1
|
Mathews Paul B, Kannan G, Jegan Raj F, Velavan Sundararajan V, Annadurai Y, Piramanayagam S, Thangaraj P. GC-MS/HPLC Profiling and Sono-Maceration Mediated Extraction of Osbeckia Parvifolia Polyphenols: In Silico and In Vitro Analysis on Anti-Proliferative Activity in Ovarian Cancer Cell Lines. Chem Biodivers 2025; 22:e202402228. [PMID: 39417207 DOI: 10.1002/cbdv.202402228] [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: 09/06/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024]
Abstract
Osbeckia parvifolia, an endemic edible plant of Western Ghats, was investigated in the present study for its polyphenolic compounds, including content, constituents, extraction through an ultrasonic-assisted maceration technique and therapeutic potential in biomedical applications. The methanolic extract (OPM) exhibited an IC50 value of 1.25 μg/mL against 2,2-Diphenyl-1-picrylhydrazyl (DPPH) radicals. Furthermore, the ethyl acetate and methanolic extracts also strongly inhibited 5-lipoxygenase, especially OPM (84.93 %), which was comparable to standard curcumin. OPM also elicited cytotoxicity in SKOV3 ovarian cancer cells (93.80 %), surpassing paclitaxel. Bio-accessibility analysis demonstrated that the release of phenolic compounds and antioxidant potential were very high (above 100 %), revealing the possibility of synergistic efficacy of polyphenolic complexes in drug development. Gas Chromatography -Mass Spectrometry (GC-MS) analysis revealed 22 bioactive polyphenolic compounds in OPM, such as epicatechin, quercetin, and psoralidin. This was confirmed by High Performance Liquid Chromatography (HPLC) and High-Pressure Thin Layer Chromatography (HPTLC) analyses, which revealed a high quantity of catechin (37.45 mg/g). Molecular docking revealed the significant binding affinity of these proteins for the ovarian oncoproteins PI3K (-8.52 kcal/mol) and Casp-8 (-8.41 kcal/mol). Adsorption, Distribution, Metabolism, Excretion and Toxicity (ADMET) profiling indicated the favorable pharmacokinetic properties of these compounds, supporting their candidacy in drug formulations against ovarian cancer.
Collapse
Affiliation(s)
- Benedict Mathews Paul
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Gowtham Kannan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Francis Jegan Raj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Vetri Velavan Sundararajan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Yamuna Annadurai
- Computational Biology Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Shanmughavel Piramanayagam
- Computational Biology Laboratory, Department of Bioinformatics, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Parimelazhagan Thangaraj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| |
Collapse
|
2
|
Zhuang H, Zhang X, Wu S, Yong P, Yan H. Opportunities and challenges of foodborne polyphenols applied to anti-aging health foods. Food Sci Biotechnol 2024; 33:3445-3461. [PMID: 39493397 PMCID: PMC11525373 DOI: 10.1007/s10068-024-01686-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/27/2024] [Accepted: 08/12/2024] [Indexed: 11/05/2024] Open
Abstract
Abstract With the increasing proportion of the global aging population, aging mechanisms and anti-aging strategies become hot topics. Nonetheless, the safety of non-natural anti-aging active molecule and the changes in physiological function that occur during aging have not been clarified. There is therefore a need to develop safer pharmaceutical interventions for anti-aging. Numerous types of research have shown that food-derived biomolecules are of great interest due to their unique contribution to anti-aging safety issues and the prevention of degenerative diseases. Among these, polyphenolic organic compounds are widely used in anti-aging research for their ability to mitigate the physiological functional changes that occur during aging. The mechanisms include the free radical theory, immune aging theory, cellular autophagy theory, epigenetic modification theory, gut microbial effects on aging theory, telomere shortening theory, etc. This review elucidates the mechanisms underlying the anti-aging effects of polyphenols found in food-derived bioactive molecules, while also addressing the challenges associated with anti-aging pharmaceuticals. The review concludes by offering insights into the current landscape of anti-aging active molecule research, aiming to serve as a valuable resource for further scholarly inquiry. Graphical abstract
Collapse
Affiliation(s)
- Hong Zhuang
- College of Food Science and Engineering, Jilin University, Changchun, 130062 Jilin China
| | - Xiaoliang Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062 Jilin China
| | - Sijia Wu
- College of Food Science and Engineering, Jilin University, Changchun, 130062 Jilin China
| | - Pang Yong
- College of Food Science and Engineering, Jilin University, Changchun, 130062 Jilin China
| | - Haiyang Yan
- College of Food Science and Engineering, Jilin University, Changchun, 130062 Jilin China
| |
Collapse
|
3
|
Ejaz B, Mujib A, Syeed R, Mamgain J, Malik MQ, Birat K, Dewir YH, Magyar-Tábori K. Phytocompounds and Regulation of Flavonoids in In Vitro-Grown Safflower Plant Tissue by Abiotic Elicitor CdCl 2. Metabolites 2024; 14:127. [PMID: 38393019 PMCID: PMC10891796 DOI: 10.3390/metabo14020127] [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: 12/28/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
In this study, a Gas chromatography-mass spectrometry (GC-MS) investigation of embryogenic callus and somatic embryo regenerated shoots of Carthamus tinctorius revealed the presence of a variety of sugars, sugar acids, sugar alcohols, fatty acids, organic acids, and amino acids of broad therapeutic value. The in vitro developed inflorescence contained a wide range of active compounds. In embryogenic calluses, important flavonoids like naringenin, myricetin, kaempferol, epicatechin gallate, rutin, pelargonidin, peonidin, and delphinidin were identified. To augment the synthesis of active compounds, the effect of cadmium chloride (CdCl2) elicitation was tested for various treatments (T1-T4) along with a control (T0). Varying concentrations of CdCl2 [0.05 mM (T1), 0.10 mM (T2), 0.15 mM (T3), and 0.20 mM (T4)] were added to the MS medium, and flavonoid accumulation was quantified through ultra-high-pressure liquid chromatography-tandem mass spectroscopy (UHPLC-MS/MS). The flavonoids naringenin, kaempferol, epicatechin gallate, pelargonidin, cyanidin, and delphinidin increased by 6.7-, 1.9-, 3.3-, 2.1-, 1.9-, and 4.4-fold, respectively, at T3, whereas quercetin, myricetin, rutin, and peonidin showed a linear increase with the increase in CdCl2 levels. The impacts of stress markers, i.e., ascorbate peroxidase (APX), catalase (CAT), and superoxide dismutase (SOD), on defense responses in triggering synthesis were also evaluated. The maximum APX and SOD activity was observed at T3, while CAT activity was at its maximum at T2. The impact of elicitor on biochemical attributes like protein, proline, sugar, and malondialdehyde (MDA) content was investigated. The maximum protein, proline, and sugar accumulation was noted at high elicitor dose T4, while the maximum MDA content was noted at T3. These elevated levels of biochemical parameters indicated stress in culture, and the amendment of CdCl2 in media thus could be a realistic approach for enhancing secondary metabolite synthesis in safflower.
Collapse
Affiliation(s)
- Bushra Ejaz
- Cellular Differentiation and Molecular Genetics Section, Department of Botany, Jamia Hamdard, New Delhi 110062, India; (B.E.); (R.S.); (J.M.); (M.Q.M.); (K.B.)
| | - Abdul Mujib
- Cellular Differentiation and Molecular Genetics Section, Department of Botany, Jamia Hamdard, New Delhi 110062, India; (B.E.); (R.S.); (J.M.); (M.Q.M.); (K.B.)
| | - Rukaya Syeed
- Cellular Differentiation and Molecular Genetics Section, Department of Botany, Jamia Hamdard, New Delhi 110062, India; (B.E.); (R.S.); (J.M.); (M.Q.M.); (K.B.)
| | - Jyoti Mamgain
- Cellular Differentiation and Molecular Genetics Section, Department of Botany, Jamia Hamdard, New Delhi 110062, India; (B.E.); (R.S.); (J.M.); (M.Q.M.); (K.B.)
| | - Moien Qadir Malik
- Cellular Differentiation and Molecular Genetics Section, Department of Botany, Jamia Hamdard, New Delhi 110062, India; (B.E.); (R.S.); (J.M.); (M.Q.M.); (K.B.)
| | - Kanchan Birat
- Cellular Differentiation and Molecular Genetics Section, Department of Botany, Jamia Hamdard, New Delhi 110062, India; (B.E.); (R.S.); (J.M.); (M.Q.M.); (K.B.)
| | - Yaser Hassan Dewir
- Plant Production Department, College of Food and Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Katalin Magyar-Tábori
- Research Institute of Nyíregyháza, Institutes for Agricultural Research and Educational Farm (IAREF), University of Debrecen, P.O. Box 12, 4400 Nyíregyháza, Hungary;
| |
Collapse
|
4
|
Gao J, Wang Y, Jia Z, Xue J, Zhou T, Zu G. (-)-Epigallocatechin-3-gallate promotes intestinal epithelial proliferation and barrier function after ischemia/reperfusion injury via activation of Nurr1. PHARMACEUTICAL BIOLOGY 2023; 61:1310-1317. [PMID: 37621064 PMCID: PMC10461505 DOI: 10.1080/13880209.2023.2245445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 06/20/2023] [Accepted: 08/02/2023] [Indexed: 08/26/2023]
Abstract
CONTEXT (-)-Epigallocatechin-3-gallate (EGCG) is involved in cell proliferation and ischemia/reperfusion (I/R) injury of several organs. OBJECTIVE To identify the role of EGCG in intestinal epithelial proliferation and barrier exposed to I/R injury. MATERIAL AND METHODS Fifty Sprague-Dawley rats were divided into sham, I/R, I/R + EGCG (12.5 mg/kg), I/R + EGCG (25 mg/kg) and I/R + EGCG (50 mg/kg). I/R group rats were subjected to intestinal ischemia for 1 h and 6 h reperfusion. The rats were supplemented with EGCG 12.5, 25 and 50 mg/kg daily for 3 days via intraperitoneal injection before surgery. We used IEC-6 to expose to hypoxia/reoxygenation (H/R) injury to mimic I/R in vivo. IEC-6 cells were divided into control, H/R and H/R + EGCG (40 μmol/L). The effects of EGCG and its mechanism was explored. RESULTS Pharmacological treatment with EGCG notably improves intestinal epithelial proliferation (12.5 mg/kg, 1.74-fold; 25 mg/kg, 2.93-fold, and 50 mg/kg, 4.33-fold) and barrier function after I/R injury. EGCG promoted cell proliferation (2.99-fold) and increased the expression of occludin (2.36-fold) and ZO-1 (1.64-fold) in IEC-6 cells after H/R injury. EGCG promoted proliferation of IEC-6 cells with ED50 values of 18.16 μmol/L. Further investigations indicated that EGCG activated Nurr1 expression in intestine after I/R injury. EGCG promote cell proliferation and increased the expression of occludin and ZO-1 in IEC-6 cells after H/R injury were abrogated in the knockdown of Nurr1 by siRNA. DISCUSSION AND CONCLUSION Our findings indicate that EGCG promotes intestinal epithelial cell proliferation and barrier function after I/R injury in vitro and in vivo via activation of Nurr1.
Collapse
Affiliation(s)
- Jiacheng Gao
- Department of Gastroenterology Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Yuhang Wang
- Department of Gastroenterology Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Zirui Jia
- Department of Gastroenterology Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Jiaming Xue
- Department of Gastroenterology Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
- Department of Graduate School, Dalian Medical University, Dalian, China
| | - Tingting Zhou
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guo Zu
- Department of Gastroenterology Surgery, The Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China
| |
Collapse
|
5
|
Wardana AP, Aminah NS, Kristanti AN, Fahmi MZ, Zahrah HI, Widiyastuti W, Ajiz HA, Zubaidah U, Wiratama PA, Takaya Y. Nano Uncaria gambir as Chemopreventive Agent Against Breast Cancer. Int J Nanomedicine 2023; 18:4471-4484. [PMID: 37555190 PMCID: PMC10406122 DOI: 10.2147/ijn.s403385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/19/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Breast cancer is one of the main causes of death in women. Uncaria gambir is an Indonesian herbal plant that can be used as an anti-cancer. However, herbal medicines have low bioavailability, which affects their bioactivity. Nanoencapsulation can increase bioavailability and stability of bioactive compounds in herbal medicines. PURPOSE This recent finding tried to unravel anti-cancer and chemopreventive of U. gambir nano-encapsulated by Na-alginate. STUDY DESIGN U. gambir bioactive compounds were isolated and characterized using UV-Vis spectrometer, FTIR, NMR and HR-MS. U. gambir extract was nanoencapsulated using Na-alginate. Anti-cancer effect was assessed by MTT assay towards T47D cell. Meanwhile, a chemopreventive analysis was carried out in breast cancer mice-induced benzo[α]pyrene. The healthy mice were divided into 8 groups comprising control and treatment. RESULTS Elucidation of U. gambir ethyl acetate extract confirmed high catechin content, 89.34% (w/w). Successful nanoencapsulation of U. gambir (G-NPs) was indicated. The particle size of G-NPs was 78.40 ± 12.25 nm. Loading efficiency (LE) and loading amount (LA) of G-NPs were 97.56 ± 0.04% and 32.52 ± 0.01%, respectively. G-NPs had an EC50 value of 10.39 ± 3.50 µg/mL, which was more toxic than the EC50 value of extract towards the T47D cell line. Administration of 200 mg/kg BW G-NPs to mice induced by benzo[α]pyrene exhibited SOD and GSH levels of 13.69 ng/mL and 455.6 ng/mL. In addition, the lowest TNF-α level was 27.96 ng/mL. A dose of 100 mg/kg BW G-NPs could best increase CAT levels by 7.18 ng/mL. There was no damage or histological abnormalities found in histological analysis of the breast tissue in the group given 200 mg/kg BW G-NPs.
Collapse
Affiliation(s)
- Andika Pramudya Wardana
- Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Nanik Siti Aminah
- Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
- Biotechnology of Tropical Medicinal Plants Research Group, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Alfinda Novi Kristanti
- Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
- Biotechnology of Tropical Medicinal Plants Research Group, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Mochamad Zakki Fahmi
- Department of Chemistry, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
| | | | - W Widiyastuti
- Department of Chemical Engineering, Faculty of Industrial Technology and Systems Engineering, Institut Teknologi Sepuluh Nopember, Surabaya, East Java, Indonesia
| | - Hendrix Abdul Ajiz
- Department of Chemical Engineering, Faculty of Industrial Technology and Systems Engineering, Institut Teknologi Sepuluh Nopember, Surabaya, East Java, Indonesia
| | - Ummi Zubaidah
- Department of Biology, Faculty of Science and Technology, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Priangga Adi Wiratama
- Department of Anatomic Pathology, Faculty of Medicine, Universitas Airlangga – RSUD Dr. Soetomo Academic General Hospital, Surabaya, East Java, Indonesia
| | | |
Collapse
|
6
|
Majrashi TA, Alshehri SA, Alsayari A, Muhsinah AB, Alrouji M, Alshahrani AM, Shamsi A, Atiya A. Insight into the Biological Roles and Mechanisms of Phytochemicals in Different Types of Cancer: Targeting Cancer Therapeutics. Nutrients 2023; 15:nu15071704. [PMID: 37049544 PMCID: PMC10097354 DOI: 10.3390/nu15071704] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
Cancer is a hard-to-treat disease with a high reoccurrence rate that affects health and lives globally. The condition has a high occurrence rate and is the second leading cause of mortality after cardiovascular disorders. Increased research and more profound knowledge of the mechanisms contributing to the disease’s onset and progression have led to drug discovery and development. Various drugs are on the market against cancer; however, the drugs face challenges of chemoresistance. The other major problem is the side effects of these drugs. Therefore, using complementary and additional medicines from natural sources is the best strategy to overcome these issues. The naturally occurring phytochemicals are a vast source of novel drugs against various ailments. The modes of action by which phytochemicals show their anti-cancer effects can be the induction of apoptosis, the onset of cell cycle arrest, kinase inhibition, and the blocking of carcinogens. This review aims to describe different phytochemicals, their classification, the role of phytochemicals as anti-cancer agents, the mode of action of phytochemicals, and their role in various types of cancer.
Collapse
Affiliation(s)
- Taghreed A. Majrashi
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger, Abha 62529, Saudi Arabia
| | - Saad Ali Alshehri
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger, Abha 62529, Saudi Arabia
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger, Abha 62529, Saudi Arabia
- Complementary and Alternative Medicine Unit, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Abdullatif Bin Muhsinah
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger, Abha 62529, Saudi Arabia
- Complementary and Alternative Medicine Unit, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Mohammad Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Asma M. Alshahrani
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Akhtar Atiya
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger, Abha 62529, Saudi Arabia
| |
Collapse
|
7
|
Dai W, Yang J, Liu X, Mei Q, Peng W, Hu X. Anti-colorectal cancer of Ardisia gigantifolia Stapf. and targets prediction via network pharmacology and molecular docking study. BMC Complement Med Ther 2023; 23:4. [PMID: 36624500 PMCID: PMC9827653 DOI: 10.1186/s12906-022-03822-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 12/09/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Ardisia gigantifolia Stapf. (AGS), a Chinese folk medicine widely grows in the south of China and several studies reported that AGS could inhibit the proliferation of breast cancer, liver cancer, and bladder cancer cell lines. However, little is known about its anti-colorectal cancer (CRC) efficiency. METHODS In the present study, a combination of MTT assay, network pharmacological analysis, bioinformatics, molecular docking, and molecular dynamics simulation study was used to investigate the active ingredients, and targets of AGS against CRC, as well as the potential mechanism. RESULTS MTT assay showed that three kinds of fractions from AGS, including the n-butanol extract (NBAGS), ethyl acetate fraction (EAAGS), and petroleum ether fraction (PEAGS) significantly inhibited the proliferation of CRC cells, with the IC50 values of 197.24, 264.85, 15.45 µg/mL on HCT116 cells, and 523.6, 323.59, 150.31 µg/mL on SW620 cells, respectively. Eleven active ingredients, including, 11-O-galloylbergenin, 11-O-protocatechuoylbergenin, 11-O-syringylbergenin, ardisiacrispin B, bergenin, epicatechin-3-gallate, gallic acid, quercetin, stigmasterol, stigmasterol-3-o-β-D-glucopyranoside were identified. A total of 173 targets related to the bioactive components and 21,572 targets related to CRC were picked out through database searching. Based on the crossover targets of AGS and CRC, a protein-protein interaction network was built up by the String database, from which it was concluded that the core targets would be SRC, MAPK1, ESR1, HSP90AA1, MAPK8. Besides, GO analysis showed that the numbers of biological process, cellular component, and molecular function of AGS against CRC were 1079, 44, and 132, respectively, and KEGG pathway enrichment indicated that 96 signaling pathways in all would probably be involved in AGS against CRC, among which MAPK signaling pathway, lipid, and atherosclerosis, proteoglycans in cancer, prostate cancer, adherens junction would probably be the major pathways. The docking study verified that AGS had multiple ingredients and multiple targets against CRC. Molecular dynamics (MD) simulation analysis showed that the binding would be stable via forming hydrogen bonds. CONCLUSION Our study showed that AGS had good anti-CRC potency with the characteristics of multi-ingredients, -targets, and -signaling pathways.
Collapse
Affiliation(s)
- Weibo Dai
- grid.411866.c0000 0000 8848 7685Pharmacology Laboratory, Zhongshan Hospital, Guangzhou University of Chinese Medicine, 528401 Zhongshan, PR China
| | - Jing Yang
- grid.411866.c0000 0000 8848 7685Pharmacology Laboratory, Zhongshan Hospital, Guangzhou University of Chinese Medicine, 528401 Zhongshan, PR China ,Zhongshan Torch Development Zone People’s Hospital, 528401 Zhongshan, PR China
| | - Xin Liu
- grid.411866.c0000 0000 8848 7685Pharmacology Laboratory, Zhongshan Hospital, Guangzhou University of Chinese Medicine, 528401 Zhongshan, PR China
| | - Quanxi Mei
- Shenzhen Baoan Authentic TCM Therapy Hospital, 518101 Shenzhen, PR China
| | - Weijie Peng
- grid.411866.c0000 0000 8848 7685Pharmacology Laboratory, Zhongshan Hospital, Guangzhou University of Chinese Medicine, 528401 Zhongshan, PR China
| | - Xianjing Hu
- grid.410560.60000 0004 1760 3078Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, 523808 Dongguan, PR China
| |
Collapse
|
8
|
Majdan M, Bobrowska-Korczak B. Active Compounds in Fruits and Inflammation in the Body. Nutrients 2022; 14:2496. [PMID: 35745226 PMCID: PMC9229651 DOI: 10.3390/nu14122496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 12/10/2022] Open
Abstract
Inflammation plays an important role in the pathogenesis of many diseases, including cardiovascular diseases, atherosclerosis, diabetes, asthma, and cancer. An appropriate diet and the active compounds contained in it can affect various stages of the inflammatory process and significantly affect the course of inflammatory diseases. Recent reports indicate that polyphenolic acids, vitamins, minerals, and other components of fruits may exhibit activity stimulating an anti-inflammatory response, which may be of importance in maintaining health and reducing the risk of disease. The article presents the latest data on the chemical composition of fruits and the health benefits arising from their anti-inflammatory and antioxidant effects. The chemical composition of fruits determines their anti-inflammatory and antioxidant properties, but the mechanisms of action are not fully understood.
Collapse
Affiliation(s)
| | - Barbara Bobrowska-Korczak
- Department of Bromatology, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland;
| |
Collapse
|
9
|
Yoshida S, Inaba H, Nomura R, Nakano K, Matsumoto-Nakano M. Green tea catechins inhibit Porphyromonas gulae LPS-induced inflammatory responses in human gingival epithelial cells: Running title. J Oral Biosci 2022; 64:352-358. [PMID: 35660639 DOI: 10.1016/j.job.2022.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To determine the anti-inflammatory effects of green tea catechins in immortalized human gingival epithelial cells (Ca9-22) stimulated with Porphyromonas gulae lipopolysaccharide (LPS). METHODS Ca9-22 cells were incubated with P. gulae LPS (10 μg/ml) with or without green tea catechins, epigallocatechin-3-gallate (EGCg), epigallocatechin (EGC), epicatechin-3-gallate (ECG), and epicatechin (EC) (each at 50 μM), for 6 or 24 hours. Real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay were used to determine the induction of cyclooxygenase 2 (COX2), tumor necrosis factor alpha (TNF-ɑ), interleukin 6 (IL-6), and IL-8. Furthermore, the expression of toll-like receptors (TLRs) 2 and 4 was examined using real-time PCR and western blotting analysis, and phosphorylation of the p38 and ERK1/2 was examined using western blotting analysis. RESULTS At the mRNA and protein levels, EGCg, EGC, ECG, and EC were found to significantly inhibit COX2, TNF-ɑ, IL-6, and IL-8. Furthermore, the levels of ERK1/2 and p38 phosphorylation induced by P. gulae LPS were decreased following the addition of each of the catechins, as well as TLR2 and 4 mRNA and protein. CONCLUSIONS These findings indicate that green tea catechins are potent inhibitors of inflammatory responses induced by P. gulae LPS, and may also be useful for prevention and/or attenuation of periodontitis.
Collapse
Affiliation(s)
- Sho Yoshida
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Inaba
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Ryota Nomura
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Michiyo Matsumoto-Nakano
- Department of Pediatric Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
10
|
Wang S, Zeng T, Zhao S, Zhu Y, Feng C, Zhan J, Li S, Ho CT, Gosslau A. Multifunctional health-promoting effects of oolong tea and its products. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
11
|
Li Z, Feng C, Dong H, Jin W, Zhang W, Zhan J, Wang S. Health promoting activities and corresponding mechanism of (–)-epicatechin-3-gallate. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2021.12.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
12
|
Hong M, Cheng L, Liu Y, Wu Z, Zhang P, Zhang X. A Natural Plant Source-Tea Polyphenols, a Potential Drug for Improving Immunity and Combating Virus. Nutrients 2022; 14:nu14030550. [PMID: 35276917 PMCID: PMC8839699 DOI: 10.3390/nu14030550] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is still in a global epidemic, which has profoundly affected people’s lives. Tea polyphenols (TP) has been reported to enhance the immunity of the body to COVID-19 and other viral infectious diseases. The inhibitory effect of TP on COVID-19 may be achieved through a series of mechanisms, including the inhibition of multiple viral targets, the blocking of cellular receptors, and the activation of transcription factors. Emerging evidence shows gastrointestinal tract is closely related to respiratory tract, therefore, the relationship between the state of the gut–lung axis microflora and immune homeostasis of the host needs further research. This article summarized that TP can improve the disorder of flora, reduce the occurrence of cytokine storm, improve immunity, and prevent COVID-19 infection. TP may be regarded as a potential and valuable source for the design of new antiviral drugs with high efficiency and low toxicity.
Collapse
Affiliation(s)
- Mengyu Hong
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (M.H.); (Y.L.); (Z.W.)
| | - Lu Cheng
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA;
| | - Yanan Liu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (M.H.); (Y.L.); (Z.W.)
| | - Zufang Wu
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (M.H.); (Y.L.); (Z.W.)
| | - Peng Zhang
- Department of Student Affairs, Xinyang Normal University, Xinyang 464000, China
- Correspondence: (P.Z.); (X.Z.)
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo 315211, China; (M.H.); (Y.L.); (Z.W.)
- Correspondence: (P.Z.); (X.Z.)
| |
Collapse
|
13
|
Mahmud S, Biswas S, Paul GK, Mita MA, Promi MM, Afrose S, Hasan MR, Zaman S, Uddin MS, Dhama K, Emran TB, Saleh MA, Simal-Gandara J. Plant-Based Phytochemical Screening by Targeting Main Protease of SARS-CoV-2 to Design Effective Potent Inhibitors. BIOLOGY 2021; 10:589. [PMID: 34206970 PMCID: PMC8301192 DOI: 10.3390/biology10070589] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Currently, a worldwide pandemic has been declared in response to the spread of coronavirus disease 2019 (COVID-19), a fatal and fast-spreading viral infection caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The low availability of efficient vaccines and treatment options has resulted in a high mortality rate, bringing the world economy to its knees. Thus, mechanistic investigations of drugs capable of counteracting this disease are in high demand. The main protease (Mpro) expressed by SARS-CoV-2 has been targeted for the development of potential drug candidates due to the crucial role played by Mpro in viral replication and transcription. We generated a phytochemical library containing 1672 phytochemicals derived from 56 plants, which have been reported as having antiviral, antibacterial, and antifungal activity. A molecular docking program was used to screen the top three candidate compounds: epicatechin-3-O-gallate, psi-taraxasterol, and catechin gallate, which had respective binding affinities of -8.4, -8.5, and -8.8 kcal/mol. Several active sites in the targeted protein, including Cys145, His41, Met49, Glu66, and Met165, were found to interact with the top three candidate compounds. The multiple simulation profile, root-mean-square deviation, root-mean-square fluctuation, radius of gyration, and solvent-accessible surface area values supported the inflexible nature of the docked protein-compound complexes. The toxicity and carcinogenicity profiles were assessed, which showed that epicatechin-3-O-gallate, psi-taraxasterol, and catechin gallate had favorable pharmacological properties with no adverse effects. These findings suggest that these compounds could be developed as part of an effective drug development pathway to treat COVID-19.
Collapse
Affiliation(s)
- Shafi Mahmud
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.M.); (G.K.P.); (S.Z.); (M.S.U.)
| | - Suvro Biswas
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.B.); (M.A.M.); (M.M.P.); (S.A.); (M.R.H.)
| | - Gobindo Kumar Paul
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.M.); (G.K.P.); (S.Z.); (M.S.U.)
| | - Mohasana Akter Mita
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.B.); (M.A.M.); (M.M.P.); (S.A.); (M.R.H.)
| | - Maria Meha Promi
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.B.); (M.A.M.); (M.M.P.); (S.A.); (M.R.H.)
| | - Shamima Afrose
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.B.); (M.A.M.); (M.M.P.); (S.A.); (M.R.H.)
| | - Md. Robiul Hasan
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.B.); (M.A.M.); (M.M.P.); (S.A.); (M.R.H.)
| | - Shahriar Zaman
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.M.); (G.K.P.); (S.Z.); (M.S.U.)
| | - Md. Salah Uddin
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.M.); (G.K.P.); (S.Z.); (M.S.U.)
| | - Kuldeep Dhama
- Division of Pathology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Md. Abu Saleh
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh; (S.M.); (G.K.P.); (S.Z.); (M.S.U.)
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo–Ourense Campus, E32004 Ourense, Spain
| |
Collapse
|
14
|
Wang S, Li Z, Ma Y, Liu Y, Lin CC, Li S, Zhan J, Ho CT. Immunomodulatory Effects of Green Tea Polyphenols. Molecules 2021; 26:molecules26123755. [PMID: 34203004 PMCID: PMC8234133 DOI: 10.3390/molecules26123755] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 01/03/2023] Open
Abstract
Green tea and its bioactive components, especially polyphenols, possess many health-promoting and disease-preventing benefits, especially anti-inflammatory, antioxidant, anticancer, and metabolic modulation effects with multi-target modes of action. However, the effect of tea polyphenols on immune function has not been well studied. Moreover, the underlying cellular and molecular mechanisms mediating immunoregulation are not well understood. This review summarizes the recent studies on the immune-potentiating effects and corresponding mechanisms of tea polyphenols, especially the main components of (-)-epigallocatechin-3-gallate (EGCG) and (-)-epicatechin-3-gallate (ECG). In addition, the benefits towards immune-related diseases, such as autoimmune diseases, cutaneous-related immune diseases, and obesity-related immune diseases, have been discussed.
Collapse
Affiliation(s)
- Shuzhen Wang
- Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains, Hubei Zhongke Research Institute of Industrial Technology, College of Life Science, Huanggang Normal University, Huanggang 438000, China; (S.W.); (Z.L.); (Y.M.); (Y.L.); (J.Z.)
| | - Zhiliang Li
- Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains, Hubei Zhongke Research Institute of Industrial Technology, College of Life Science, Huanggang Normal University, Huanggang 438000, China; (S.W.); (Z.L.); (Y.M.); (Y.L.); (J.Z.)
| | - Yuting Ma
- Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains, Hubei Zhongke Research Institute of Industrial Technology, College of Life Science, Huanggang Normal University, Huanggang 438000, China; (S.W.); (Z.L.); (Y.M.); (Y.L.); (J.Z.)
| | - Yan Liu
- Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains, Hubei Zhongke Research Institute of Industrial Technology, College of Life Science, Huanggang Normal University, Huanggang 438000, China; (S.W.); (Z.L.); (Y.M.); (Y.L.); (J.Z.)
| | - Chi-Chen Lin
- Institute of Biomedical Science, The iEGG and Animal Biotechnology Center, National Chung-Hsing University, Taichung 402, Taiwan;
| | - Shiming Li
- Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains, Hubei Zhongke Research Institute of Industrial Technology, College of Life Science, Huanggang Normal University, Huanggang 438000, China; (S.W.); (Z.L.); (Y.M.); (Y.L.); (J.Z.)
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
- Correspondence: (S.L.); (C.-T.H.)
| | - Jianfeng Zhan
- Hubei Collaborative Innovation Center for the Characteristic Resources Exploitation of Dabie Mountains, Hubei Zhongke Research Institute of Industrial Technology, College of Life Science, Huanggang Normal University, Huanggang 438000, China; (S.W.); (Z.L.); (Y.M.); (Y.L.); (J.Z.)
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
- Correspondence: (S.L.); (C.-T.H.)
| |
Collapse
|
15
|
Jiang Y, Jiang Z, Ma L, Huang Q. Advances in Nanodelivery of Green Tea Catechins to Enhance the Anticancer Activity. Molecules 2021; 26:3301. [PMID: 34072700 PMCID: PMC8198522 DOI: 10.3390/molecules26113301] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the leading causes of death globally. A variety of phenolic compounds display preventative and therapeutic effects against cancers. Green teas are rich in phenolics. Catechins are the most dominant phenolic component in green teas. Studies have shown that catechins have anticancer activity in various cancer models. The anticancer activity of catechins, however, may be compromised due to their low oral bioavailability. Nanodelivery emerges as a promising way to improve the oral bioavailability and anticancer activity of catechins. Research in this area has been actively conducted in recent decades. This review provides the molecular mechanisms of the anticancer effects of catechins, the factors that limit the oral bioavailability of catechins, and the latest advances of delivering catechins using nanodelivery systems through different routes to enhance their anticancer activity.
Collapse
Affiliation(s)
- Yike Jiang
- Shenzhen Bay Laboratory, Institute of Biomedical Health Technology and Engineering, Shenzhen 518132, China;
| | - Ziyi Jiang
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China;
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Lan Ma
- Shenzhen Bay Laboratory, Institute of Biomedical Health Technology and Engineering, Shenzhen 518132, China;
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China;
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Qingrong Huang
- Department of Food Science, Rutgers University, 65 Dudley Road, New Brunswick, NJ 08901, USA
| |
Collapse
|
16
|
Dziąbowska-Grabias K, Sztanke M, Zając P, Celejewski M, Kurek K, Szkutnicki S, Korga P, Bulikowski W, Sztanke K. Antioxidant Therapy in Inflammatory Bowel Diseases. Antioxidants (Basel) 2021; 10:antiox10030412. [PMID: 33803138 PMCID: PMC8000291 DOI: 10.3390/antiox10030412] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic, incurable diseases of the digestive tract, the etiology of which remains unclear to this day. IBD result in significant repercussions on the quality of patients’ life. There is a continuous increase in the incidence and prevalence of IBD worldwide, and it is becoming a significant public health burden. Pharmaceuticals commonly used in IBD management, for example, mesalamine, sulfasalazine, corticosteroids, and others, expose patients to diverse, potentially detrimental side effects and frequently do not provide sufficient disease control. The chronic inflammation underlies the etiology of IBD and closely associates with oxidative/nitrosative stress and a vast generation of reactive oxygen/nitrogen species. Relative to this, several substances with antioxidant and anti-inflammatory properties are now intensively researched as possible adjunctive or independent treatment options in IBD. Representatives of several different groups, including natural and chemical compounds will be characterized in this dissertation.
Collapse
Affiliation(s)
- Katarzyna Dziąbowska-Grabias
- Department of Gastroenterology, 1st Military Research Hospital, and Polyclinic of Lublin, 20-049 Lublin, Poland; (K.D.-G.); (P.Z.); (M.C.)
| | - Małgorzata Sztanke
- Department of Medical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence: ; Tel.: +48-814-486-195
| | - Przemysław Zając
- Department of Gastroenterology, 1st Military Research Hospital, and Polyclinic of Lublin, 20-049 Lublin, Poland; (K.D.-G.); (P.Z.); (M.C.)
| | - Michał Celejewski
- Department of Gastroenterology, 1st Military Research Hospital, and Polyclinic of Lublin, 20-049 Lublin, Poland; (K.D.-G.); (P.Z.); (M.C.)
| | - Katarzyna Kurek
- Department of Pneumonology, Oncology, and Allergology, Medical University of Lublin, 20-090 Lublin, Poland; (K.K.); (S.S.)
| | - Stanisław Szkutnicki
- Department of Pneumonology, Oncology, and Allergology, Medical University of Lublin, 20-090 Lublin, Poland; (K.K.); (S.S.)
| | - Patryk Korga
- Department of Gastroenterology, 10ft Military Research Hospital, and Polyclinic of Bydgoszcz, 85-681 Bydgoszcz, Poland;
| | | | - Krzysztof Sztanke
- Laboratory of Bioorganic Synthesis and Analysis, Chair and Department of Medical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland;
| |
Collapse
|
17
|
Wang F, Han Y, Xi S, Lu Y. Catechins reduce inflammation in lipopolysaccharide-stimulated dental pulp cells by inhibiting activation of the NF-κB pathway. Oral Dis 2020; 26:815-821. [PMID: 31999881 DOI: 10.1111/odi.13290] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 01/02/2020] [Accepted: 01/17/2020] [Indexed: 12/19/2022]
Abstract
AIM To ascertain the anti-inflammation mechanism of catechins in lipopolysaccharide-treated human dental pulp cells (HDPCs). METHODS Expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 was measured using quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assays. The anti-inflammatory mechanism was explored by examining activation of nuclear factor-kappa B (NF-κB) signaling using qPCR, Western blotting, and immunofluorescence staining. RESULTS Human dental pulp cells proliferation was not affected by treatment with epigallocatechin (ECG) or epigallocatechin 3-gallate (EGCG). mRNA expression of the pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 was decreased significantly in ECG- and EGCG-treated HDPCs. Subsequently, the effects of ECG and EGCG upon activation of NF-κB signaling were evaluated by Western blotting and immunofluorescence staining. Expression of p-p65 protein in HDPCs treated with ECG, EGCG, or an NF-κB inhibitor (Bay 11-7082) was lower than that in HDPCs treated with lipopolysaccharide, data that were consistent with the location of p65 protein according to immunofluorescence staining. CONCLUSIONS Catechin could reduce lipopolysaccharide-stimulated inflammation in HDPCs by inhibiting activation of the NF-κB pathway.
Collapse
Affiliation(s)
- Fang Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ying Han
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shuang Xi
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yi Lu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
18
|
Tung YC, Chou YC, Hung WL, Cheng AC, Yu RC, Ho CT, Pan MH. Polymethoxyflavones: Chemistry and Molecular Mechanisms for Cancer Prevention and Treatment. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s40495-019-00170-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
19
|
Zhao CN, Zhang JJ, Li Y, Meng X, Li HB. Microwave-Assisted Extraction of Phenolic Compounds from Melastoma sanguineum Fruit: Optimization and Identification. Molecules 2018; 23:molecules23102498. [PMID: 30274261 PMCID: PMC6222716 DOI: 10.3390/molecules23102498] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/18/2018] [Accepted: 09/27/2018] [Indexed: 12/25/2022] Open
Abstract
A microwave-assisted extraction (MAE) technology optimized by response surface methodology (RSM) was established to extract phenolic compounds from the fruit of Melastoma sanguineum. The effects of solvent composition, ratio of solvent to material, temperature, time and microwave power on phenol yield were evaluated in single-factor tests. The three parameters exerting main impacts on phenol yield were further optimized by RSM. Under optimal extraction conditions (31.33% ethanol, solvent/material ratio of 32.21 mL/g, 52.24 °C, 45 min and 500 W), the total phenolic content was 39.02 ± 0.73 mg gallic acid equivalent (GAE)/g dry weight (DW). This MAE method performed better in comparison with two conventional methods, those being maceration (25.79 ± 1.03 mg GAE/g DW) and Soxhlet extraction (18.40 ± 1.34 mg GAE/g DW), using lower process temperature, shorter irradiation time, and lower organic solvent consumption. In addition, five flavonoids (epicatechin gallate, epicatechin, rutin, pigallocatechin and quercetin) and two phenolic acids (protocatechuic acid and chlorogenic acid) in the extract were identified and quantified using UPLC-MS/MS.
Collapse
Affiliation(s)
- Cai-Ning Zhao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangdong Engineering Technology Research Center of Nutrition Translation, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Jiao-Jiao Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangdong Engineering Technology Research Center of Nutrition Translation, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Ya Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangdong Engineering Technology Research Center of Nutrition Translation, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Xiao Meng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangdong Engineering Technology Research Center of Nutrition Translation, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangdong Engineering Technology Research Center of Nutrition Translation, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
- South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-Sen University, Guangzhou 510006, China.
| |
Collapse
|
20
|
Xu M, Zheng M, Liu G, Zhang M, Kang J. Screening of break point cluster region Abelson tyrosine kinase inhibitors by capillary electrophoresis. J Chromatogr A 2018; 1537:128-134. [DOI: 10.1016/j.chroma.2018.01.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 12/31/2022]
|
21
|
Jiao L, Bi L, Lu Y, Wang Q, Gong Y, Shi J, Xu L. Cancer chemoprevention and therapy using chinese herbal medicine. Biol Proced Online 2018; 20:1. [PMID: 29321719 PMCID: PMC5757296 DOI: 10.1186/s12575-017-0066-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 12/20/2017] [Indexed: 12/20/2022] Open
Abstract
Traditional Chinese medicine (TCM) plays an indispensable role in cancer prevention and treatment. Chinese herbal medicine (CHM) is a key component of TCM and has been practiced for thousands of years. A number of naturally occurring products from Chinese herbs extracts exhibit strong inhibitory properties against carcinogenesis, including CHM single-herb extracts, CHM-derived active components, and CHM formulas (the polyherbal combinations), which regulate JAK/STAT, MAPK, and NF-ҡB pathways. The present review aims to report the cancer-preventive effect of CHM with evidence from cell-line, animal, epidemiological, and clinical experiments. We also present several issues that have yet to be resolved. In the future, cancer prevention by CHM will face unprecedented opportunities and challenges.
Collapse
Affiliation(s)
- Lijing Jiao
- Institute of Clinical Immunology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437 China.,Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110, Ganhe Road, Hongkou District, Shanghai, 200437 China
| | - Ling Bi
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110, Ganhe Road, Hongkou District, Shanghai, 200437 China
| | - Yan Lu
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110, Ganhe Road, Hongkou District, Shanghai, 200437 China
| | - Qin Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110, Ganhe Road, Hongkou District, Shanghai, 200437 China
| | - Yabin Gong
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110, Ganhe Road, Hongkou District, Shanghai, 200437 China
| | - Jun Shi
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110, Ganhe Road, Hongkou District, Shanghai, 200437 China
| | - Ling Xu
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, No.110, Ganhe Road, Hongkou District, Shanghai, 200437 China
| |
Collapse
|
22
|
Sinha D, Biswas J, Nabavi SM, Bishayee A. Tea phytochemicals for breast cancer prevention and intervention: From bench to bedside and beyond. Semin Cancer Biol 2017; 46:33-54. [DOI: 10.1016/j.semcancer.2017.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/25/2017] [Accepted: 04/01/2017] [Indexed: 02/06/2023]
|
23
|
Xie J, Yu H, Song S, Fang C, Wang X, Bai Z, Ma X, Hao S, Zhao HY, Sheng J. Pu-erh Tea Water Extract Mediates Cell Cycle Arrest and Apoptosis in MDA-MB-231 Human Breast Cancer Cells. Front Pharmacol 2017; 8:190. [PMID: 28428754 PMCID: PMC5382226 DOI: 10.3389/fphar.2017.00190] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 03/23/2017] [Indexed: 12/24/2022] Open
Abstract
Pu-erh tea is believed to have health benefits, the growth inhibition activity of Pu-erh tea on breast cancer cell has not been investigated. In this study, we examined the activity of Pu-erh tea water extract on apoptosis and cell cycle arrest in the human breast adenocarcinoma cell line MDA-MB-231 and clarified its underlying mechanism of action. We found that Pu-erh tea extract inhibited cell proliferation and induced apoptosis in a dose-dependent manner. We also found that Pu-erh tea extract inhibited tumor cell growth within 24 h via accumulation of cells in S phase. Further experiments showed that at 24 h, Pu-erh tea extract up-regulated the expressions of P-p53 (Ser15), p21 and P-JNK and down-regulated the expressions of PCNA, CyclinD1 and CyclinE at the protein level in MDA-MB-231 cells. In particular, the JNK-specific inhibitor SP600125 restored the induction of P-JNK, P-p53 (Ser15), p21, CyclinD1 and CyclinE by Pu-erh tea extract. Our results indicate that Pu-erh tea water extract inhibits cell proliferation of MDA-MB-231 cells through the induction of apoptosis and the stimulation of cell cycle arrest, which is mediated via activation of the JNK-related pathway.
Collapse
Affiliation(s)
- Jing Xie
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China.,College of Animal Science and Technology, Yunnan Agricultural UniversityKunming, China.,Key Laboratory of Agricultural Biodiversity and Plant Disease Management of China Education Ministry, Yunnan Agricultural UniversityKunming, China
| | - Haishuang Yu
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China
| | - Shuang Song
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China
| | - Chongye Fang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China
| | - Xuanjun Wang
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China
| | - Zhongbin Bai
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China
| | - Xiao Ma
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China
| | - Shumei Hao
- State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan UniversityKunming, China
| | - Hong-Ye Zhao
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan UniversityKunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan UniversityKunming, China
| | - Jun Sheng
- Key Laboratory of Pu-er Tea Science, Ministry of Education, Yunnan Agricultural UniversityKunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan UniversityKunming, China
| |
Collapse
|
24
|
Fan FY, Sang LX, Jiang M. Catechins and Their Therapeutic Benefits to Inflammatory Bowel Disease. Molecules 2017; 22:E484. [PMID: 28335502 PMCID: PMC6155401 DOI: 10.3390/molecules22030484] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 02/19/2017] [Accepted: 02/28/2017] [Indexed: 02/07/2023] Open
Abstract
Catechins are natural polyphenolic phytochemicals that exist in food and medicinal plants, such as tea, legume and rubiaceae. An increasing number of studies have associated the intake of catechins-rich foods with the prevention and treatment of chronic diseases in humans, such as inflammatory bowel disease (IBD). Some studies have demonstrated that catechins could significantly inhibit the excessive oxidative stress through direct or indirect antioxidant effects and promote the activation of the antioxidative substances such as glutathione peroxidases (GPO) and glutathione (GSH), reducing the oxidative damages to the colon. In addition, catechins can also regulate the infiltration and proliferation of immune related-cells, such as neutrophils, colonic epithelial cells, macrophages, and T lymphocytes, helping reduce the inflammatory relations and provide benefits to IBD. Perhaps catechins can further inhibit the deterioration of intestinal lesions through regulating the cell gap junctions. Furthermore, catechins can exert their significant anti-inflammatory properties by regulating the activation or deactivation of inflammation-related oxidative stress-related cell signaling pathways, such as nuclear factor-kappa B (NF-κB), mitogen activated protein kinases (MAPKs), transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2), signal transducer and the activator of transcription 1/3 (STAT1/3) pathways. Finally, catechins can also stabilize the structure of the gastrointestinal micro-ecological environment via promoting the proliferation of beneficial intestinal bacteria and regulating the balance of intestinal flora, so as to relieve the IBD. Furthermore, catechins may regulate the tight junctions (TJ) in the epithelium. This paper elaborates the currently known possible molecular mechanisms of catechins in favor of IBD.
Collapse
Affiliation(s)
- Fei-Yan Fan
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China.
| | - Li-Xuan Sang
- Department of Geriatrics, First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China.
| | - Min Jiang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang 110001, China.
| |
Collapse
|
25
|
Li KK, Peng JM, Zhu W, Cheng BH, Li CM. Gallocatechin gallate (GCG) inhibits 3T3-L1 differentiation and lipopolysaccharide induced inflammation through MAPK and NF-κB signaling. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.01.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
26
|
Zhou T, Xu DP, Lin SJ, Li Y, Zheng J, Zhou Y, Zhang JJ, Li HB. Ultrasound-Assisted Extraction and Identification of Natural Antioxidants from the Fruit of Melastoma sanguineum Sims. Molecules 2017; 22:E306. [PMID: 28218717 PMCID: PMC6155708 DOI: 10.3390/molecules22020306] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 02/10/2017] [Accepted: 02/15/2017] [Indexed: 12/19/2022] Open
Abstract
The fruit of Melastoma sanguineum Sims is an edible and sweet wild fruit. In our previous study, the fruit was found to have a strong antioxidant property. In this study, an ultrasound-assisted extraction (UAE) method was developed to extract natural antioxidants from the fruit of Melastoma sanguineum Sims, and a response surface methodology was used to optimize the conditions of UAE to maximize the extraction efficiency. The influence of five independent extraction parameters (ethanol concentration, solvent/material ratio, extracting time, temperature, and ultrasound power) on the extraction efficiency were investigated using a single factor experiment, and then a central composite rotatable design was used to investigate the interaction of three key parameters. The results showed that the optimal extraction conditions were 42.98% ethanol, 28.29 mL/g solvent/material ratio, 34.29 min extracting time, 60 °C temperature, and 600 W ultrasound power. Under these conditions, the Trolox equivalent antioxidant capacity (TEAC) value of the extracts was 1074.61 ± 32.56 μmol Trolox/g dry weight (DW). Compared with conventional maceration (723.27 ± 11.61 μmol Trolox/g DW) and Soxhlet extraction methods (518.37 ± 23.23 μmol Trolox/g DW), the UAE method improved the extraction efficiency, in a shorter period of time. In addition, epicatechin gallate, epicatechin, rutin, epigallocatechin, protocatechuic acid, chlorogenic acid, and quercetin, were identified and quantified in the fruit extracts of Melastoma sanguineum Sims by UPLC-MS/MS.
Collapse
Affiliation(s)
- Tong Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Dong-Ping Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Sheng-Jun Lin
- Zhongshan Center for Disease Control and Prevention, Zhongshan 528403, China.
| | - Ya Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Jie Zheng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Yue Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Jiao-Jiao Zhang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China.
- South China Sea Bioresource Exploitation and Utilization Collaborative Innovation Center, Sun Yat-Sen University, Guangzhou 510006, China.
| |
Collapse
|
27
|
Huang SF, Horng CT, Hsieh YS, Hsieh YH, Chu SC, Chen PN. Epicatechin-3-gallate reverses TGF-β1-induced epithelial-to-mesenchymal transition and inhibits cell invasion and protease activities in human lung cancer cells. Food Chem Toxicol 2016; 94:1-10. [DOI: 10.1016/j.fct.2016.05.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/21/2016] [Accepted: 05/12/2016] [Indexed: 12/25/2022]
|
28
|
Shih LJ, Chen TF, Lin CK, Liu HS, Kao YH. Green tea (-)-epigallocatechin gallate inhibits the growth of human villous trophoblasts via the ERK, p38, AMP-activated protein kinase, and protein kinase B pathways. Am J Physiol Cell Physiol 2016; 311:C308-21. [PMID: 27147558 DOI: 10.1152/ajpcell.00003.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 06/20/2016] [Indexed: 11/22/2022]
Abstract
Green tea catechins, especially (-)-epigallocatechin gallate (EGCG), have been reported to circulate in the placenta of animals and blood of humans after consumption. Whether EGCG regulates activity of human villous trophoblasts (HVT) is unknown. This study investigated the pathways involved in EGCG modulation of trophoblast mitogenesis. EGCG inhibited trophoblast proliferation in a dose-dependent and time-dependent manner, as indicated by the number of cells and incorporation of bromodeoxyuridine (BrdU). EGCG was more effective than other green tea catechins in inhibiting cell growth. EGCG also increased the phosphorylation of the MAPK pathway proteins, ERK1/2, and p38, but not JNK. Furthermore, EGCG had no effects on the total amounts of ERK1/2, p38 MAPK, and JNK proteins. This suggests that EGCG selectively affects particular MAPK subfamilies. Pretreatment with specific inhibitors of ERK1/2, p38 MAPK, and AMP-activated protein kinase (AMPK) antagonized EGCG-induced decreases in both cell number and BrdU incorporation. These inhibitors also blocked EGCG-induced increases in the levels of phospho-ERK1/2, phospho-p38, and phospho-AMPK proteins, respectively. Moreover, EGCG was similar to the phosphatidylinositol 3-kinase inhibitors wortmannin and LY-294002 to decrease protein kinase B (AKT) phosphorylation, cell number, and BrdU incorporation. These data imply that EGCG inhibits the growth of HVT through the ERK, p38, AMPK, and AKT pathways.
Collapse
Affiliation(s)
- Li-Jane Shih
- Department of Life Sciences, National Central University, Jhongli, Taoyuan, Taiwan; and Taoyuan Armed Forces General Hospital, Longtan, Taoyuan, Taiwan
| | - Tz-Fang Chen
- Taoyuan Armed Forces General Hospital, Longtan, Taoyuan, Taiwan
| | - Cheng-Kuo Lin
- Taoyuan Armed Forces General Hospital, Longtan, Taoyuan, Taiwan
| | - Hang-Shen Liu
- Taoyuan Armed Forces General Hospital, Longtan, Taoyuan, Taiwan
| | - Yung-Hsi Kao
- Department of Life Sciences, National Central University, Jhongli, Taoyuan, Taiwan; and
| |
Collapse
|
29
|
Chiou YS, Huang Q, Ho CT, Wang YJ, Pan MH. Directly interact with Keap1 and LPS is involved in the anti-inflammatory mechanisms of (-)-epicatechin-3-gallate in LPS-induced macrophages and endotoxemia. Free Radic Biol Med 2016; 94:1-16. [PMID: 26878775 DOI: 10.1016/j.freeradbiomed.2016.02.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 01/24/2016] [Accepted: 02/10/2016] [Indexed: 12/30/2022]
Abstract
Disruption of the Kelch-like ECH-associated protein 1 (Keap1)-Nuclear factor erythroid-derived factor 2-related factor 2 (Nrf2) interaction has emerged as a promising strategy to reduce oxidative stress-induced inflammation. However, its roles in regulating downstream events, including the cross talk between Nrf2 and nuclear factor-kappa B (NF-κB), are not well defined. The objective of this study was to elucidate the mechanistic connection between Keap1-Nrf2 signaling and the transcription factor NF-κB and to investigate the function of (-)-epicatechin-3-gallate (ECG) in the repression of multiple inflammatory mediators. ECG attenuated lipopolysaccharide (LPS)-induced inflammatory mediator expression and intracellular reactive oxygen species (ROS) generation through the induction of Nrf2/antioxidant response element (ARE)-driven glutathione (GSH) and hemeoxygenase-1 (HO-1) levels, interference with NF-κB and Nfr2/ARE transcriptional activities, and suppression of the MAPKs (JNK1/2 and p38) and PI3K/Akt signaling pathways. Importantly, anti-inflammatory effects of ECG partly require activation of ERK1/2 signaling to mediate HO-1 expression and Nrf2/ARE signaling activation. Furthermore, ECG may directly interact intracellularly with the Kelch repeat domains of Keap1 and bind to extracellular LPS, thereby promoting the nuclear accumulation of the Nrf2 protein and blockading the activation of LPS-induced downstream target signaling pathways. Consistent with in vitro studies, ECG attenuates pathological syndromes of LPS-induced sepsis and systemic inflammation. Our results identified ECG as a novel Keap1-Nrf2 interaction disruptor and LPS-induced TLR4 activation inhibitor, thereby providing an innovative strategy to prevent or treat immune, oxidative stress and inflammatory-related diseases.
Collapse
Affiliation(s)
- Yi-Shiou Chiou
- Department of Environmental and Occupational Health, National Cheng Kung University Medical College, Tainan 704, Taiwan; Institute of Food Science and Technology, National Taiwan University, No.1, Section 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Qingrong Huang
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901-8520, USA
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901-8520, USA
| | - Ying-Jan Wang
- Department of Environmental and Occupational Health, National Cheng Kung University Medical College, Tainan 704, Taiwan; Department of Biomedical Informatics, Asia University, Taichung 413, Taiwan.
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, No.1, Section 4, Roosevelt Road, Taipei 10617, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung 413, Taiwan.
| |
Collapse
|
30
|
Shih LJ, Lin YR, Lin CK, Liu HS, Kao YH. Green tea (-)-epigallocatechin gallate induced growth inhibition of human placental choriocarcinoma cells. Placenta 2016; 41:1-9. [DOI: 10.1016/j.placenta.2016.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/09/2016] [Accepted: 02/28/2016] [Indexed: 12/27/2022]
|
31
|
Isolation of eugenyl β-primeveroside from Camellia sasanqua and its anticancer activity in PC3 prostate cancer cells. J Food Drug Anal 2016; 24:105-111. [PMID: 28911392 PMCID: PMC9345418 DOI: 10.1016/j.jfda.2015.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/01/2015] [Accepted: 06/16/2015] [Indexed: 12/26/2022] Open
Abstract
Most studies of tea trees have focused on their ornamental properties, there are fewer published studies on their medical values. The purpose of this study was to compare the chemical constituents and the biological potential of the water extract of leaves in eight species of Camellia including Camellia sinensis. Among eight Camellia species, Camellia sasanqua showed potent anticancer activities in prostate cancer PC3 cells. In addition to catechins, the major component, eugenyl β-primeveroside was detected in C. sasanqua. Eugenyl β-primeveroside blocked the progression of cell cycle at G1 phase by inducing p53 expression and further upregulating p21 expression. Moreover, eugenyl β-primeveroside induced apoptosis in PC3 prostate cancer cells. Our results suggest that C. sasanqua may have anticancer potential.
Collapse
|
32
|
Block KI, Gyllenhaal C, Lowe L, Amedei A, Amin ARMR, Amin A, Aquilano K, Arbiser J, Arreola A, Arzumanyan A, Ashraf SS, Azmi AS, Benencia F, Bhakta D, Bilsland A, Bishayee A, Blain SW, Block PB, Boosani CS, Carey TE, Carnero A, Carotenuto M, Casey SC, Chakrabarti M, Chaturvedi R, Chen GZ, Chen H, Chen S, Chen YC, Choi BK, Ciriolo MR, Coley HM, Collins AR, Connell M, Crawford S, Curran CS, Dabrosin C, Damia G, Dasgupta S, DeBerardinis RJ, Decker WK, Dhawan P, Diehl AME, Dong JT, Dou QP, Drew JE, Elkord E, El-Rayes B, Feitelson MA, Felsher DW, Ferguson LR, Fimognari C, Firestone GL, Frezza C, Fujii H, Fuster MM, Generali D, Georgakilas AG, Gieseler F, Gilbertson M, Green MF, Grue B, Guha G, Halicka D, Helferich WG, Heneberg P, Hentosh P, Hirschey MD, Hofseth LJ, Holcombe RF, Honoki K, Hsu HY, Huang GS, Jensen LD, Jiang WG, Jones LW, Karpowicz PA, Keith WN, Kerkar SP, Khan GN, Khatami M, Ko YH, Kucuk O, Kulathinal RJ, Kumar NB, Kwon BS, Le A, Lea MA, Lee HY, Lichtor T, Lin LT, Locasale JW, Lokeshwar BL, Longo VD, Lyssiotis CA, MacKenzie KL, Malhotra M, Marino M, Martinez-Chantar ML, Matheu A, et alBlock KI, Gyllenhaal C, Lowe L, Amedei A, Amin ARMR, Amin A, Aquilano K, Arbiser J, Arreola A, Arzumanyan A, Ashraf SS, Azmi AS, Benencia F, Bhakta D, Bilsland A, Bishayee A, Blain SW, Block PB, Boosani CS, Carey TE, Carnero A, Carotenuto M, Casey SC, Chakrabarti M, Chaturvedi R, Chen GZ, Chen H, Chen S, Chen YC, Choi BK, Ciriolo MR, Coley HM, Collins AR, Connell M, Crawford S, Curran CS, Dabrosin C, Damia G, Dasgupta S, DeBerardinis RJ, Decker WK, Dhawan P, Diehl AME, Dong JT, Dou QP, Drew JE, Elkord E, El-Rayes B, Feitelson MA, Felsher DW, Ferguson LR, Fimognari C, Firestone GL, Frezza C, Fujii H, Fuster MM, Generali D, Georgakilas AG, Gieseler F, Gilbertson M, Green MF, Grue B, Guha G, Halicka D, Helferich WG, Heneberg P, Hentosh P, Hirschey MD, Hofseth LJ, Holcombe RF, Honoki K, Hsu HY, Huang GS, Jensen LD, Jiang WG, Jones LW, Karpowicz PA, Keith WN, Kerkar SP, Khan GN, Khatami M, Ko YH, Kucuk O, Kulathinal RJ, Kumar NB, Kwon BS, Le A, Lea MA, Lee HY, Lichtor T, Lin LT, Locasale JW, Lokeshwar BL, Longo VD, Lyssiotis CA, MacKenzie KL, Malhotra M, Marino M, Martinez-Chantar ML, Matheu A, Maxwell C, McDonnell E, Meeker AK, Mehrmohamadi M, Mehta K, Michelotti GA, Mohammad RM, Mohammed SI, Morre DJ, Muralidhar V, Muqbil I, Murphy MP, Nagaraju GP, Nahta R, Niccolai E, Nowsheen S, Panis C, Pantano F, Parslow VR, Pawelec G, Pedersen PL, Poore B, Poudyal D, Prakash S, Prince M, Raffaghello L, Rathmell JC, Rathmell WK, Ray SK, Reichrath J, Rezazadeh S, Ribatti D, Ricciardiello L, Robey RB, Rodier F, Rupasinghe HPV, Russo GL, Ryan EP, Samadi AK, Sanchez-Garcia I, Sanders AJ, Santini D, Sarkar M, Sasada T, Saxena NK, Shackelford RE, Shantha Kumara HMC, Sharma D, Shin DM, Sidransky D, Siegelin MD, Signori E, Singh N, Sivanand S, Sliva D, Smythe C, Spagnuolo C, Stafforini DM, Stagg J, Subbarayan PR, Sundin T, Talib WH, Thompson SK, Tran PT, Ungefroren H, Vander Heiden MG, Venkateswaran V, Vinay DS, Vlachostergios PJ, Wang Z, Wellen KE, Whelan RL, Yang ES, Yang H, Yang X, Yaswen P, Yedjou C, Yin X, Zhu J, Zollo M. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin Cancer Biol 2015; 35 Suppl:S276-S304. [PMID: 26590477 PMCID: PMC4819002 DOI: 10.1016/j.semcancer.2015.09.007] [Show More Authors] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 08/12/2015] [Accepted: 09/14/2015] [Indexed: 12/14/2022]
Abstract
Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.
Collapse
Affiliation(s)
- Keith I Block
- Block Center for Integrative Cancer Treatment, Skokie, IL, United States.
| | | | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada; Lancaster Environment Centre, Lancaster University, Bailrigg, Lancaster, United Kingdom.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - A R M Ruhul Amin
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Jack Arbiser
- Winship Cancer Institute of Emory University, Atlanta, GA, United States; Atlanta Veterans Administration Medical Center, Atlanta, GA, United States; Department of Dermatology, Emory University School of Medicine, Emory University, Atlanta, GA, United States
| | - Alexandra Arreola
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Penny B Block
- Block Center for Integrative Cancer Treatment, Skokie, IL, United States
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Thomas E Carey
- Head and Neck Cancer Biology Laboratory, University of Michigan, Ann Arbor, MI, United States
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, Consejo Superior de Investigaciones Cientificas, Seville, Spain
| | - Marianeve Carotenuto
- Centro di Ingegneria Genetica e Biotecnologia Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, Federico II, Via Pansini 5, 80131 Naples, Italy
| | - Stephanie C Casey
- Stanford University, Division of Oncology, Department of Medicine and Pathology, Stanford, CA, United States
| | - Mrinmay Chakrabarti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, School of Medicine, Columbia, SC, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Georgia Zhuo Chen
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Helen Chen
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | - Yi Charlie Chen
- Department of Biology, Alderson Broaddus University, Philippi, WV, United States
| | - Beom K Choi
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi, Republic of Korea
| | | | - Helen M Coley
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, United Kingdom
| | - Andrew R Collins
- Department of Nutrition, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Sarah Crawford
- Cancer Biology Research Laboratory, Southern Connecticut State University, New Haven, CT, United States
| | - Colleen S Curran
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Charlotta Dabrosin
- Department of Oncology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Giovanna Damia
- Department of Oncology, Istituto Di Ricovero e Cura a Carattere Scientifico - Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Santanu Dasgupta
- Department of Cellular and Molecular Biology, the University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas - Southwestern Medical Center, Dallas, TX, United States
| | - William K Decker
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Punita Dhawan
- Department of Surgery and Cancer Biology, Division of Surgical Oncology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Anna Mae E Diehl
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Jin-Tang Dong
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Q Ping Dou
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Janice E Drew
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Eyad Elkord
- College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bassel El-Rayes
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, United States
| | - Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Dean W Felsher
- Stanford University, Division of Oncology, Department of Medicine and Pathology, Stanford, CA, United States
| | - Lynnette R Ferguson
- Discipline of Nutrition and Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Gary L Firestone
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Christian Frezza
- Medical Research Council Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, United Kingdom
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Mark M Fuster
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, CA, United States
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy; Molecular Therapy and Pharmacogenomics Unit, Azienda Ospedaliera Istituti Ospitalieri di Cremona, Cremona, Italy
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Frank Gieseler
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | - Michelle F Green
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Brendan Grue
- Departments of Environmental Science, Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Dorota Halicka
- Department of Pathology, New York Medical College, Valhalla, NY, United States
| | | | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Patricia Hentosh
- School of Medical Laboratory and Radiation Sciences, Old Dominion University, Norfolk, VA, United States
| | - Matthew D Hirschey
- Department of Medicine, Duke University Medical Center, Durham, NC, United States; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Lorne J Hofseth
- College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Hualien, Taiwan
| | - Gloria S Huang
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, United States
| | - Lasse D Jensen
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden; Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Wen G Jiang
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Lee W Jones
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, United States
| | | | | | - Sid P Kerkar
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | | | - Mahin Khatami
- Inflammation and Cancer Research, National Cancer Institute (Retired), National Institutes of Health, Bethesda, MD, United States
| | - Young H Ko
- University of Maryland BioPark, Innovation Center, KoDiscovery, Baltimore, MD, United States
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Nagi B Kumar
- Moffitt Cancer Center, University of South Florida College of Medicine, Tampa, FL, United States
| | - Byoung S Kwon
- Cancer Immunology Branch, Division of Cancer Biology, National Cancer Center, Goyang, Gyeonggi, Republic of Korea; Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Anne Le
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael A Lea
- New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Ho-Young Lee
- College of Pharmacy, Seoul National University, South Korea
| | - Terry Lichtor
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, United States
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jason W Locasale
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Bal L Lokeshwar
- Department of Medicine, Georgia Regents University Cancer Center, Augusta, GA, United States
| | - Valter D Longo
- Andrus Gerontology Center, Division of Biogerontology, University of Southern California, Los Angeles, CA, United States
| | - Costas A Lyssiotis
- Department of Molecular and Integrative Physiology and Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, United States
| | - Karen L MacKenzie
- Children's Cancer Institute Australia, Kensington, New South Wales, Australia
| | - Meenakshi Malhotra
- Department of Biomedical Engineering, McGill University, Montréal, Canada
| | - Maria Marino
- Department of Science, University Roma Tre, Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | | | - Christopher Maxwell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, British Columbia, Canada
| | - Eoin McDonnell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mahya Mehrmohamadi
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, United States
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Gregory A Michelotti
- Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - D James Morre
- Mor-NuCo, Inc, Purdue Research Park, West Lafayette, IN, United States
| | - Vinayak Muralidhar
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA, United States; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Irfana Muqbil
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Wellcome Trust-MRC Building, Hills Road, Cambridge, United Kingdom
| | | | - Rita Nahta
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Francesco Pantano
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Virginia R Parslow
- Discipline of Nutrition and Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Graham Pawelec
- Center for Medical Research, University of Tübingen, Tübingen, Germany
| | - Peter L Pedersen
- Departments of Biological Chemistry and Oncology, Member at Large, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| | - Brad Poore
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Deepak Poudyal
- College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Satya Prakash
- Department of Biomedical Engineering, McGill University, Montréal, Canada
| | - Mark Prince
- Department of Otolaryngology-Head and Neck, Medical School, University of Michigan, Ann Arbor, MI, United States
| | | | - Jeffrey C Rathmell
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, United States
| | - W Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina, School of Medicine, Columbia, SC, United States
| | - Jörg Reichrath
- Center for Clinical and Experimental Photodermatology, Clinic for Dermatology, Venerology and Allergology, The Saarland University Hospital, Homburg, Germany
| | - Sarallah Rezazadeh
- Department of Biology, University of Rochester, Rochester, NY, United States
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy & National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - R Brooks Robey
- White River Junction Veterans Affairs Medical Center, White River Junction, VT, United States; Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Francis Rodier
- Centre de Rechercher du Centre Hospitalier de l'Université de Montréal and Institut du Cancer de Montréal, Montréal, Quebec, Canada; Université de Montréal, Département de Radiologie, Radio-Oncologie et Médicine Nucléaire, Montréal, Quebec, Canada
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gian Luigi Russo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | | | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
| | - Andrew J Sanders
- Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Daniele Santini
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Malancha Sarkar
- Department of Biology, University of Miami, Miami, FL, United States
| | - Tetsuro Sasada
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Neeraj K Saxena
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University, Health Shreveport, Shreveport, LA, United States
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Dong M Shin
- Winship Cancer Institute of Emory University, Atlanta, GA, United States
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Markus David Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, United States
| | - Emanuela Signori
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Sharanya Sivanand
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Sliva
- DSTest Laboratories, Purdue Research Park, Indianapolis, IN, United States
| | - Carl Smythe
- Department of Biomedical Science, Sheffield Cancer Research Centre, University of Sheffield, Sheffield, United Kingdom
| | - Carmela Spagnuolo
- Institute of Food Sciences National Research Council, Avellino, Italy
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Faculté de Pharmacie et Institut du Cancer de Montréal, Montréal, Quebec, Canada
| | - Pochi R Subbarayan
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Tabetha Sundin
- Department of Molecular Diagnostics, Sentara Healthcare, Norfolk, VA, United States
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | - Sarah K Thompson
- Department of Surgery, Royal Adelaide Hospital, Adelaide, Australia
| | - Phuoc T Tran
- Departments of Radiation Oncology & Molecular Radiation Sciences, Oncology and Urology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vasundara Venkateswaran
- Department of Surgery, University of Toronto, Division of Urology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Dass S Vinay
- Section of Clinical Immunology, Allergy, and Rheumatology, Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA, United States
| | - Panagiotis J Vlachostergios
- Department of Internal Medicine, New York University Lutheran Medical Center, Brooklyn, New York, NY, United States
| | - Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kathryn E Wellen
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Huanjie Yang
- The School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| | - Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA, United States
| | - Clement Yedjou
- Department of Biology, Jackson State University, Jackson, MS, United States
| | - Xin Yin
- Medicine and Research Services, Veterans Affairs San Diego Healthcare System & University of California, San Diego, CA, United States
| | - Jiyue Zhu
- Washington State University College of Pharmacy, Spokane, WA, United States
| | - Massimo Zollo
- Centro di Ingegneria Genetica e Biotecnologia Avanzate, Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, Federico II, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
33
|
Thipubon P, Tipsuwan W, Uthaipibull C, Santitherakul S, Srichairatanakool S. Anti-malarial effect of 1-(N-acetyl-6-aminohexyl)-3-hydroxy-2-methylpyridin-4-one and green tea extract on erythrocyte-stage Plasmodium berghei in mice. Asian Pac J Trop Biomed 2015. [DOI: 10.1016/j.apjtb.2015.07.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
34
|
Chen L, Ye HL, Zhang G, Yao WM, Chen XZ, Liang G. Effect of (-)-epigallocatechin-3- O-gallate on autophagic signaling in HepG2 cells. Shijie Huaren Xiaohua Zazhi 2015; 23:3022-3028. [DOI: 10.11569/wcjd.v23.i19.3022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the changes of autophagy in hepatocellular carcinoma (HCC) HepG2 cells in response to (-)-epigallocatechin-3-O-gallate (EGCG), and to explore its impact on cell proliferation and death.
METHODS: HepG2 cells were routinely cultured and re-plated in Dulbecco's modified eagle's medium (DMEM) in the presence of EGCG of different concentrations. Transmission electron microscopic technique was used to record the formation of autophagosomes in HepG2 cells. Real-time RT-PCR and Western blot were used to detect the mRNA and protein expression of autophagy-related genes, respectively. MTT and trypan blue assays were carried out to determine the cellular proliferation and death. Autophagic intervention experiment was performed to evaluate whether changes in autophagy are involved in the anti-cancer efficacy of EGCG in HCC.
RESULTS: The proliferation of HepG2 cells was significantly inhibited by EGCG and was negatively related to the concentrations of this compound (r = -0.9341, P < 0.001). Doses of EGCG that could effectively inhibit the proliferation of HepG2 cells significantly decreased the mRNA and protein expression of Beclin1 and Atg5, with increments of P62 named autophagic substrate as well as substantially reduced numbers of autophagasomes found in these cells. Moreover, up-regulating autophagy with rapamycin was found to apparently impair the effect of EGCG in killing HepG2 cells (t = 9.95, P < 0.01), while 3-MA, an autophagy inhibitor, dramatically exaggerated the anti-cancer effects of EGCG (t = 22.82, P < 0.01).
CONCLUSION: EGCG substantially inhibits cell proliferation and promotes cell death in HCC cells via down-regulation of autophagy, which indicates a novel critical pharmacological mechanism of EGCG for hepatoma therapy.
Collapse
|
35
|
(−)-Epicatechin-3-gallate (a polyphenol from green tea) potentiates doxorubicin-induced apoptosis in H9C2 cardiomyocytes. Biotechnol Lett 2015; 37:1937-43. [PMID: 26063620 DOI: 10.1007/s10529-015-1879-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 06/03/2015] [Indexed: 01/08/2023]
|
36
|
Hajrezaie M, Paydar M, Looi CY, Moghadamtousi SZ, Hassandarvish P, Salga MS, Karimian H, Shams K, Zahedifard M, Majid NA, Ali HM, Abdulla MA. Apoptotic effect of novel Schiff based CdCl₂(C₁₄H₂₁N₃O₂) complex is mediated via activation of the mitochondrial pathway in colon cancer cells. Sci Rep 2015; 5:9097. [PMID: 25764970 PMCID: PMC4649862 DOI: 10.1038/srep09097] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 01/21/2015] [Indexed: 01/06/2023] Open
Abstract
The development of metal-based agents has had a tremendous role in the present progress in cancer chemotherapy. One well-known example of metal-based agents is Schiff based metal complexes, which hold great promise for cancer therapy. Based on the potential of Schiff based complexes for the induction of apoptosis, this study aimed to examine the cytotoxic and apoptotic activity of a CdCl2(C14H21N3O2) complex on HT-29 cells. The complex exerted a potent suppressive effect on HT-29 cells with an IC50 value of 2.57 ± 0.39 after 72 h of treatment. The collapse of the mitochondrial membrane potential and the elevated release of cytochrome c from the mitochondria to the cytosol indicate the involvement of the intrinsic pathway in the induction of apoptosis. The role of the mitochondria-dependent apoptotic pathway was further proved by the significant activation of the initiator caspase-9 and the executioner caspases-3 and -7. In addition, the activation of caspase-8, which is associated with the suppression of NF-κB translocation to the nucleus, also revealed the involvement of the extrinsic pathway in the induced apoptosis. The results suggest that the CdCl2(C14H21N3O2) complex is able to induce the apoptosis of colon cancer cells and is a potential candidate for future cancer studies.
Collapse
Affiliation(s)
- Maryam Hajrezaie
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mohammadjavad Paydar
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Pouya Hassandarvish
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | | | - Hamed Karimian
- Department of Pharmacology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Keivan Shams
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Maryam Zahedifard
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hapipah Mohd Ali
- Department of Chemistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Chiu CT, Hsuan SW, Lin HH, Hsu CC, Chou FP, Chen JH. Hibiscus sabdariffa leaf polyphenolic extract induces human melanoma cell death, apoptosis, and autophagy. J Food Sci 2015; 80:H649-58. [PMID: 25694272 DOI: 10.1111/1750-3841.12790] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/22/2014] [Indexed: 12/31/2022]
Abstract
Melanoma is the least common but most fatal form of skin cancer. Previous studies have indicated that an aqueous extract of Hibiscus sabdariffa leaves possess hypoglycemic, hypolipidemic, and antioxidant effects. In this study, we want to investigate the anticancer activity of Hibiscus leaf polyphenolic (HLP) extract in melanoma cells. First, HLP was exhibited to be rich in epicatechin gallate (ECG) and other polyphenols. Apoptotic and autophagic activities of HLP and ECG were further evaluated by DAPI stain, cell-cycle analysis, and acidic vascular organelle (AVO) stain. Our results revealed that both HLP and ECG induced the caspases cleavages, Bcl-2 family proteins regulation, and Fas/FasL activation in A375 cells. In addition, we also revealed that the cells presented AVO-positive after HLP treatments. HLP could increase the expressions of autophagy-related proteins autophagy-related gene 5 (ATG5), Beclin1, and light chain 3-II (LC3-II), and induce autophagic cell death in A375 cells. These data indicated that the anticancer effect of HLP, partly contributed by ECG, in A375 cells. HLP potentially could be developed as an antimelanoma agent.
Collapse
Affiliation(s)
- Chun-Tang Chiu
- Inst. of Biochemistry and Biotechnology, Chung Shan Medical Univ, No. 110, Sec. 1, Chien Kuo N. Rd., Taichung, 402, Taiwan
| | | | | | | | | | | |
Collapse
|
38
|
Zhou H, Chen JX, Yang CS, Yang MQ, Deng Y, Wang H. Gene regulation mediated by microRNAs in response to green tea polyphenol EGCG in mouse lung cancer. BMC Genomics 2014; 15 Suppl 11:S3. [PMID: 25559244 PMCID: PMC4304179 DOI: 10.1186/1471-2164-15-s11-s3] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Epigallocatechin-3-gallate (EGCG) has been demonstrated to inhibit cancer in experimental studies through its antioxidant activity and modulations on cellular functions by binding specific proteins. We demonstrated previously that EGCG upregulates the expression of microRNA (i.e. miR-210) by binding HIF-1α, resulting in reduced cell proliferation and anchorage-independent growth. However, the binding affinities of EGCG to HIF-1α and many other targets are higher than the EGCG plasma peak level in experimental animals administered with high dose of EGCG, raising a concern whether the microRNA regulation by HIF-1α is involved in the anti-cancer activity of EGCG in vivo. RESULTS We employed functional genomic approaches to elucidate the role of microRNA in the EGCG inhibition of tobacco carcinogen-induced lung tumors in A/J mice. By analysing the microRNA profiles, we found modest changes in the expression levels of 21 microRNAs. By correlating these 21 microRNAs with the mRNA expression profiles using the computation methods, we identified 26 potential targeted genes of the 21 microRNAs. Further exploration using pathway analysis revealed that the most impacted pathways of EGCG treatment are the regulatory networks associated to AKT, NF-κB, MAP kinases, and cell cycle, and the identified miRNA targets are involved in the networks of AKT, MAP kinases and cell cycle regulation CONCLUSIONS These results demonstrate that the miRNA-mediated regulation is actively involved in the major aspects of the anti-cancer activity of EGCG in vivo.
Collapse
|
39
|
Pluchino LA, Wang HCR. Chronic exposure to combined carcinogens enhances breast cell carcinogenesis with mesenchymal and stem-like cell properties. PLoS One 2014; 9:e108698. [PMID: 25372613 PMCID: PMC4220909 DOI: 10.1371/journal.pone.0108698] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 09/01/2014] [Indexed: 01/16/2023] Open
Abstract
Breast cancer is the most common type of cancer affecting women in North America and Europe. More than 85% of breast cancers are sporadic and attributable to long-term exposure to small quantities of multiple carcinogens. To understand how multiple carcinogens act together to induce cellular carcinogenesis, we studied the activity of environmental carcinogens 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and benzo[a]pyrene (B[a]P), and dietary carcinogen 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) using our breast cell carcinogenesis model. Our study revealed, for the first time, that combined NNK and B[a]P enhanced breast cell carcinogenesis chronically induced by PhIP in both non-cancerous and cancerous breast cells. Co-exposure was more potent than sequential exposure to combined NNK and B[a]P followed by PhIP in inducing carcinogenesis. Initiation of carcinogenesis was measured by transient endpoints induced in a single exposure, while progression of carcinogenesis was measured by acquisition of constitutive endpoints in cumulative exposures. Transient endpoints included DNA damage, Ras-Erk-Nox pathway activation, reactive oxygen species elevation, and increased cellular proliferation. Constitutive endpoints included various cancer-associated properties and signaling modulators, as well as enrichment of cancer stem-like cell population and activation of the epithelial-to-mesenchymal transition program. Using transient and constitutive endpoints as targets, we detected that a combination of the green tea catechins ECG and EGCG, at non-cytotoxic levels, was more effective than individual agents in intervention of cellular carcinogenesis induced by combined NNK, B[a]P, and PhIP. Thus, use of combined ECG and EGCG should be seriously considered for early intervention of breast cell carcinogenesis associated with long-term exposure to environmental and dietary carcinogens.
Collapse
Affiliation(s)
- Lenora Ann Pluchino
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America; Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Hwa-Chain Robert Wang
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America; Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee, United States of America
| |
Collapse
|
40
|
Wu JC, Tsai ML, Lai CS, Wang YJ, Ho CT, Pan MH. Chemopreventative effects of tetrahydrocurcumin on human diseases. Food Funct 2014; 5:12-7. [DOI: 10.1039/c3fo60370a] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
41
|
Wang S, Zhang J, Chen M, Wang Y. Delivering flavonoids into solid tumors using nanotechnologies. Expert Opin Drug Deliv 2013; 10:1411-28. [PMID: 23862581 DOI: 10.1517/17425247.2013.807795] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Long-term epidemiological studies have demonstrated that regular ingestion of flavonoids contained in dietary sources is associated with a reduced risk for many chronic diseases including cancer. However, although flavonoids are largely consumed in the diet and high concentrations may exist in the intestine after oral administration, the plasma/tissue concentrations of flavonoids are lower than their effective therapeutic doses due to poor bioavailability, resulting in the limited efficacy of flavonoids in various clinical studies. Therefore, the application of nanotechnology to deliver flavonoids to tumor sites has received considerable attention in recent years. AREAS COVERED In this review, after a general review of the potential benefits of flavonoids in cancer therapy and several key factors affecting their bioavailability, the current efforts in improving the delivery efficacy of promising candidates that are particularly important in the human diet, namely quercetin, epigallocatechin-3-gallate (EGCG) and genistein were focused on. Finally, the challenges of developing flavonoid delivery systems that improve flavonoid bioavailability and their anticancer therapy potentials were summarized. EXPERT OPINION The design of suitable molecular carriers for flavonoids is an area of research that is in rapid progress. A large number of unheeded promising favonoids are suffering from poor in vivo parameters, their potential benefits deserves further research. Furthermore, more effort should be placed on developing active targeting systems, evaluating the efficacy and toxicity of novel flavonoid delivery systems through small and large scale clinical trials.
Collapse
Affiliation(s)
- Shengpeng Wang
- Assistant Professor, University of Macau, Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine , Av. Padre Tomas Pereira S.J, Taipa, Macau, 999078 , China
| | | | | | | |
Collapse
|
42
|
Hou IC, Amarnani S, Chong MT, Bishayee A. Green tea and the risk of gastric cancer: epidemiological evidence. World J Gastroenterol 2013; 19:3713-22. [PMID: 23840110 PMCID: PMC3699047 DOI: 10.3748/wjg.v19.i24.3713] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/14/2013] [Accepted: 05/09/2013] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer death in the world. Numerous efforts are being made to find chemoprotective agents able to reduce its risk. Amongst these, green tea has been reported to have a protective effect against stomach cancer. This article aims to critically evaluate all epidemiological studies reporting an association between green tea consumption and GC risk. MEDLINE, EBSCOHOST and Google Scholar were used to search for clinical trials of green tea and its correlation to stomach cancer. Studies include cohort and case-control studies. Outcome of interests are inverse association, no association, and positive association. Seventeen epidemiologic studies were reviewed. Eleven studies were conducted in Japan, five in China, and one with Japanese descendent in Hawaii. Ten case-control studies and seven cohort studies were included. The relative risks or odds ratio of GC for the highest level of green tea consumption was compared. Seven studies suggested no association, eight an inverse association, and one a positive association. One study had shown a significantly lowered GC risk when tea was served warm to cold. Another study also showed a significantly risk with lukewarm tea. All studies that analyzed men and women separately have suggested a reduced risk in women than in men, albeit no significant difference. This review demonstrates that there is insufficient information to support green tea consumption reduces the risk of GC. More studies on the subject matter are warranted.
Collapse
|
43
|
Chun KS, Kim EH, Lee S, Hahm KB. Chemoprevention of gastrointestinal cancer: the reality and the dream. Gut Liver 2013; 7:137-49. [PMID: 23560148 PMCID: PMC3607766 DOI: 10.5009/gnl.2013.7.2.137] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 07/03/2012] [Accepted: 09/17/2012] [Indexed: 12/17/2022] Open
Abstract
Despite substantial progress in screening, early diagnosis, and the development of noninvasive technology, gastrointestinal (GI) cancer remains a major cause of cancer-associated mortality. Chemoprevention is thought to be a realistic approach for reducing the global burden of GI cancer, and efforts have been made to search for chemopreventive agents that suppress acid reflux, GI inflammation and the eradication of Helicobacter pylori. Thus, proton pump inhibitors, statins, monoclonal antibodies targeting tumor necrosis factor-alpha, and nonsteroidal anti-inflammatory agents have been investigated for their potential to prevent GI cancer. Besides the development of these synthetic agents, a wide variety of the natural products present in a plant-based diet, which are commonly called phytoceuticals, have also sparked hope for the chemoprevention of GI cancer. To perform successful searches of chemopreventive agents for GI cancer, it is of the utmost importance to understand the factors contributing to GI carcinogenesis. Emerging evidence has highlighted the role of chronic inflammation in inducing genomic instability and telomere shortening and affecting polyamine metabolism and DNA repair, which may help in the search for new chemopreventive agents for GI cancer.
Collapse
|
44
|
Liu LC, Tsao TCY, Hsu SR, Wang HC, Tsai TC, Kao JY, Way TD. EGCG inhibits transforming growth factor-β-mediated epithelial-to-mesenchymal transition via the inhibition of Smad2 and Erk1/2 signaling pathways in nonsmall cell lung cancer cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:9863-9873. [PMID: 22957988 DOI: 10.1021/jf303690x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Transforming growth factor-β (TGF-β)-mediated epithelial mesenchymal transition (EMT) of human lung cancer cells may contribute to lung cancer metastasis. It has been reported that EGCG can inhibit tumorigenesis and cancer cell growth in lung cancer; however, the effect of EGCG on EMT in nonsmall cell lung cancer (NSCLC) cells has not been investigated. In this study, we found that NSCLC cells A549 and H1299 were converted to the fibroblastic phenotype in response to TGF-β. Epithelial marker E-cadherin was down-regulated, and mesenchymal marker vimentin was up-regulated simultaneously. Our results illustrated that TGF-β was able to induce EMT in NSCLC cells, and EGCG would reverse TGF-β-induced morphological changes, up-regulate the expression of E-cadherin, and down-regulate the expression of vimentin. Immunofluorescent staining also demonstrated that E-cadherin was up-regulated and that vimentin was down-regulated by EGCG pretreatment. Moreover, wound-healing and the in vitro invasion assay showed that EGCG could inhibit TGF-β-induced migration and invasion of NSCLC cells. By using the dual-luciferase reporter assay, we demonstrated that EGCG inhibited TGF-β-induced EMT at the transcriptional level. EGCG decreased the phosphorylation of Smad2 and Erk1/2, inhibited the nuclear translocation of Smad2, and repressed the expression of transcription factors ZEB1, Snail, Slug, and Twist, and up-regulated the expression of E-cadherin. In summary, our results suggest that EGCG can inhibit TGF-β-induced EMT via down-regulation of phosphorylated Smad2 and Erk1/2 in NSCLC cells.
Collapse
Affiliation(s)
- Liang-Chih Liu
- Institute of Biochemistry, College of Life Science, National Chung Hsing University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
45
|
PARK SEUNGBIN, BAE JONGWOON, KIM JONGMIN, LEE SEUNGGEE, HAN MYOUNGSEOK. Antiproliferative and apoptotic effect of epigallocatechin-3-gallate on Ishikawa cells is accompanied by sex steroid receptor downregulation. Int J Mol Med 2012; 30:1211-8. [DOI: 10.3892/ijmm.2012.1104] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 07/09/2012] [Indexed: 11/05/2022] Open
|
46
|
Hussain A, Harish G, Prabhu SA, Mohsin J, Khan MA, Rizvi TA, Sharma C. Inhibitory effect of genistein on the invasive potential of human cervical cancer cells via modulation of matrix metalloproteinase-9 and tissue inhibitors of matrix metalloproteinase-1 expression. Cancer Epidemiol 2012; 36:e387-93. [PMID: 22884883 DOI: 10.1016/j.canep.2012.07.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 06/14/2012] [Accepted: 07/10/2012] [Indexed: 01/11/2023]
Abstract
BACKGROUND One of the most challenging stumbling blocks for the treatment of cancer is the ability of cancer cells to break the natural barriers and spread from its site of origin to non-adjacent regional and distant sites, accounting for high cancer mortality rates. Gamut experimental and epidemiological data advocate the use of pharmacological or nutritional interventions to inhibit or delay various stage(s) of cancer such as invasion and metastasis. Genistein, a promising chemopreventive agent, has gained considerable attention for its powerful anti-carcinogenic, anti-angiogenic and chemosensitizing activities. METHODS In this study, the cytotoxic potential of genistein on HeLa cells by cell viability assay and the mode of cell death induced by genistein were determined by nuclear morphological examination, DNA laddering assay and cell cycle analysis. Moreover, to establish its inhibitory effect on migration of HeLa cells, scratch wound assay was performed and these results were correlated with the expression of genes involved in invasion and migration (MMP-9 and TIMP-1) by RT-PCR. RESULTS The exposure of HeLa cells to genistein resulted in significant dose- and time-dependent growth inhibition, which was found to be mediated by apoptosis and cell cycle arrest at G(2)/M phase. In addition, it induced migration-inhibition in a time-dependent manner by modulating the expression of MMP-9 and TIMP-1. CONCLUSION Our results signify that genistein may be an effective anti-neoplastic agent to prevent cancer cell growth and invasion and metastasis. Therefore therapeutic strategies utilizing genistein could be developed to substantially reduce cancer morbidity and mortality.
Collapse
Affiliation(s)
- Arif Hussain
- Department of Biotechnology, Manipal University, PO Box 345050, Dubai, United Arab Emirates
| | | | | | | | | | | | | |
Collapse
|
47
|
Chan CK, Goh BH, Kamarudin MNA, Kadir HA. Aqueous fraction of Nephelium ramboutan-ake rind induces mitochondrial-mediated apoptosis in HT-29 human colorectal adenocarcinoma cells. Molecules 2012; 17:6633-57. [PMID: 22728359 PMCID: PMC6268511 DOI: 10.3390/molecules17066633] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 05/23/2012] [Accepted: 05/25/2012] [Indexed: 01/04/2023] Open
Abstract
The aim of this study was to investigate the cytotoxic and apoptotic effects of Nephelium ramboutan-ake (pulasan) rind in selected human cancer cell lines. The crude ethanol extract and fractions (ethyl acetate and aqueous) of N. ramboutan-ake inhibited the growth of HT-29, HCT-116, MDA-MB-231, Ca Ski cells according to MTT assays. The N. ramboutan-ake aqueous fraction (NRAF) was found to exert the greatest cytotoxic effect against HT-29 in a dose-dependent manner. Evidence of apoptotic cell death was revealed by features such as chromatin condensation, nuclear fragmentation and apoptotic body formation. The result from a TUNEL assay strongly suggested that NRAF brings about DNA fragmentation in HT-29 cells. Phosphatidylserine (PS) externalization on the outer leaflet of plasma membranes was detected with annexin V-FITC/PI binding, confirming the early stage of apoptosis. The mitochondrial permeability transition is an important step in the induction of cellular apoptosis, and the results clearly suggested that NRAF led to collapse of mitochondrial transmembrane potential in HT-29 cells. This attenuation of mitochondrial membrane potential (Δψm) was accompanied by increased production of ROS and depletion of GSH, an increase of Bax protein expression, and induced-activation of caspase-3/7 and caspase-9. These combined results suggest that NRAF induces mitochondrial-mediated apoptosis.
Collapse
Affiliation(s)
| | | | | | - Habsah Abdul Kadir
- Biomolecular Research Group, Biochemistry Program, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
48
|
SUZUKI Y, MIYOSHI N, ISEMURA M. Health-promoting effects of green tea. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2012; 88:88-101. [PMID: 22450537 PMCID: PMC3365247 DOI: 10.2183/pjab.88.88] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Green tea is manufactured from the leaves of the plant Camellia sinensis Theaceae and has been regarded to possess anti-cancer, anti-obesity, anti-atherosclerotic, anti-diabetic, anti-bacterial, and anti-viral effects. Many of the beneficial effects of green tea are related to the activities of (-)-epigallocatechin gallate (EGCG), a major component of green tea catechins. For about 20 years, we have engaged in studies to reveal the biological activities and action mechanisms of green tea and EGCG. This review summarizes several lines of evidence to indicate the health-promoting properties of green tea mainly based on our own experimental findings.
Collapse
Affiliation(s)
- Yasuo SUZUKI
- Faculty of Human Life Sciences, Nagoya Keizai University, Inuyama, Japan
| | - Noriyuki MIYOSHI
- Graduate School of Nutritional and Environmental Sciences and Global COE Program, University of Shizuoka, Shizuoka, Japan
| | - Mamoru ISEMURA
- Graduate School of Nutritional and Environmental Sciences and Global COE Program, University of Shizuoka, Shizuoka, Japan
- Correspondence should be addressed: M. Isemura, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan (e-mail: )
| |
Collapse
|