1
|
Gelman IH. Metastasis suppressor genes in clinical practice: are they druggable? Cancer Metastasis Rev 2023; 42:1169-1188. [PMID: 37749308 DOI: 10.1007/s10555-023-10135-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/01/2023] [Indexed: 09/27/2023]
Abstract
Since the identification of NM23 (now called NME1) as the first metastasis suppressor gene (MSG), a small number of other gene products and non-coding RNAs have been identified that suppress specific parameters of the metastatic cascade, yet which have little or no ability to regulate primary tumor initiation or maintenance. MSG can regulate various pathways or cell biological functions such as those controlling mitogen-activated protein kinase pathway mediators, cell-cell and cell-extracellular matrix protein adhesion, cytoskeletal architecture, G-protein-coupled receptors, apoptosis, and transcriptional complexes. One defining facet of this gene class is that their expression is typically downregulated, not mutated, in metastasis, such that any effective therapeutic intervention would involve their re-expression. This review will address the therapeutic targeting of MSG, once thought to be a daunting task only facilitated by ectopically re-expressing MSG in metastatic cells in vivo. Examples will be cited of attempts to identify actionable oncogenic pathways that might suppress the formation or progression of metastases through the re-expression of specific metastasis suppressors.
Collapse
Affiliation(s)
- Irwin H Gelman
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
2
|
Lai TH, Ahmed M, Hwang JS, Bahar ME, Pham TM, Yang J, Kim W, Maulidi RF, Lee DK, Kim DH, Kim HJ, Kim DR. Manipulating RKIP reverses the metastatic potential of breast cancer cells. Front Oncol 2023; 13:1189350. [PMID: 37469399 PMCID: PMC10352845 DOI: 10.3389/fonc.2023.1189350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023] Open
Abstract
Breast cancer is a common tumor type among women, with a high fatality due to metastasis. Metastasis suppressors encode proteins that inhibit the metastatic cascade independent of the primary tumor growth. Raf kinase inhibitory protein (RKIP) is one of the promising metastasis suppressor candidates. RKIP is reduced or lost in aggressive variants of different types of cancer. A few pre-clinical or clinical studies have capitalized on this protein as a possible therapeutic target. In this article, we employed two breast cancer cells to highlight the role of RKIP as an antimetastatic gene. One is the low metastatic MCF-7 with high RKIP expression, and the other is MDA-MB-231 highly metastatic cell with low RKIP expression. We used high-throughput data to explore how RKIP is lost in human tissues and its effect on cell mobility. Based on our previous work recapitulating the links between RKIP and SNAI, we experimentally manipulated RKIP in the cell models through its novel upstream NME1 and investigated the subsequent genotypic and phenotypic changes. We also demonstrated that RKIP explained the uneven migration abilities of the two cell types. Furthermore, we identified the regulatory circuit that might carry the effect of an existing drug, Epirubicin, on activating gene transcription. In conclusion, we propose and test a potential strategy to reverse the metastatic capability of breast cancer cells by chemically manipulating RKIP expression.
Collapse
Affiliation(s)
- Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Mahmoud Ahmed
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Md Entaz Bahar
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Trang Minh Pham
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Jinsung Yang
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Wanil Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Rizi Firman Maulidi
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Dong-Kun Lee
- Department of Physiology and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Dong-Hee Kim
- Department of Orthopaedic Surgery and Institute of Health Sciences, Gyeongsang National University Hospital, and Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| |
Collapse
|
3
|
Gao Y, Lu Y, Zhang N, Udenigwe CC, Zhang Y, Fu Y. Preparation, pungency and bioactivity of gingerols from ginger ( Zingiber officinale Roscoe): a review. Crit Rev Food Sci Nutr 2022; 64:2708-2733. [PMID: 36135317 DOI: 10.1080/10408398.2022.2124951] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ginger has been widely used for different purposes, such as condiment, functional food, drugs, and cosmetics. Gingerols, the main pungent component in ginger, possess a variety of bioactivities. To fully understand the significance of gingerols in the food and pharmaceutical industry, this paper first recaps the composition and physiochemical properties of gingerols, and the major extraction and synthesis methods. Furthermore, the pungency and bioactivity of gingerols are reviewed. In addition, the food application of gingerols and future perspectives are discussed. Gingerols, characterized by a 3-methoxy-4-hydroxyphenyl moiety, are divided into gingerols, shogaols, paradols, zingerone, gingerdiones and gingerdiols. At present, gingerols are extracted by conventional, innovative, and integrated extraction methods, and synthesized by chemical, biological and in vitro cell synthesis methods. Gingerols can activate transient receptor potential vanilloid type 1 (TRPV1) and induce signal transduction, thereby exhibiting its pungent properties and bioactivity. By targeted mediation of various cell signaling pathways, gingerols display potential anticancer, antibacterial, blood glucose regulatory, hepato- and renal-protective, gastrointestinal regulatory, nerve regulatory, and cardiovascular protective effects. This review contributes to the application of gingerols as functional ingredients in the food and pharmaceutical industry.
Collapse
Affiliation(s)
- Yuge Gao
- College of Food Science, Southwest University, Chongqing, China
- Westa College, Southwest University, Chongqing, China
| | - Yujia Lu
- Department of Epidemiology, Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Na Zhang
- Key Laboratory of Food Science and Engineering of Heilongjiang Province, College of Food Engineering, Harbin University of Commerce, Harbin, China
| | - Chibuike C Udenigwe
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing, China
| | - Yu Fu
- College of Food Science, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing, China
| |
Collapse
|
4
|
Engür-Öztürk S, Dikmen M. Proteasome inhibitor immunotherapy for the epithelial to mesenchymal transition: assessing the A549 lung cancer cell microenvironment and the role of M1, M2a and M2c ‘hydrocortisone-polarised’ macrophages. Mol Biol Rep 2022; 49:4777-4793. [DOI: 10.1007/s11033-022-07329-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/02/2022] [Indexed: 02/06/2023]
|
5
|
Zhang M, Zhao R, Wang D, Wang L, Zhang Q, Wei S, Lu F, Peng W, Wu C. Ginger (Zingiber officinale Rosc.) and its bioactive components are potential resources for health beneficial agents. Phytother Res 2021; 35:711-742. [PMID: 32954562 DOI: 10.1002/ptr.6858] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/17/2020] [Accepted: 08/02/2020] [Indexed: 12/25/2022]
Abstract
Zingiber officinale Rosc. (Zingiberacae), commonly known as ginger, is a perennial and herbaceous plant with long cultivation history. Ginger rhizome is one of the most popular food spices with unique pungent flavor and is prescribed as a well-known traditional Chinese herbal medicine. To date, over 160 constituents, including volatile oil, gingerol analogues, diarylheptanoids, phenylalkanoids, sulfonates, steroids, and monoterpenoid glycosides compounds, have been isolated and identified from ginger. Increasing evidence has revealed that ginger possesses a broad range of biological activities, especially gastrointestinal-protective, anti-cancer, and obesity-preventive effects. In addition, gingerol analogues such as 6-gingerol and 6-shogaol can be rapidly eliminated in the serum and detected as glucuronide and sulfate conjugates. Structural variation would be useful to improve the metabolic characteristics and bioactivities of lead compounds derived from ginger. Furthermore, some clinical trials have indicated that ginger can be consumed for attenuating nausea and vomiting during early pregnancy; however, there is not sufficient data available to rule out its potential toxicity, which should be monitored especially over longer periods. This review provides an up-to-date understanding of the scientific evidence on the development of ginger and its active compounds as health beneficial agents in future clinical trials.
Collapse
Affiliation(s)
- Mengmeng Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Zhao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shujun Wei
- Basic Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
Mathiyazhagan J, Kodiveri Muthukaliannan G. Combined Zingiber officinale and Terminalia chebula Induces Apoptosis and Modulates mTOR and hTERT Gene Expressions in MCF-7 Cell Line. Nutr Cancer 2020; 73:1207-1216. [PMID: 32664754 DOI: 10.1080/01635581.2020.1792518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022]
Abstract
In this study, we evaluated the cytotoxicity and apoptotic activity of Zingiber officinale (ZO), Terminalia chebula (TC) alone, and in combination (ZO:TC-1:4). The presence of major bioactive compounds in ZO (6-gingerol and 6-shogaol) and TC (gallic acid, ellagic acid, and chebulinic acid) were evaluated by high performance liquid chromatography. The IC50 values of ZO, TC, and ZOTC (1:4) was estimated to be 88.5, 108.5, and 53.5 μg/mL, respectively. The cell death and cytomorphology changes upon treatment were observed. At these concentrations, ZO, TC, and ZOTC showed reduced mitochondrial membrane potential, increased reactive oxygen species, and apoptotic activities. It was also reported to downregulate mTOR and hTERT gene expression levels which are the primary genes for cell proliferation and growth. This first report on ZOTC combination has the potential to develop as a therapeutic agent for breast cancer.
Collapse
Affiliation(s)
- Jayasindu Mathiyazhagan
- School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | | |
Collapse
|
7
|
NF-κB interaction long non-coding RNA inhibits migration, invasion and epithelial-mesenchymal transition of cervical cancer cells through inhibiting NF-κB signaling pathways. Exp Ther Med 2020; 20:1039-1047. [PMID: 32765657 PMCID: PMC7388573 DOI: 10.3892/etm.2020.8752] [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: 06/18/2019] [Accepted: 03/17/2020] [Indexed: 12/17/2022] Open
Abstract
The long non-coding RNA (lncRNA) NF-κB interaction lncRNA (NKILA) has been found to exert tumor suppressive effects in numerous types of carcinoma; however, the relationship between NKILA and cervical cancer (CC) remains largely unclear. The present study aimed to investigate the effects of NKILA on the proliferation and metastasis of CC cell lines, in addition to the related molecular mechanisms. Reverse transcription-quantitative PCR was used to detect the expression levels of NKILA in cancer tissues and cell lines. The constructed overexpression vector, pcDNA3.1NKILA, and its corresponding negative control sequence were transfected into CaSki cells and short hairpin RNA targeting NKILA and the corresponding negative control sequence were transfected into C-33A cells. Subsequently, the proliferative, migratory and invasive ability, as well as the process of epithelial-mesenchymal transition (EMT) of C-33 A and CaSki cells were analyzed by performing Cell Counting Kit-8, wound healing, Matrigel invasion and western blot assays, respectively. The expression levels of proteins were detected using western blot analysis. The expression levels of NKILA were decreased in CC tissues and CC cell lines (SiHa, C-33A, CaSki and HeLa) and the downregulation of NKILA expression using shRNA was observed to significantly increase the proliferation of CC cells. Conversely, the upregulation of NKILA inhibited the proliferation of CC cells, in addition to significantly inhibiting the migration and invasion of CaSki cells, whereas the knockdown of NKILA promoted the invasion of C-33A cells. Thus, it was hypothesized that NKILA may inhibit the migration and invasion of CC cells via regulation of EMT processes, which was reflected by the expression of ZO-1, E-cadherin, N-cadherin and Vimentin. Furthermore, the overexpression of NKILA significantly inhibited the activation of NF-κB in CaSki cells, whereas the knockdown of NKILA expression promoted the degradation of inhibitory protein-κB and promoted the transfer of p65 into the nucleus in C-33A cells. In conclusion, the results from the present study suggested that NKILA may be involved in the inhibition of migration and invasion in CC cells through regulating EMT processes, which may be related to its inhibition of NF-κB activation.
Collapse
|
8
|
Chen CY, Kao CL, Yeh HC, Song PL, Li HT, Li WJ. A New Norsesquiterpenoid from the Rhizomes of Curcuma longa. Chem Nat Compd 2020. [DOI: 10.1007/s10600-020-02947-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
9
|
Wong KM, Song J, Saini V, Wong YH. Small Molecules as Drugs to Upregulate Metastasis Suppressors in Cancer Cells. Curr Med Chem 2019; 26:5876-5899. [PMID: 29788870 DOI: 10.2174/0929867325666180522090842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/20/2018] [Accepted: 05/18/2018] [Indexed: 12/26/2022]
Abstract
It is well-recognized that the majority of cancer-related deaths is attributed to metastasis, which can arise from virtually any type of tumor. Metastasis is a complex multistep process wherein cancer cells must break away from the primary tumor, intravasate into the circulatory or lymphatic systems, extravasate, proliferate and eventually colonize secondary sites. Since these molecular processes involve the coordinated actions of numerous proteins, targeted disruptions of key players along these pathways represent possible therapeutic interventions to impede metastasis formation and reduce cancer mortality. A diverse group of proteins with demonstrated ability to inhibit metastatic colonization have been identified and they are collectively known as metastasis suppressors. Given that the metastasis suppressors are often downregulated in tumors, drug-induced re-expression or upregulation of these proteins represents a promising approach to limit metastasis. Indeed, over 40 compounds are known to exhibit efficacy in upregulating the expression of metastasis suppressors via transcriptional or post-transcriptional mechanisms, and the most promising ones are being evaluated for their translational potentials. These small molecules range from natural products to drugs in clinical use and they apparently target different molecular pathways, reflecting the diverse nature of the metastasis suppressors. In this review, we provide an overview of the different classes of compounds known to possess the ability to upregulate one or more metastasis suppressors, with an emphasis on their mechanisms of action and therapeutic potentials.
Collapse
Affiliation(s)
- Ka Ming Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Jiaxing Song
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Vasu Saini
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Yung H Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.,State Key Laboratory of Molecular Neuroscience, and the Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
10
|
Lechner JF, Stoner GD. Gingers and Their Purified Components as Cancer Chemopreventative Agents. Molecules 2019; 24:E2859. [PMID: 31394732 PMCID: PMC6719158 DOI: 10.3390/molecules24162859] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 07/29/2019] [Accepted: 08/05/2019] [Indexed: 12/17/2022] Open
Abstract
Chemoprevention by ingested substituents is the process through which nutraceuticals and/or their bioactive components antagonize carcinogenesis. Carcinogenesis is the course of action whereby a normal cell is transformed into a neoplastic cell. This latter action involves several steps, starting with initiation and followed by promotion and progression. Driving these stages is continued oxidative stress and inflammation, which in turn, causes a myriad of aberrant gene expressions and mutations within the transforming cell population and abnormal gene expressions by the cells within the surrounding lesion. Chemoprevention of cancer with bioreactive foods or their extracted/purified components occurs primarily via normalizing these inappropriate gene activities. Various foods/agents have been shown to affect different gene expressions. In this review, we discuss how the chemoprevention activities of gingers antagonize cancer development.
Collapse
Affiliation(s)
- John F Lechner
- Retired from Department of Medicine, Division of Medical Oncology, Ohio State University, Columbus 43210, OH, USA.
| | - Gary D Stoner
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| |
Collapse
|
11
|
Almatroudi A, Alsahli MA, Alrumaihi F, Allemailem KS, Rahmani AH. Ginger: A Novel Strategy to Battle Cancer through Modulating Cell Signalling Pathways: A Review. Curr Pharm Biotechnol 2019; 20:5-16. [PMID: 30659535 DOI: 10.2174/1389201020666190119142331] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/17/2018] [Accepted: 01/07/2019] [Indexed: 12/24/2022]
Abstract
Numerous studies have been performed in understanding the development of cancer. Though, the mechanism of action of genes in the development of cancer remains to be explained. The current mode of treatment of cancer shows adverse effects on normal cells and also alter the cell signalling pathways. However, ginger and its active compound have fascinated research based on animal model and laboratories during the past decade due to its potentiality in killing cancer cells. Ginger is a mixture of various compounds including gingerol, paradol, zingiberene and shogaol and such compounds are the main players in diseases management. Most of the health-promoting effects of ginger and its active compound can be attributed due to its antioxidant and anti-tumour activity. Besides, the active compound of ginger has proven its role in cancer management through its modulatory effect on tumour suppressor genes, cell cycle, apoptosis, transcription factors, angiogenesis and growth factor. In this review, the role of ginger and its active compound in the inhibition of cancer growth through modulating cell signalling pathways will be reviewed and discussed.
Collapse
Affiliation(s)
- Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Arshad H Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| |
Collapse
|
12
|
Das B, Sarkar N, Bishayee A, Sinha D. Dietary phytochemicals in the regulation of epithelial to mesenchymal transition and associated enzymes: A promising anticancer therapeutic approach. Semin Cancer Biol 2018; 56:196-218. [PMID: 30472212 DOI: 10.1016/j.semcancer.2018.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 01/06/2023]
Abstract
Epithelial to mesenchymal transition (EMT) is a biological phenomenon that plays a primordial role for initiation of metastasis. It renders cancer cells with increased self-renewal and tumor-initiating capabilities and exacerbated resistance to apoptosis and chemotherapy. Hence, regulation of EMT stands out to be an important strategy in controlling the behavior of malignant cells. Despite the enormous amount of preclinical data on the implication of EMT in cancer progression, there is still lack of routine clinical translation at therapeutic levels. The need of EMT-modulating drugs with high efficacy and low cytotoxicity has led to studies involving the evaluation of the efficacy of a plethora of various classes of phytochemicals present in dietary sources of fruits and vegetables. This review summarizes the role of these different classes of phytochemicals, their natural/synthetic analogs, and their nano-formulations in regulation of EMT in various preclinical models through attenuation of primary signaling pathways. Numerous proteins, transcription factors and enzymes targeted by various classes of phytochemicals in repression of EMT has been presented in this review. Additionally, we have critically analyzed the existing literature and provided views on new direction for accelerating the discovery of novel drug candidates which could be cautiously administered without concomitant effects.
Collapse
Affiliation(s)
- Bornita Das
- Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, West Bengal, India
| | - Nivedita Sarkar
- Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, West Bengal, India
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| | - Dona Sinha
- Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, West Bengal, India.
| |
Collapse
|
13
|
Huang GC, Kao CL, Li WJ, Huang ST, Li HT, Chen CY. A New Phenylalkanoid from the Rhizomes of Alpinia galanga. Chem Nat Compd 2018. [DOI: 10.1007/s10600-018-2558-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
14
|
RKIP: A Key Regulator in Tumor Metastasis Initiation and Resistance to Apoptosis: Therapeutic Targeting and Impact. Cancers (Basel) 2018; 10:cancers10090287. [PMID: 30149591 PMCID: PMC6162400 DOI: 10.3390/cancers10090287] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/12/2018] [Accepted: 08/18/2018] [Indexed: 02/07/2023] Open
Abstract
RAF-kinase inhibitor protein (RKIP) is a well-established tumor suppressor that is frequently downregulated in a plethora of solid and hematological malignancies. RKIP exerts antimetastatic and pro-apoptotic properties in cancer cells, via modulation of signaling pathways and gene products involved in tumor survival and spread. Here we review the contribution of RKIP in the regulation of early metastatic steps such as epithelial–mesenchymal transition (EMT), migration, and invasion, as well as in tumor sensitivity to conventional therapeutics and immuno-mediated cytotoxicity. We further provide updated justification for targeting RKIP as a strategy to overcome tumor chemo/immuno-resistance and suppress metastasis, through the use of agents able to modulate RKIP expression in cancer cells.
Collapse
|
15
|
Jung MY, Lee MK, Park HJ, Oh EB, Shin JY, Park JS, Jung SY, Oh JH, Choi DS. Heat-induced conversion of gingerols to shogaols in ginger as affected by heat type (dry or moist heat), sample type (fresh or dried), temperature and time. Food Sci Biotechnol 2018; 27:687-693. [PMID: 30263794 PMCID: PMC6049668 DOI: 10.1007/s10068-017-0301-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 11/25/2022] Open
Abstract
The impact of heat type, sample type, temperature and time on the heat-induced conversion of gingerols to shogaols in ginger were studied by an UHPLC-ESI-MS/MS. Heat treatments greatly induced the conversion of gingerols to shogaols in ginger. As the temperature increased, the faster conversion of gingerols into shogaols were observed. However, the efficiency of the heat-induced conversion differed greatly with the heat types. Moist heat treatment induced significantly higher quantity of shogaols than dry heat treatment. The moist heat treatment at 120 °C for 360 min induced the highest conversion, reaching to 2991 mg 6-shogaol per kg ginger. In addition, dry-heat induced conversion was affected by the sample type. The dry-heat treatment on dried powder induced significantly higher quantity of shogaols than that on sliced fresh ginger. This represents the first systematic comparative study on the heat and sample types on the heat-induced conversion of gingerols into shogaols in ginger.
Collapse
Affiliation(s)
- Mun Yhung Jung
- Department of Food and Biotechnology, Graduate School, Woosuk University, Samnye-Eup, Wanju-Gun, Jeonbuk Province 55338 Republic of Korea
| | - Min Kyoung Lee
- Department of Food and Biotechnology, Graduate School, Woosuk University, Samnye-Eup, Wanju-Gun, Jeonbuk Province 55338 Republic of Korea
| | - Hee Jeong Park
- Department of Food and Biotechnology, Graduate School, Woosuk University, Samnye-Eup, Wanju-Gun, Jeonbuk Province 55338 Republic of Korea
| | - Eun-Bi Oh
- Department of Food and Biotechnology, Graduate School, Woosuk University, Samnye-Eup, Wanju-Gun, Jeonbuk Province 55338 Republic of Korea
| | - Je Young Shin
- Department of Food and Biotechnology, Graduate School, Woosuk University, Samnye-Eup, Wanju-Gun, Jeonbuk Province 55338 Republic of Korea
| | - Ji Su Park
- Department of Food and Biotechnology, Graduate School, Woosuk University, Samnye-Eup, Wanju-Gun, Jeonbuk Province 55338 Republic of Korea
- Food Analysis Center, Korea Food Research Institute, Wanju-Gun, Jeonbuk Province 55365 Republic of Korea
| | - Su Young Jung
- Healthy Local Food Branding Agency, Wanju, Jeonbuk Republic of Korea
| | - Jung-Hee Oh
- Healthy Local Food Branding Agency, Wanju, Jeonbuk Republic of Korea
| | - Dong-Seong Choi
- Department of Food and Biotechnology, Graduate School, Woosuk University, Samnye-Eup, Wanju-Gun, Jeonbuk Province 55338 Republic of Korea
| |
Collapse
|
16
|
Wang HM, Kao CL, Li WJ, Li HT, Chen CY. Two New Phenylalkanoids from the Rhizomes of Zingiber officinale. Chem Nat Compd 2018. [DOI: 10.1007/s10600-018-2246-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
17
|
Inverse correlation between the metastasis suppressor RKIP and the metastasis inducer YY1: Contrasting roles in the regulation of chemo/immuno-resistance in cancer. Drug Resist Updat 2017; 30:28-38. [PMID: 28363333 DOI: 10.1016/j.drup.2017.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/28/2016] [Accepted: 01/04/2017] [Indexed: 02/06/2023]
Abstract
Several gene products have been postulated to mediate inherent and/or acquired anticancer drug resistance and tumor metastasis. Among these, the metastasis suppressor and chemo-immuno-sensitizing gene product, Raf Kinase Inhibitor Protein (RKIP), is poorly expressed in many cancers. In contrast, the metastasis inducer and chemo-immuno-resistant factor Yin Yang 1 (YY1) is overexpressed in many cancers. This inverse relationship between RKIP and YY1 expression suggests that these two gene products may be regulated via cross-talks of molecular signaling pathways, culminating in the expression of different phenotypes based on their targets. Analyses of the molecular regulation of the expression patterns of RKIP and YY1 as well as epigenetic, post-transcriptional, and post-translational regulation revealed the existence of several effector mechanisms and crosstalk pathways, of which five pathways of relevance have been identified and analyzed. The five examined cross-talk pathways include the following loops: RKIP/NF-κB/Snail/YY1, p38/MAPK/RKIP/GSK3β/Snail/YY1, RKIP/Smurf2/YY1/Snail, RKIP/MAPK/Myc/Let-7/HMGA2/Snail/YY1, as well as RKIP/GPCR/STAT3/miR-34/YY1. Each loop is comprised of multiple interactions and cascades that provide evidence for YY1's negative regulation of RKIP expression and vice versa. These loops elucidate potential prognostic motifs and targets for therapeutic intervention. Chiefly, these findings suggest that targeted inhibition of YY1 by specific small molecule inhibitors and/or the specific induction of RKIP expression and activity are potential therapeutic strategies to block tumor growth and metastasis in many cancers, as well as to overcome anticancer drug resistance. These strategies present potential alternatives for their synergistic uses in combination with low doses of conventional chemo-immunotherapeutics and hence, increasing survival, reducing toxicity, and improving quality of life.
Collapse
|
18
|
Subramani R, Lakshmanaswamy R. Complementary and Alternative Medicine and Breast Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 151:231-274. [DOI: 10.1016/bs.pmbts.2017.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
19
|
Park SY, Jung MY. UHPLC-ESI-MS/MS for the Quantification of Eight Major Gingerols and Shogaols in Ginger Products: Effects of Ionization Polarity and Mobile Phase Modifier on the Sensitivity. J Food Sci 2016; 81:C2457-C2465. [PMID: 27589022 DOI: 10.1111/1750-3841.13429] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Revised: 07/15/2016] [Accepted: 07/27/2016] [Indexed: 11/30/2022]
Abstract
We developed and validated an improved ultra high performance liquid chromatography (UHPLC)-electrospray ionization (ESI)-tandem mass spectrometry (MS/MS) method for the fast separation and quantification of 8 major gingerols and shogaols (4-, 6-, 8-, 10-, and 12-gingerols, and 6-, 8-, and 10-shogaols) in gingerol products. The ionization polarity and mobile phase modifier greatly affected the mass ion profiles and sensitivity of the analytes. A mobile phase modifier of 0.05 mM ammonium formate dramatically decreased the sodium adduct ions and greatly increased the protonated ions of the gingerols and shogaols. Positive ion UHPLC-MS/MS with the ammonium formate in mobile phase showed greatly higher (4.5- to 15.7-fold) sensitivities than negative ion UHPLC. The positive ion UHPLC-MS/MS method showed excellent linearities (r2 > 0.999), low limits of detection (LOD = 2.5 to 8.2 pg), high accuracy and precision, and no considerable matrix effect. Baseline separation of the 8 target compounds was achieved in 1 min by the UHPLC with a short C18 core-shell column. The method was successfully applied to analyze the compounds in fresh and dried powdered gingers, and dietary supplements. The total contents of the major compounds in the fresh gingers, dried powdered gingers, and dietary supplements were in the range of 1114 to 1478, 4380 to 11324, and 2915 to 29998 mg/kg, respectively. There was about 10-fold variation of the content in the dietary supplements with the commercial brands. This represents the 1st report on the ionization polarity and mobile phase modifier on the sensitivity of gingerols and shogaols in LC-mass spectrometry. Furthermore, the established method provided great improvement in chromatographic separation of the target gingerols.
Collapse
Affiliation(s)
- Su Yeon Park
- Dept. of Food and Biotechnology, Graduate School, College of Food Science, Woosuk Univ, Samnye-eup, Wanju-gun, Jeonbuk Province, 55338, Republic of Korea
| | - Mun Yhung Jung
- Dept. of Food and Biotechnology, Graduate School, College of Food Science, Woosuk Univ, Samnye-eup, Wanju-gun, Jeonbuk Province, 55338, Republic of Korea.
| |
Collapse
|
20
|
Marvibaigi M, Amini N, Supriyanto E, Abdul Majid FA, Kumar Jaganathan S, Jamil S, Hamzehalipour Almaki J, Nasiri R. Antioxidant Activity and ROS-Dependent Apoptotic Effect of Scurrula ferruginea (Jack) Danser Methanol Extract in Human Breast Cancer Cell MDA-MB-231. PLoS One 2016; 11:e0158942. [PMID: 27410459 PMCID: PMC4943642 DOI: 10.1371/journal.pone.0158942] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/24/2016] [Indexed: 01/04/2023] Open
Abstract
Scurrula ferruginea (Jack) Danser is one of the mistletoe species belonging to Loranthaceae family, which grows on the branches of many deciduous trees in tropical countries. This study evaluated the antioxidant activities of S. ferruginea extracts. The cytotoxic activity of the selected extracts, which showed potent antioxidant activities, and high phenolic and flavonoid contents, were investigated in human breast cancer cell line (MDA-MB-231) and non-cancer human skin fibroblast cells (HSF-1184). The activities and characteristics varied depending on the different parts of S. ferruginea, solvent polarity, and concentrations of extracts. The stem methanol extract showed the highest amount of both phenolic (273.51 ± 4.84 mg gallic acid/g extract) and flavonoid contents (163.41 ± 4.62 mg catechin/g extract) and strong DPPH• radical scavenging (IC50 = 27.81 μg/mL) and metal chelation activity (IC50 = 80.20 μg/mL). The stem aqueous extract showed the highest ABTS•+ scavenging ability. The stem methanol and aqueous extracts exhibited dose-dependent cytotoxic activity against MDA-MB-231 cells with IC50 of 19.27 and 50.35 μg/mL, respectively. Furthermore, the extracts inhibited the migration and colony formation of MDA-MB-231 cells in a concentration-dependent manner. Morphological observations revealed hallmark properties of apoptosis in treated cells. The methanol extract induced an increase in ROS generation and mitochondrial depolarization in MDA-MB-231 cells, suggesting its potent apoptotic activity. The present study demonstrated that the S. ferruginea methanol extract mediated MDA-MB-231 cell growth inhibition via induction of apoptosis which was confirmed by Western blot analysis. It may be a potential anticancer agent; however, its in vivo anticancer activity needs to be investigated.
Collapse
Affiliation(s)
- Mohsen Marvibaigi
- IJN-UTM Cardiovascular Engineering Center, Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Neda Amini
- IJN-UTM Cardiovascular Engineering Center, Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Eko Supriyanto
- IJN-UTM Cardiovascular Engineering Center, Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Fadzilah Adibah Abdul Majid
- Bioprocess Engineering Department, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu, Malaysia
| | - Saravana Kumar Jaganathan
- IJN-UTM Cardiovascular Engineering Center, Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Shajarahtunnur Jamil
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, UTM Johor Bahru, Johor, Malaysia
| | - Javad Hamzehalipour Almaki
- Bioprocess Engineering Department, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Rozita Nasiri
- Bioprocess Engineering Department, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| |
Collapse
|
21
|
Rajkumar K, Nichita A, Anoor PK, Raju S, Singh SS, Burgula S. Understanding perspectives of signalling mechanisms regulating PEBP1 function. Cell Biochem Funct 2016; 34:394-403. [PMID: 27385268 DOI: 10.1002/cbf.3198] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 05/13/2016] [Accepted: 05/19/2016] [Indexed: 12/21/2022]
Abstract
UNLABELLED Phosphatidylethanolamine-binding protein 1 (PEBP1), also known as Raf kinase inhibitor protein, belongs to PEBP family of proteins. It is known to interact with many proteins that are mainly involved in pathways that monitor cell proliferation and differentiation. PEBP1 in many cells interacts with several pathways, namely MAPK, GRK2, NF-кB, etc. that keeps the cell proliferation and differentiation in check. This protein is expressed by many cells in humans, including neurons where it is predominantly involved in production of choline acetyltransferase. Deregulated PEBP1 is known to cause cancer, diabetic nephropathy and neurodegenerative diseases like Alzheimer's and dementia. Recent research led to the discovery of many drugs that mainly target the interaction of PEBP1 with its partners. These compounds are known to bind PEBP1 in its conserved domain which abrogate its association with interacting partners in several different pathways. We outline here the latest developments in understanding of PEBP1 function in maintaining cell integrity. Copyright © 2016 John Wiley & Sons, Ltd. SIGNIFICANCE OF THE STUDY Phosphatidylethanolamine-binding protein is crucial in regulation of MAPK and PKC pathways. Its diverse roles, including regulating these pathways keep cell differentiation and proliferation in check. This review outlines some latest findings which greatly add to our current knowledge of phosphatidylethanolamine-binding protein.
Collapse
Affiliation(s)
- Karthik Rajkumar
- Department of Microbiology, Osmania University, Hyderabad, India
| | - Aare Nichita
- Department of Microbiology, Osmania University, Hyderabad, India
| | | | - Swathi Raju
- Department of Microbiology, Osmania University, Hyderabad, India
| | | | | |
Collapse
|
22
|
Saxena R, Rida PCG, Kucuk O, Aneja R. Ginger augmented chemotherapy: A novel multitarget nontoxic approach for cancer management. Mol Nutr Food Res 2016; 60:1364-73. [PMID: 26842968 DOI: 10.1002/mnfr.201500955] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/11/2016] [Accepted: 01/12/2016] [Indexed: 01/22/2023]
Abstract
Cancer, referred to as the 'disease of civilization', continues to haunt humanity due to its dreadful manifestations and limited success of therapeutic interventions such as chemotherapy in curing the disease. Although effective, chemotherapy has repeatedly demonstrated inadequacy in disease management due to its debilitating side effects arising from its deleterious nonspecific effects on normal healthy cells. In addition, development of chemoresistance due to mono-targeting often results in cessation of chemotherapy. This urgently demands development and implementation of multitargeted alternative therapies with mild or no side effects. One extremely promising strategy that yet remains untapped in the clinic is augmenting chemotherapy with dietary phytochemicals or extracts. Ginger, depository of numerous bioactive molecules, not only targets cancer cells but can also mitigate chemotherapy-associated side effects. Consequently, combination therapy involving ginger extract and chemotherapeutic agents may offer the advantage of being efficacious with reduced toxicity. Here we discuss the remarkable and often overlooked potential of ginger extract to manage cancer, the possibility of developing ginger-based combinational therapies, and the major roadblocks along with strategies to overcome them in clinical translation of such inventions. We are optimistic that clinical implementation of such combination regimens would be a much sought after modality in cancer management.
Collapse
Affiliation(s)
- Roopali Saxena
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | | | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
23
|
Song J, Feng L, Zhong R, Xia Z, Zhang L, Cui L, Yan H, Jia X, Zhang Z. Icariside II inhibits the EMT of NSCLC cells in inflammatory microenvironment via down-regulation of Akt/NF-κB signaling pathway. Mol Carcinog 2016; 56:36-48. [DOI: 10.1002/mc.22471] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Jie Song
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine; Nanjing University of Chinese Medicine; Jiangsu Nanjing China
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Liang Feng
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Rongling Zhong
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Zhi Xia
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Li Zhang
- Clinical Laboratory; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Li Cui
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Hongmei Yan
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Xiaobin Jia
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine; Nanjing University of Chinese Medicine; Jiangsu Nanjing China
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| | - Zhenhai Zhang
- Key Laboratory of Delivery Systems of Chinese Meteria Medica; Jiangsu Provincial Academy of Chinese Medicine; Jiangsu Nanjing China
| |
Collapse
|
24
|
Park JY, Shin MS, Kim SN, Kim HY, Kim KH, Shin KS, Kang KS. Polysaccharides from Korean Citrus hallabong peels inhibit angiogenesis and breast cancer cell migration. Int J Biol Macromol 2016; 85:522-9. [PMID: 26778161 DOI: 10.1016/j.ijbiomac.2016.01.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/02/2015] [Accepted: 01/04/2016] [Indexed: 01/07/2023]
Abstract
Although the peel of the hallabong (Citrus sphaerocarpa) fruit is rich in polysaccharides, which are valuable dietary ingredients for human health, it is normally wasted. The present study aimed to utilize the peel waste and identify properties it may have against breast cancer metastasis. Hallabong peel extract containing crude polysaccharides was fractionated by gel permeation chromatography to produce four different polysaccharide fractions (HBE-I, -II, -III, and -IV). The HBE polysaccharides significantly blocked tube formation of human umbilical vein vascular endothelial cells (HUVECs), at a concentration of 12.5 or 25 μg/mL. Tube formation appeared to be more sensitive to HBE-II than to other HBE polysaccharides. HBE-II also inhibited breast cancer cell migration, through downregulation of matrix metalloproteinase-9 (MMP-9) in MDA-MB-231 triple-negative breast cancer cells. Therefore, inhibition of tube formation and MMP-9-mediated migration observed in HUVEC and MDA-MB-231 cells, respectively, are likely to be important therapeutic targets in triple-negative breast cancer metastasis.
Collapse
Affiliation(s)
- J Y Park
- College of Korean Medicine, Gachon University, Seongnam 461-701, Republic of Korea
| | - M S Shin
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung 210-340, Republic of Korea; Department of Food Science and Biotechnology, Kyonggi University, Suwon 443-760, Republic of Korea
| | - S N Kim
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung 210-340, Republic of Korea
| | - H Y Kim
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 660-758, Republic of Korea
| | - K H Kim
- Natural Product Research Laboratory, School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Republic of Korea
| | - K S Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 443-760, Republic of Korea.
| | - K S Kang
- College of Korean Medicine, Gachon University, Seongnam 461-701, Republic of Korea.
| |
Collapse
|
25
|
Labbozzetta M, Poma P, Vivona N, Gulino A, D'Alessandro N, Notarbartolo M. Epigenetic changes and nuclear factor-κB activation, but not microRNA-224, downregulate Raf-1 kinase inhibitor protein in triple-negative breast cancer SUM 159 cells. Oncol Lett 2015; 10:3807-3815. [PMID: 26788213 DOI: 10.3892/ol.2015.3787] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 07/07/2015] [Indexed: 01/16/2023] Open
Abstract
Raf-1 kinase inhibitor protein (RKIP) is a tumor suppressor and metastasis inhibitor, which enhances drug-induced apoptosis of cancer cells. Downregulation of RKIP may be significant in the biology of highly aggressive and drug-resistant tumors, for example triple-negative breast cancers (TNBCs). Potential causes for the low levels of RKIP expressed by SUM 159 TNBC cells were investigated in the present study. Bisulphite modification, methylation specific-polymerase chain reaction (PCR) and a TransAM NF-κB assay were performed and the results suggested that various mechanisms, including methylation of the gene promoter, histone deacetylation and nuclear factor-κB (NF-κB) activation, but not targeting by microRNA-224 (miR/miRNA-224), as determined by transfection of pre-miR-224 miRNA precursor or anti-miR-224 miRNA inhibitor, may downregulate RKIP in these cells. Furthermore, reverse transcription-quantitative PCR, western blotting, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphophenyl)-2H-tetrazolium cell growth assay and flow cytometry revealed that in SUM 159 cells, the demethylating agent 5-aza-2'-deoxycytidine (5-AZA), the histone deacetylase inhibitor trichostatin A (TSA) and the NF-κB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) enhanced RKIP expression and resulted in significant cell growth inhibition and induction of apoptosis. 5-AZA and TSA mainly produced additive antitumor effects, while the combination of DHMEQ and TSA exhibited significant synergy in cell growth inhibition and induction of apoptosis assays. Increasing evidence that aberrant activation of NF-κB signaling is a frequent characteristic of TNBC highlights the fact that this transcription factor may be a useful target for treatment of such tumors. In addition to DHMEQ, proteasome inhibitors may also represent valuable therapeutic resources in this context. Notably, proteasome inhibitors, in addition to the inhibition of NF-κB activation, may also restore RKIP levels by inhibiting proteasome degradation of the ubiquitinated protein. The current results contribute to the understanding of the molecular mechanisms of RKIP downregulation in TNBC and suggest possible novel therapeutic approaches for the treatment of these types of cancer.
Collapse
Affiliation(s)
- Manuela Labbozzetta
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care 'G. D'Alessandro', University of Palermo, Palermo 90127, Italy
| | - Paola Poma
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care 'G. D'Alessandro', University of Palermo, Palermo 90127, Italy
| | - Nicoletta Vivona
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care 'G. D'Alessandro', University of Palermo, Palermo 90127, Italy
| | - Alessandro Gulino
- Tumor Immunology Unit, Department of Health Sciences and Mother and Child Care 'G. D'Alessandro', University of Palermo, Palermo 90127, Italy
| | - Natale D'Alessandro
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care 'G. D'Alessandro', University of Palermo, Palermo 90127, Italy
| | - Monica Notarbartolo
- Pharmacology Unit, Department of Health Sciences and Mother and Child Care 'G. D'Alessandro', University of Palermo, Palermo 90127, Italy
| |
Collapse
|
26
|
The biological complexity of RKIP signaling in human cancers. Exp Mol Med 2015; 47:e185. [PMID: 26403261 PMCID: PMC4650930 DOI: 10.1038/emm.2015.70] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 12/17/2022] Open
Abstract
The Raf kinase inhibitory protein (RKIP) has been demonstrated to modulate different intracellular signaling pathways in cancers. Studies have shown that RKIP is frequently downregulated in cancers; therefore, attempts have been made to upregulate the expression of RKIP using natural and synthetic agents for the treatment of human malignancies. Moreover, various regulators such as specific proteins and microRNAs (miRNAs) that are involved in the regulation of RKIP expression have also been identified. RKIP mechanistically modulates the apoptotic regulators of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) signaling. Because of its critical role in human cancers, RKIP has drawn much research attention, and our understanding is expanding rapidly. Here, we summarize some of the biological complexities of RKIP regulation. However, we restrict our discussion to selected tumors by focusing on TRAIL, miRNAs and natural agents. Emerging evidence suggests a role for natural agents in RKIP regulation in cancer cells; therefore, naturally occurring agents may serve as cancer-targeting agents for cancer treatment. Although the literature suggests some advancement in our knowledge of RKIP biology, it is incomplete with regard to its preclinical and clinical efficacy; thus, further research is warranted. Furthermore, the mechanism by which chemotherapeutic drugs and novel compounds modulate RKIP and how nanotechnologically delivered RKIP can be therapeutically exploited remain to be determined.
Collapse
|
27
|
Tang L, Yue B, Cheng Y, Yao H, Ma X, Tian Q, Ge L, Liu Z, Han X. Inhibition of invasion and metastasis by DMBT, a novel trehalose derivative, through Akt/GSK-3β/β-catenin pathway in B16BL6 cells. Chem Biol Interact 2014; 222:7-17. [PMID: 25148938 DOI: 10.1016/j.cbi.2014.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 07/14/2014] [Accepted: 08/11/2014] [Indexed: 12/19/2022]
Abstract
Invasion, either directly or via metastasis formation, is the main cause of death in cancer patients. Development of efficient anti-invasive agents is an important research challenge. 6,6'-bis (2,3-dimethoxybenzoyl)-a, a-d-trehalose (DMBT), one of brartemicin analogs, was found to be the most potent anti-invasive agent, but the underlying mechanisms are poorly understood. Our current study was to explore the effects of DMBT on invasion and metastasis in B16BL6 cells. Antiproliferation assay and trypan blue exclusion assay showed that no obvious inhibitory or cytotoxic effect of DMBT was found in B16BL6 cells. Wound healing demonstrated that DMBT could inhibit cell migration compared with the normal group. Transwell experiments showed that DMBT could significantly inhibit invasion to the reconstituted basement membrane (P<0.01). We examined the effects of lung metastasis produced by highly metastatic B16BL6 melanoma cells by using experimental metastasis models and BLI analysis. DMBT could significantly suppress lung metastasis in mice. Results from immunohistochemical staining, Western blotting and real-time PCR indicated that the chemopreventive effect of DMBT was attributed to the inhibition of the VEGF and MMP-9 through Akt/GSK-3β/β-catenin and Akt/mTOR signaling pathways. These results suggested that DMBT could be a promising lead molecule for the anti-metastasis and serve as a therapeutic agent to inhibit cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Linlin Tang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Bin Yue
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Yanna Cheng
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China; Key Laboratory of Chemical Biology of Natural Products, Ministry of Education, China
| | - Hong Yao
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Xiaowen Ma
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Qi Tian
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Lianping Ge
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Zhaopeng Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China; Key Laboratory of Chemical Biology of Natural Products, Ministry of Education, China
| | - Xiuzhen Han
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, China; Key Laboratory of Chemical Biology of Natural Products, Ministry of Education, China.
| |
Collapse
|
28
|
Liu BY, Huang JA, Liu SQ, Li SY, Xu CY, Liang MZ, Tan L, Qin MB. Role of NF-κB in induction of epithelial-mesenchymal transition, migration and invasion of human colon cancer cells. Shijie Huaren Xiaohua Zazhi 2014; 22:3403-3409. [DOI: 10.11569/wcjd.v22.i23.3403] [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 role of nuclear factor-kappa B (NF-κB) in the induction of epithelial-mesenchymal transition (EMT), migration and invasion of human colon cancer cell line HCT116.
METHODS: HCT116 cells were divided into three groups and treated with 20 ng/mL of tumor necrosis factor-α (TNF-α) (NF-κB activation group), 20 μmol/L of ammonium pyrrolidinedithiocarbamate (PDTC) (NF-κB suppression group), and equal volume of culture medium (control group), respectively. Four days later, cell morphological changes associated with EMT were observed under a phase contrast microscope, and the migration ability and invasiveness were assessed by Transwell chamber assays. The protein expression of p65, P-p65, E-cadherin and N-cadherin was analyzed by Western blot, and the mRNA expression of E-cadherin and N-cadherin was detected by quantitative real-time PCR.
RESULTS: TNF-α up-regulated the expression of P-p65 and N-cadherin, suppressed the expression of E-cadherin, and caused a complete EMT-phenotype, which manifested as the formation of large filopodia-like processes and spindle-cell shape. Also, TNF-α promoted cell invasion and migration. In contrast, PDTC down-regulated the expression of P-p65 and N-cadherin, up-regulated the expression of E-cadherin, inhibited the occurrence of typical EMT phenomenon, and suppressed cell invasion and migration. Compared with the control group, TNF-α group and PDTC group showed significantly different cell invasion (97.75 ± 3.77 vs 118.50 ± 1.95, 51.00 ± 1.83, P < 0.05 for both), cell migration (140.00 ± 4.32 vs 167.00 ± 6.36, 80.00 ± 2.53, P < 0.05 for both), and mRNA expression of N-cadherin (1.00 ± 0.00 vs 3.90 ± 0.47, 0.08 ± 0.02, P < 0.05 for both) and E-cadherin (1.00 ± 0.00 vs 0.26 ± 0.08, 6.03 ± 0.59, P < 0.05 for both).
CONCLUSION: NF-κB induces the occurrence of typical EMT phenomenon and promotes cell invasion and migration in human colon cancer cell line HCT116.
Collapse
|
29
|
Li DX, Cai HY, Wang X, Feng YL, Cai SW. Promoter methylation of Raf kinase inhibitory protein: A significant prognostic indicator for patients with gastric adenocarcinoma. Exp Ther Med 2014; 8:844-850. [PMID: 25120612 PMCID: PMC4113522 DOI: 10.3892/etm.2014.1833] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 06/20/2014] [Indexed: 12/23/2022] Open
Abstract
DNA methylation has an important role in the development of carcinomas. As a metastasis suppressor gene, Raf kinase inhibitory protein (RKIP) suppresses tumor cell invasion and metastasis. In the present study, the associations between RKIP protein expression and promoter methylation with clinicopathological parameters, prognosis and survival rates in gastric adenocarcinoma were investigated. RKIP protein expression and promoter methylation were measured in 135 cases of surgically resected gastric adenocarcinoma specimens and corresponding normal tissues using immunohistochemistry and methylation-specific polymerase chain reaction, respectively. Kaplan-Meier analyses were performed to analyze the patient survival rate. Prognostic factors were determined using multivariate Cox analysis. RKIP promoter methylation was detected in 48.9% of gastric carcinoma tissues and 5.17% of adjacent tissues (P<0.05). RKIP protein expression was detected in 43.0% of gastric carcinoma tissues and 91.1% of adjacent tissues (P<0.05). The protein expression levels and promoter methylation of RKIP were shown to correlate with pathological staging, Union for International Cancer Control-stage, tumor differentiation and lymph node metastasis (P<0.05). In addition, the protein expression of RKIP in gastric carcinomas was demonstrated to be associated with promoter methylation of RKIP. Survival analysis of gastric carcinoma patients revealed that promoter methylation in RKIP-positive tumors correlated with a significantly shorter survival time when compared with RKIP-negative tumors (P=0.0002, using the log-rank test). Using multivariate Cox analysis, promoter methylation of RKIP was shown to be an independent prognostic factor (P=0.033). These results indicated that abnormal promoter methylation of RKIP may be one cause of downregulated RKIP expression. Downregulation of RKIP expression was shown to correlate with the incidence and development of gastric carcinomas. Thus, abnormal promoter methylation of RKIP may be a valuable biomarker for estimating gastric carcinoma prognosis.
Collapse
Affiliation(s)
- Dong-Xia Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Hai-Yang Cai
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xia Wang
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| | - Yan-Ling Feng
- Department of Pathology, Public Health Clinical Center of Fudan University, Shanghai 201508, P.R. China
| | - Song-Wang Cai
- Department of Cardiothoracic Surgery, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
30
|
Sun CC, Xu HM, Yuan Y, Gao ZH, Lou HX, Qu XJ. Riccardin D, a Macrocyclic Bisbibenzy, Inhibits Human Breast Cancer Growth through the Suppression of Telomerase Activity. Basic Clin Pharmacol Toxicol 2014; 115:488-98. [DOI: 10.1111/bcpt.12267] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 05/02/2014] [Indexed: 12/28/2022]
Affiliation(s)
- Cui-Cui Sun
- Department of Pharmacology; Key Laboratory of Chemical Biology; School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Hui-Min Xu
- Faculty of Radiologic Sciences; School of Medicine; Qingdao University; Qingdao China
| | - Yi Yuan
- Department of Pharmacology; Key Laboratory of Chemical Biology; School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Zu-Hua Gao
- Department of Pharmacology; School of Chemical Biology & Pharmaceutical Sciences; Capital Medical University; Beijing China
- Department of Pathology; McGill University; Montreal QC Canada
| | - Hong-Xiang Lou
- Department of Pharmacology; Key Laboratory of Chemical Biology; School of Pharmaceutical Sciences; Shandong University; Jinan China
| | - Xian-Jun Qu
- Department of Pharmacology; Key Laboratory of Chemical Biology; School of Pharmaceutical Sciences; Shandong University; Jinan China
- Department of Pharmacology; School of Chemical Biology & Pharmaceutical Sciences; Capital Medical University; Beijing China
| |
Collapse
|
31
|
Linalool exhibits cytotoxic effects by activating antitumor immunity. Molecules 2014; 19:6694-706. [PMID: 24858101 PMCID: PMC6271996 DOI: 10.3390/molecules19056694] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/05/2014] [Accepted: 05/19/2014] [Indexed: 12/18/2022] Open
Abstract
According to recent studies, the Plantaginaceae, which are traditional Chinese herbal remedies, have potential for use in viral infection treatment and cancer therapy. Linalool and p-coumaric acid are two of the biologically active compounds that can be isolated from the Plantaginaceae. This study mainly focused on investigating the bioactivity of linalool as well as the bioactivity of p-coumaric acid in terms of their cytotoxic effects on cancer cells. Whether the mechanisms of such effects are generated through apoptosis and immunoregulatory activity were also investigated. By using WST-1 analysis, it was shown that linalool and p-coumaric acid have good inhibitory effects against breast, colorectal and liver cancer cells. The IC50 values of linalool for those cancer cell types were 224 μM, 222 μM, and 290 μM, respectively, and the IC50 values of p-coumaric acid were 693 μM, 215 μM and 87 μM, respectively. Cell cycle analysis also confirmed that linalool and p-coumaric acid can lead to apoptosis. By using flow cytometry, it was determined that treatment with linalool rather than p-coumaric acid significantly increased the sub-G1 phase and that there were more cells concentrated in the G1 phase. Furthermore, by using cytokine array analysis, we found that linalool can stimulate IFN-γ, IL-13, IL-2, IL-21, IL-21R, IL-4, IL-6sR and TNF-α secretion. This demonstrated that in addition to the bidirectional regulation capabilities found in linalool, it also induces Th1 cellular immune response in T-47D cells. These results showed that linalool holds great potential for use in cancer therapy, and we believe that it could provide an alternative way to take action against tumors.
Collapse
|
32
|
Gemifloxacin, a fluoroquinolone antimicrobial drug, inhibits migration and invasion of human colon cancer cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:159786. [PMID: 24386633 PMCID: PMC3872387 DOI: 10.1155/2013/159786] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/16/2013] [Accepted: 11/18/2013] [Indexed: 02/07/2023]
Abstract
Gemifloxacin (GMF) is an orally administered broad-spectrum fluoroquinolone antimicrobial agent used to treat acute bacterial exacerbation of pneumonia and bronchitis. Although fluoroquinolone antibiotics have also been found to have anti-inflammatory and anticancer effects, studies on the effect of GMF on treating colon cancer have been relatively rare. To the best of our knowledge, this is the first report to describe the antimetastasis activities of GMF in colon cancer and the possible mechanisms involved. Results have shown that GMF inhibits the migration and invasion of colon cancer SW620 and LoVo cells and causes epithelial mesenchymal transition (EMT). In addition, GMF suppresses the activation of NF-κB and cell migration and invasion induced by TNF-α and inhibits the TAK1/TAB2 interaction, resulting in decreased IκB phosphorylation and NF-κB nuclear translocation in SW620 cells. Furthermore, Snail, a critical transcriptional factor of EMT, was downregulated after GMF treatment. Overexpression of Snail by cDNA transfection significantly decreases the inhibitory effect of GMF on EMT and cell migration and invasion. In conclusion, GMF may be a novel anticancer agent for the treatment of metastasis in colon cancer.
Collapse
|
33
|
Wu GS, Song YL, Yin ZQ, Guo JJ, Wang SP, Zhao WW, Chen XP, Zhang QW, Lu JJ, Wang YT. Ganoderiol A-enriched extract suppresses migration and adhesion of MDA-MB-231 cells by inhibiting FAK-SRC-paxillin cascade pathway. PLoS One 2013; 8:e76620. [PMID: 24204647 PMCID: PMC3812178 DOI: 10.1371/journal.pone.0076620] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 08/27/2013] [Indexed: 12/14/2022] Open
Abstract
Cell adhesion, migration and invasion are critical steps for carcinogenesis and cancer metastasis. Ganoderma lucidum, also called Lingzhi in China, is a traditional Chinese medicine, which exhibits anti-proliferation, anti-inflammation and anti-metastasis properties. Herein, GAEE, G. lucidum extract mainly contains ganoderiol A (GA), dihydrogenated GA and GA isomer, was shown to inhibit the abilities of adhesion and migration, while have a slight influence on that of invasion in highly metastatic breast cancer MDA-MB-231 cells at non-toxic doses. Further investigation revealed that GAEE decreased the active forms of focal adhesion kinase (FAK) and disrupted the interaction between FAK and SRC, which lead to deactivating of paxillin. Moreover, GAEE treatment downregulated the expressions of RhoA, Rac1, and Cdc42, and decreased the interaction between neural Wiskott-Aldrich Syndrome protein (N-WASP) and Cdc42, which impair cell migration and actin assembly. To our knowledge, this is the first report to show that G.lucidum triterpenoids could suppress cell migration and adhesion through FAK-SRC-paxillin signaling pathway. Our study also suggests that GAEE may be a potential agent for treatment of breast cancer.
Collapse
Affiliation(s)
- Guo-Sheng Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Al-Mulla F, Bitar MS, Taqi Z, Yeung KC. RKIP: much more than Raf kinase inhibitory protein. J Cell Physiol 2013; 228:1688-702. [PMID: 23359513 DOI: 10.1002/jcp.24335] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/16/2013] [Indexed: 12/11/2022]
Abstract
From its discovery as a phosphatidylethanolamine-binding protein in bovine brain to its designation as a physiological inhibitor of Raf kinase protein, RKIP has emerged as a critical molecule for maintaining subdued, well-orchestrated cellular responses to stimuli. The disruption of RKIP in a wide range of pathologies, including cancer, Alzheimer's disease, and pancreatitis, makes it an exciting target for individualized therapy and disease-specific interventions. This review attempts to highlight recent advances in the RKIP field underscoring its potential role as a master modulator of many pivotal intracellular signaling cascades that control cellular growth, motility, apoptosis, genomic integrity, and therapeutic resistance. Specific biological and functional niches are highlighted to focus future research towards an enhanced understanding of the multiple roles of RKIP in health and disease.
Collapse
Affiliation(s)
- Fahd Al-Mulla
- Faculty of Medicine, Department of Pathology, Kuwait University Health Sciences Centre, Safat, Kuwait.
| | | | | | | |
Collapse
|
36
|
Inhibition of angiogenesis and invasion by DMBT is mediated by downregulation of VEGF and MMP-9 through Akt pathway in MDA-MB-231 breast cancer cells. Food Chem Toxicol 2013; 56:204-13. [DOI: 10.1016/j.fct.2013.02.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 02/06/2013] [Accepted: 02/14/2013] [Indexed: 01/28/2023]
|
37
|
Arai Y, Iinuma H, Ikeda Y, Igarashi M, Hatano M, Kinoshita N, Ukaji T, Simizu S, Umezawa K. Migracins A and B, new inhibitors of cancer cell migration, produced by Streptomyces sp. J Antibiot (Tokyo) 2012; 66:225-30. [DOI: 10.1038/ja.2012.112] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Hu S, Luo Q, Cun B, Hu D, Ge S, Fan X, Chen F. The pharmacological NF-κB inhibitor BAY11-7082 induces cell apoptosis and inhibits the migration of human uveal melanoma cells. Int J Mol Sci 2012; 13:15653-67. [PMID: 23443086 PMCID: PMC3546654 DOI: 10.3390/ijms131215653] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 11/12/2012] [Accepted: 11/19/2012] [Indexed: 01/27/2023] Open
Abstract
Uveal melanomas are highly metastatic and have high rate of recurrence due to the lack of effective systemic therapy. The identification of important survival pathways in uveal melanomas provides novel therapeutic targets for effective treatment. In the present study, we found that the NF-κB signaling pathway was constitutively and highly activated in uveal melanoma cells. Treatment with the pharmacological NF-κB specific inhibitor BAY11-7082 markedly decreased the nuclear translocation of NF-κB. In a dose-dependent setting, BAY11-7082 inhibited the proliferation and growth of uveal melanoma cells by inducing apoptosis without effect on cell cycle. The migration capacity of uveal melanoma cells was also significantly suppressed by BAY11-7082 treatment. Mechanistically, BAY11-7082 increased the activity of caspase 3 and reduced the expression of anti-apoptotic protein Bcl-2, but did not influence the expression of pro-apoptotic protein Bax. Furthermore, BAY11-7082 induced uveal melanoma cell apoptosis and inhibited xenograft tumor growth in vivo. Collectively, the present study identified NF-κB as an important survival signal for uveal melanoma cells and suggested that administration of specific NF-κB inhibitor BAY11-7082 could serve as an effective treatment for patients with uveal melanoma.
Collapse
Affiliation(s)
- Shuiqing Hu
- Department of Clinical Laboratories, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; E-Mails: (S.H.); (Q.L.); (D.H.)
| | - Qingqiong Luo
- Department of Clinical Laboratories, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; E-Mails: (S.H.); (Q.L.); (D.H.)
| | - Biyun Cun
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; E-Mails: (B.C.); (S.G.)
| | - Dan Hu
- Department of Clinical Laboratories, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; E-Mails: (S.H.); (Q.L.); (D.H.)
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; E-Mails: (B.C.); (S.G.)
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; E-Mails: (B.C.); (S.G.)
- Authors to whom correspondence should be addressed; E-Mails: (X.F.); (F.C.); Tel: +86-21-63135606 (X.F.); Fax: +86-21-63137148 (X.F.); Tel.: +86-21-63080932 (F.C.); Fax: +86-21-63136856 (F.C.)
| | - Fuxiang Chen
- Department of Clinical Laboratories, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China; E-Mails: (S.H.); (Q.L.); (D.H.)
- Authors to whom correspondence should be addressed; E-Mails: (X.F.); (F.C.); Tel: +86-21-63135606 (X.F.); Fax: +86-21-63137148 (X.F.); Tel.: +86-21-63080932 (F.C.); Fax: +86-21-63136856 (F.C.)
| |
Collapse
|
39
|
Park JS, Jung MY. Development of high-performance liquid chromatography-time-of-flight mass spectrometry for the simultaneous characterization and quantitative analysis of gingerol-related compounds in ginger products. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:10015-26. [PMID: 22985300 DOI: 10.1021/jf302944p] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Liquid chromatography-time-of-flight mass spectrometry (LC-TOF/MS) was established for the simultaneous separation, identification, and quantification of gingerol-related compounds in ginger products. The established method has been shown to provide a satisfactory linearity (r > 0.999) in a wide range (5-5000 ng/mL), low limits of detection and quantification, high precision, and inter- and intraday repeatability. The detection sensitivity of gingerols and shogaols by TOF/MS was 70-100 times higher than conventional UV detection at 288 nm. In this study, 19 ginerol-related compounds in the samples were identified and quantified by the established LC-TOF/MS method. The dried ginger powder products contained the highest quantity of gingerol-related compounds (7126.3-13789.0 μg/g), followed by fresh ginger products (2007.9-2790.0 μg/g), powdered ginger tea products (77.29-81.75 μg/g), and hot water ginger extracts (54.59-123.23 μg/mL). Shogaols were not found in fresh gingers. This paper represents the first report on the LC-TOF/MS analysis for the simultaneous characterization and quantification of gingerol-related compounds in ginger products.
Collapse
Affiliation(s)
- Ji Su Park
- College of Food Science, Woosuk University , Samrea-Up, Wanju-Kun, Jeonbuk Province 565-701, Republic of Korea
| | | |
Collapse
|