1
|
Casari G, Romaldi B, Scirè A, Minnelli C, Marzioni D, Ferretti G, Armeni T. Epigenetic Properties of Compounds Contained in Functional Foods Against Cancer. Biomolecules 2024; 15:15. [PMID: 39858410 PMCID: PMC11762081 DOI: 10.3390/biom15010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Epigenetics encompasses reversible and heritable genomic changes in histones, DNA expression, and non-coding RNAs that occur without modifying the nucleotide DNA sequence. These changes play a critical role in modulating cell function in both healthy and pathological conditions. Dysregulated epigenetic mechanisms are implicated in various diseases, including cardiovascular disorders, neurodegenerative diseases, obesity, and mainly cancer. Therefore, to develop innovative therapeutic strategies, research for compounds able to modulate the complex epigenetic landscape of cancer is rapidly surging. Dietary phytochemicals, mostly flavonoids but also tetraterpenoids, organosulfur compounds, and isothiocyanates, represent biologically active molecules found in vegetables, fruits, medicinal plants, and beverages. These natural organic compounds exhibit epigenetic modulatory properties by influencing the activity of epigenetics key enzymes, such as DNA methyltransferases, histone acetyltransferases and deacetylases, and histone methyltransferases and demethylases. Due to the reversibility of the modifications that they induce, their minimal adverse effects, and their potent epigenetic regulatory activity, dietary phytochemicals hold significant promise as antitumor agents and warrant further investigation. This review aims to consolidate current data on the diverse epigenetic effects of the six major flavonoid subclasses, as well as other natural compounds, in the context of cancer. The goal is to identify new therapeutic epigenetic targets for drug development, whether as stand-alone treatments or in combination with conventional antitumor approaches.
Collapse
Affiliation(s)
- Giulia Casari
- Department of Clinical and Specialist Sciences (DISCO), Università Politecnica delle Marche, 60131 Ancona, Italy; (G.C.); (B.R.); (G.F.)
| | - Brenda Romaldi
- Department of Clinical and Specialist Sciences (DISCO), Università Politecnica delle Marche, 60131 Ancona, Italy; (G.C.); (B.R.); (G.F.)
| | - Andrea Scirè
- Department of Life and Environmental Sciences (DISVA), Università Politecnica delle Marche, 60131 Ancona, Italy; (A.S.); (C.M.)
| | - Cristina Minnelli
- Department of Life and Environmental Sciences (DISVA), Università Politecnica delle Marche, 60131 Ancona, Italy; (A.S.); (C.M.)
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60131 Ancona, Italy;
| | - Gianna Ferretti
- Department of Clinical and Specialist Sciences (DISCO), Università Politecnica delle Marche, 60131 Ancona, Italy; (G.C.); (B.R.); (G.F.)
| | - Tatiana Armeni
- Department of Clinical and Specialist Sciences (DISCO), Università Politecnica delle Marche, 60131 Ancona, Italy; (G.C.); (B.R.); (G.F.)
| |
Collapse
|
2
|
Sidhu D, Vasundhara M, Dey P. The intestinal-level metabolic benefits of green tea catechins: Mechanistic insights from pre-clinical and clinical studies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155207. [PMID: 38000106 DOI: 10.1016/j.phymed.2023.155207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/11/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023]
Abstract
BACKGROUND The intestinal-level host-microbiota interaction has been implicated in the pathogenesis of chronic diseases. The current review is intended to provide a comprehensive insight into deciphering whether intestinal-level bioactivities mediate the overall metabolic health benefits of green tea catechins. PURPOSE We have comprehensively discussed pre-clinical and clinical evidences of intestinal-level changes in metabolism, microbiota, and metabolome due to catechin-rich green tea treatments, ultimately limiting metabolic diseases. Exclusive emphasis has been given to purified catechins and green tea, and discussions on extraintestinal mechanisms of metabolic health benefits were avoided. METHODS A literature search for relevant pre-clinical and clinical studies was performed in various online databases (e.g., PubMed) using specific keywords (e.g., catechin, intestine, microbiota). Out of all the referred literature, ∼15% belonged to 2021-2023, ∼51% were from 2011-2020, and ∼32% from 2000-2010. RESULT The metabolic health benefits of green tea catechins are indeed influenced by the intestinal-level bioactivities, including reduction of mucosal inflammation and oxidative stress, attenuation of gut barrier dysfunction, decrease in intestinal lipid absorption and metabolism, favorable modulation of mucosal nuclear receptor signaling, alterations of the luminal global metabolome, and mitigation of the gut dysbiosis. The results from the recent clinical studies support the pre-clinical evidences. The challenges and pitfalls of the currently available knowledge on catechin bioactivities have been discussed, and constructive directions to harness the translational benefits of green tea through future interventions have been provided. CONCLUSION The metabolism, metabolome, and microbiota at the intestinal epithelia play critical roles in catechin metabolism, pharmacokinetics, bioavailability, and bioactivities. Especially the reciprocal interaction between the catechins and the gut microbiota dictates the metabolic benefits of catechins.
Collapse
Affiliation(s)
- Dwinder Sidhu
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala 147004, India
| | - M Vasundhara
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala 147004, India.
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala 147004, India.
| |
Collapse
|
3
|
Kour S, Biswas I, Sheoran S, Arora S, Sheela P, Duppala SK, Murthy DK, Pawar SC, Singh H, Kumar D, Prabhu D, Vuree S, Kumar R. Artificial intelligence and nanotechnology for cervical cancer treatment: Current status and future perspectives. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
4
|
Hunsu VO, Facey COB, Fields JZ, Boman BM. Retinoids as Chemo-Preventive and Molecular-Targeted Anti-Cancer Therapies. Int J Mol Sci 2021; 22:7731. [PMID: 34299349 PMCID: PMC8304138 DOI: 10.3390/ijms22147731] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Retinoic acid (RA) agents possess anti-tumor activity through their ability to induce cellular differentiation. However, retinoids have not yet been translated into effective systemic treatments for most solid tumors. RA signaling is mediated by the following two nuclear retinoic receptor subtypes: the retinoic acid receptor (RAR) and the retinoic X receptor (RXR), and their isoforms. The identification of mutations in retinoid receptors and other RA signaling pathway genes in human cancers offers opportunities for target discovery, drug design, and personalized medicine for distinct molecular retinoid subtypes. For example, chromosomal translocation involving RARA occurs in acute promyelocytic leukemia (APL), and all-trans retinoic acid (ATRA) is a highly effective and even curative therapeutic for APL patients. Thus, retinoid-based target discovery presents an important line of attack toward designing new, more effective strategies for treating other cancer types. Here, we review retinoid signaling, provide an update on retinoid agents and the current clinical research on retinoids in cancer, and discuss how the retinoid pathway genotype affects the ability of retinoid agents to inhibit the growth of colorectal cancer (CRC) cells. We also deliberate on why retinoid agents have not shown clinical efficacy against solid tumors and discuss alternative strategies that could overcome the lack of efficacy.
Collapse
Affiliation(s)
- Victoria O. Hunsu
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (V.O.H.); (C.O.B.F.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19713, USA
| | - Caroline O. B. Facey
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (V.O.H.); (C.O.B.F.)
| | | | - Bruce M. Boman
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE 19713, USA; (V.O.H.); (C.O.B.F.)
- Department of Biological Sciences, University of Delaware, Newark, DE 19713, USA
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
5
|
O.B. Facey C, M. Boman B. Retinoids in Treatment of Colorectal Cancer. COLORECTAL CANCER 2021. [DOI: 10.5772/intechopen.93699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Retinoids are vitamin A metabolites best known for their role in embryonic development. Indeed, retinoid acid (RA) signaling plays a key role in regulating the development of the embryo body-plan by controlling embryonic stem cells (SCs). Retinoids function through their ability to induce cellular differentiation. Mutations in RA signaling pathway genes occur in most human cancers. The classic example is the chromosomal translocation involving RA receptor alpha in acute promyelocytic leukemia (APL). Because all-trans retinoic acid (ATRA) is a highly effective and often curative treatment for APL patients, determining if retinoids are efficacious for other cancer types is imperative. We review the current research on retinoids in colorectal cancer (CRC) and provide bioinformatics analyses of RA signaling. Our results show that most RA pathway genes are overexpressed and often mutated in CRC. Moreover, aberrant expression of many RA signaling proteins predicts decreased CRC patient survival. We also review aldehyde dehydrogenase (ALDH) expression in CRC because ALDH is a key enzyme in RA signaling, which regulates colonic SCs. Further investigation of RA signaling mechanisms that regulate colon SCs and how dysregulation contributes to the SC overpopulation that drives CRC growth should provide insight into strategies for designing new SC-targeted therapies for CRC.
Collapse
|
6
|
Wang C, Li H, Wu L, Jiao X, Jin Z, Zhu Y, Fang Z, Zhang X, Huang H, Zhao L. Coiled-Coil Domain-Containing 68 Downregulation Promotes Colorectal Cancer Cell Growth by Inhibiting ITCH-Mediated CDK4 Degradation. Front Oncol 2021; 11:668743. [PMID: 33968776 PMCID: PMC8100586 DOI: 10.3389/fonc.2021.668743] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/30/2021] [Indexed: 12/31/2022] Open
Abstract
Coiled-coil domain-containing 68 (CCDC68) plays different roles in cancer and is predicted as a tumor suppressor in human colorectal cancer (CRC). However, the specific role of CCDC68 in CRC and the underlying mechanisms remain unknown. Here, we showed that CCDC68 expression was lower in CRC than that in corresponding normal tissues, and CCDC68 level was positively correlated with disease-free survival. Ectopic expression of CCDC68 decreased CRC cell proliferation in vitro and suppressed the growth of CRC xenograft tumors in vivo. CCDC68 caused G0/G1 cell cycle arrest, downregulated CDK4, and upregulated ITCH, the E3 ubiquitin ligase responsible for CDK4 protein degradation. This increased CDK4 degradation, which decreased CDK4 protein levels and inhibited CRC tumor growth. Collectively, the present results identify a novel CDK4 regulatory axis consisting of CCDC68 and ITCH, which suggest that CCDC68 is a promising target for the treatment of CRC.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongyan Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lei Wu
- Department of General Surgery, Heze Municipal Hospital, Heze, China
| | - Xueli Jiao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zihui Jin
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yujie Zhu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ziling Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaodong Zhang
- Department of Colorectal anal surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haishan Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lingling Zhao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Ghasemi S, Xu S, Nabavi SM, Amirkhani MA, Sureda A, Tejada S, Lorigooini Z. Epigenetic targeting of cancer stem cells by polyphenols (cancer stem cells targeting). Phytother Res 2021; 35:3649-3664. [PMID: 33619811 DOI: 10.1002/ptr.7059] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/29/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022]
Abstract
Epigenetic alterations are one of the main factors that disrupt the expression of genes and consequently, they have an important role in the carcinogenicity and the progression of different cancers. Cancer stem cells (CSCs) are accountable for the recurrence, metastasis, and therapeutic failure of cancer. The noticeable and specific pathways in CSCs can be organized by epigenetic mechanisms such as DNA methylation, chromatin remodeling, regulatory RNAs, among others. Since epigenetics modifications can be changed and reversed, it is a possible tool for cancer control and treatment. Epigenetic therapies against CSCs are emerging as a very new strategy with a good future expectation to treat cancer patients. Phenolic compounds are a vast group of substances with anticarcinogenic functions, antiinflammatory, and antioxidative activities. It seems these characteristics are related to neutralizing CSCs development, their microenvironment, and metabolism through epigenetic mechanisms. In the current work, the types of epigenetic changes known in these cells are introduced. In addition, some studies about the use of polyphenols acting through a variety of epigenetic mechanisms to counteract these cells will be reviewed. The reported results seem to indicate that the use of these phenolic compounds may be useful for CSCs defeat.
Collapse
Affiliation(s)
- Sorayya Ghasemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, New York, USA
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Amir Amirkhani
- Stem Cell and Regenerative Medicine Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain.,CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Tejada
- CIBEROBN (Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, Madrid, Spain.,Laboratory of neurophysiology. Biology Department, University of Balearic Islands & Health Research Institute of the Balearic Islands (IdISBa), Palma de Mallorca, Spain
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
8
|
Kaleem M, Alhosin M, Khan K, Ahmad W, Hosawi S, Nur SM, Choudhry H, Zamzami MA, Al-Abbasi FA, Javed MDN. Epigenetic Basis of Polyphenols in Cancer Prevention and Therapy. POLYPHENOLS-BASED NANOTHERAPEUTICS FOR CANCER MANAGEMENT 2021:189-238. [DOI: 10.1007/978-981-16-4935-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
|
9
|
Exploring the Antihyperglycemic Chemical Composition and Mechanisms of Tea Using Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8871088. [PMID: 33343682 PMCID: PMC7725569 DOI: 10.1155/2020/8871088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/11/2020] [Accepted: 10/19/2020] [Indexed: 11/18/2022]
Abstract
Tea, a widely consumed beverage, has long been utilized for promoting human health with a close correlation to hyperglycemia. The Tea Metabolome Database (TMDB), the most complete and comprehensive curated collection of tea compounds data containing 1271 identified small molecule compounds from the tea plant (Camellia sinensis), was established previously by our research team. More recently, our studies have found that various tea types possess an antihyperglycemic effect in mice. However, the bioactive ingredients from tea have potential antihyperglycemic activity and their underlying molecular mechanisms remain unclear. In this study, we used a molecular docking approach to investigate the potential interactions between a selected 747 constituents contained in tea and 11 key protein targets of clinical antihyperglycemic drugs. According to our results, the main antihyperglycemic targets of tea composition were consistent with those of the drug rosiglitazone. The screening results showed that GCG, ECG3'Me, TMDB-01443, and CG had great target binding capacity. The results indicated that these chemicals of tea might affect hyperglycemia by acting on protein targets of rosiglitazone.
Collapse
|
10
|
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related morbidity and mortality worldwide. Although targeted therapy in combination with chemotherapy in CRC prolongs the overall survival of patients with metastatic disease, acquired resistance and relapse hinder their clinical benefits. Moreover, patients with some specific genetic profile are unlikely to benefit from targeted therapy, suggesting the need for safe and effective treatment strategies. Retinoids, comprising of natural and synthetic analogs, are a class of chemical compounds that regulate cellular proliferation, differentiation, and cell death. Retinoids have been used in the clinic for several leukemias and solid tumors, either as single agents or in combination therapy. Furthermore, retinoids have shown potent chemotherapeutic and chemopreventive properties in different cancer models, including CRC. In this review, we summarize the major preclinical findings in CRC in which natural and synthetic retinoids showed promising antitumor activities and stress on the proposed mechanisms of action. Understanding of the retinoids' antitumor mechanisms would provide insights to support and warrant their development in the management of CRC.
Collapse
|
11
|
Xu B, Jiang X, Xiong J, Lan J, Tian Y, Zhong L, Wang X, Xu N, Cao H, Zhang W, Zhang H, Hong X, Zhan YY, Zhang Y, Hu T. Structure-Activity Relationship Study Enables the Discovery of a Novel Berberine Analogue as the RXRα Activator to Inhibit Colon Cancer. J Med Chem 2020; 63:5841-5855. [PMID: 32391701 DOI: 10.1021/acs.jmedchem.0c00088] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We reported recently that berberine (Ber), a traditional oriental medicine to treat gastroenteritis, binds and activates retinoid X receptor α (RXRα) for suppressing the growth of colon cancer cells. Here, we extended our studies based on the binding mode of Ber with RXRα by design, synthesis, and biological evaluation of a focused library of 15 novel Ber analogues. Among them, 3,9-dimethoxy-5,6-dihydroisoquinolino[3,2-a]isoquinolin-7-ium chloride (B-12) was identified as the optimal RXRα activator. More efficiently than Ber, B-12 bound and altered the conformation of RXRα/LBD, thereby suppressing the Wnt/β-catenin pathway and colon cancer cell growth via RXRα mediation. In addition, B-12 not only preserved Ber's tumor selectivity but also greatly improved its bioavailability. Remarkably, in mice, B-12 did not show obvious side effects including hypertriglyceridemia as other RXRα agonists or induce hepatorenal toxicity. Together, our study describes an approach for the rational design of Ber-derived RXRα activators as novel effective antineoplastic agents for colon cancer.
Collapse
Affiliation(s)
- Beibei Xu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Xunjin Jiang
- Department of Chemistry and Key Laboratory of Chemical Biology of Fujian Province, iChEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Jing Xiong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Jun Lan
- Department of Biological Sciences, Tsinghua University, Beijing 100084, China
| | - Yuan Tian
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Linhai Zhong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Xinquan Wang
- Department of Biological Sciences, Tsinghua University, Beijing 100084, China
| | - Ning Xu
- Department of Biological Sciences, Tsinghua University, Beijing 100084, China
| | - Hanwei Cao
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Wenqing Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Hao Zhang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou 510632, China
| | - Xiaoting Hong
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yan-Yan Zhan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yandong Zhang
- Department of Chemistry and Key Laboratory of Chemical Biology of Fujian Province, iChEM, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, Fujian 361005, China
| | - Tianhui Hu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361005, China
| |
Collapse
|
12
|
Yu S, Zhu L, Wang K, Yan Y, He J, Ren Y. Green tea consumption and risk of breast cancer: A systematic review and updated meta-analysis of case-control studies. Medicine (Baltimore) 2019; 98:e16147. [PMID: 31277115 PMCID: PMC6635178 DOI: 10.1097/md.0000000000016147] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND As the most popular beverage in East Asia, green tea (GT) has various biological activities effects such as anti-mutation, anti-oxidation, and anti-tumor. In this study, we aimed to evaluate whether GT consumption could be an effective way to decrease the risk of breast cancer. METHODS We had performed a systematic review and updated meta-analysis of published case-control studies to evaluate the association between GT intake and the risk of breast cancer. Searching strategies were performed by the following keywords "Breast cancer," "breast neoplasm," and "green tea," with derivations and different combinations. The following databases were searched: PubMed, Cochrane Library, EMBASE, Web of science, China National Knowledge Infrastructure, WanFang, and China Biology Medicine disc. Studies published in both English and Chinese were considered for inclusion. Risk of bias was assessed through the Newcastle-Ottawa Scale (NOS). All data were analyzed through using Review Manager 5.1 software. RESULTS Fourteen studies fulfilled inclusion criteria for meta-analysis, yielding a total of 14,058 breast cancer patients and 15,043 control subjects. Individuals with the habit of drinking GT were found to have a negative association with the risk of future breast cancer (odds ratio 0.83; 95% confidence interval: 0.72-0.96) despite significant heterogeneity. In subgroup analyses, the negative correlation was still found in studies using registry-based controls, NOS grades ≥6 and the number of cases <500. CONCLUSIONS GT consumption may have a decreased incidence of breast cancer despite significant heterogeneity. However, owing to the quality of available studies, more properly designed trials are warranted to clarify the association between GT consumption and breast cancer.
Collapse
|
13
|
Modarai SR, Gupta A, Opdenaker LM, Kowash R, Masters G, Viswanathan V, Zhang T, Fields JZ, Boman BM. The anti-cancer effect of retinoic acid signaling in CRC occurs via decreased growth of ALDH+ colon cancer stem cells and increased differentiation of stem cells. Oncotarget 2018; 9:34658-34669. [PMID: 30410666 PMCID: PMC6205182 DOI: 10.18632/oncotarget.26157] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/15/2018] [Indexed: 01/09/2023] Open
Abstract
Background Tumorigenesis is driven by stem cell (SC) overpopulation. Because ALDH is both a marker for SCs in many tissues and a key enzyme in retinoid acid (RA) signaling, we studied RA signaling in normal and malignant colonic SCs. Hypothesis RA signaling regulates growth and differentiation of ALDH+ colonic SCs; dysregulation of RA signaling contributes to SC overpopulation and colorectal cancer (CRC) development. Methods We analyzed normal and malignant colonic tissues and CRC cell lines to see if retinoid receptors (RXR & RAR) are exclusively expressed in ALDH+ SCs, and if RA signaling changes during CRC development. We determined whether RA signaling regulates cancer SC (CSC) proliferation, differentiation, sphere formation, and population size. Results RXR & RAR were expressed in ALDH+ colonic SCs, but not in MCM2+ proliferative cells. Western blotting/immunostaining of CRCs revealed that RA signaling components become overexpressed in parallel with ALDH overexpression, which coincides with the known overpopulation of ALDH+ SCs that occurs during, and drives, CRC development. Treatment of SCs with all-trans retinoic acid (ATRA) decreased proliferation, sphere formation and ALDH+ SC population size, and induced differentiation along the neuroendocrine cell (NEC) lineage. Conclusions Retinoid signaling, by regulating ALDH+ colonic CSCs, decreases SC proliferation, sphere formation, and population size, and increases SC differentiation to NECs. Dysregulation of RA signaling in colonic SCs likely contributes to overpopulation of ALDH+ SCs and CRC growth. Implications That retinoid receptors RXR and RAR are selectively expressed in ALDH+ SCs indicates RA signaling mainly occurs via ALDH+ SCs, which provides a mechanism to selectively target CSCs.
Collapse
Affiliation(s)
- Shirin R Modarai
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.,Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA
| | - Anindita Gupta
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.,Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA
| | - Lynn M Opdenaker
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.,Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA
| | - Ryan Kowash
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA.,Department of Biological Sciences, Dickinson College, Carlisle, PA, USA
| | - Gabriel Masters
- Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA.,Biochemistry Department, Hamilton College, Clinton, NY, USA
| | - Vignesh Viswanathan
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.,Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA
| | - Tao Zhang
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.,Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA.,Genetic and Preventive Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,Research Pediatric Development, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Bruce M Boman
- Department of Biological Sciences, University of Delaware, Newark, DE, USA.,Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Christiana Care Health System, Newark, DE, USA.,Genetic and Preventive Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
14
|
Wang L, Yu K, Zhang X, Yu S. Dual functional roles of the MyD88 signaling in colorectal cancer development. Biomed Pharmacother 2018; 107:177-184. [PMID: 30086464 DOI: 10.1016/j.biopha.2018.07.139] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022] Open
Abstract
The myeloid differentiation factor 88 (MyD88), an adaptor protein in regulation of the innate immunity, functions to regulate immune responses against viral and bacterial infections in the human body. Toll-like receptors (TLRs) and interleukin 1 receptors (IL-1R) can recognize microbes or endogenous ligands and then recruit MyD88 to activate the MyD88-dependent pathway, while MyD88 mutation associated with lymphoma development and altered MyD88 signaling also involved in cancer-associated cell intrinsic and extrinsic inflammation progression and carcinogenesis. Detection of MyD88 expression was to predict prognosis of various human cancers, e.g., lymphoid, liver, and colorectal cancers. In human cancers, MyD88 protein acts as a bridge between the inflammatory signaling from the TLR/IL-1R and Ras oncogenic signaling pathway. However, the MyD88 signaling played dual functional roles in colorectal cancer, i.e., the tumor-promoting role that enhances cancer inflammation and intestinal flora imbalance to induce tumor invasion and tumor cell self-renewal, and the anti-tumor role that helps to maintain the host-microbiota homeostasis to induce tumor cell cycle arrest and immune responses against cancer cells. This review precisely discusses the up to date literature for these contrasting effects of MyD88 signaling on colorectal cancer development and progression.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Kewei Yu
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Xiang Zhang
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Shuwen Yu
- Department of Pharmacy, Jinan Central Hospital Affiliated to Shandong University, Jinan, China.
| |
Collapse
|
15
|
Morris J, Moseley VR, Cabang AB, Coleman K, Wei W, Garrett-Mayer E, Wargovich MJ. Reduction in promotor methylation utilizing EGCG (epigallocatechin-3-gallate) restores RXRα expression in human colon cancer cells. Oncotarget 2018; 7:35313-26. [PMID: 27167203 PMCID: PMC5085231 DOI: 10.18632/oncotarget.9204] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/16/2016] [Indexed: 12/16/2022] Open
Abstract
Silencing of regulatory genes through hypermethylation of CpG islands is an important mechanism in tumorigenesis. In colon cancer, RXRα, an important dimerization partner with other nuclear transcription factors, is silenced through this mechanism. We previously found that colon tumors in ApcMin/+ mice had diminished levels of RXRα protein and expression levels of this gene were restored by treatment with a green tea intervention, due to reduced promoter methylation of RXRα. We hypothesized that CIMP+ cell lines, which epigenetically silence key regulatory genes would also evidence silencing of RXRα and EGCG treatment would restore its expression. We indeed found EGCG to restore RXRα activity levels in the human cell lines, in a dose dependent manner and reduced RXRα promoter methylation. EGCG induced methylation changes in several other colon cancer related genes but did not cause a decrease in global methylation. Numerous epidemiological reports have shown the benefits of green tea consumption in reducing colon cancer risk but to date no studies have shown that the risk reduction may be related to the epigenetic restoration by tea polyphenols. Our results show that EGCG modulates the reversal of gene silencing involved in colon carcinogenesis providing a possible avenue for colon cancer prevention and treatment.
Collapse
Affiliation(s)
- Jay Morris
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Vondina R Moseley
- Department of Cell & Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - April B Cabang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Katie Coleman
- Department of Cell & Molecular Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Wei Wei
- Department of Public Health Science, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Elizabeth Garrett-Mayer
- Department of Biostatistics & Epidemiology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael J Wargovich
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
16
|
Berberine binds RXRα to suppress β-catenin signaling in colon cancer cells. Oncogene 2017; 36:6906-6918. [PMID: 28846104 PMCID: PMC5735301 DOI: 10.1038/onc.2017.296] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/22/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Berberine, an isoquinoline alkaloid, is a traditional oriental medicine used to treat diarrhea and gastroenteritis. Recently, we reported that it could inhibit the growth of intestinal polyp in animals and in patients with the familial adenomatous polyposis by downregulating β-catenin signaling. However, the intracellular target mediating the effects of berberine remains elusive. Here, we provide evidence that berberine inhibits β-catenin function via directly binding to a unique region comprising residues Gln275, Arg316 and Arg371 in nuclear receptor retinoid X receptor alpha (RXRα), where berberine concomitantly binding to and synergistically activating RXRα with 9-cis-retinoic acid (9-cis-RA), a natural ligand binding to the classical ligand-binding pocket of RXRα. Berberine binding promotes RXRα interaction with nuclear β-catenin, leading to c-Cbl mediated degradation of β-catenin, and consequently inhibits the proliferation of colon cancer cells. Furthermore, berberine suppresses the growth of human colon carcinoma xenograft in nude mice in an RXRα-dependent manner. Together, our study not only identifies RXRα as a direct protein target for berberine but also dissects their binding mode and validates that berberine indeed suppresses β-catenin signaling and cell growth in colon cancer via binding RXRα, which provide new strategies for the design of new RXRα-based antitumor agents and drug combinations.
Collapse
|
17
|
Ratovitski EA. Anticancer Natural Compounds as Epigenetic Modulators of Gene Expression. Curr Genomics 2017; 18:175-205. [PMID: 28367075 PMCID: PMC5345332 DOI: 10.2174/1389202917666160803165229] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/24/2015] [Accepted: 11/29/2015] [Indexed: 11/30/2022] Open
Abstract
Accumulating evidence shows that hallmarks of cancer include: "genetic and epigenetic alterations leading to inactivation of cancer suppressors, overexpression of oncogenes, deregulation of intracellular signaling cascades, alterations of cancer cell metabolism, failure to undergo cancer cell death, induction of epithelial to mesenchymal transition, invasiveness, metastasis, deregulation of immune response and changes in cancer microenvironment, which underpin cancer development". Natural compounds as bioactive ingredients isolated from natural sources (plants, fungi, marine life forms) have revolutionized the field of anticancer therapeutics and rapid developments in preclinical studies are encouraging. Natural compounds could affect the epigenetic molecular mechanisms that modulate gene expression, as well as DNA damage and repair mechanisms. The current review will describe the latest achievements in using naturally produced compounds targeting epigenetic regulators and modulators of gene transcription in vitro and in vivo to generate novel anticancer therapeutics.
Collapse
Affiliation(s)
- Edward A. Ratovitski
- Head and Neck Cancer Research Division, Department of Otolaryngology/Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
18
|
Khoogar R, Kim BC, Morris J, Wargovich MJ. Chemoprevention in gastrointestinal physiology and disease. Targeting the progression of cancer with natural products: a focus on gastrointestinal cancer. Am J Physiol Gastrointest Liver Physiol 2016; 310:G629-44. [PMID: 26893159 PMCID: PMC4867331 DOI: 10.1152/ajpgi.00201.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 02/11/2016] [Indexed: 01/31/2023]
Abstract
The last decade has witnessed remarkable progress in the utilization of natural products for the prevention and treatment of human cancer. Many agents now in the pipeline for clinical trial testing have evolved from our understanding of how human nutritional patterns account for widespread differences in cancer risk. In this review, we have focused on many of these promising agents arguing that they may provide a new strategy for cancer control: natural products once thought to be only preventive in their mode of action now are being explored for efficacy in tandem with cancer therapeutics. Natural products may reduce off-target toxicity of therapeutics while making cancers more amenable to therapy. On the horizon is the use of certain natural products, in their own right, as mitigants of late-stage cancer, a new frontier for small-molecule natural product drug discovery.
Collapse
Affiliation(s)
- Roxane Khoogar
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| | - Byung-Chang Kim
- 2Center for Colorectal Center, Center for Cancer Prevention and Detection, Research Institute and Hospital, National Cancer Center, Ilsan-ro, Illsandong-gu, Goyang-si Gyeonggi-do, Republic of Korea
| | - Jay Morris
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| | - Michael J. Wargovich
- 1Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| |
Collapse
|
19
|
Hu Y, Le Leu RK, Christophersen CT, Somashekar R, Conlon MA, Meng XQ, Winter JM, Woodman RJ, McKinnon R, Young GP. Manipulation of the gut microbiota using resistant starch is associated with protection against colitis-associated colorectal cancer in rats. Carcinogenesis 2016; 37:366-375. [PMID: 26905582 DOI: 10.1093/carcin/bgw019] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/05/2016] [Indexed: 12/13/2022] Open
Abstract
This study evaluated whether dietary resistant starch (RS) and green tea extract (GTE), which have anti-inflammatory and anticancer properties, protect against colitis-associated colorectal cancer (CAC) using a rat model, also investigated potential mechanisms of action of these agents including their effects on the gut microbiota. Rats were fed a control diet or diets containing 10% RS, 0.5% GTE or a combination of the two (RS + GTE). CAC was initiated with 2 weekly azoxymethane (AOM) injections (10mg/kg) followed by 2% dextran sodium sulphate in drinking water for 7 days after 2 weeks on diets. Rats were killed 20 weeks after the first AOM. Colon tissues and tumours were examined for histopathology by H&E, gene/protein expression by PCR and immunohistochemistry and digesta for analyses of fermentation products and microbiota populations. RS and RS + GTE (but not GTE) diets significantly (P< 0.05) decreased tumour multiplicity and adenocarcinoma formation, relative to the control diet. Effects of RS + GTE were not different from RS alone. RS diet caused significant shifts in microbial composition/diversity, with increases in Parabacteroides, Barnesiella, Ruminococcus, Marvinbryantia and Bifidobacterium as primary contributors to the shift. RS-containing diets increased short chain fatty acids (SCFA) and expression of the SCFA receptor GPR43 mRNA, and reduced inflammation (COX-2, NF-kB, TNF-α and IL-1β mRNA) and cell proliferation P< 0.05. GTE had no effect. This is the first study that demonstrates chemopreventive effects of RS (but not GTE) in a rodent CAC model, suggesting RS might have benefit to patients with ulcerative colitis who are at an increased risk of developing CRC.
Collapse
Affiliation(s)
- Ying Hu
- Flinders Centre for Innovation in Cancer , Flinders University of South Australia , Bedford Park , South Australia , Australia
| | - Richard K Le Leu
- Flinders Centre for Innovation in Cancer, Flinders University of South Australia, Bedford Park, South Australia, Australia.,CSIRO Food and Nutrition, Adelaide, South Australia, Australia
| | - Claus T Christophersen
- CSIRO Food and Nutrition, Adelaide, South Australia, Australia.,School of Medical and Health Science, Edith Cowan University, Western Australia, Australia, and
| | - Roshini Somashekar
- Flinders Centre for Innovation in Cancer , Flinders University of South Australia , Bedford Park , South Australia , Australia
| | - Michael A Conlon
- CSIRO Food and Nutrition , Adelaide , South Australia , Australia
| | - Xing Q Meng
- Biostatistics, Flinders Prevention, Promotion and Primary Health Care , General Practice , Flinders University of South Australia , Bedford Park , South Australia , Australia
| | - Jean M Winter
- Flinders Centre for Innovation in Cancer , Flinders University of South Australia , Bedford Park , South Australia , Australia
| | - Richard J Woodman
- Biostatistics, Flinders Prevention, Promotion and Primary Health Care , General Practice , Flinders University of South Australia , Bedford Park , South Australia , Australia
| | - Ross McKinnon
- Flinders Centre for Innovation in Cancer , Flinders University of South Australia , Bedford Park , South Australia , Australia
| | - Graeme P Young
- Flinders Centre for Innovation in Cancer , Flinders University of South Australia , Bedford Park , South Australia , Australia
| |
Collapse
|
20
|
Morris J, Fang Y, De Mukhopdhyay K, Wargovich MJ. Natural Agents Used in Chemoprevention of Aerodigestive and GI Cancers. ACTA ACUST UNITED AC 2016; 2:11-20. [PMID: 27134816 DOI: 10.1007/s40495-016-0047-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Aerodigestive cancers are on an increasing level in both occurrence and mortality. A major cause in many of these cancers is disruption of the inflammatory pathway, leading to increased cell proliferation, and epigenetic silencing of normal regulatory genes. Here we review the research on several natural products: silibinin, silymarin, quercetin, neem & nimbolide, gingerol, epigallatecatechin-3- gallate, curcumin, genistein and resveratrol conducted on aerodigestive cancers. These types of cancers are primarily those from oral cavity, esophagus/windpipe, stomach, small and large intestine, colon/rectum and bile/pancreas tissues. We report on the utilization in vivo and in vitro systems to research these dose effects on the inflammatory and epigenetic pathway components within the aerodigestive cancer. To follow up on the basic research we will discuss remaining research questions and future directions involving these natural products as putative stand alone or in combination with clinical agents.
Collapse
Affiliation(s)
- Jay Morris
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Yuan Fang
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Keya De Mukhopdhyay
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Michael J Wargovich
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| |
Collapse
|
21
|
Amin ARMR, Karpowicz PA, Carey TE, Arbiser J, Nahta R, Chen ZG, Dong JT, Kucuk O, Khan GN, Huang GS, Mi S, Lee HY, Reichrath J, Honoki K, Georgakilas AG, Amedei A, Amin A, Helferich B, Boosani CS, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Keith WN, Bhakta D, Halicka D, Niccolai E, Fujii H, Aquilano K, Ashraf SS, Nowsheen S, Yang X, Bilsland A, Shin DM. Evasion of anti-growth signaling: A key step in tumorigenesis and potential target for treatment and prophylaxis by natural compounds. Semin Cancer Biol 2015; 35 Suppl:S55-S77. [PMID: 25749195 PMCID: PMC4561219 DOI: 10.1016/j.semcancer.2015.02.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 12/14/2022]
Abstract
The evasion of anti-growth signaling is an important characteristic of cancer cells. In order to continue to proliferate, cancer cells must somehow uncouple themselves from the many signals that exist to slow down cell growth. Here, we define the anti-growth signaling process, and review several important pathways involved in growth signaling: p53, phosphatase and tensin homolog (PTEN), retinoblastoma protein (Rb), Hippo, growth differentiation factor 15 (GDF15), AT-rich interactive domain 1A (ARID1A), Notch, insulin-like growth factor (IGF), and Krüppel-like factor 5 (KLF5) pathways. Aberrations in these processes in cancer cells involve mutations and thus the suppression of genes that prevent growth, as well as mutation and activation of genes involved in driving cell growth. Using these pathways as examples, we prioritize molecular targets that might be leveraged to promote anti-growth signaling in cancer cells. Interestingly, naturally occurring phytochemicals found in human diets (either singly or as mixtures) may promote anti-growth signaling, and do so without the potentially adverse effects associated with synthetic chemicals. We review examples of naturally occurring phytochemicals that may be applied to prevent cancer by antagonizing growth signaling, and propose one phytochemical for each pathway. These are: epigallocatechin-3-gallate (EGCG) for the Rb pathway, luteolin for p53, curcumin for PTEN, porphyrins for Hippo, genistein for GDF15, resveratrol for ARID1A, withaferin A for Notch and diguelin for the IGF1-receptor pathway. The coordination of anti-growth signaling and natural compound studies will provide insight into the future application of these compounds in the clinical setting.
Collapse
Affiliation(s)
| | - Phillip A Karpowicz
- Department of Biological Sciences, University of Windsor, 401 Sunset Ave., Room 327, Windsor, Ontario, N9B 3P4, Canada
| | | | - Jack Arbiser
- Winship Cancer Institute of Emory University, Atlanta, GA, USA; Atlanta Veterans Administration Health Center, Atlanta, GA, USA
| | - Rita Nahta
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Zhuo G Chen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Jin-Tang Dong
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | | | - Shijun Mi
- Albert Einstein College of Medicine, New York, NY, USA
| | - Ho-Young Lee
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | | | | | | | | | - Amr Amin
- UAE University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Urbana Champaign, IL, USA
| | | | | | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | | | | | | | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | | | | | | | | | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Medical School, Mayo Graduate School, Mayo Clinic, Rochester, MN, USA
| | - Xujuan Yang
- University of Illinois at Urbana Champaign, Urbana Champaign, IL, USA
| | | | - Dong M Shin
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
22
|
Busch C, Burkard M, Leischner C, Lauer UM, Frank J, Venturelli S. Epigenetic activities of flavonoids in the prevention and treatment of cancer. Clin Epigenetics 2015; 7:64. [PMID: 26161152 PMCID: PMC4497414 DOI: 10.1186/s13148-015-0095-z] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/17/2015] [Indexed: 02/07/2023] Open
Abstract
Aberrant epigenetic modifications are described in an increasing number of pathological conditions, including neurodegenerative diseases, cardiovascular diseases, diabetes mellitus type 2, obesity and cancer. The general reversibility of epigenetic changes makes them an attractive and promising target e.g. in the treatment of cancer. Thus, a growing number of epigenetically active compounds are currently tested in clinical trials for their therapeutic potential. Interestingly, many phytochemicals present in plant foods, particularly flavonoids, are suggested to be able to alter epigenetic cellular mechanisms. Flavonoids are natural phenol compounds that form a large group of secondary plant metabolites with interesting biological activities. They can be categorized into six major subclasses, which display diverse properties affecting the two best characterized epigenetic mechanisms: modulation of the DNA methylation status and histone acetylation. High dietary flavonoid intake has strongly been suggested to reduce the risk of numerous cancer entities in a large body of epidemiological studies. Established health-promoting effects of diets rich in fruit and vegetables are faced by efforts to use purified flavonoids as supplements or pharmaceuticals, whereupon data on the latter applications remain controversial. The purpose of this review is to give an overview of current research on flavonoids to further elucidate their potential in cancer prevention and therapy, thereby focusing on their distinct epigenetic activities.
Collapse
Affiliation(s)
- Christian Busch
- Division of Dermatologic Oncology, Department of Dermatology and Allergology, Medical University Hospital, Tuebingen, Germany
| | - Markus Burkard
- Division of Dermatologic Oncology, Department of Dermatology and Allergology, Medical University Hospital, Tuebingen, Germany ; Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Christian Leischner
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Ulrich M Lauer
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| | - Jan Frank
- Institute of Biological Chemistry and Nutrition, University of Hohenheim, Stuttgart, Germany
| | - Sascha Venturelli
- Department of Internal Medicine I, Medical University Hospital, Otfried-Mueller-Str. 27, 72076 Tuebingen, Germany
| |
Collapse
|
23
|
Giardina C, Nakanishi M, Khan A, Kuratnik A, Xu W, Brenner B, Rosenberg DW. Regulation of VDR Expression in Apc-Mutant Mice, Human Colon Cancers and Adenomas. Cancer Prev Res (Phila) 2015; 8:387-99. [PMID: 25873367 DOI: 10.1158/1940-6207.capr-14-0371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/20/2015] [Indexed: 02/06/2023]
Abstract
One variable that may affect the ability of vitamin D to reduce colon cancer risk is the expression of its high-affinity receptor, VDR. Here, we show that vitamin D does not reduce tumor formation in Apc(Δ14/+) mice and that VDR expression is lost in the majority of the colon tumor cells. The extent of VDR loss corresponded inversely to the level of β-catenin nuclear localization and could be observed in early lesions composed of just a few crypts. Analysis of reported VDR regulators showed that the repressing class I histone deacetylases (HDAC) were significantly elevated in the tumors (up to 4-fold), whereas the VDR-activating retinoid X receptors (RXR) were downregulated (∼50%). Expression of the Slug repressor was also increased, but was found primarily in stromal cells. Analysis of epigenetically active compounds on colon cell lines and intestinal organoids showed that HDAC inhibitors were particularly adept at stimulating VDR expression. Treatment of tumor-bearing Apc(Δ14/+) mice with the HDAC inhibitor panobinostat increased VDR expression in the tumors and normal mucosa. The RXR agonist bexarotene failed to activate VDR expression, indicating that RXR ligands were not limiting. Analysis of human microarray data indicated that VDR mRNA is frequently downregulated in colon adenomas, which correlated positively with RXRA expression and inversely with HDAC 2 and 8 expression. Human adenomas showed variable VDR protein expression levels, both between and within individual lesions. Determining the mechanisms of VDR regulation in colon neoplasms may significantly enhance our ability to use vitamin D as a cancer prevention agent.
Collapse
Affiliation(s)
- Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut.
| | - Masako Nakanishi
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Awaad Khan
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Anton Kuratnik
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Wanli Xu
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Bruce Brenner
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - Daniel W Rosenberg
- Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
24
|
Henning SM, Wang P, Carpenter CL, Heber D. Epigenetic effects of green tea polyphenols in cancer. Epigenomics 2014; 5:729-41. [PMID: 24283885 DOI: 10.2217/epi.13.57] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Epigenetics describes heritable alterations of gene expression and chromatin organization without changes in DNA sequence. Both hypermethylation and hypomethylation of DNA can affect gene expression and the multistep process of carcinogenesis. Epigenetic changes are reversible and may be targeted by dietary interventions. Bioactive compounds from green tea (GT) such as (-)-epigallocatechin gallate have been shown to alter DNA methyltransferase activity in studies of esophageal, oral, skin, Tregs, lung, breast and prostate cancer cells, which may contribute to the chemopreventive effect of GT. Three out of four mouse model studies have confirmed the inhibitory effect of (-)-epigallocatechin gallate on DNA methylation. A human study demonstrated that decreased methylation of CDX2 and BMP-2 in gastric carcinoma was associated with higher GT consumption. It is the goal of this review to summarize our current knowledge of the potential of GT to alter epigenetic processes, which may be useful in chemoprevention.
Collapse
Affiliation(s)
- Susanne M Henning
- Center for Human Nutrition, David Geffen School of Medicine, University of California Los Angeles, 900 Veteran Avenue, Warren Hall 14-166, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
25
|
Hun Lee J, Shu L, Fuentes F, Su ZY, Tony Kong AN. Cancer chemoprevention by traditional chinese herbal medicine and dietary phytochemicals: targeting nrf2-mediated oxidative stress/anti-inflammatory responses, epigenetics, and cancer stem cells. J Tradit Complement Med 2014; 3:69-79. [PMID: 24716158 PMCID: PMC3924975 DOI: 10.4103/2225-4110.107700] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Excessive oxidative stress induced by reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive metabolites of carcinogens alters cellular homeostasis, leading to genetic/epigenetic changes, genomic instability, neoplastic transformation, and cancer initiation/progression. As a protective mechanism against oxidative stress, antioxidant/detoxifying enzymes reduce these reactive species and protect normal cells from endo-/exogenous oxidative damage. The transcription factor nuclear factor-erythroid 2 p45 (NF-E2)-related factor 2 (Nrf2), a master regulator of the antioxidative stress response, plays a critical role in the expression of many cytoprotective enzymes, including NAD(P)H:quinine oxidoreductase (NQO1), heme oxygenase-1 (HO-1), UDP-glucuronosyltransferase (UGT), and glutathione S-transferase (GST). Recent studies demonstrated that many dietary phytochemicals derived from various vegetables, fruits, spices, and herbal medicines induce Nrf2-mediated antioxidant/detoxifying enzymes, restore aberrant epigenetic alterations, and eliminate cancer stem cells (CSCs). The Nrf2-mediated antioxidant response prevents many age-related diseases, including cancer. Owing to their fundamental contribution to carcinogenesis, epigenetic modifications and CSCs are novel targets of dietary phytochemicals and traditional Chinese herbal medicine (TCHM). In this review, we summarize cancer chemoprevention by dietary phytochemicals, including TCHM, which have great potential as a safer and more effective strategy for preventing cancer.
Collapse
Affiliation(s)
- Jong Hun Lee
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Limin Shu
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Francisco Fuentes
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA ; Department of Desert Agriculture and Biotechnology, Arturo Prat University, PO box 121, Iquique, Chile
| | - Zheng-Yuan Su
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Ah-Ng Tony Kong
- Center for Cancer Prevention Research, Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| |
Collapse
|
26
|
Elevated methylation of the RXRA promoter region may be responsible for its downregulated expression in the myocardium of patients with TOF. Pediatr Res 2014; 75:588-94. [PMID: 24513686 DOI: 10.1038/pr.2014.17] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 11/21/2013] [Indexed: 11/08/2022]
Abstract
BACKGROUND As an important component of retinoid acid signaling pathway, the retinoid X receptor α (RXRA) is considered to play an important role in the pathogenesis of tetralogy of Fallot (TOF). METHODS The expression level of RXRA mRNA and the methylation status of the RXRA promoter region in 26 patients with TOF and 6 controls were detected using real-time PCR and bisulfite-specific PCR and cloning-based sequencing, respectively. Dual-luciferase reporter assays, combined with in vitro methylation assay, were performed to determine the transcriptional regulatory activity of unmethylated and methylated CpG regions in the RXRA promoter. RESULTS The mRNA expression of RXRA in the right ventricular outflow tract (RVOT) myocardium was significantly decreased in patients with TOF compared with that in the controls. The methylation status of region -1453 to -1000 containing CpG sites 1-23 in the RXRA promoter region was higher in patients with TOF than that in the controls. This region contained several transcription factor sites. In addition, dual-luciferase reporter assays combined with methylation assay in vitro showed that this region had transcriptional regulatory activity, which can be depressed by the methylation of this region. CONCLUSION The elevated methylation at RXRA promoter may be responsible for the downregulated mRNA expression in RVOT myocardium of patients with TOF.
Collapse
|
27
|
The importance of the retinoid X receptor alpha in modulating inflammatory signaling in acute murine colitis. Dig Dis Sci 2014; 59:753-9. [PMID: 24146318 PMCID: PMC4009739 DOI: 10.1007/s10620-013-2902-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2013] [Accepted: 09/27/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND In order for vitamin D to signal and regulate inflammatory pathways, it must bind to its receptor (VDR) which must heterodimerize with the retinoid X receptor alpha (RXRα). Although the role that vitamin D signaling plays in the development and progression of colitis, a disease characterized by excessive inflammation, has been suggested, little research has been done on determining the role that RXRα plays in acute colitis development. AIMS This study sought to determine the effects that reduced availability of RXRα would have on the development of acute murine colitis. Expression of inflammatory markers, VDR and RXRα were investigated to determine if the reduction in expression of RXRα in RXRα(+/-) mice would result in increased inflammatory signaling and receptor downregulation as compared to their wild-type littermates. METHODS An acute murine model of colitis, the axozymethane (AOM) and dextran sulfate sodium (DSS) model was utilized in wild-type and RXRα(+/-) mice. Gross manifestations of colitis measured included weight loss and colitis score. Immunblots and real-time PCR were performed for inflammatory markers and receptor expression. RESULTS RXRα(+/-) mice induced with AOM/DSS colitis demonstrated increased gene expression of Snail and Snail2, transcription factors downstream of inflammatory mediators, as compared to their wild-type littermates. CONCLUSIONS This demonstrates the importance of RXRα in regulating inflammation in acute colitis and also identifies RXRα expression as a new consideration when developing successful interventions for acute colitis due to the requirement of numerous receptors for RXRα.
Collapse
|
28
|
Thakur VS, Deb G, Babcook MA, Gupta S. Plant phytochemicals as epigenetic modulators: role in cancer chemoprevention. AAPS JOURNAL 2013; 16:151-63. [PMID: 24307610 DOI: 10.1208/s12248-013-9548-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 11/18/2013] [Indexed: 12/18/2022]
Abstract
In recent years, "nutri-epigenetics," which focuses on the influence of dietary agents on epigenetic mechanism(s), has emerged as an exciting novel area in epigenetics research. Targeting of aberrant epigenetic modifications has gained considerable attention in cancer chemoprevention research because, unlike genetic changes, epigenetic alterations are reversible and occur during early carcinogenesis. Aberrant epigenetic mechanisms, such as promoter DNA methylation, histone modifications, and miRNA-mediated post-transcriptional alterations, can silence critical tumor suppressor genes, such as transcription factors, cell cycle regulators, nuclear receptors, signal transducers, and apoptosis-inducing and DNA repair gene products, and ultimately contribute to carcinogenesis. In an effort to identify and develop anticancer agents which cause minimal harm to normal cells while effectively killing cancer cells, a number of naturally occurring phytochemicals in food and medicinal plants have been investigated. This review highlights the potential role of plant-derived phytochemicals in targeting epigenetic alterations that occur during carcinogenesis, by modulating the activity or expression of DNA methyltransferases, histone modifying enzymes, and miRNAs. We present in detail the epigenetic mode of action of various phytochemicals and discuss their potential as safe and clinically useful chemopreventive strategies.
Collapse
Affiliation(s)
- Vijay S Thakur
- Department of Urology, Case Western Reserve University, University Hospitals Case Medical Center, 10900 Euclid Avenue, Cleveland, Ohio, 44106, USA
| | | | | | | |
Collapse
|
29
|
Moseley VR, Morris J, Knackstedt RW, Wargovich MJ. Green tea polyphenol epigallocatechin 3-gallate, contributes to the degradation of DNMT3A and HDAC3 in HCT 116 human colon cancer cells. Anticancer Res 2013; 33:5325-5333. [PMID: 24324066 PMCID: PMC4017340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
BACKGROUND Colon cancer is still the second leading cause of cancer deaths in the United States. Epigenetic gene silencing involving DNA methyltransferases (DNMTs) and histone deacetylases (HDACs) plays an important role in the progression of colon cancer. MATERIALS AND METHODS In the present study we found that the sensitivity of colon cancer cells to methylation plays a role in its response to alternative therapy involving the green tea polyphenol, epigallocatechin 3-gallate. HDAC and DNMT protein expression were reduced when methylation-sensitive HCT 116 human colon cancer cells was treated with EGCG, but was relatively stable in the HT-29 cell line. This decrease in expression may be partially explained by our finding that DNMT3A and HDAC3 are degraded in the methylation-sensitive colon cancer cells in part by inhibiting their association with the E3 ubiquitin ligase, UHRF1. CONCLUSION These findings provide a rationale for the development of a targeted therapy for methylation-sensitive colon cancer that can include EGCG in combination with other DNMT and HDAC inhibitors.
Collapse
|
30
|
Lambert JD. Does tea prevent cancer? Evidence from laboratory and human intervention studies. Am J Clin Nutr 2013; 98:1667S-1675S. [PMID: 24172300 DOI: 10.3945/ajcn.113.059352] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tea (Camellia sinensis) is a widely consumed beverage and has been extensively studied for its cancer-preventive activity. Both the polyphenolic constituents as well as the caffeine in tea have been implicated as potential cancer-preventive compounds; the relative importance seems to depend on the cancer type. Green tea and the green tea catechin have been shown to inhibit tumorigenesis at a number of organ sites and to be effective when administered either during the initiation or postinitiation phases of carcinogenesis. Black tea, although not as well studied as green tea, has also shown cancer-preventive effects in laboratory models. A number of potential mechanisms have been proposed to account for the cancer-preventive effects of tea, including modulation of phase II metabolism, alterations in redox environment, inhibition of growth factor signaling, and others. In addition to the laboratory studies, there is a growing body of human intervention studies suggesting that tea can slow cancer progression and modify biomarkers relevant to carcinogenesis. Although available data are promising, many questions remain with regard to the dose-response relations of tea constituents in various models, the primary mechanisms of action, and the potential for combination chemoprevention strategies that involve tea as well as other dietary or pharmaceutical agents. The present review examines the available data from laboratory animal and human intervention studies on tea and cancer prevention. These data were evaluated, and areas for further research are identified.
Collapse
Affiliation(s)
- Joshua D Lambert
- Center of Excellence for Plant and Mushroom Foods for Health, the Department of Food Science, Pennsylvania State University, University Park, PA
| |
Collapse
|
31
|
Cao H, Song S, Zhang H, Zhang Y, Qu R, Yang B, Jing Y, Hu T, Yan F, Wang B. Chemopreventive effects of berberine on intestinal tumor development in Apcmin/+ mice. BMC Gastroenterol 2013; 13:163. [PMID: 24279644 PMCID: PMC4222594 DOI: 10.1186/1471-230x-13-163] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 11/23/2013] [Indexed: 02/08/2023] Open
Abstract
Background Berberine, an isoquinoline alkaloid, has shown inhibitory effects on growth of several tumor cell lines in vitro. The aim of this study was to investigate chemopreventive effects of berberine on intestinal tumor development in Apcmin/+ mice. Methods Four-week old Apcmin/+ mice were treated with 0.05% or 0.1% berberine in drinking water for twelve weeks. The number and the size of tumors were measured to evaluate intestinal tumor development. Tissue sections were prepared for PCNA and Ki-67 immunostaining to detect cell proliferation, and TUNEL assay and cleaved caspase-3 immunostaining for apoptosis. Western blot analysis and immunostaining were performed to detect the activation of Wnt and epidermal growth factor receptor (EGFR) signaling pathways and COX-2 expression in the intestinal tumor cells. The prostaglandin E2 level in the small intestine was detected using ELISA. Results Compared with untreated Apcmin/+ mice, the total numbers of tumors in the small intestine and the colon were reduced by 39.6% and 62.5% in 0.05% and 0.1% berberine-treated mice, respectively. The numbers of tumors in proximal, middle, and distal segments of the small intestine in 0.1% berberine-treated mice were significantly reduced by 53.7%, 55.3%, and 76.5% respectively. Berberine treatment also decreased the numbers of all sizes of tumors (>2 mm, 1–2 mm, and <1 mm) in the small intestine. Berberine suppressed tumor cell proliferation and increased apoptosis. Furthermore, berberine decreased the activation levels of Wnt and EGFR signaling pathways, and down-regulated COX-2 expression in intestinal tumor cells and prostaglandin E2 production in the small intestine. Conclusions Berberine inhibits intestinal tumor development, which is correlated with its activity to suppress tumor cell proliferation and increase apoptosis in Apcmin/+ mice. Down-regulation of Wnt and EGFR signaling pathways and COX-2 expression by berberine may be involved in its anti-tumorigenic effects.
Collapse
Affiliation(s)
- Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, 154 Anshan Road in Heping District, Tianjin 300052, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
van der Weyden L, Adams DJ. Cancer of mice and men: old twists and new tails. J Pathol 2013; 230:4-16. [PMID: 23436574 DOI: 10.1002/path.4184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 01/28/2013] [Accepted: 02/16/2013] [Indexed: 12/18/2022]
Abstract
In this review we set out to celebrate the contribution that mouse models of human cancer have made to our understanding of the fundamental mechanisms driving tumourigenesis. We take the opportunity to look forward to how the mouse will be used to model cancer and the tools and technologies that will be applied, and indulge in looking back at the key advances the mouse has made possible.
Collapse
|
33
|
Knackstedt RW, Moseley VR, Wargovich MJ. Epigenetic mechanisms underlying diet-sourced compounds in the prevention and treatment of gastrointestinal cancer. Anticancer Agents Med Chem 2013; 12:1203-10. [PMID: 22931412 DOI: 10.2174/187152012803833053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 06/06/2012] [Accepted: 06/07/2012] [Indexed: 12/31/2022]
Abstract
The development of colon cancer, the third most diagnosed cancer and third leading cause of cancer deaths in the United States, can be influenced by genetic predispositions and environmental exposures. As 80% of colon cancer cases are sporadic in nature, much interest lies in determining risk factors that may foster its development, as well as identifying compounds that could inhibit colon cancer development or halt progression. A major risk factor for sporadic colon cancer is a high fat, Western diet which has been linked to a cancer-prone, pro-inflammatory state. Cultures which place an emphasis on fresh fruits and vegetables demonstrate lower colon cancer incidences. Diet not only has the potential to encourage colon cancer development, but recent evidence demonstrates that certain dietary natural products can halt colon cancer development and progression via epigenetic regulation. Epigenetic dysregulation may contribute to inflammation-driven diseases, such as cancer, and can lead to the inappropriate silencing of genes necessary to inhibit cancer development. Natural compounds have shown the ability to reverse epigenetic dysregulation in in vitro and in vivo models. As current allopathic medicines aimed at reversing epigenetic silencing are accompanied with the risk of toxicity and side effects, much interest lies in being able to harness the disease preventing properties in natural products. Here, we discuss the epidemiology of colon cancer, describe the need for natural approaches to inhibit disease development and highlight natural products which have been shown to inhibit gastrointestinal cancer initiation and progression in vitro or in vivo through epigenetic modulation.
Collapse
Affiliation(s)
- Rebecca W Knackstedt
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
34
|
Hu Y, McIntosh GH, Le Leu RK, Nyskohus LS, Woodman RJ, Young GP. Combination of selenium and green tea improves the efficacy of chemoprevention in a rat colorectal cancer model by modulating genetic and epigenetic biomarkers. PLoS One 2013; 8:e64362. [PMID: 23717604 PMCID: PMC3662759 DOI: 10.1371/journal.pone.0064362] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 04/12/2013] [Indexed: 02/08/2023] Open
Abstract
Dietary supplementation of selenium and green tea holds promise in cancer prevention. In this study, we evaluated the efficacies of selenium and green tea administered individually and in combination against colorectal cancer in an azoxymethane (AOM)-induced rat colonic carcinogenesis model and determined the underlying mechanisms of the protection. Four-week old Sprague-Dawley male rats were fed with diets containing 0.5% green tea extract, 1 ppm selenium as selenium-enriched milk protein, or combination of 1 ppm selenium and 0.5% green tea extract. Animals received 2 AOM (15 mg/kg) treatments to induce colonic oncogenesis. Rats were killed 8 or 30 wk later after the last AOM to examine the effect of dietary intervention on aberrant crypt foci (ACF) formation or tumor development. On sacrifice, colons were examined for ACF and tumors, the mRNA levels of SFRP5 and Cyclin D1, and the proteins levels of ß-catenin, COX-2, Ki-67, DNMT1 and acetyl histone H3. The combination of selenium and green tea resulted in a significant additive inhibition of large ACF formation, this effect was greater than either selenium or green tea alone, P<0.01; the combination also had a significant additive inhibition effect on all tumor endpoints, the effect of the combination diet on tumor incidence, multiplicity and size was greater than selenium or green tea alone, P<0.01. Rats fed the combination diet showed marked reduction of DNMT1 expression and induction of histone H3 acetylation, which were accompanied by restoration of SFRP5 mRNA in normal-appearing colonic crypts. The combination diet also significantly reduced ß-catenin nuclear translocation, Cyclin D1 expression and cell proliferation. These data show, for the first time, that combination of selenium and green tea is more effective in suppressing colorectal oncogenesis than either agent alone. The preventive effect is associated with regulation of genetic and epigenetic biomarkers implicated in colonic carcinogenesis.
Collapse
Affiliation(s)
- Ying Hu
- Flinders Centre for Innovation in Cancer, Flinders University of South Australia, Adelaide, Australia.
| | | | | | | | | | | |
Collapse
|
35
|
Shankar S, Kumar D, Srivastava RK. Epigenetic modifications by dietary phytochemicals: implications for personalized nutrition. Pharmacol Ther 2013; 138:1-17. [PMID: 23159372 PMCID: PMC4153856 DOI: 10.1016/j.pharmthera.2012.11.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 11/01/2012] [Indexed: 12/20/2022]
Abstract
In the last two decades, the study of epigenetic modification emerged as one of the major areas of cancer treatment targeted by dietary phytochemicals. Recent studies with various types of cancers revealed that the epigenetic modifications are associated with the food source corresponds to dietary phytochemicals. The dietary phytochemicals have been used in Asian countries for thousands of years to cure several diseases including cancer. They have been reported to modulate the several biological processes including histone modification, DNA methylation and non-coding microRNA expression. These events play a vital role in carcinogenesis. Various studies suggest that a number of dietary compounds present in vegetables, spices and other herbal products have epigenetic targets in cancer cells. Dietary phytochemicals have been reported to repair DNA damage by enhancing histone acetylation that helps to restrain cell death, and also alter DNA methylation. These phytochemicals are able to modulate epigenetic modifications and their targets to cure several cancers. Epigenetic aberrations dynamically contribute to cancer pathogenesis. Given the individualized traits of epigenetic biomarkers, the personalized nutrition will help us to prevent various types of cancer. In this review, we will discuss the effect of dietary phytochemicals on genetic and epigenetic modifications and how these modifications help to prevent various types of cancers and improve health outcomes.
Collapse
Affiliation(s)
- Sharmila Shankar
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, The University of Kansas Cancer Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Dhruv Kumar
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Medical Center, The University of Kansas Cancer Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Rakesh K. Srivastava
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Medical Center, The University of Kansas Cancer Center, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| |
Collapse
|
36
|
Cho Y, Turner ND, Davidson LA, Chapkin RS, Carroll RJ, Lupton JR. A chemoprotective fish oil/pectin diet enhances apoptosis via Bcl-2 promoter methylation in rat azoxymethane-induced carcinomas. Exp Biol Med (Maywood) 2013; 237:1387-93. [PMID: 23354397 DOI: 10.1258/ebm.2012.012244] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have demonstrated that diets containing fish oil and pectin (FO/P) reduce colon tumor incidence relative to control (corn oil and cellulose [CO/C]) in part by inducing apoptosis of DNA-damaged colon cells. Relative to FO/P, CO/C promotes colonocyte expression of the antiapoptotic modulator, Bcl-2, and Bcl-2 promoter methylation is altered in colon cancer. To determine if FO/P, compared with CO/C, limits Bcl-2 expression by enhancing promoter methylation in colon tumors, we examined Bcl-2 promoter methylation, mRNA levels, colonocyte apoptosis and colon tumor incidence in azoxymethane (AOM)-injected rats. Rats were provided diets containing FO/P or CO/C, and were terminated 16 and 34 weeks after AOM injection. DNA isolated from paraformaldehyde-fixed colon tumors and uninvolved tissue was bisulfite modified and amplified by quantitative reverese transcriptase-polymerase chain reaction to assess DNA methylation in Bcl-2 cytosine-guanosine islands. FO/P increased Bcl-2 promoter methylation (P = 0.009) in tumor tissues and colonocyte apoptosis (P = 0.020) relative to CO/C. An inverse correlation between Bcl-2 DNA methylation and Bcl-2 mRNA levels was observed in the tumors. We conclude that dietary FO/P promotes apoptosis in part by enhancing Bcl-2 promoter methylation. These Bcl-2 promoter methylation responses, measured in vivo, contribute to our understanding of the mechanisms involved in chemoprevention of colon cancer by diets containing FO/P.
Collapse
Affiliation(s)
- Youngmi Cho
- Faculty of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
Tea is one of the most consumed beverages worldwide, and green tea is the least processed from the buds of the Camellia sinensis plant. The most abundant component of green tea is (-)-epigallocatechin-3-gallate (EGCG), which has been the focus of many cell culture, animal and clinical trials, revealing that EGCG possesses antiproliferative, antimutagenic, antioxidant, antibacterial, antiviral and chemopreventive effects. In this review we briefly summarize the mechanism of action(s) of the green tea component EGCG, highlighting recent advances in the epigenetic regulation by EGCG. Additionally, we provide an overview of mouse chemoprevention studies and EGCG chemoprevention clinical trials.
Collapse
Affiliation(s)
- Laura Schramm
- Department of Biological Sciences, St. John's University, NY 11439, USA
| |
Collapse
|
38
|
|
39
|
McClellan JL, Davis JM, Steiner JL, Enos RT, Jung SH, Carson JA, Pena MM, Carnevale KA, Berger FG, Murphy EA. Linking tumor-associated macrophages, inflammation, and intestinal tumorigenesis: role of MCP-1. Am J Physiol Gastrointest Liver Physiol 2012; 303:G1087-95. [PMID: 23019193 PMCID: PMC3517651 DOI: 10.1152/ajpgi.00252.2012] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Tumor-associated macrophages are associated with poor prognosis in certain cancers. Monocyte chemoattractant protein 1 (MCP-1) is thought to be the most important chemokine for recruitment of macrophages to the tumor microenvironment. However, its role on tumorigenesis in a genetic mouse model of colon cancer has not been explored. We examined the role of MCP-1 on tumor-associated macrophages, inflammation, and intestinal tumorigenesis. Male Apc(Min/+), Apc(Min/+)/MCP-1(-/-) or wild-type mice were euthanized at 18 wk of age and intestines were analyzed for polyp burden, apoptosis, proliferation, β-catenin, macrophage number and phenotype, markers for cytotoxic T lymphocytes and regulatory T cells, and inflammatory mediators. MCP-1 deficiency decreased overall polyp number by 20% and specifically large polyp number by 45% (P < 0.05). This was consistent with an increase in apoptotic cells (P < 0.05), but there was no change detected in proliferation or β-catenin. MCP-1 deficiency decreased F4/80-positive cells in both the polyp tissue and surrounding intestinal tissue (P < 0.05) as well as expression of markers associated with M1 (IL-12 and IL-23) and M2 macrophages (IL-13, CD206, TGF-β, and CCL17) (P < 0.05). MCP-1 knockout was also associated with increased cytotoxic T lymphocytes and decreased regulatory T cells (P < 0.05). In addition, MCP-1(-/-) offset the increased mRNA expression of IL-1β and IL-6 in intestinal tissue and IL-1β and TNF-α in polyp tissue (P < 0.05), and prevented the decrease in SOCS1 expression (P < 0.05). We demonstrate that MCP-1 is an important mediator of tumor growth and immune regulation that may serve as an important biomarker and/or therapeutic target in colon cancer.
Collapse
Affiliation(s)
- Jamie L. McClellan
- 1Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina; ,2Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - J. Mark Davis
- 2Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - Jennifer L. Steiner
- 2Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - Reilly T. Enos
- 2Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - Seung H. Jung
- 2Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina;
| | - James A. Carson
- 2Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina; ,4Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Maria M. Pena
- 3Department of Biological Sciences, University of South Carolina, Columbia, South Carolina; and ,4Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Kevin A. Carnevale
- 1Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina;
| | - Franklin G. Berger
- 3Department of Biological Sciences, University of South Carolina, Columbia, South Carolina; and ,4Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - E. Angela Murphy
- 1Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina;
| |
Collapse
|
40
|
Pogribny IP, Beland FA. DNA methylome alterations in chemical carcinogenesis. Cancer Lett 2012; 334:39-45. [PMID: 23010082 DOI: 10.1016/j.canlet.2012.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 08/28/2012] [Accepted: 09/14/2012] [Indexed: 01/30/2023]
Abstract
Carcinogenesis, a complex multifactorial process of the transformation of normal cells into malignant cells, is characterized by many biologically significant and interdependent alterations triggered by the mutational and/or non-mutational (i.e., epigenetic) events. One of these events, specific to all types of cancer, is alterations in DNA methylation. This review summarizes the current knowledge of the role of DNA methylation changes induced by various genotoxic chemicals (carcinogenic agents that interact with DNA) and non-genotoxic carcinogens (chemicals causing tumor by mechanisms other than directly damaging DNA) in the lung, colorectal, liver, and hematologic carcinogenesis. It also emphasizes the potential role for epigenetic changes to serve as markers for carcinogen exposure and carcinogen risk assessment.
Collapse
Affiliation(s)
- Igor P Pogribny
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA.
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA.
| |
Collapse
|
41
|
Mao GE, Harris DM, Moro A, Heber D, Roy-Burman P, Zhang ZF, Rao J. A joint effect of new Western diet and retinoid X receptor α prostate-specific knockout with development of high-grade prostatic intraepithelial neoplasia in mice--a preliminary study. Prostate 2012; 72:1052-9. [PMID: 22314496 PMCID: PMC4321893 DOI: 10.1002/pros.22496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 01/11/2012] [Indexed: 12/31/2022]
Abstract
BACKGROUND The "New Western-style Diet" (NWD) characterized by high in fat and low in fiber, vitamin D, calcium, and methyl donors--are considered as a risk factor for prostate cancer. Previous studies have shown that premalignant lesions of human prostate have decreased expression of the Retinoid X Receptor alpha (RXRα). This study was to determine the effect of diet in RXRα knockout mice in developing high-grade prostate intraepithelial neoplasia (mPIN). METHODS Male mice (n = 54) with or without the RXRα prostate null mutation were fed either NWD or AIN-76A control diet for 10 months; prostates were harvested at 11 months of age and examined for prostate mPIN. RESULTS mPIN was seen in 79% of RXRα prostate null mice fed NWD (n = 19), 30.8% RXRα prostate null mice fed AIN-76A (n = 13), 42.9% RXRα wild-type mice fed NWD (n = 14), and 12.5% RXRα wild-type mice fed AIN-76A (n = 8). Unconditional Logistic analysis showed a significant joint effect of NWD and RXRα status in developing mPIN 26.3 (95% CI: 2.5-280), but interaction was not significant owing to the small sample size 1.6 (0.09-27.7, P = 0.7441). CONCLUSION This study provides preliminary data to support a joint RXRα-diet effect in prostate carcinogenesis.
Collapse
Affiliation(s)
- Gloria E. Mao
- Center for Human Nutrition, Los Angeles, California
- Department of Epidemiology, UCLA School of Public Health, UCLA, Los Angeles, California
| | | | - Aune Moro
- Center for Human Nutrition, Los Angeles, California
| | - David Heber
- Center for Human Nutrition, Los Angeles, California
| | - Pradip Roy-Burman
- Department of Pathology, Biochemistry and Molecular Biology, University of Southern California School of Medicine, Los Angeles, California
| | - Zuo-Feng Zhang
- Department of Epidemiology, UCLA School of Public Health, UCLA, Los Angeles, California
| | - Jianyu Rao
- Department of Epidemiology, UCLA School of Public Health, UCLA, Los Angeles, California
- Department of Pathology and Laboratory Medicine, UCLA David Geffen School of Medicine and Jonsson Comprehensive Cancer Center, Los Angeles, California
| |
Collapse
|
42
|
Epigenetic impact of dietary polyphenols in cancer chemoprevention: Lifelong remodeling of our epigenomes. Pharmacol Res 2012; 65:565-76. [DOI: 10.1016/j.phrs.2012.03.007] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 03/10/2012] [Accepted: 03/13/2012] [Indexed: 02/07/2023]
|
43
|
Ong TP, Moreno FS, Ross SA. Targeting the epigenome with bioactive food components for cancer prevention. JOURNAL OF NUTRIGENETICS AND NUTRIGENOMICS 2012; 4:275-92. [PMID: 22353664 DOI: 10.1159/000334585] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Epigenetic processes participate in cancer development and likely influence cancer prevention. Global DNA hypomethylation, gene promoter hypermethylation and aberrant histone post-translational modifications are hallmarks of neoplastic cells which have been associated with genomic instability and altered gene expression. Because epigenetic deregulation occurs early in carcinogenesis and is potentially reversible, intervention strategies targeting the epigenome have been proposed for cancer prevention. Bioactive food components (BFCs) with anticancer potential, including folate, polyphenols, selenium, retinoids, fatty acids, isothiocyanates and allyl compounds, influence DNA methylation and histone modification processes. Such activities have been shown to affect the expression of genes involved in cell proliferation, death and differentiation that are frequently altered in cancer. Although the epigenome represents a promising target for cancer prevention with BFCs, few studies have addressed the influence of dietary components on these mechanisms in vivo, particularly on the phenotype of humans, and thus the exact mechanisms whereby diet mediates an effect on cancer prevention remains unclear. Primary factors that should be elucidated include the effective doses and dose timing of BFCs to attain epigenetic effects. Because diet-epigenome interactions are likely to occur in utero, the impact of early-life nutrition on cancer risk programming should be further investigated.
Collapse
Affiliation(s)
- Thomas Prates Ong
- Laboratory of Diet, Nutrition and Cancer, Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| | | | | |
Collapse
|
44
|
Lee HS, Herceg Z. The epigenome and cancer prevention: A complex story of dietary supplementation. Cancer Lett 2012; 342:275-84. [PMID: 22266189 DOI: 10.1016/j.canlet.2012.01.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 01/08/2012] [Accepted: 01/15/2012] [Indexed: 12/16/2022]
Abstract
Epigenetic changes have been implicated in virtually all types of human malignancies. In contrast to genetic changes, epigenetic changes occur in a gradual manner during the tumorigenic process and they are potentially reversible. Because epigenetic changes have frequently been detected in high-risk populations, they are attractive targets to prevent the initiation of premalignant lesions or their advance to a malignant stage. A wide range of chemical entities has been found capable of altering the epigenome in animal models and humans. Epidemiological and laboratory-based studies suggested that these agents may have an anti-neoplastic effect against different cancer types. Several of these agents have been tested as dietary supplements, often with conflicting results. In this review, we discuss recent developments in our understanding of agents capable of modulating the epigenome and their potential to prevent human cancer when administered as dietary supplements.
Collapse
Affiliation(s)
- Ho-Sun Lee
- Epigenetics Group, International Agency for Research on Cancer (IARC), 150 Cours Albert-Thomas, 69372 Lyon Cedex 08, France
| | | |
Collapse
|
45
|
Gerhauser C. Cancer chemoprevention and nutriepigenetics: state of the art and future challenges. Top Curr Chem (Cham) 2012; 329:73-132. [PMID: 22955508 DOI: 10.1007/128_2012_360] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term "epigenetics" refers to modifications in gene expression caused by heritable, but potentially reversible, changes in DNA methylation and chromatin structure. Epigenetic alterations have been identified as promising new targets for cancer prevention strategies as they occur early during carcinogenesis and represent potentially initiating events for cancer development. Over the past few years, nutriepigenetics - the influence of dietary components on mechanisms influencing the epigenome - has emerged as an exciting new field in current epigenetic research. During carcinogenesis, major cellular functions and pathways, including drug metabolism, cell cycle regulation, potential to repair DNA damage or to induce apoptosis, response to inflammatory stimuli, cell signalling, and cell growth control and differentiation become deregulated. Recent evidence now indicates that epigenetic alterations contribute to these cellular defects, for example epigenetic silencing of detoxifying enzymes, tumor suppressor genes, cell cycle regulators, apoptosis-inducing and DNA repair genes, nuclear receptors, signal transducers and transcription factors by promoter methylation, and modifications of histones and non-histone proteins such as p53, NF-κB, and the chaperone HSP90 by acetylation or methylation.The present review will summarize the potential of natural chemopreventive agents to counteract these cancer-related epigenetic alterations by influencing the activity or expression of DNA methyltransferases and histone modifying enzymes. Chemopreventive agents that target the epigenome include micronutrients (folate, retinoic acid, and selenium compounds), butyrate, polyphenols from green tea, apples, coffee, black raspberries, and other dietary sources, genistein and soy isoflavones, curcumin, resveratrol, dihydrocoumarin, nordihydroguaiaretic acid (NDGA), lycopene, anacardic acid, garcinol, constituents of Allium species and cruciferous vegetables, including indol-3-carbinol (I3C), diindolylmethane (DIM), sulforaphane, phenylethyl isothiocyanate (PEITC), phenylhexyl isothiocyanate (PHI), diallyldisulfide (DADS) and its metabolite allyl mercaptan (AM), cambinol, and relatively unexplored modulators of histone lysine methylation (chaetocin, polyamine analogs). So far, data are still mainly derived from in vitro investigations, and results of animal models or human intervention studies are limited that demonstrate the functional relevance of epigenetic mechanisms for health promoting or cancer preventive efficacy of natural products. Also, most studies have focused on single candidate genes or mechanisms. With the emergence of novel technologies such as next-generation sequencing, future research has the potential to explore nutriepigenomics at a genome-wide level to understand better the importance of epigenetic mechanisms for gene regulation in cancer chemoprevention.
Collapse
Affiliation(s)
- Clarissa Gerhauser
- Division Epigenomics and Cancer Risk Factors, German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
46
|
Kim M, Bae M, Na H, Yang M. Environmental toxicants--induced epigenetic alterations and their reversers. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2012; 30:323-367. [PMID: 23167630 DOI: 10.1080/10590501.2012.731959] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Epigenetics has been emphasized in the postgenome era to clarify obscure health risks of environmental toxicants including endocrine disrupting chemicals (EDCs). In addition, mixed exposure in real life can modify health consequences of the toxicants. Particularly, some nutritional and dietary materials modify individual susceptibility through changes in the epigenome. Therefore, we focused on some environmental toxicants that induce epigenetic alterations, and introduced chemopreventive materials to reverse the toxicants-induced epigenetic alterations. Methodologically, we used global and specific DNA methylation as epigenetic end points and searched epigenetic modulators in food. We reviewed various epigenetic end points induced by environmental toxicants including alcohol, asbestos, nanomaterials, benzene, EDCs, metals, and ionizing radiation. The epigenetic end points can be summarized into global hypomethylation and specific hypermethylation at diverse tumor suppress genes. Exposure timing, dose, sex, or organ specificity should be considered to use the epigenetic end points as biomarkers for exposure to the epimutagenic toxicants. Particularly, neonatal exposure to the epimutagens can influence their future adult health because of characteristics of the epimutagens, which disrupt epigenetic regulation in imprinting, organogenesis, development, etc. Considering interaction between epimutagenic toxicants and their reversers in food, we suggest that multiple exposures to them can alleviate or mask epigenetic toxicity in real life. Our present review provides useful information to find new end points of environmental toxicants and to prevention from environment-related diseases.
Collapse
Affiliation(s)
- Minju Kim
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Yongsan-gu, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
47
|
Giardina C, Madigan JP, Tierney CAG, Brenner BM, Rosenberg DW. Vitamin D resistance and colon cancer prevention. Carcinogenesis 2011; 33:475-82. [PMID: 22180570 DOI: 10.1093/carcin/bgr301] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Observational studies have been largely consistent in showing an inverse association between vitamin D and an individual's risk of developing colorectal cancer. Vitamin D protection is further supported by a range of preclinical colon cancer models, including carcinogen, genetic and dietary models. A large number of mechanistic studies in both humans and rodents point to vitamin D preventing cancer by regulating cell proliferation. Counterbalancing this mostly positive data are the results of human intervention studies in which supplemental vitamin D was found to be ineffective for reducing colon cancer risk. One explanation for these discrepancies is the timing of vitamin D intervention. It is possible that colon lesions may progress to a stage where they become unresponsive to vitamin D. Such a somatic loss in vitamin D responsiveness bears the hallmarks of an epigenetic change. Here, we review data supporting the chemopreventive effectiveness of vitamin D and discuss how gene silencing and other molecular changes somatically acquired during colon cancer development may limit the protection that may otherwise be afforded by vitamin D via dietary intervention. Finally, we discuss how understanding the mechanisms by which vitamin D protection is lost might be used to devise strategies to enhance its chemopreventive actions.
Collapse
Affiliation(s)
- Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, 91 North Eagleville Road, U3215, Storrs, CT 06269, USA.
| | | | | | | | | |
Collapse
|
48
|
Singh BN, Shankar S, Srivastava RK. Green tea catechin, epigallocatechin-3-gallate (EGCG): mechanisms, perspectives and clinical applications. Biochem Pharmacol 2011; 82:1807-21. [PMID: 21827739 DOI: 10.1016/j.bcp.2011.07.093] [Citation(s) in RCA: 1047] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/22/2011] [Accepted: 07/25/2011] [Indexed: 12/22/2022]
Abstract
An expanding body of preclinical evidence suggests EGCG, the major catechin found in green tea (Camellia sinensis), has the potential to impact a variety of human diseases. Apparently, EGCG functions as a powerful antioxidant, preventing oxidative damage in healthy cells, but also as an antiangiogenic and antitumor agent and as a modulator of tumor cell response to chemotherapy. Much of the cancer chemopreventive properties of green tea are mediated by EGCG that induces apoptosis and promotes cell growth arrest by altering the expression of cell cycle regulatory proteins, activating killer caspases, and suppressing oncogenic transcription factors and pluripotency maintain factors. In vitro studies have demonstrated that EGCG blocks carcinogenesis by affecting a wide array of signal transduction pathways including JAK/STAT, MAPK, PI3K/AKT, Wnt and Notch. EGCG stimulates telomere fragmentation through inhibiting telomerase activity. Various clinical studies have revealed that treatment by EGCG inhibits tumor incidence and multiplicity in different organ sites such as liver, stomach, skin, lung, mammary gland and colon. Recent work demonstrated that EGCG reduced DNMTs, proteases, and DHFR activities, which would affect transcription of TSGs and protein synthesis. EGCG has great potential in cancer prevention because of its safety, low cost and bioavailability. In this review, we discuss its cancer preventive properties and its mechanism of action at numerous points regulating cancer cell growth, survival, angiogenesis and metastasis. Therefore, non-toxic natural agent could be useful either alone or in combination with conventional therapeutics for the prevention of tumor progression and/or treatment of human malignancies.
Collapse
Affiliation(s)
- Brahma N Singh
- Department of Pharmacology, Toxicology and Therapeutics, and Medicine, The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | | | | |
Collapse
|
49
|
Zhang F, Meng F, Li H, Dong Y, Yang W, Han A. Suppression of retinoid X receptor alpha and aberrant β-catenin expression significantly associates with progression of colorectal carcinoma. Eur J Cancer 2011; 47:2060-7. [PMID: 21561764 DOI: 10.1016/j.ejca.2011.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Revised: 04/01/2011] [Accepted: 04/05/2011] [Indexed: 01/27/2023]
Abstract
To investigate retinoid X receptor alpha (RXRα) and β-catenin expression and their relationship with the clinicopathological features of colorectal carcinoma (CRC). Real-time PCR and western blot analyses revealed that β-catenin and RXRα expression at both mRNA and protein levels in four pairs of fresh CRC and adjacent non-tumour tissues (ANT) dramatically was increased and decreased in CRC compared with ANT, respectively. Furthermore, RXRα expression at both mRNA and protein levels was downregulated in higher histological grade CRC. Immunohistochemistry staining in 120 cases of CRC and 60 cases of lymph node metastatic carcinoma of CRC showed that RXRα expression was significantly suppressed in CRC compared with ANT (P<0.001) and low expression of RXRα in CRC was significantly associated with histological grade (P<0.001), TNM stage (P=0.022) and N classification (P=0.002). The aberrant (accumulated cytoplasm or/and nuclei) expression of β-catenin was higher in CRC than that in ANT (P<0.001) and associated with histological grade (P=0.001) and N classification (P=0.002). Moreover, there was a close relationship between low RXRα expression and aberrant β-catenin expression in CRC (P=0.032). Taken together with our previous study, aberrant β-catenin expression upregulated by suppression of RXRα may play a crucial role in pathogenesis and progression of CRC.
Collapse
Affiliation(s)
- Fenfen Zhang
- Department of Pathology, The First Affiliated Hospital and Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
50
|
Yang CS, Wang H. Mechanistic issues concerning cancer prevention by tea catechins. Mol Nutr Food Res 2011; 55:819-31. [PMID: 21538856 DOI: 10.1002/mnfr.201100036] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 03/04/2011] [Accepted: 03/24/2011] [Indexed: 12/12/2022]
Abstract
The cancer preventive activities of tea (Camellia sinensis, Theaceae) have been demonstrated in animal models for cancers at different organ sites and suggested by some epidemiological studies. Many mechanisms for cancer prevention have been proposed based on studies in cell lines, which demonstrated the modulation of signal transduction and metabolic pathways by (-)-epigallocatechin-3-gallate (EGCG), the most abundant and active polyphenol in green tea. These molecular events may result in cellular changes, such as enhancement of apoptosis, suppression of cell proliferation, and inhibition of angiogenesis. Nevertheless, it is not known whether these are the molecular mechanisms of inhibition of carcinogenesis in animals and humans. This article discusses the key issues involved in extrapolating results from cell line studies to mechanistic information in vivo and in translating animal studies to human cancer prevention.
Collapse
Affiliation(s)
- Chung S Yang
- Department of Chemical Biology and Center for Cancer Prevention Research, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854-8020, USA.
| | | |
Collapse
|