, Sağlam H, Köroğlu K, Karaaslan M. Compositional analysis, biological activity, and food protecting ability of ethanolic extract of
Quercus infectoria
gall. J FOOD PROCESS PRES 2020. [DOI: 10.1111/jfpp.14692] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Bülent Başyiğit
- Engineering Faculty, Food Engineering Department Harran University Şanlıurfa Turkey
| | - Hidayet Sağlam
- Engineering‐Architecture Faculty, Food Engineering Department Kilis 7 Aralık University Kilis Turkey
| | - Kübra Köroğlu
- Engineering Faculty, Food Engineering Department Harran University Şanlıurfa Turkey
| | - Mehmet Karaaslan
- Engineering Faculty, Food Engineering Department Harran University Şanlıurfa Turkey
| |
Collapse
|
43
|
Wu X, Liu L, Zhang H. miR‑802 inhibits the epithelial‑mesenchymal transition, migration and invasion of cervical cancer by regulating BTF3. Mol Med Rep 2020; 22:1883-1891. [PMID: 32582971 PMCID: PMC7411396 DOI: 10.3892/mmr.2020.11267] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
MicroRNA (miR)-802 has been discovered to be involved in the occurrence and development of numerous types of tumor; however, studies into the role of miR‑802 in cervical cancer are limited. Therefore, the present study aimed to investigate the regulatory effects of miR‑802 in cervical cancer cells. miR‑802 expression levels in cervical cancer tissue and cells were analyzed using reverse transcription‑quantitative (RT‑q)PCR, a dual‑reporter luciferase activity assay was used to identify the direct target gene of miR‑802, and RT‑qPCR and western blotting were performed to determine the relationship between miR‑802 and basic transcription factor 3 (BTF3). Cell viability, and migration and invasion were analyzed using Cell Counting Kit‑8 and Transwell assays, respectively. Finally, the expression levels of metastasis‑associated proteins, N‑cadherin and E‑cadherin, were determined using RT‑qPCR and western blotting. Decreased expression levels of miR‑802 were found in cervical cancer tissues and cells, and the overexpression of miR‑802 inhibited cell viability, migration and invasion. Moreover, miR‑802 was discovered to directly target BTF3 to inhibit its expression. Notably, the overexpression miR‑802 markedly reversed the promotive effect of BTF3 on cell viability, in addition to the migratory and invasive abilities of the cells. Simultaneously, the overexpression of miR‑802 significantly suppressed epithelial‑mesenchymal transition, and the expression levels of matrix metallopeptidase (MMP)2 and MMP9 in cells through regulating BTF3. In conclusion, the present study revealed that miR‑802 may suppress cervical cancer progression by decreasing BTF3 expression levels, indicating that it may represent a potential therapeutic target for the treatment and prognosis of patients with cervical cancer.
Collapse
Affiliation(s)
- Xiuhui Wu
- Department of Gynecology, Jingmen No.1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Leng Liu
- Department of Gynecology, Jingmen No.1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Hongxia Zhang
- Department of Breast Surgery, Xiantao First People's Hospital, Xiantao, Hubei 433000, P.R. China
| |
Collapse
|
44
|
Zeng Z, Lin C, Wang S, Wang P, Xu W, Ma W, Wang J, Xiang Q, Liu Y, Yang J, Ye F, Xie K, Xu J, Luo Y, Liu SL, Liu H. Suppressive activities of mangiferin on human epithelial ovarian cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 76:153267. [PMID: 32570111 DOI: 10.1016/j.phymed.2020.153267] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 05/31/2020] [Accepted: 06/13/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Epithelial carcinoma is a subtype of ovarian cancers, with the highest lethality among all ovarian cancer subtypes. Hitherto surgical excision combined with chemotherapy has been the most extensively employed method in clinical treatment. However, the disease relapses very frequently, calling for more effective therapies. Mangiferin, a natural xanthone glucoside, has displayed promising anti-cancer activities by in vitro studies, but its therapeutic value in epithelial ovarian cancer treatment, either by in vivo or in vitro studies, remained to be known. PURPOSE This study aimed to determine the suppressive activities of mangiferin on human epithelial ovarian cancer and elucidate the underlying molecular mechanisms. STUDY DESIGN AND METHODS We employed the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and the crystal violet assay to determine the half maximal inhibitory concentration (IC50) values of mangiferin with paclitaxel as a positive control and the inhibitory effects of mangiferin on the proliferation of two human epithelial ovarian cancer cell lines. Wound healing and Transwell assays were used to determine anti-metastastic activities of mangiferin. ES-2 xenograft nude mouse model was used for the in vivo experiments. Western blotting, enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) assays were carried out for evaluating the expression level of matrix metalloproteinase 2 (MMP2) and matrix metalloproteinase 9 (MMP9). RESULTS In the present study, we demonstrated by both in vitro and in vivo assays that mangiferin suppressed the progress of epithelial ovarian cancer in a dose-dependent manner. In the animals treated with mangiferin, the tumor volume and weight were reduced significantly. Analyses of involved molecular events demonstrated that mangiferin down-regulated the expression of metastasis-associated proteins MMP2 and MMP9. CONCLUSION Mangiferin strongly inhibited the progression of human epithelial ovarian cancer by down-regulating MMP2 and MMP9.
Collapse
Affiliation(s)
- Zheng Zeng
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada
| | - Caiji Lin
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Siwen Wang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Pengfei Wang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Wenwen Xu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Wenqing Ma
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Jiali Wang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Qian Xiang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Yiting Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Jiaming Yang
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Fan Ye
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Kaihong Xie
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Jian Xu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Yao Luo
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China
| | - Shu-Lin Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China; Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, T2N 4N1, Canada.
| | - Huidi Liu
- Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), College of Pharmacy, Harbin Medical University, No. 157, Baojian Road, Harbin, China; HMU-UCCSM Centre for Infection and Genomics, Harbin, 150081, China; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang, China; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada.
| |
Collapse
|
45
|
Rizeq B, Gupta I, Ilesanmi J, AlSafran M, Rahman MDM, Ouhtit A. The Power of Phytochemicals Combination in Cancer Chemoprevention. J Cancer 2020; 11:4521-4533. [PMID: 32489469 PMCID: PMC7255361 DOI: 10.7150/jca.34374] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 12/03/2019] [Indexed: 12/25/2022] Open
Abstract
Conventional therapies for cancer treatment have posed many challenges, including toxicity, multidrug resistance and economic expenses. In contrast, complementary alternative medicine (CAM), employing phytochemicals have recently received increased attention owing to their capability to modulate a myriad of molecular mechanisms with a less toxic effect. Increasing evidence from preclinical and clinical studies suggest that phytochemicals can favorably modulate several signaling pathways involved in cancer development and progression. Combinations of phytochemicals promote cell death, inhibit cell proliferation and invasion, sensitize cancerous cells, and boost the immune system, thus making them striking alternatives in cancer therapy. We previously investigated the effect of six phytochemicals (Indol-3-Carbinol, Resveratrol, C-phycocyanin, Isoflavone, Curcumin and Quercetin), at their bioavailable levels on breast cancer cell lines and were compared to primary cell lines over a period of 6 days. This study showed the compounds had a synergestic effect in inhibiting cell proliferation, reducing cellular migration and invasion, inducing both cell cycle arrest and apoptosis. Despite the vast number of basic science and preclinical cancer studies involving phytochemicals, the number of CAM clinical trials in cancer treatment still remains nascent. In this review, we summarize findings from preclinical and clinical studies, including our work involving use of phytochemicals, individually as well as in combination and further discuss the potential of these phytochemicals to pave way to integrate CAM in primary health care.
Collapse
Affiliation(s)
- Balsam Rizeq
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Ishita Gupta
- College of Medicine, Qatar University, Doha, Qatar
| | - Josephine Ilesanmi
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Mohammed AlSafran
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - MD Mizanur Rahman
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Allal Ouhtit
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
46
|
Formulation Strategies to Improve Oral Bioavailability of Ellagic Acid. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10103353] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ellagic acid, a polyphenolic compound present in fruit and berries, has recently been the object of extensive research for its antioxidant activity, which might be useful for the prevention and treatment of cancer, cardiovascular pathologies, and neurodegenerative disorders. Its protective role justifies numerous attempts to include it in functional food preparations and in dietary supplements, and not only to limit the unpleasant collateral effects of chemotherapy. However, ellagic acid use as a chemopreventive agent has been debated because of its poor bioavailability associated with low solubility, limited permeability, first pass effect, and interindividual variability in gut microbial transformations. To overcome these drawbacks, various strategies for oral administration including solid dispersions, micro and nanoparticles, inclusion complexes, self-emulsifying systems, and polymorphs were proposed. Here, we listed an updated description of pursued micro and nanotechnological approaches focusing on the fabrication processes and the features of the obtained products, as well as on the positive results yielded by in vitro and in vivo studies in comparison to the raw material. The micro and nanosized formulations here described might be exploited for pharmaceutical delivery of this active, as well as for the production of nutritional supplements or for the enrichment of novel foods.
Collapse
|
47
|
Mete M, Ünsal ÜÜ, Aydemir I, Sönmez PK, Tuglu MI. Punicic Acid Inhibits Glioblastoma Migration and Proliferation via the PI3K/AKT1/mTOR Signaling Pathway. Anticancer Agents Med Chem 2020; 19:1120-1131. [PMID: 30950355 DOI: 10.2174/1871520619666190405112507] [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: 12/25/2018] [Revised: 01/08/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Punicic Acid (PA) is a polyunsaturated fatty acid that accounts for approximately 70%- 80% of Pomegranate Seed Oil (PSO). PA possesses strong antioxidant, anti-inflammatory, anti-atherogenic effects, and anti-tumorigenic properties. Pomegranate extracts have been shown to have anticancer activity in many studies. However, there is no evidence for the effect of PSO on T98 glioblastoma cells. Therefore, the present study was the first to investigate the mechanisms induced by PA on T98 cells, which is one of the major compounds extracted from PSO. METHODS The effects of PA on cell viability; oxidative stress; and migration, proliferation, and apoptosis at the IC50 dose were studied. RESULTS The proliferation and migration were inhibited in the treated group compared to the non-treated group by 9.85µl/ml PA. The difference was statistically significant (***p<0.001). Furthermore, PA-induced apoptosis in the T98 glioblastoma cells compared to non-treated group and the difference was statistically significant (***p<0.001). Apoptosis was determined via immunocytochemistry staining of caspase-3, caspase-9 and TUNEL methods. Apoptosis was checked by flow cytometry (using caspase 3 methods) and Scanning Electron Microscopy Analysis. We also investigated the potential signaling pathway underlying this apoptotic effect. The immunocytochemical stainings of PI3K/ Akt-1/ mTOR-1 demonstrated that Akt-1 staining was increased with PA treatment similar to mTOR-1 and PI3K staining (***p<0.001). These increases were statistically significant compared to the non-treated group. CONCLUSION PA exhibited exceptional abilities as an anticancer agent against GBM cells. The use of punicic acid in combination with other drugs used in the treatment of glioblastoma may increase the efficacy of the treatment. This study provided a basis for future investigation of its use in preclinical and clinical studies.
Collapse
Affiliation(s)
- Mesut Mete
- Neurosurgery Department, School of Medicine, Celal Bayar University, Manisa, Turkey
| | - Ülkün Ünlü Ünsal
- Neurosurgery Department, School of Medicine, Koc University, Istanbul, Turkey
| | - Işıl Aydemir
- Histology-Embryology Department, School of Medicine, Nigde Omer Halisdemir University, Nigde, Turkey
| | - Pınar K Sönmez
- Histology-Embryology Department, School of Medicine, Celal Bayar University, Manisa, Turkey
| | - Mehmet I Tuglu
- Histology-Embryology Department, School of Medicine, Celal Bayar University, Manisa, Turkey
| |
Collapse
|
48
|
Luteolin-7-O-Glucoside Inhibits Oral Cancer Cell Migration and Invasion by Regulating Matrix Metalloproteinase-2 Expression and Extracellular Signal-Regulated Kinase Pathway. Biomolecules 2020; 10:biom10040502. [PMID: 32224968 PMCID: PMC7226481 DOI: 10.3390/biom10040502] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
Oral squamous cell carcinoma is the sixth most common type of cancer globally, which is associated with high rates of cancer-related deaths. Metastasis to distant organs is the main reason behind worst prognostic outcome of oral cancer. In the present study, we aimed at evaluating the effects of a natural plant flavonoid, luteolin-7-O-glucoside, on oral cancer cell migration and invasion. The study findings showed that in addition to preventing cell proliferation, luteolin-7-O-glucoside caused a significant reduction in oral cancer cell migration and invasion. Mechanistically, luteolin-7-O-glucoside caused a reduction in cancer metastasis by reducing p38 phosphorylation and downregulating matrix metalloproteinase (MMP)-2 expression. Using a p38 inhibitor, SB203580, we proved that luteolin-7-O-glucoside exerts anti-migratory effects by suppressing p38-mediated increased expression of MMP-2. This is the first study to demonstrate the luteolin-7-O-glucoside inhibits cell migration and invasion by regulating MMP-2 expression and extracellular signal-regulated kinase pathway in human oral cancer cell. The study identifies luteolin-7-O-glucoside as a potential anti-cancer candidate that can be utilized clinically for improving oral cancer prognosis.
Collapse
|
49
|
Chaves FM, Pavan ICB, da Silva LGS, de Freitas LB, Rostagno MA, Antunes AEC, Bezerra RMN, Simabuco FM. Pomegranate Juice and Peel Extracts are Able to Inhibit Proliferation, Migration and Colony Formation of Prostate Cancer Cell Lines and Modulate the Akt/mTOR/S6K Signaling Pathway. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2020; 75:54-62. [PMID: 31838616 DOI: 10.1007/s11130-019-00776-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pomegranate (Punica granatum) is known to contain polyphenols with many potential health benefits, including anti-tumoral, anti-inflammatory, and anti-microbial properties. It has been used in popular medicine for cancer treatment, which still represents the major cause of cancer-related deaths in men worldwide. Importantly, pomegranate peels are valuable by-products of the food industry that are rich in polyphenols. Here we report a comparison between juice and peel aqueous extracts in prostate cancer DU-145 and PC-3 cell lines. Both extracts were able to inhibit the proliferation, migration and colony formation of those cells, although peel extracts presented more robust effects compared to juice. Besides, the growth-related mTOR/S6K signaling pathway presented strong inhibition after pomegranate extracts treatment. This study presents evidence that both juice and isolated peel extracts from promegate fruit have important anti-cancer effects against prostate cancer cells, modulating the mTOR/S6K signaling pathway.
Collapse
Affiliation(s)
- Fernanda Machado Chaves
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
- Dairy, Probiotics and Prebiotics Laboratory (LLPP), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
| | - Lidia Broglio de Freitas
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mauricio Ariel Rostagno
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
| | - Adriane Elisabete Costa Antunes
- Dairy, Probiotics and Prebiotics Laboratory (LLPP), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rosângela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), R. Pedro Zaccaria, 1300, Jardim São Paulo, CEP 13484-350, Limeira, São Paulo, Brazil.
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.
| |
Collapse
|
50
|
Baradaran Rahimi V, Ghadiri M, Ramezani M, Askari VR. Antiinflammatory and anti‐cancer activities of pomegranate and its constituent, ellagic acid: Evidence from cellular, animal, and clinical studies. Phytother Res 2020; 34:685-720. [DOI: 10.1002/ptr.6565] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 08/05/2019] [Accepted: 11/10/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Vafa Baradaran Rahimi
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical Sciences Mashhad Iran
| | - Mobarakeh Ghadiri
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical Sciences Mashhad Iran
| | - Mobina Ramezani
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical Sciences Mashhad Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal PlantsMashhad University of Medical Sciences Mashhad Iran
- Neurogenic Inflammation Research CenterMashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|