1
|
Keyvani‐Ghamsari S, Rahimi M, Khorsandi K. An update on the potential mechanism of gallic acid as an antibacterial and anticancer agent. Food Sci Nutr 2023; 11:5856-5872. [PMID: 37823155 PMCID: PMC10563697 DOI: 10.1002/fsn3.3615] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 10/13/2023] Open
Abstract
Drug resistance to antibacterial and anticancer drugs is one of the most important global problems in the treatment field that is constantly expanding and hinders the recovery and survival of patients. Therefore, it is necessary to identify compounds that have antibacterial and anticancer properties or increase the effectiveness of existing drugs. One of these approaches is using natural compounds that have few side effects and are effective. Gallic acid (GA) has been identified as one of the most important plant polyphenols that health-promoting effects in various aspects such as bacterial and viral infections, cancer, inflammatory, neuropsychological, gastrointestinal, and metabolic disease. Various studies have shown that GA inhibits bacterial growth by altering membrane structure, and bacterial metabolism, and inhibits biofilm formation. Also, GA inhibits cancer cell growth by targeting different signaling pathways in apoptosis, increasing reactive oxygen species (ROS) production, targeting the cell cycle, and inhibiting oncogenes and matrix metalloproteinases (MMPs) expression. Due to the powerful function of GA against bacteria and cancer cells. In this review, we describe the latest findings in the field of the sources and chemical properties of GA, its pharmacological properties and bioavailability, the antibacterial and anticancer activities of GA, and its derivatives alone, in combination with other drugs and in the form of nanoformulation. This review can be a comprehensive perspective for scientists to use medicinal compounds containing GA in future research and expand its clinical applications.
Collapse
Affiliation(s)
- Saeedeh Keyvani‐Ghamsari
- Clinical Cares and Health Promotion Research Center, Karaj BranchIslamic Azad UniversityKarajIran
| | - Maryam Rahimi
- Clinical Cares and Health Promotion Research Center, Karaj BranchIslamic Azad UniversityKarajIran
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research CenterYara Institute, ACECRTehranIran
| |
Collapse
|
2
|
Hao Q, Wu Y, Vadgama JV, Wang P. Phytochemicals in Inhibition of Prostate Cancer: Evidence from Molecular Mechanisms Studies. Biomolecules 2022; 12:1306. [PMID: 36139145 PMCID: PMC9496067 DOI: 10.3390/biom12091306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022] Open
Abstract
Prostate cancer is one of the leading causes of death for men worldwide. The development of resistance, toxicity, and side effects of conventional therapies have made prostate cancer treatment become more intensive and aggressive. Many phytochemicals isolated from plants have shown to be tumor cytotoxic. In vitro laboratory studies have revealed that natural compounds can affect cancer cell proliferation by modulating many crucial cellular signaling pathways frequently dysregulated in prostate cancer. A multitude of natural compounds have been found to induce cell cycle arrest, promote apoptosis, inhibit cancer cell growth, and suppress angiogenesis. In addition, combinatorial use of natural compounds with hormone and/or chemotherapeutic drugs seems to be a promising strategy to enhance the therapeutic effect in a less toxic manner, as suggested by pre-clinical studies. In this context, we systematically reviewed the currently available literature of naturally occurring compounds isolated from vegetables, fruits, teas, and herbs, with their relevant mechanisms of action in prostate cancer. As there is increasing data on how phytochemicals interfere with diverse molecular pathways in prostate cancer, this review discusses and emphasizes the implicated molecular pathways of cell proliferation, cell cycle control, apoptosis, and autophagy as important processes that control tumor angiogenesis, invasion, and metastasis. In conclusion, the elucidation of the natural compounds' chemical structure-based anti-cancer mechanisms will facilitate drug development and the optimization of drug combinations. Phytochemicals, as anti-cancer agents in the treatment of prostate cancer, can have significant health benefits for humans.
Collapse
Affiliation(s)
- Qiongyu Hao
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | - Yanyuan Wu
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Jaydutt V. Vadgama
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Piwen Wang
- Division of Cancer Research and Training, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| |
Collapse
|
3
|
Kumar NB, Hogue S, Pow-Sang J, Poch M, Manley BJ, Li R, Dhillon J, Yu A, Byrd DA. Effects of Green Tea Catechins on Prostate Cancer Chemoprevention: The Role of the Gut Microbiome. Cancers (Basel) 2022; 14:3988. [PMID: 36010981 PMCID: PMC9406482 DOI: 10.3390/cancers14163988] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 01/22/2023] Open
Abstract
Accumulating evidence supports green tea catechins (GTCs) in chemoprevention for prostate cancer (PCa), a leading cause of cancer morbidity and mortality among men. GTCs include (-)-epigallocatechin-3-gallate, which may modulate the molecular pathways implicated in prostate carcinogenesis. Prior studies of GTCs suggested that they are bioavailable, safe, and effective for modulating clinical and biological markers implicated in prostate carcinogenesis. GTCs may be of particular benefit to those with low-grade PCas typically managed with careful monitoring via active surveillance (AS). Though AS is recommended, it has limitations including potential under-grading, variations in eligibility, and anxiety reported by men while on AS. Secondary chemoprevention of low-grade PCas using GTCs may help address these limitations. When administrated orally, the gut microbiome enzymatically transforms GTC structure, altering its bioavailability, bioactivity, and toxicity. In addition to xenobiotic metabolism, the gut microbiome has multiple other physiological effects potentially involved in PCa progression, including regulating inflammation, hormones, and other known/unknown pathways. Therefore, it is important to consider not only the independent roles of GTCs and the gut microbiome in the context of PCa chemoprevention, but how gut microbes may relate to individual responses to GTCs, which, in turn, can enhance clinical decision-making.
Collapse
Affiliation(s)
- Nagi B. Kumar
- Cancer Epidemiology Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Stephanie Hogue
- Cancer Epidemiology Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Julio Pow-Sang
- Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Michael Poch
- Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Brandon J. Manley
- Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Roger Li
- Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jasreman Dhillon
- Anatomic Pathology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Alice Yu
- Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Doratha A. Byrd
- Cancer Epidemiology Program, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
4
|
New Co-Crystals/Salts of Gallic Acid and Substituted Pyridines: An Effect of Ortho-Substituents on the Formation of an Acid–Pyridine Heterosynthon. CRYSTALS 2022. [DOI: 10.3390/cryst12040497] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Co-crystallization of gallic acid with pyridines and their polyaromatic analogue, quinoline, ortho-substituted by various proton-donating groups able to form hydrogen bonds, produced the only reported co-crystal of gallic acid with an ortho-substituted pyridine, 2-hydroxypyridine, as its preferred pyridone-2 tautomer, and four new crystalline products of gallic acid. These co-crystals, or gallate salts depending on the choice of the pyridine-containing compound, as predicted by the pKa rule, were identified by X-ray diffraction to feature the popular acid–pyridine heterosynthon found in most of the two-component systems of gallic acid that lack ortho-substituents in the pyridine-containing compound. This single-point heterosynthon is, however, modified by one or two proton-donating ortho-substituents, which sometimes may transform into the proton acceptors in an adopted tautomer or zwitterion, to produce its two- or other multi-point variants, including a very rare four-point heterosynthon. The hydrogen bonds they form with the gallic acid species in the appropriate co-crystals/salts strongly favors the formation of the acid–pyridine heterosynthon over the acid–acid homosynthon. In the competitive conditions of multi-component systems, such a modification might be used to reduce supramolecular-synthon-based polymorphism to produce new pharmaceuticals and other crystalline materials with designed properties.
Collapse
|
5
|
Hazafa A, Iqbal MO, Javaid U, Tareen MBK, Amna D, Ramzan A, Piracha S, Naeem M. Inhibitory effect of polyphenols (phenolic acids, lignans, and stilbenes) on cancer by regulating signal transduction pathways: a review. Clin Transl Oncol 2022; 24:432-445. [PMID: 34609675 DOI: 10.1007/s12094-021-02709-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/11/2021] [Indexed: 02/07/2023]
Abstract
Natural products, especially polyphenols (phenolic acids, lignans, and stilbenes) are suggested to be more potent anticancer drugs because of their no or less adverse effects, excess availability, high accuracy, and secure mode of action. In the present review, potential anticancer mechanisms of action of some polyphenols including phenolic acids, lignans, and stilbenes are discussed based on clinical, epidemiological, in vivo, and in vitro studies. The emerging evidence revealed that phenolic acids, lignans, and stilbenes induced apoptosis in the treatment of breast (MCF-7), colon (Caco-2), lung (SKLU-1), prostate (DU-145 and LNCaP), hepatocellular (hepG-2), and cervical (A-431) cancer cells, cell cycle arrest (S/G2/M/G1-phases) in gastric (MKN-45 and MKN-74), colorectal (HCT-116), bladder (T-24 and 5637), oral (H-400), leukemic (HL-60 and MOLT-4) and colon (Caco-2) cancer cells, and inhibit cell proliferation against the prostate (PC-3), liver (LI-90), breast (T47D and MDA-MB-231), colon (HT-29 and Caco-2), cervical (HTB-35), and MIC-1 cancer cells through caspase-3, MAPK, AMPK, Akt, NF-κB, Wnt, CD95, and SIRT1 pathways. Based on accumulated data, we suggested that polyphenols could be considered as a viable therapeutic option in the treatment of cancer cells in the near future.
Collapse
Affiliation(s)
- A Hazafa
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, 38040, Pakistan.
| | - M O Iqbal
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - U Javaid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - M B K Tareen
- College of Food Science & Technology, Huazhong Agricultural University, Huazhong, China
| | - D Amna
- Institute of Food Science & Nutrition, Bahauddin Zakariya University, Multan, Pakistan
| | - A Ramzan
- Department of Botany, University of Agriculture Faisalabad, Faisalabad, 38040, Pakistan
| | - S Piracha
- Department of Chemistry, University of Agriculture Faisalabad, Faisalabad, 38040, Pakistan
| | - M Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| |
Collapse
|
6
|
Jiang Y, Pei J, Zheng Y, Miao YJ, Duan BZ, Huang LF. Gallic Acid: A Potential Anti-Cancer Agent. Chin J Integr Med 2021; 28:661-671. [PMID: 34755289 DOI: 10.1007/s11655-021-3345-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 10/19/2022]
Abstract
Cancer is one of the most devastating diseases worldwide and definitive therapeutics for treating cancer are not yet available despite extensive research efforts. The key challenges include limiting factors connected with traditional chemotherapeutics, primarily drug resistance, low response rates, and adverse side-effects. Therefore, there is a high demand for novel anti-cancer drugs that are both potent and safe for cancer prevention and treatment. Gallic acid (GA), a natural botanic phenolic compound, can mediate various therapeutic properties that are involved in anti-inflammation, anti-obesity, and anti-cancer activities. More recently, GA has been shown to exert anti-cancer activities via several biological pathways that include migration, metastasis, apoptosis, cell cycle arrest, angiogenesis, and oncogene expression. This review discusses two aspects, one is the anti-cancer potential of GA against different types of cancer and the underlying molecular mechanisms, the other is the bibliometric analysis of GA in cancer and tumor research. The results indicated that lung cancer, prostate cancer, stomach cancer, and colon adenocarcinoma may become a hot topic in further research. Overall, this review provides evidence that GA represents a promising novel, potent, and safe anti-cancer drug candidate for treating cancer.
Collapse
Affiliation(s)
- Yuan Jiang
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.,State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.,College of Pharmaceutical Science, Dali University, Dali, Yunnan Province, 671000, China
| | - Jin Pei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yan Zheng
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Yu-Jing Miao
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Bao-Zhong Duan
- College of Pharmaceutical Science, Dali University, Dali, Yunnan Province, 671000, China
| | - Lin-Fang Huang
- Key Laboratory of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People's Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China. .,State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
7
|
Collard M, Gallagher PE, Tallant EA. A Polyphenol-Rich Extract From Muscadine Grapes Inhibits Triple-Negative Breast Tumor Growth. Integr Cancer Ther 2021; 19:1534735420917444. [PMID: 32578460 PMCID: PMC7315667 DOI: 10.1177/1534735420917444] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that tends to affect young women and has a high propensity to metastasize. No targeted treatments are available for this type of breast cancer due to a lack of estrogen or progesterone receptors or overexpression of human epidermal growth factor receptor type 2 overexpression. Currently, patients have no therapeutic options once standard of care is complete, indicating a need for safe and effective therapies to slow or prevent the progression of TNBC to metastatic disease. Studies showed that isolated polyphenols or polyphenol-rich muscadine grape extracts polyphenols inhibit the proliferation of various cancer cells including breast cancer. A proprietary muscadine grape extract (MGE) was administered to nude mice with human MDA-MB-231 TNBC atumors for 4 weeks to determine the effect of the extract on tumor growth. MGE decreased tumor volume in association with a reduction in the proliferative markers Ki67 and cyclin D1. To determine the molecular mechanisms for the MGE-induced reduction in tumor growth, mouse 4T1, MDA-MB-231, or human BT-549 TNBC cells were treated with MGE, and various signaling pathways were investigated. MGE reduced c-Met, differentially abrogated ERK/MAPK and AKT signaling, and decreased a downstream targets of ERK/MAPK and AKT pathways, cyclin D1. Cyclin D1 reduction was associated with retinoblastoma activation and cell cycle arrest in MDA-MB-231 TNBC cells. MGE-regulated molecular signaling pathways were functionally associated with a dose-dependent reduction in cell proliferation. The pluripotency of MGE and high index of safety and tolerability suggest that the extract may serve as a therapeutic to reduce TNBC progression to metastatic disease.
Collapse
Affiliation(s)
| | | | - E Ann Tallant
- Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
8
|
Polyphenol Extract from Evening Primrose ( Oenothera paradoxa) Inhibits Invasion Properties of Human Malignant Pleural Mesothelioma Cells. Biomolecules 2020; 10:biom10111574. [PMID: 33228230 PMCID: PMC7699585 DOI: 10.3390/biom10111574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 01/10/2023] Open
Abstract
Extracts from the defatted evening primrose (Oenothera paradoxa Hudziok) seeds are the source of a range of stable polyphenolic compounds, including ellagic acid, gallic acid, and catechin. Our studies evaluate, for the first time, the influence of evening primrose isopropanol extract (EPE) on malignant pleural mesothelioma (MPM) cells. MPM is rarely diagnosed, its high aggressiveness and frequently noted chemoresistance limit its treatment schemes and it is characterized by low prognostic features. Here, we demonstrate that EPE inhibited MPM growth in a dose-dependent manner in cells with increased invasion properties. Moreover, EPE treatment resulted in cell cycle arrest in the G2/M phase and increased apoptosis in invasive MPM cell lines. Additionally, EPE strongly limited invasion and MMP-7 secretion in MPM cancer cells. Our original data provide evidence about the potential anti-invasive effects of EPE in MPM therapy treatment.
Collapse
|
9
|
Zhou X, Huang N, Chen W, Xiaoling T, Mahdavi B, Raoofi A, Mahdian D, Atabati H. HPLC phenolic profile and induction of apoptosis by Linum usitatissimum extract in LNCaP cells by caspase3 and Bax pathways. AMB Express 2020; 10:203. [PMID: 33169228 PMCID: PMC7652951 DOI: 10.1186/s13568-020-01138-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Linum usitatissimum is a candidate as a remedy to treat prostate problems in some folklore medicines. In this study, we have reported the phenolic and flavonoid constituents, antioxidant activity, and potential of the plant extract against prostate cancer cells. The phenolic and flavonoid compound profile of the extract were established using HPLC analysis. While the total phenolic and flavonoid content (TPC and TFC) were analyzed using classic methods. The antioxidant activity of the extract was also evaluated. MTT assay and flow cytometry technique was used to evaluate antiproliferation activity and induction apoptosis of the plant extract on prostate cancer cells of LNCaP. We also evaluated the gene expression of Bax and caspase-3 using the real-time qPCR assay. HPLC result revealed that L. usitatissimum extract (LUE) was rich in phenolic acids such as gallic, ferulic, and vanillic acid with the amount of 3.56, 2.12, 1.24 μg/g extract respectively. 383.4 mg GAE/g and 47.1 mgRuE/g were calculated for total phenolic and flavonoid content. LUE exhibited radical scavenging activity with IC50 = 19.3 ± 1.1 µg/mL. LUE chelated ferrous ions with IC50 = 121.1 ± 1.3 µg/mL. LUE showed anti-proliferative activity on LNCaP cells with the IC50 values of 8.3, 6.3, and 5.4 μg/mL after 24, 48, and 72 h treatment. LUE also increased cell mortality by inducing apoptosis (15.3-29.8%). The real-time qPCR results exhibited an increase in gene expression of Bax and caspase-3. Our in vitro study demonstrates that L. usitatissimum can be considered as an effective agent to inhibit the growth and invasion the human prostate cancer cells.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Urology, Beijing Tiantan Hospital,Capital Medical University, Beijing, 100070, China
| | - Ningou Huang
- Department of Pharmacy, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210031, Jiangsu, China
| | - Wenxin Chen
- Department of Urology, Occupational Disease Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830000, Xinjiang, China
| | - Tang Xiaoling
- Jiangxi Research Institute of Traditional Chinese Medicine, Nanchang, 330046, Jiangxi, China.
| | - Behnam Mahdavi
- Department of Chemistry, Faculty of Science, Hakim Sabzevari University, 96179-76487, Sabzevar, Iran
| | - Amir Raoofi
- Cellular and Molecular Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Mahdian
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Hadi Atabati
- Cellular and Molecular Research Center, Department of Anatomy, Sabzevar University of Medical Sciences, Sabzevar, Iran
| |
Collapse
|
10
|
Lee MML, Chan BD, Wong WY, Qu Z, Chan MS, Leung TW, Lin Y, Mok DKW, Chen S, Tai WCS. Anti-cancer Activity of Centipeda minima Extract in Triple Negative Breast Cancer via Inhibition of AKT, NF-κB, and STAT3 Signaling Pathways. Front Oncol 2020; 10:491. [PMID: 32328465 PMCID: PMC7160338 DOI: 10.3389/fonc.2020.00491] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 03/18/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in females worldwide. Estimates from the World Health Organization (WHO) International Agency for Research on Cancer, suggest that globally, there were around 2.1 million new breast cancer cases and 627,000 deaths due to breast cancer in 2018. Among the subtypes of breast cancer, triple negative breast cancer (TNBC) is the most aggressive and carries the poorest prognosis, largest recurrence, and lowest survival rate. Major treatment options for TNBC patients are mainly constrained to chemotherapy, which can be accompanied by severe side effects. Therefore, development of novel and effective anti-cancer drugs for the treatment of TNBC are urgently required. Centipeda minima is a well-known traditional Chinese herbal medicine that has historically been used to treat rhinitis, sinusitis, relieve pain, and reduce swelling. Recent studies have shown that Centipeda minima exhibited efficacy against certain cancers, however, to date, no studies have been conducted on its effects in breast cancer. Here, we aimed to investigate the anti-cancer activity of the total extract of Centipeda minima (CME), and its underlying mechanism, in TNBC. In MDA-MB-231, we found that CME could significantly reduce cell viability and proliferation, induce apoptosis and inhibit cancer cell migration and invasion, in a dose and time-dependent manner. We showed that CME may potentially act via inhibition of multiple signaling pathways, including the EGFR, PI3K/AKT/mTOR, NF-κB, and STAT3 pathways. Treatment with CME also led to in vitro downregulation of MMP-9 activity and inhibition of metastasis. Further, we demonstrated that CME could significantly reduce tumor burden in MDA-MB-231 xenograft mice, without any appreciable side effects. Based on our findings, CME is a promising candidate for development as a therapeutic with high efficacy against TNBC.
Collapse
Affiliation(s)
- Magnolia Muk-Lan Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Brandon Dow Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Wing-Yan Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zhao Qu
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Man-Sum Chan
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Tsz-Wing Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yushan Lin
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Daniel Kam-Wah Mok
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Sibao Chen
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.,Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - William Chi-Shing Tai
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China.,State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
11
|
Arora I, Sharma M, Tollefsbol TO. Combinatorial Epigenetics Impact of Polyphenols and Phytochemicals in Cancer Prevention and Therapy. Int J Mol Sci 2019; 20:ijms20184567. [PMID: 31540128 PMCID: PMC6769666 DOI: 10.3390/ijms20184567] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/08/2019] [Accepted: 09/11/2019] [Indexed: 12/24/2022] Open
Abstract
Polyphenols are potent micronutrients that can be found in large quantities in various food sources and spices. These compounds, also known as phenolics due to their phenolic structure, play a vital nutrient-based role in the prevention of various diseases such as diabetes, cardiovascular diseases, neurodegenerative diseases, liver disease, and cancers. However, the function of polyphenols in disease prevention and therapy depends on their dietary consumption and biological properties. According to American Cancer Society statistics, there will be an expected rise of 23.6 million new cancer cases by 2030. Due to the severity of the increased risk, it is important to evaluate various preventive measures associated with cancer. Relatively recently, numerous studies have indicated that various dietary polyphenols and phytochemicals possess properties of modifying epigenetic mechanisms that modulate gene expression resulting in regulation of cancer. These polyphenols and phytochemicals, when administrated in a dose-dependent and combinatorial-based manner, can have an enhanced effect on epigenetic changes, which play a crucial role in cancer prevention and therapy. Hence, this review will focus on the mechanisms of combined polyphenols and phytochemicals that can impact various epigenetic modifications such as DNA methylation and histone modifications as well as regulation of non-coding miRNAs expression for treatment and prevention of various types of cancer.
Collapse
Affiliation(s)
- Itika Arora
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Manvi Sharma
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Center for Healthy Aging, University of Alabama Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA.
- Comprehensive Cancer Center, University of Alabama Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA.
- Nutrition Obesity Research Center, University of Alabama Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Diabetes Center, University of Alabama Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA.
| |
Collapse
|
12
|
Cisplatin Synergistically Enhances Antitumor Potency of Conditionally Replicating Adenovirus via p53 Dependent or Independent Pathways in Human Lung Carcinoma. Int J Mol Sci 2019; 20:ijms20051125. [PMID: 30841620 PMCID: PMC6429304 DOI: 10.3390/ijms20051125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/27/2019] [Indexed: 01/31/2023] Open
Abstract
Cisplatin is ranked as one of the most powerful and commonly prescribed anti-tumor chemotherapeutic agents which improve survival in many solid tumors including non-small cell lung cancer. However, the treatment of advanced lung cancer is restricted due to chemotherapy resistance. Here, we developed and investigated survivin promoter regulating conditionally replicating adenovirus (CRAd) for its anti-tumor potential alone or in combination with cisplatin in two lung cancer cells, H23, H2126, and their resistant cells, H23/CPR, H2126/CPR. To measure the expression of genes which regulate resistance, adenoviral transduction, metastasis, and apoptosis in cancer cells, RT-PCR and Western blotting were performed. The anti-tumor efficacy of the treatments was evaluated through flow cytometry, MTT and transwell assays. This study demonstrated that co-treatment with cisplatin and CRAd exerts synergistic anti-tumor effects on chemotherapy sensitive lung cancer cells and monotherapy of CRAd could be a practical approach to deal with chemotherapy resistance. Combined treatment induced stronger apoptosis by suppressing the anti-apoptotic molecule Bcl-2, and reversed epithelial to mesenchymal transition. In conclusion, cisplatin synergistically increased the tumor-killing of CRAd by (1) increasing CRAd transduction via enhanced CAR expression and (2) increasing p53 dependent or independent apoptosis of lung cancer cell lines. Also, CRAd alone proved to be a very efficient anti-tumor agent in cancer cells resistant to cisplatin owing to upregulated CAR levels. In an exciting outcome, we have revealed novel therapeutic opportunities to exploit intrinsic and acquired resistance to enhance the therapeutic index of anti-tumor treatment in lung cancer.
Collapse
|
13
|
Molecular Mechanisms and Bioavailability of Polyphenols in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20051062. [PMID: 30823649 PMCID: PMC6429226 DOI: 10.3390/ijms20051062] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 02/20/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is the one of the most frequently diagnosed cancers among men over the age of 50. Several lines of evidence support the observation that polyphenols have preventive and therapeutic effects in prostate cancer. Moreover, prostate cancer is ideal for chemoprevention due to its long latency. We propose here an equilibrated lifestyle with a diet rich in polyphenols as prophylactic attempts to slow down the progression of localized prostate cancer or prevent the occurrence of the disease. In this review, we will first summarize the molecular mechanisms of polyphenols in prostate cancer with a focus on the antioxidant and pro-oxidant effects, androgen receptors (AR), key molecules involved in AR signaling and their transactivation pathways, cell cycle, apoptosis, angiogenesis, metastasis, genetic aspects, and epigenetic mechanisms. The relevance of the molecular mechanisms is discussed in light of current bioavailability data regarding the activity of polyphenols in prostate cancer. We also highlight strategies for improving the bioavailability of polyphenols. We hope that this review will lead to further research regarding the bioavailability and the role of polyphenols in prostate cancer prevention and treatment.
Collapse
|
14
|
Sheng H, Li X, Xu Y. Knockdown of FOXP1 promotes the development of lung adenocarcinoma. Cancer Biol Ther 2018; 20:537-545. [PMID: 30409062 DOI: 10.1080/15384047.2018.1537999] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Lung cancer is one of the most common cancers in the world, which accounts for about 27% of all cancer deaths. However, the mechanisms underlying the pathogenesis of lung cancer cells remain largely elusive. In this study, we examined the role of the Forkhead box protein P1 (FOXP1) in lung cancer development. Our Oncomine analysis shows that FOXP1 is downregulated in lung adenocarcinoma compared with normal lung tissue. Knockdown of FOXP1 promotes the growth and invasion of PC9 and A549 cells by regulating genes of chemokine signaling molecules, including CCR1, ADCY5, GNG7, VAV3, and PLCB1. Simultaneous knockdown of CCR1 and FOXP1 attenuated FOXP1 knockdown-induced increase of lung cancer cell growth. Finally, knockdown of FOXP1 in PC9 cells promotes the tumorigenesis via CCR1 signaling in xenograft mouse model. Taken together, our data suggest that FOXP1 plays important roles in preventing lung adenocarcinoma development via suppressing chemokine signaling pathways.
Collapse
Affiliation(s)
- Hua Sheng
- a Department of Pulmonary and Critical Care Medicine, Huadong Hospital, Fudan University , Shanghai , China
| | - Xiangyang Li
- a Department of Pulmonary and Critical Care Medicine, Huadong Hospital, Fudan University , Shanghai , China
| | - Yi Xu
- a Department of Pulmonary and Critical Care Medicine, Huadong Hospital, Fudan University , Shanghai , China
| |
Collapse
|
15
|
Sheng J, Yin M, Sun Z, Kang X, Liu D, Jiang K, Xu J, Zhao F, Guo Q, Zheng W. SPC24 promotes osteosarcoma progression by increasing EGFR/MAPK signaling. Oncotarget 2017; 8:105276-105283. [PMID: 29285250 PMCID: PMC5739637 DOI: 10.18632/oncotarget.22167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 08/29/2017] [Indexed: 12/18/2022] Open
Abstract
In this study, we investigated the role of the spindle checkpoint protein SPC24 in osteosarcoma progression. SPC24 knockdown in 143B and U2OS osteosarcoma cells decreased cell growth, survival and invasiveness. The SPC24 knockdown cells also exhibited low EGFR, Ras and phospho-ERK levels and high E-cadherin levels, suggesting inhibition of EGFR/Ras/ERK signaling and epithelial-to-mesenchymal transitioning. Xenografted SPC24 knockdown osteosarcoma cells showed reduced tumor growth in nude mice with decreased EGFR and phospho-ERK levels and increased E-cadherin levels. By contrast, human osteosarcoma tissue samples showed high SPC24 and phospho-ERK levels and low E-cadherin levels. These results suggest SPC24 promotes osteosarcoma progression by increasing EGFR/Ras/ERK signaling.
Collapse
Affiliation(s)
- Jun Sheng
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Mengchen Yin
- Department of Orthopedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhengwang Sun
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Xia Kang
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Da Liu
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Kai Jiang
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Jia Xu
- Department of Personnel Office, Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang, China
| | - Feixing Zhao
- Department of Pathology, Zhuji People's Hospital of Zhejiang Province, Zhuji, Zhejiang, China
| | - Qunfeng Guo
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Wei Zheng
- Department of Orthopedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| |
Collapse
|
16
|
SPC24 is critical for anaplastic thyroid cancer progression. Oncotarget 2017; 8:21884-21891. [PMID: 28423533 PMCID: PMC5400631 DOI: 10.18632/oncotarget.15670] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/27/2017] [Indexed: 12/18/2022] Open
Abstract
In the past 2 decades, the incidence of thyroid cancer has been rapidly increasing worldwide. Anaplastic thyroid cancer (ATC) is the most lethal of all thyroid cancers and one of the most aggressive human carcinomas. SPC24 is an important component of the mitotic checkpoint machinery in the tumorigenesis and high levels of SPC24 have been found in colorectal and hepatocellular carcinomas, but its role in anaplastic thyroid cancer is still unclear. Our results showed that SPC24 was high expressed in human thyroid cancer samples. In addition, knockingdown endogenous SPC24 could repress cell growth, inhibit cell invasive ability and promote apoptosis in different ATC cells. Next, in vivo xenograft studies indicated that the SPC24 knockdown cells has decreased tumor size compared to the controls. This conclusion is also endorsed by our studies using human thyroid cancer samples. Taken together, our data demonstrates that SPC24 can serve as a promising prognostic biomarker of ATC cells and it is a novel strategy which could be developed by targeting SPC24 in future.
Collapse
|
17
|
Protective Effect of Green Tea ( Camellia sinensis (L.) Kuntze) against Prostate Cancer: From In Vitro Data to Algerian Patients. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:1691568. [PMID: 28133488 PMCID: PMC5241931 DOI: 10.1155/2017/1691568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/19/2016] [Accepted: 12/05/2016] [Indexed: 02/06/2023]
Abstract
Green tea (GT) has been studied for its effects as antioxidant and cancer-preventive agent. Epidemiological studies showed that GT consumption decreases the risk for prostate cancer (PC). To investigate whether erythrocyte oxidative stress (OS) is associated with PC and whether daily consumption of GT improves the oxidative phenotype, we performed a study in a group of Algerian PC patients, preceded by an in vitro study to characterize composition and antioxidant/antiproliferative activities of the GT used. This contained a high content of phenolic and flavonoid compounds, demonstrating in vitro antioxidant activity and significant antiproliferative effect on human prostate cancer PC-3 cell line. Seventy PC patients and 120 age-matched healthy subjects participated in the study, with glutathione (GSH), malondialdehyde (MDA), and catalase activity evaluated before and after GT consumption. The results showed a reduced GSH and catalase activity and a high level of MDA in erythrocytes from PC patients. The consumption of 2-3 cups per day of GT during 6 months significantly increased GSH concentration and catalase activity and decreased MDA concentration. In conclusion, GT significantly decreased OS in Algerian PC patients. Regular consumption of GT for a long period may prevent men from developing PC or at least delay its progression.
Collapse
|
18
|
Kim H, Banerjee N, Barnes RC, Pfent CM, Talcott ST, Dashwood RH, Mertens-Talcott SU. Mango polyphenolics reduce inflammation in intestinal colitis-involvement of the miR-126/PI3K/AKT/mTOR axis in vitro and in vivo. Mol Carcinog 2017; 56:197-207. [PMID: 27061150 PMCID: PMC5053910 DOI: 10.1002/mc.22484] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 03/07/2016] [Accepted: 03/18/2016] [Indexed: 12/19/2022]
Abstract
This study sought to elucidate the mechanisms underlying the anti-inflammatory effect of mango (Mangifera Indica L.) polyphenolics containing gallic acid and gallotanins, and the role of the miR-126/PI3K/AKT/mTOR signaling axis in vitro and in vivo. Polyphenolics extracted from mango (var. Keitt) were investigated in lipopolysaccharide (LPS)-treated CCD-18Co cells. Rats received either a beverage with mango polyphenolics or a control beverage, and were exposed to three cycles of 3% dextran sodium sulfate (DSS) followed by a 2-wk recovery period. The mango extract (10 mg GAE/L) suppressed the protein expression of NF-κB, p-NF-κB, PI3K (p85β), HIF-1α, p70S6K1, and RPS6 in LPS-treated CCD-18Co cells. LPS reduced miR-126 expression, whereas, the mango extract induced miR-126 expression in a dose-dependent manner. The relationship between miR-126 and its target, PI3K (p85β), was confirmed by treating cells with miR-126 antagomiR where mango polyphenols reversed the effects of the antagomiR. In vivo, mango beverage protected against DSS-induced colonic inflammation (47%, P = 0.05) and decreased the Ki-67 labeling index in the central and basal regions compared to the control. Mango beverage significantly attenuated the expression of pro-inflammatory cytokines such as TNF-α, IL-1β, and iNOS at the mRNA and protein level. Moreover, the expression of PI3K, AKT, and mTOR was reduced, whereas, miR-126 was upregulated by the mango treatment. These results suggest that mango polyphenols attenuated inflammatory response by modulating the PI3K/AKT/mTOR pathway at least in part through upregulation of miRNA-126 expression both in vitro and in vivo; thus, mango polyphenolics might be relevant as preventive agents in ulcerative colitis. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hyemee Kim
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Nivedita Banerjee
- Interdisciplinary Program of Toxicology, Texas A&M University, College Station, Texas
| | - Ryan C Barnes
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Catherine M Pfent
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
| | - Stephen T Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Roderick H Dashwood
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
- Center for Epigenetics and Disease Prevention, Texas A&M Health Science Center, Houston, Texas
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Susanne U Mertens-Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| |
Collapse
|
19
|
Natural Polyphenols for Prevention and Treatment of Cancer. Nutrients 2016; 8:nu8080515. [PMID: 27556486 PMCID: PMC4997428 DOI: 10.3390/nu8080515] [Citation(s) in RCA: 384] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/12/2016] [Accepted: 08/12/2016] [Indexed: 02/06/2023] Open
Abstract
There is much epidemiological evidence that a diet rich in fruits and vegetables could lower the risk of certain cancers. The effect has been attributed, in part, to natural polyphenols. Besides, numerous studies have demonstrated that natural polyphenols could be used for the prevention and treatment of cancer. Potential mechanisms included antioxidant, anti-inflammation as well as the modulation of multiple molecular events involved in carcinogenesis. The current review summarized the anticancer efficacy of major polyphenol classes (flavonoids, phenolic acids, lignans and stilbenes) and discussed the potential mechanisms of action, which were based on epidemiological, in vitro, in vivo and clinical studies within the past five years.
Collapse
|
20
|
Huang Q, Wei H, Wu Z, Li L, Yao L, Sun Z, Li L, Lin Z, Xu W, Han S, Cao W, Xu Y, Song D, Yang X, Xiao J. Preferentially Expressed Antigen of Melanoma Prevents Lung Cancer Metastasis. PLoS One 2016; 11:e0149640. [PMID: 27391090 PMCID: PMC4938541 DOI: 10.1371/journal.pone.0149640] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/03/2016] [Indexed: 12/28/2022] Open
Abstract
Lung cancer is the most common cause of cancer death worldwide. The poor survival rate is largely due to the extensive local invasion and metastasis. However, the mechanisms underlying the invasion and metastasis of lung cancer cells remain largely elusive. In this study, we examined the role of preferentially expressed antigen of melanoma (PRAME) in lung cancer metastasis. Our results show that PRAME is downregulated in lung adenocarcinoma and lung bone metastasis compared with normal human lung. Knockdown of PRAME decreases the expression of E-Cadherin and promotes the proliferation, invasion, and metastasis of lung cancer cells by regulating multiple critical genes, most of which are related to cell migration, including MMP1, CCL2, CTGF, and PLAU. Clinical data analysis reveals that the expression of MMP1 correlates with the clinical features and outcome of lung adenocarcinoma. Taken together, our data demonstrate that PRAME plays a role in preventing the invasion and metastasis of lung adenocarcinoma and novel diagnostic or therapeutic strategies can be developed by targeting PRAME.
Collapse
Affiliation(s)
- Quan Huang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Haifeng Wei
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
- * E-mail: (YFX); (DWS); (HFW); (XHY); (JRX)
| | - Zhipeng Wu
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Lin Li
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Liangfang Yao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Zhengwang Sun
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Lei Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Zaijun Lin
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Wei Xu
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Shuai Han
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
| | - Wenjiao Cao
- The International Peace Maternity& Child Health Hospital of China welfare institute (IPMCH), Shanghai, China
| | - Yunfei Xu
- Urology Department, Tenth People's Hospital of Tongji University, Shanghai, PR China
- * E-mail: (YFX); (DWS); (HFW); (XHY); (JRX)
| | - Dianwen Song
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
- * E-mail: (YFX); (DWS); (HFW); (XHY); (JRX)
| | - Xinghai Yang
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
- * E-mail: (YFX); (DWS); (HFW); (XHY); (JRX)
| | - Jianru Xiao
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, No. 415 Fengyang Road, Shanghai, 200003, China
- * E-mail: (YFX); (DWS); (HFW); (XHY); (JRX)
| |
Collapse
|
21
|
Di J, Cao H, Tang J, Lu Z, Gao K, Zhu Z, Zheng J. Rap2B promotes cell proliferation, migration and invasion in prostate cancer. Med Oncol 2016; 33:58. [PMID: 27154636 DOI: 10.1007/s12032-016-0771-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/26/2016] [Indexed: 10/21/2022]
Abstract
Rap2B, a member of the Ras family of small GTP-binding proteins, reportedly presents a high level of expression in various human tumors and plays a significant role in the development of tumor. However, the function of Rap2B in prostate cancer (PCa) remains unclear. We elucidated the stimulative role of Rap2B in PCa cell proliferation, migration and invasion by means of the CCK-8 cell proliferation assay, cell cycle analysis and transwell migration assay. Western blot analysis uncovered that elevated Rap2B leads to increased phosphorylation levels of FAK, suggesting that FAK-dependent pathway might be responsible for the effect of Rap2B on PCa cells migration and invasion. Inversely, FAK-specific inhibitor (PF-573228) can abort Rap2B-induced FAK phosphorylation. In vivo experiment confirmed that Rap2B positively regulated PCa growth and metastasis, as well as the expression of phosphorylated FAK. Collectively, these findings shed light on Rap2B as a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Jiehui Di
- Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Huan Cao
- Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Juangjuan Tang
- Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Oncology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Zheng Lu
- Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Keyu Gao
- Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Zhesi Zhu
- Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, People's Republic of China.
| |
Collapse
|
22
|
Hsieh CL, Lin CH, Chen KC, Peng CC, Peng RY. The teratogenicity and the action mechanism of gallic acid relating with brain and cervical muscles. PLoS One 2015; 10:e0119516. [PMID: 26030624 PMCID: PMC4452303 DOI: 10.1371/journal.pone.0119516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 01/29/2015] [Indexed: 12/30/2022] Open
Abstract
Gallic acid (3,4,5-trihydroxybenzoic acid) (GA) and other flavanoids are extensively used in nutraceuticals because of their antioxidant and antiinflammatory properties. While examining whether GA is effective in alleviating valproic-acid-induced teratogenesis in a chicken embryo model (CEM), we observed embryo hemorrhage and liposis in the musculi longissimus cervicis. We conducted this study to determine whether GA is inherently teratogenic and the extent to which the risk can be transferred to fetuses. A CEM was used to administer GA at 2, 6, 10, and 14 μM. GA at 2 μM did not exhibit cytotoxicity. At 6, 10, and 14 μM, GA caused severe decreases in body and liver weights, causing -5.6%, -21.3%, and -27.5% body weights and 4.0, 3.8, and 3.2-g, liver weights, respectively, in day-1 chicks. The optimal alive birth rate (or damaging rate) reached 33.3%, 39.4%, and 29.2% at 6, 10, and 14 μM GA, respectively. The damaged tissue was primarily cervical muscle (musculi longissimus cervicis), as evidenced by liposis, Zenker’s necrosis, and hemolysis. The erythrocyte, hemoglobin, eosinophil, lymphocyte, and monocyte counts were severely reduced and PPAR-α was downregulated, whereas the Ras/Raf/JAK/STAT pathway was upregulated. The GA dose required to induce teratogenesis was ≥ 6 μM (1.02 mg/kg), which can be easily consumed by pregnant women in typical teas such as Chinese Pu-’Er and Chinese black teas, indicating a potential risk to human fetuses. GA at doses ≥ 1.02 mg/kg of body weight potentially causes characteristic cerebral hemolysis and liposis in the musculi longissimus cervicis. The mechanism of action of GA is multidisciplinary: The liposis can be ascribed to downregulation of PPAR-α; the erythrocyte hemolysis can be attributed to its unique autooxidative and prooxidant behavior and the inhibition of carbonic anhydrase; and the proliferation and differentiation deficits can be attributed to the upregulation of the Ras/Raf/JAK/STAT pathway.
Collapse
Affiliation(s)
- Chiu Lan Hsieh
- Graduate Institute of Biotechnology, Changhua University of Education, Changhua, Taiwan
| | - Chien-Hong Lin
- Graduate Institute of Biotechnology, Changhua University of Education, Changhua, Taiwan
| | - Kuan Chou Chen
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- * E-mail: (KCC); (CCP)
| | - Chiung-Chi Peng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- * E-mail: (KCC); (CCP)
| | - Robert Y. Peng
- Research Institute of Biotechnology, Hungkuang University, Taichung City, Taiwan
| |
Collapse
|
23
|
Huang YL, Chu YL, Ho CT, Chung JG, Lai CI, Su YC, Kuo YH, Sheen LY. Antcin K, an Active Triterpenoid from the Fruiting Bodies of Basswood-Cultivated Antrodia cinnamomea, Inhibits Metastasis via Suppression of Integrin-Mediated Adhesion, Migration, and Invasion in Human Hepatoma Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:4561-4569. [PMID: 25911944 DOI: 10.1021/jf5059304] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Previous research demonstrated that the ethyl acetate extract from Antrodia cinnamomea suppresses the invasive potential of human breast and hepatoma cells, but the effective compounds are not identified. The main bioactive compounds of A. cinnamomea are ergostane-type triterpenoids, and the content of antcin K is the highest. The objective of this study was to evaluate the antimetastatic activity and mechanisms of antcin K purified from the fruiting body of basswood-cultivated A. cinnamomea on human liver cancer Hep 3B cells. The results showed that adhesion, migration, and invasion of Hep 3B cells were effectively inhibited by antcin K within 24 h of treatment. Antcin K not only reduced the protein expression and activity of MMP-2 and MMP-9 but also down-regulated vimentin and up-regulated E-cadherin in Hep 3B cells. In depth investigation for the molecular mechanism revealed that antcin K could reduce the protein expression of integrin β1, β3, α5, and αv and suppress phosphorylation of FAK, Src, PI3K, AKT, MEK, ERK, and JNK. These results suggested that antcin K was able to inhibit the metastasis of human hepatoma cells through suppression of integrin-mediated adhesion, migration, and invasion. Coupled with these findings, antcin K has a good potential to reduce the risk of liver cancer metastasis.
Collapse
Affiliation(s)
| | | | - Chi-Tang Ho
- ΔDepartment of Food Science, Rutgers University, New Brunswick, New Jersey, United States
| | - Jing-Gung Chung
- §Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | | | | | - Yueh-Hsiung Kuo
- ΠDepartment of Chinese Pharmaceutical Sciences and Chinese Medicine Resourceσ, China Medical University, Taichung, Taiwan
| | | |
Collapse
|
24
|
Pavese JM, Ogden IM, Voll EA, Huang X, Xu L, Jovanovic B, Bergan RC. Mitogen-activated protein kinase kinase 4 (MAP2K4) promotes human prostate cancer metastasis. PLoS One 2014; 9:e102289. [PMID: 25019290 PMCID: PMC4096757 DOI: 10.1371/journal.pone.0102289] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/17/2014] [Indexed: 11/19/2022] Open
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in the US. Death from PCa primarily results from metastasis. Mitogen-activated protein kinase kinase 4 (MAP2K4) is overexpressed in invasive PCa lesions in humans, and can be inhibited by small molecule therapeutics that demonstrate favorable activity in phase II studies. However, MAP2K4's role in regulating metastatic behavior is controversial and unknown. To investigate, we engineered human PCa cell lines which overexpress either wild type or constitutive active MAP2K4. Orthotopic implantation into mice demonstrated MAP2K4 increases formation of distant metastasis. Constitutive active MAP2K4, though not wild type, increases tumor size and circulating tumor cells in the blood and bone marrow. Complementary in vitro studies establish stable MAP2K4 overexpression promotes cell invasion, but does not affect cell growth or migration. MAP2K4 overexpression increases the expression of heat shock protein 27 (HSP27) protein and protease production, with the largest effect upon matrix metalloproteinase 2 (MMP-2), both in vitro and in mouse tumor samples. Further, MAP2K4-mediated increases in cell invasion are dependent upon heat shock protein 27 (HSP27) and MMP-2, but not upon MAP2K4's immediate downstream targets, p38 MAPK or JNK. We demonstrate that MAP2K4 increases human PCa metastasis, and prolonged over expression induces long term changes in cell signaling pathways leading to independence from p38 MAPK and JNK. These findings provide a mechanistic explanation for human studies linking increases in HSP27 and MMP-2 to progression to metastatic disease. MAP2K4 is validated as an important therapeutic target for inhibiting human PCa metastasis.
Collapse
Affiliation(s)
- Janet M. Pavese
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Irene M. Ogden
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Eric A. Voll
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Xiaoke Huang
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Li Xu
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Borko Jovanovic
- Department of Preventative Medicine, Northwestern University, Chicago, Illinois, United States of America
- Robert H. Lurie Cancer Center, Northwestern University, Chicago, Illinois, United States of America
| | - Raymond C. Bergan
- Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Department of Preventative Medicine, Northwestern University, Chicago, Illinois, United States of America
- Robert H. Lurie Cancer Center, Northwestern University, Chicago, Illinois, United States of America
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
25
|
Wei C, Bai WK, Wang Y, Hu B. Combined treatment of PC-3 cells with ultrasound and microbubbles suppresses invasion and migration. Oncol Lett 2014; 8:1372-1376. [PMID: 25120726 PMCID: PMC4114620 DOI: 10.3892/ol.2014.2310] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 06/12/2014] [Indexed: 01/25/2023] Open
Abstract
The aim of the present study was to investigate whether ultrasound treatment combined with microbubbles inhibits cell invasion and migration in androgen-independent prostate cancer (PCa) cells and to identify the probable mechanism. Ultrasound was used in continuous wave mode at a frequency of 21 kHz and with a spatial-average temporal-average intensity of 46 mW/cm2. Ultrasound combined with microbubbles (200 μl; SonoVue) was administered to androgen-independent human PCa PC-3 cells for 30 sec. The PC-3 cells were divided into three groups: The control group, the ultrasound group (US) and the ultrasound combined with microbubbles group (US + MB). Following treatment for 12, 24, 48 and 72 h, cell counting kit-8 was used to assess cell viability. Cell invasion and migration was measured 12 h after treatment using Transwell migration assays. Quantitative polymerase chain reaction and western blot analysis were used to evaluate the expression of the migration-associated proteins, matrix metalloproteinase (MMP)-2 and MMP-9. Cell reproduction levels in the US and US + MB groups were significantly suppressed when compared with the control group (P<0.01) following 24 h of treatment and this suppression was significantly higher in the US + MB group than in the US group (P<0.01). However, no significant differences in cell reproduction levels between the three groups were identified at 12 h (P>0.05). Ultrasound combined with microbubbles significantly suppressed the level of invasion and migration in the PC-3 cells compared with the control group (190.83±14.63 vs. 509.67±18.62, P<0.01; and 86.67±10.60 vs. 271.33±65.14; P<0.01, respectively). Furthermore, combined treatment with ultrasound and microbubbles suppressed the expression of MMP-2 and MMP-9. In conclusion, it was found that ultrasound combined with microbubbles suppressed invasion and migration in human PCa PC-3 cells via downregulation of MMP-2 and MMP-9.
Collapse
Affiliation(s)
- Cong Wei
- Department of Ultrasound in Medicine, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai 200233, P.R. China
| | - Wen-Kun Bai
- Department of Ultrasound in Medicine, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai 200233, P.R. China
| | - Yu Wang
- Department of Ultrasound in Medicine, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai 200233, P.R. China
| | - Bing Hu
- Department of Ultrasound in Medicine, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Institute of Ultrasound in Medicine, Shanghai 200233, P.R. China
| |
Collapse
|
26
|
Shohrati M, Haji Hosseini R, Esfandiari MA, Najafian N, Najafian B, Golbedagh A. Serum matrix metalloproteinase levels in patients exposed to sulfur mustard. IRANIAN RED CRESCENT MEDICAL JOURNAL 2014; 16:e15129. [PMID: 24829780 PMCID: PMC4005442 DOI: 10.5812/ircmj.15129] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/04/2013] [Accepted: 12/07/2013] [Indexed: 02/04/2023]
Abstract
Background: Matrix metalloproteinases (MMPs) are a group of endopeptidases which comprised of various types. These proteolytic enzymes are zinc-dependent and play role in degradation of extracellular matrix (ECM). Various types of cells such as macrophages, fibroblasts, neutrophils, synovial cells and some epithelial cells secrete MMPs. According to previous studies on bronchiolitis and respiratory tract lesions in these patients and unknown pathophysiology mechanism up to date, this cross–sectional study was performed. Objectives: The aim of this study was to compare the serum MMP level in patients with chemical injuries and normal people and also determine the role of these parameters in pulmonary disorders . Materials and Methods: In this cross–sectional study, 25 Iranian patients exposed to the sulfur mustard and 25 unexposed participants as the control group were enrolled. Serum samples were collected from two groups and stored at -70˚C until the measurement of MMPs and TIMPs. ELISA kit was used for measurement of MMP and TIMP based on the kit's instruction. For validations in measurement, all samples were analyzed duplicate and in some cases triplicate. Results: The mean level of MMP-9 in serum of chemically-injured group was 1592.42 and this amount in normal group was 679.72 .So there was a significant difference between two groups (P = 0.001) and the mean level of MMP-8 in serum of patients group was 49.10 and in normal group was 35.53. Then there was no significant difference between two groups (P = 0.197). The mean levels of MMP-1 and MMP-2 was not significantly different (P value > 0.05) in the patient and normal groups. And also the mean levels of TIMP-1 and TIMP-2 was not significantly different (P > 0.05) in the patients and normal groups. Conclusions: In summary, serum MMPs in chemically-injured has shown no significant difference with normal people except for the MMP-9.
Collapse
Affiliation(s)
- Majid Shohrati
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
- Corresponding Author: Majid Shohrati, Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran. Tel/Fax: +98-2182482523, E-mail:
| | | | | | | | - Bita Najafian
- Department of Pediatrics, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | - Abbas Golbedagh
- Department of Biochemistry, Payame Noor University, Tehran, IR Iran
| |
Collapse
|
27
|
Zhang L, Lokeshwar BL. Medicinal properties of the Jamaican pepper plant Pimenta dioica and Allspice. Curr Drug Targets 2013; 13:1900-6. [PMID: 23140298 DOI: 10.2174/138945012804545641] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 10/24/2012] [Accepted: 11/03/2012] [Indexed: 11/22/2022]
Abstract
The Caribbean tropical tree, Pimenta dioica has been used for a variety of human endeavors, such as in perfumery industry, food spice, as a natural pesticide, and in folk medicine. Discovered in Jamaica during the voyages of Christopher Columbus, the dried unripe berries of P. dioica also known as Allspice can be found in all continents with unique names in over 50 languages. Systematic investigation of aromatic constituents of Pimenta leaves and its unripe berries, Allspice, have resulted in discovery of many and novel aromatic compounds, mostly glycosides and polyphenols that show antibacterial, hypotensive, anti-neuralgic and analgesic properties. Recent studies have shown two of the known compounds isolated from Allspice, Eugenol and Gallic acid have selective antiproliferative and anti-tumor properties on human cancer cells and their animal models. New characterization of novel compounds such as Ericifolin from the aqueous extract of Allspice berries show potent anti-prostate cancer and anti-breast cancer properties that can be verified in vitro as well as in vivo. Considering its purity, mostly available as "organically grown" berries, availability at low cost, wide acceptance in culinary delights of many cultures world-wide, Allspice may have an additional space in most households, in their medicine cabinets.
Collapse
Affiliation(s)
- Lei Zhang
- Sheila and David Fuente Graduate Program in Cancer Biology, Leonard Miller School of Medicine, University of Miami, Miami, FL33136, USA
| | | |
Collapse
|
28
|
Suppressions of Migration and Invasion by Cantharidin in TSGH-8301 Human Bladder Carcinoma Cells through the Inhibitions of Matrix Metalloproteinase-2/-9 Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:190281. [PMID: 23431332 PMCID: PMC3568914 DOI: 10.1155/2013/190281] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/10/2012] [Accepted: 12/11/2012] [Indexed: 12/04/2022]
Abstract
Cancer metastasis becomes an initial cause of cancer death in human population. In many cancers, it has been shown that the high levels of matrix metalloproteinase (MMP)-2 and/or MMP-9 are associated with the invasive phenotypes of cancer cells. In this study, we investigated the effects of cantharidin, a derivative of blister beetles which is one of the traditional Chinese medicines, on the adhesion, migration, and invasion of human bladder cancer TSGH-8301 cells. Cantharidin effectively suppressed TSGH-8301 cell adhesion, migration, and invasion in a concentration-dependent manner. Results from Western blotting, RT-PCR, and gelatin zymography assays indicated that cantharidin blocked the protein levels, gene expression (mRNA), and activities of MMP-2 and -9 in TSGH-8301 cells. Cantharidin also significantly suppressed the protein expressions of p-p38 and p-JNK1/2 in TSGH-8301 cells. Taken together, cantharidin was suggested to present antimetastatic potential via suppressing the levels of MMP-2 and MMP-9 expression that might be mediated by targeting the p38 and JNK1/2 MAPKs pathway in TSGH-8301 human bladder cancer cells.
Collapse
|
29
|
Liang CZ, Zhang X, Li H, Tao YQ, Tao LJ, Yang ZR, Zhou XP, Shi ZL, Tao HM. Gallic acid induces the apoptosis of human osteosarcoma cells in vitro and in vivo via the regulation of mitogen-activated protein kinase pathways. Cancer Biother Radiopharm 2012; 27:701-10. [PMID: 22849560 DOI: 10.1089/cbr.2012.1245] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
To examine the antitumor effects of gallic acid (GA) on osteosarcoma, two human osteosarcoma cell lines U-2OS and MNNG/HOS were treated by GA and subjected to cell proliferation and apoptosis assays. In addition, MNNG/HOS xenograft tumors were established in nude BALB/c mice to evaluate the anticancer capacity of GA in vivo. The results showed that GA inhibited the proliferation and induced the apoptosis of osteosarcoma cells, accompanied by the upregulation of p-38 activation and the downregulation of c-Jun N-terminal kinase (JNK) and extracellular signal regulated kinase (ERK1/2) activation. Additionally, p38 MAPK inhibitor abrogated GA-induced growth inhibition of osteosarcoma cells, whereas JNK or ERK1/2 inhibitors sensitized osteosarcoma cells to GA-induced growth inhibition. In vivo studies further showed that GA administration decreased xenograft tumor growth in a dose-dependent manner. Immunohistochemistry analysis demonstrated the downregulation of PCNA and CD31 expression and upregulation of apoptosis in MNNG/HOS tumor tissues following GA treatment. This study demonstrates the antitumor efficacy of GA for osteosarcoma that is mediated by the modulation of cell proliferation, apoptosis, and angiogenesis. Our findings suggest that GA could be a potent agent for osteosarcoma intervention.
Collapse
Affiliation(s)
- Cheng-zhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Triggering apoptotic death of human malignant melanoma a375.s2 cells by bufalin: involvement of caspase cascade-dependent and independent mitochondrial signaling pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:591241. [PMID: 22719785 PMCID: PMC3376545 DOI: 10.1155/2012/591241] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 03/26/2012] [Indexed: 12/18/2022]
Abstract
Bufalin was obtained from the skin and parotid venom glands of toad and has been shown to induce cytotoxic effects in various types of cancer cell lines, but there is no report to show that whether bufalin affects human skin cancer cells. The aim of this investigation was to study the effects of bufalin on human malignant melanoma A375.S2 cells and to elucidate possible mechanisms involved in induction of apoptosis. A375.S2 cells were treated with different concentrations of bufalin for a specific time period and investigated for effects on apoptotic analyses. Our results indicated that cells after exposure to bufalin significantly decreased cell viability, and induced cell morphological changes and chromatin condensation in a concentration-dependent manner. Flow cytometric assays indicated that bufalin promoted ROS productions, loss of mitochondrial membrane potential (ΔΨm), intracellular Ca2+ release, and nitric oxide (NO) formations in A375.S2 cells. Additionally, the apoptotic induction of bufalin on A375.S2 cells resulted from mitochondrial dysfunction-related responses (disruption of the ΔΨm and releases of cytochrome c, AIF, and Endo G), and activations of caspase-3, caspase-8 and caspase-9 expressions. Based on those observations, we suggest that bufalin-triggered apoptosis in A375.S2 cells is correlated with extrinsic- and mitochondria-mediated multiple signal pathways.
Collapse
|
31
|
Activations of Both Extrinsic and Intrinsic Pathways in HCT 116 Human Colorectal Cancer Cells Contribute to Apoptosis through p53-Mediated ATM/Fas Signaling by Emilia sonchifolia Extract, a Folklore Medicinal Plant. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:178178. [PMID: 22474491 PMCID: PMC3303801 DOI: 10.1155/2012/178178] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 10/21/2011] [Accepted: 10/28/2011] [Indexed: 12/23/2022]
Abstract
Emilia sonchifolia (L.) DC (Compositae), an herbaceous plant found in Taiwan and India, is used as folk medicine. The clinical applications include inflammation, rheumatism, cough, cuts fever, dysentery, analgesic, and antibacteria. The activities of Emilia sonchifolia extract (ESE) on colorectal cancer cell death have not been fully investigated. The purpose of this study explored the induction of apoptosis and its molecular mechanisms in ESE-treated HCT 116 human colorectal cancer cells in vitro. The methanolic ESE was characterized, and γ-humulene was formed as the major constituent (63.86%). ESE induced cell growth inhibition in a concentration- and time-dependent response by MTT assay. Apoptotic cells (DNA fragmentation, an apoptotic catachrestic) were found after ESE treatment by TUNEL assay and DNA gel electrophoresis. Alternatively, ESE stimulated the activities of caspase-3, -8, and -9 and their specific caspase inhibitors protected against ESE-induced cytotoxicity. ESE promoted the mitochondria-dependent and death-receptor-associated protein levels. Also, ESE increased ROS production and upregulated the levels of ATM, p53, and Fas in HCT 116 cells. Strikingly, p53 siRNA reversed ESE-reduced viability involved in p53-mediated ATM/Fas signaling in HCT 116 cells. In summary, our result is the first report suggesting that ESE may be potentially efficacious in the treatment of colorectal cancer.
Collapse
|
32
|
Cucurbitacin E Induces G(2)/M Phase Arrest through STAT3/p53/p21 Signaling and Provokes Apoptosis via Fas/CD95 and Mitochondria-Dependent Pathways in Human Bladder Cancer T24 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:952762. [PMID: 22272214 PMCID: PMC3261502 DOI: 10.1155/2012/952762] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 09/25/2011] [Accepted: 10/02/2011] [Indexed: 11/28/2022]
Abstract
Cucurbitacin E, a tetracyclic triterpenes compound extracted from cucurbitaceous plants, has been shown to exhibit anticancer and anti-inflammatory activities. The purpose of this study was to elucidate whether cucurbitacin E promotes cell cycle arrest and induces apoptosis in T24 cells and further to explore the underlying molecular mechanisms. The effects of cucurbitacin E on T24 cell's growth and accompanied morphological changes were examined by MTT assay and a phase-contrast microscope. DNA content, mitochondrial membrane potential (ΔΨm) and annexin V/PI staining were determined by flow cytometry. The protein levels were measured by Western blotting. Our results demonstrated that cucurbitacin E-induced G2/M arrest was associated with a marked increase in the levels of p53, p21 and a decrease in phospho-signal transducer and activator of transcription 3 (STAT3), cyclin-dependent kinase 1 (CDK1) and cyclin B. Cucurbitacin E-triggered apoptosis was accompanied with up-regulation of Fas/CD95, truncated BID (t-BID) and a loss of ΔΨm, resulting in the releases of cytochrome c, apoptotic protease activating factor 1 (Apaf-1) and apoptosis-inducing factor (AIF), and sequential activation of caspase-8, caspase-9, and caspase-3. Our findings provided the first evidence that STAT3/p53/p21 signaling, Fas/CD95 and mitochondria-dependent pathways play critical roles in cucurbitacin E-induced G2/M phase arrest and apoptosis of T24 cells.
Collapse
|
33
|
Dong FY, Wu J, Tian HY, Ye QM, Jiang RW. Gallic acid pyridine monosolvate. Acta Crystallogr Sect E Struct Rep Online 2012; 67:o3096. [PMID: 22220100 PMCID: PMC3247482 DOI: 10.1107/s1600536811043868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Accepted: 10/22/2011] [Indexed: 11/30/2022]
Abstract
In the title compound (systenatic name: 3,4,5-trihydroxybenzoic acid pyridine monosolvate), C5H5N·C7H6O5, the gallic acid molecule is essentially planar (r.m.s deviation = 0.0766 Å for non-H atoms) and is linked to the pyridine molecule by an O—H⋯N hydrogen bond. An intramolecular O—H⋯O hydrogen bond occurs in the gallic acid molecule. The gallic acid and pyridine mean planes make a dihedral angle 12.6 (3)°. Intermolecular O—H⋯O and O—H⋯N hydrogen bonding involving the hydroxy and carboxyl groups and the pyridine molecule, and π–π interactions between inversion-related pyridines [centroid–centroid distance = 3.459 (6) Å] and between pyridine and benzene rings [centroid–centroid distance = 3.548 (6) Å], lead to a three-dimensional network in the crystal.
Collapse
Affiliation(s)
- Fu-Yue Dong
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Institute of Traditional Chinese Medicine and Natural Products, Jinan University, Guangzhou 510632, People's Republic of China
| | | | | | | | | |
Collapse
|
34
|
Ji BC, Yu CC, Yang ST, Hsia TC, Yang JS, Lai KC, Ko YC, Lin JJ, Lai TY, Chung JG. Induction of DNA damage by deguelin is mediated through reducing DNA repair genes in human non-small cell lung cancer NCI-H460 cells. Oncol Rep 2012; 27:959-64. [PMID: 22227970 PMCID: PMC3583480 DOI: 10.3892/or.2012.1622] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 11/03/2011] [Indexed: 12/13/2022] Open
Abstract
It has been shown that deguelin, one of the compounds of rotenoids from flavonoid family, induced cytotoxic effects through induction of cell cycle arrest and apoptosis in many types of human cancer cell lines, but deguelin-affected DNA damage and repair gene expression (mRNA) are not clarified yet. We investigated the effects of deguelin on DNA damage and associated gene expression in human lung cancer NCI-H460 cells in vitro. DNA damage was assayed by using the comet assay and DNA gel electrophoresis and the results indicated that NCI-H460 cells treated with 0, 50, 250 and 500 nM deguelin led to a longer DNA migration smear based on the single cell electrophoresis and DNA fragmentation occurred based on the examination of DNA gel electrophoresis. DNA damage and repair gene expression (mRNA) were evaluated by using real-time PCR assay and the results indicated that 50 and 250 nM deguelin for a 24-h exposure in NCI-H460 cells, decreased the gene levels of breast cancer 1, early onset (BRCA1), DNA-dependent serine/threonine protein kinase (DNA-PK), O6-methylguanine-DNA methyltransferase (MGMT), p53, ataxia telangiectasia mutated (ATM) and ataxia-telangiectasia and Rad3-related (ATR) mRNA expressions. Collectively, the present study showed that deguelin caused DNA damage and inhibited DNA damage and repair gene expressions, which might be due to deguelin-inhibited cell growth in vitro.
Collapse
Affiliation(s)
- Bin-Chuan Ji
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua 500, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Bernert B, Porsch H, Heldin P. Hyaluronan synthase 2 (HAS2) promotes breast cancer cell invasion by suppression of tissue metalloproteinase inhibitor 1 (TIMP-1). J Biol Chem 2011; 286:42349-42359. [PMID: 22016393 DOI: 10.1074/jbc.m111.278598] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Invasion and metastasis are the primary causes of breast cancer mortality, and increased knowledge about the molecular mechanisms involved in these processes is highly desirable. High levels of hyaluronan in breast tumors have been correlated with poor patient survival. The involvement of hyaluronan in the early invasive phase of a clone of breast cancer cell line MDA-MB-231 that forms bone metastases was studied using an in vivo-like basement membrane model. The metastatic to bone tumor cells exhibited a 7-fold higher hyaluronan-synthesizing capacity compared with MDA-MB-231 cells predominately due to an increased expression of hyaluronan synthase 2 (HAS2). We found that knockdown of HAS2 completely suppressed the invasive capability of these cells by the induction of tissue metalloproteinase inhibitor 1 (TIMP-1) and dephosphorylation of focal adhesion kinase. HAS2 knockdown-mediated inhibition of basement membrane remodeling was rescued by HAS2 overexpression, transfection with TIMP-1 siRNA, or addition of TIMP-1-blocking antibodies. Moreover, knockdown of HAS2 suppressed the EGF-mediated induction of the focal adhesion kinase/PI3K/Akt signaling pathway. Thus, this study provides new insights into a possible mechanism whereby HAS2 enhances breast cancer invasion.
Collapse
Affiliation(s)
- Berit Bernert
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala University, SE-75124 Uppsala, Sweden
| | - Helena Porsch
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala University, SE-75124 Uppsala, Sweden
| | - Paraskevi Heldin
- Ludwig Institute for Cancer Research, Biomedical Center, Uppsala University, SE-75124 Uppsala, Sweden.
| |
Collapse
|