Abstract
Cancer development takes 10–50 years and epigenetics plays an important role. Recent evidence suggests that ~80% of human cancers are linked to environmental factors impinging upon genetics/epigenetics. Since advanced metastasized cancers are resistant to radiation/chemotherapeutic drugs, cancer prevention by relatively non-toxic “epigenetic modifiers” will be logical. Many dietary phytochemicals possess powerful antioxidant and anti-inflammatory properties that are hallmarks of cancer prevention. Dietary phytochemicals can regulate gene expression of the cellular genome via epigenetic mechanisms. In this review, we will summarize preclinical studies that demonstrate epigenetic mechanisms of dietary phytochemicals in skin, colorectal, and prostate cancer prevention. Key examples of the importance of epigenetic regulation in carcinogenesis include hypermethylation of the NRF2 promoter region in cancer cells, resulting in inhibition of NRF2-ARE signaling. Many dietary phytochemicals demethylate NRF2 promoter region and restore NRF2 signaling. Phytochemicals can also inhibit inflammatory responses via hyper-methylation of inflammation-relevant genes to block gene expression. Altogether, dietary phytochemicals are excellent candidates for cancer prevention due to their low toxicity, potent antioxidant and anti-inflammatory properties, and powerful epigenetic effects in reversing pro-carcinogenic events.
Keywords: Skin cancer, colorectal cancer, prostate cancer, dietary phytochemicals, epigenetic modulation, anti-oxidation, anti-inflammation
Introduction
Is cancer a preventable disease? Cancer is a complex chronic disease and cancer development comprises multistep processes 1, 2, involving initiation, promotion, progression, and metastasis 3, 4. Recent evidence suggests that ~80% of human cancers are linked to environmental factors impinging upon the fidelity of the underlying genetic information 4–7. Cancer development is a long process that typically takes 10–50 years (depending on the tissue/organ), and excessive oxidative stress and inflammation are regarded as vital determinants driving cancer development 8–10. Since advanced metastasized cancers are resistant to radiation and chemotherapeutic drugs, prevention of early stages of cancer by relatively non-toxic, potent antioxidative and anti-inflammatory dietary phytochemicals would be logical.
During cancer development, particularly during the “promotion” stage, epigenetics has been postulated to play a vital role in driving cellular transformation such as stem cells forming benign microscopic tumors 11. Feinberg et al. in the 1980s reported that most if not all tumors could be associated with widespread losses and some gains of DNA methylation throughout the genome 12, 13. This concept was reviewed recently 6, 14 and implicated the role of epigenetics during cancer development.
Epigenetics influences many cellular activities, including cell growth and disease development 15. Epigenetic mechanisms mediate gene activation/inhibition in response to environmental cues driving three major processes: DNA methylation, histone modification, and noncoding RNA expression16, 17. DNA methylation is a chemical modification via DNA methyltransferases (DNMTs) that add methyl groups to DNA molecules to modulate gene expression. Abnormal DNA methylation, such as hypermethylation and hypomethylation is considered a hallmark of cancer development. Hypermethylation of key tumor suppressor genes results in the “silencing” of the genes which drive cancer development18, 19. Histone modifications play the role of the “switch” and are post-translational modifications (acetylation/deacetylation and methylation/ demethylation) of histones by which the linkage between DNA molecules and histone proteins changes to either tighten or loosen the interaction, further affecting the gene expression20. MicroRNAs (miRNAs) are one of the noncoding RNAs involved in epigenetics and can be classified into tumor-promoting and tumor-suppressing miRNAs, which are upregulated and downregulated during cancer development21, 22. The interwind of cellular signaling pathways and epigenetics can also affect genomic alteration in cells23. For instance, the KEAP1-NRF2 signaling pathway is associated with the cellular defense system against oxidative stress. Hypermethylation of the NRF2 promoter is observed in several types of tumors and causes cancer progression24.
Phytochemicals are bioactive ingredients derived from various plants and herbs 25. Dietary phytochemicals stem from vegetables, fruits, grains, and culinary herbs; therefore, phytochemicals in general have low toxicity when consumed long term 26, 27. They can exert antioxidant, anti-inflammatory, and anti-angiogenic properties, as well as function against cancer development via regulating receptors, ion channels, ion pumps, cytoskeletons, and transcriptional machinery in cells28–30. Additionally, recent research reveals that many dietary phytochemicals would possess epigenetic-modifying abilities 31. This review aims to summarize preclinical evidence of cancer prevention elicited by dietary phytochemicals via epigenetic mechanisms in skin, colorectal, and prostate cancers.
Natural sources of dietary phytochemicals
Dietary phytochemicals are derived from natural sources, for example, vegetables, fruits, and medical herbs. Phytochemicals are divided into primary and secondary metabolites produced by plant metabolism and they can perform bioactive activities upon oral ingestion and they play a vital role in disease prevention and reduction in risk factors via their innate attribution 32. In general, phytochemicals can be classified into five main categories: (1) phenolic phytochemicals; (2) terpenoids; (3) glucosinolates; (4) polyacetylene; (5) phytosterols and phytostanols33.
Phenolic phytochemicals account for 45% of known phytochemicals and are further classified as phenolic acids, flavonoids, stilbenes, and lignans 33. Under flavonoids, there are two subgroups, anthocyanins and anthoxanthins. Anthoxanthins include flavonols, flavones, flavanones, flavanols, and isoflavones 34 and unlike anthocyanins that are responsible for the emission of red, blue, and purple colors in vegetables, anthoxanthins, covering fiver major subclasses, are colorless or white to yellow compounds 33, 35. Flavonoids possess antioxidant and anti-inflammatory activities that contribute to, inhibition of carcinogenesis, involving induction of cell cycle arrest or apoptosis, regulation of the host immune system, and changes in cellular signaling pathways 36. Examples of phenolic phytochemicals include curcumin, luteolin, resveratrol, pelargonidin, and more.
Terpenoids are bioactive compounds of essential oils that can be extracted from roots, seeds, and other parts of plants 37. Biochemically, terpenoids are terpenes modified with various functional groups and they are secondary metabolites of aromatic and medicinal plants 38. Terpenoids have important biological activities, such as antioxidant, anti-inflammatory, neuroprotective abilities, and anti-cancer effects, including induction of apoptosis, inhibiting proliferation, and inhibiting tumor growth 39. Fucoxanthin, ursolic acid, and corosolic acid are examples of terpenoids.
Glucosinolates are sulfur-containing glucosides that are widely found in cruciferous vegetables and oilseeds 33. The bioactive compounds, indole, allylic sulfur compounds, and isothiocyanates, are encompassed within glucosinolates 40. The most common example is sulforaphane, which is a well-known activator of the NRF2-ARE signaling pathway and an HDAC modulator 41.
Phytosterols and phytostanols are plant sterols and the human body is unable to synthesize them; therefore, the only source is from dietary ingestion, such as vegetable oils, cereals, nuts, and seeds 42. The health effects of phytosterols and phytostanols include lowering cholesterol levels, cancer prevention, and immunomodulation 33. Polyacetylenes are emerging phytochemicals involved in cancer prevention and are found in apiaceous vegetables. Polyacetylenes have antioxidant and anti-inflammatory properties. The involving cellular mechanisms and molecular pathways of polyacetylene action are identified as the NF-κB pathway, antioxidant response elements, regulation of the cell cycle, and apoptosis 43.
Considering the overall strategy in cancer prevention, consuming vegetables and fruits is a convenient and logical way to ingest a wide range of phytochemicals that can provide cancer prevention in the general population. For higher-risk individuals, pharmacological doses of a combination of bioactive phytochemicals with low toxicity would be a logical approach 44, 45.
Skin cancer prevention by dietary phytochemicals
The development of skin cancer is initiated by diverse drivers, such as DNA damage, chronic inflammation, suppression of the immune system, photoaging, and/or mutations. The process is accompanied by genetic and epigenetic changes, which further trigger multiple signaling pathways that drive skin carcinogenesis 46. Non-melanomatous skin cancer (NMSC) is one of the most prevalent cancers in the world47. Basal cell carcinoma and squamous cell carcinoma are the two most commonly diagnosed types of NMSC accounting for 99% of all NMSC cases48. Exposure to ultraviolet (UV) radiation and environmental chemicals/pollutants, for example, arsenic and benzo[a]pyrene (B[a]P), are the most common known causes of NMSC49. Recently, our research revealed that exposures to UV or benzo[a]pyrene (B[a]P) and 12-O-tetradecanoylphorbol-13-acetate (TPA) led to DNA methylomic and transcriptomic changes at different stages of skin carcinogenesis50, 51. Additionally, increasing research supports alterations in cellular metabolism related to the epigenetic machinery is further linked to skin cancer development.
Polyphenols
Several polyphenols have activity against skin cancer. Tea catechins are known to re-activate silenced tumor suppressor genes, p16INK4a, and Cip1/p21 via epigenetic regulations including decreased global DNA methylation and increased histone acetylation (H3-Lys 9 and 14; H4-Lys 15,12 and 16) levels in A431 epidermoid carcinoma cells 52.
The topical application of apigenin has long been recognized as a potential chemoprevention strategy against skin carcinogenesis induced by TPA or UV irradiation in susceptible mouse strains 53, 54. Later studies with apigenin have revealed a demethylation effect coupled with attenuated DNMT and HDAC activity at 15 CpG sites in the NRF2 promoter in JB6 P+ cells 55.
Pelargonidin has been shown to possess excellent potential in blocking TPA-induced cell transformation by reducing protein levels of genes encoding DNMTs and HDACs 56 and further decreasing DNA methylation in the NRF2 promoter region of JB6 P+ cells 56.
Isothiocyanates
Several isothiocyanates have demonstrated potential in restoring the epigenetic landscape that contributes to skin carcinogenesis 57, 58. Sulforaphane (SFN), a well-studied anti-cancer phytochemical, has been shown to reactivate NRF2 through the downregulation of DNMTs and HDACs in TPA-exposed JB6 P+ epidermal cells 41. Both sulforaphane and tea polyphenols were found to suppress the expression of Bmi-1 and Ezh2, the highly expressed polycomb group proteins (PcG) and regulators of chromatin remodeling in skin cancer 59–62.
We reported the chemopreventive epigenetic effects of moringa isothiocyanate (MIC-1) in TPA-challenged JB6 P+ cells. We identified differentially methylated regions and differentially expressed genes, including the cancer-related genes Tmpt, Tubb3, and Muc2, the GTPases Gchfr and Igtp, and the cell cycle-related gene Cdc7 63. The Muc2 gene is reported to be induced by inflammatory factors (IL-1β, TNF𝛼) via the NF-κB pathway64. The correlative analysis between transcriptome gene expression and the DNA methylome showed that MIC-1 treatment led to hypermethylation and downregulation of the Muc2 gene63.
Terpenoids
We found that the synthetic triterpenoid CDDO (2-cyano 2,3-dioxoolean-1,9-dien-28-oic acid) blocked TPA-induced skin cell transformation dose-dependently at methylome and transcriptome levels in JB6 P+ cells. Five-day treatment (CDDO dose of 50 nM) revealed that LYL1 basic helix-loop-helix family members- Lyl1, Lad1, and Dennd2d genes-were the most significantly differentially expressed genes after CDDO treatment. The TPA-induced methylation status of Tmem253, Bco2, and Madd also was reversed by CDDO. Furthermore, CDDO significantly restored the NRF2-ARE pathway of Nqo1 that was inhibited by TPA by decreasing methylation of its CpG promoter65. We also determined that fucoxanthin (FX) reversed the TPA-induced transformation of JB6 P+ cells by decreasing methylation of the NRF2 promoter region and significantly reducing DNMT activity but not affecting HDAC activity 66.
An integrative study of the CpG methylome and RNA transcriptome after treatment with ursolic acid (UA) showed increased activity of antioxidant, anti-inflammatory, and anticancer pathways in UVB-induced nonmelanoma skin carcinogenesis. Yang et al. observed that central antioxidant genes, such as NRF2 and NQO1 were upregulated by UA treatment in the early phase of UVB-induced carcinogenesis, and significant hypomethylation of CpG sites of these genes was also revealed by the methylation analysis 67.
An epigenetic study with corosolic acid (CA) revealed novel molecular targets for the prevention of early stages of skin cancer. The results of methylation sequencing showed that biomarkers such as Smad-3, Tasp1, Uri1, Nsg2, Madd, Dusp22, and Rassf were hypermethylated by TPA challenge and were hypomethylated by CA treatment 68.
Lotus leaf ethanolic extract
Lotus leaf ethanolic extract is known to reduce the neoplastic transformation of JB6 P+ cells, potentially by activating the NRF2 pathway and regulating epigenetic DNA methylation and histone acetylation 69. Several canonical signaling pathways were unveiled by Ingenuity Pathway Analysis (IPA) analysis indicating that the application of these phytochemicals was able to inhibit inflammatory response pathways (NF-κB signaling, IL-1 signaling) and activate NRF2-mediated antioxidative response in the skin cancer model63, 68. The studies of using phytochemicals to prevent skin cancer carcinogenesis are listed in Table 1.
Table 1.
Phytochemicals | Cellular/ Animal Model | Epigenetic Mechanism | Reference |
---|---|---|---|
Tea catechins | A431 cells | Restore silenced tumor suppressor genes, p16INK4a and Cip1/p21 via decreasing global DNA methylation and increasing histone acetylation | 52 |
Apigenin | JB6 P+ cells | Demethylation coupled with reduction of DNMT and HDAC activity. | 55 |
Pelargonidin | TPA-induced JB6 P+ cells | Reduce protein levels of genes encoding DNMTs and HDACs and decrease DNA methylation in the Nrf2 promoter region. | 56 |
Sulforaphane | TPA-induced JB6 P+ cells | Reactive Nrf2 via downregulation of DNMTs and HDACs | 41 |
Moringa isothiocyanate | TPA-induced JB6 P+ cells | MIC-1 treatment led to hypermethylated and downregualted on the Muc2 gene. | 63 |
CDDO | TPA-induced JB6 P+ cells | Restore Nrf2-ARE pathway by reducing methylation of Nqo1 promoter region. | 65 |
Fucoxanthin | TPA-induced JB6 P+ cells | Decreasse methylation of the Nrf2 promoter region and significantly reduce DNMT activity. | 66 |
Ursolic acid | UVB-exposed female SKH-1 hairless mice | CpG sites of key antioxidant genes (Nrf2, Nqo1) were significantly reversed and hypomethylated by UA treatment in early UVB-induced carcinogenesis. | 67 |
Corosolic acid | TPA-induced JB6 P+ cells | Potential biomarker genes for early skin carcinogenesis such as Smad-3, Tasp1, Uri1, Nsg2, and more were hypermehtylated and hypomethylated by CA treatment. | 68 |
Lotus leaf ethanolic extract | JB6 P+ cells | Activate the NRF2 pathway and regulate epigenetic DNA methylation and histone acetylation. | 69 |
Colorectal cancer prevention by dietary phytochemicals
Colorectal cancer (CRC) is the third most commonly diagnosed cancer in the world and has a high incidence and mortality rate70. In the United States, according to the American Cancer Society (ACS), nearly double the number of young adults under 55 are being diagnosed with CRC today compared to a decade ago, and more are dying from the disease each year, hence there is an urgent need to identify causing factors and preventive strategy. BRAF mutations, microsatellite instability, KRAS mutations, and PIK3CA mutations all are implicated in CRC development. Dysregulation of the gene products such as growth factors, growth factor receptors, protein kinase, inflammatory cytokines, inflammatory enzymes, proapoptotic proteins, anti-apoptotic proteins, tumor suppressors, transcription factors, and their relevant signaling pathways are reported to be involved in carcinogenesis71–73.
One widely used rodent model is azoxymethane (AOM) and dextran sulfate sodium (DSS) driven colitis- associated CRC. AOM functions as a carcinogen inducing aberrant crypt foci, and DSS is an inflammatory agent damaging the colonic epithelium74, 75. Therefore, the AOM/DSS-driven mouse model is often used to induce inflammation and tumor initiation in CRC. Intractable epigenetic alteration has been linked to CRC development. In particular, aberrant methylation of the regulators is observed in an AOM/DSS-induced CRC or a DSS-induced inflammation mouse model76. Lipopolysaccharide (LPS) is a unique component of the outer cell membrane of the gram-negative bacteria and it induces inflammation via activating the Toll-like receptor 4 (TLR4)-mediated signaling pathways in intestinal epithelia cells77. Moreover, CRC patients are observed having higher LPS concentration in blood and CRC tissues than the healthy people. Consequently, LPS-challenged cellular model are often used in vitro78, 79.
Polyphenols
Guo et al. demonstrated that curcumin (CUR) reduced methylation of the promoter region of the tumor suppressor gene - DLEC1 in human colon cancer HT29 cells through its demethylating effects. In addition, curcumin decreased protein expression of DNA methyltransferases and histone deacetylases80. Guo et al. subsequently confirmed the effect of curcumin on the epigenome on the inflammatory response in the AOM-DSS mouse model of colorectal cancer. Through a series of experiments including SureSelect methyl-seq and RNA-seq, they found that curcumin restored AOM-DSS-induced hypomethylation of Tnf. The hypomethylated state of inflammation-relevant genes such as Duoxa2, Gja1, Icam1, Igfbp4, Itgb2, Lgals9, and Pf4 were also reversed by curcumin, inhibiting the abnormally high expression of these genes induced by AOM/DSS81.
In a study of the flavonoid dietary phytochemical luteolin (LUT), the epigenetic regulation of the NRF2-ARE pathway by LUT in HCT116 cells was studied. LUT was able to reduce the methylation of the NRF2 promoter region, leading to significant changes in the mRNA and protein expression levels of genes in NRF2 and its related genes (Ho-1 and Nqo1). Additionally, LUT treatment reduced protein level and enzymatic activities of DNMTs and HDACs which were associated with regulation of NRF2. All these results pointed to the potential anticancer activity of LUT82.
Resveratrol, found in peanuts, red grapes, blue barriers, and rasberries, has been shown to prevent inflammation-driven colorectal cancer by altering the expression of miRNA-101b and miRNA-455 and down-regulating inflammatory stress markers such as p5383, 84. Altamemi et al. reported that resveratrol treatment reduced protein levels of IL-6 and TNF-𝛼 and raised the expression of two anti-inflammatory miRNAs, miRNA-101b and miRNA- 455 in a DSS-induced colitis mouse model 84.
Isothiocyanates
In human colon cancer HCT116 and SW8409 cell lines, sulforaphane (SFN) or its analogs affect HAT/HDAC activity and decrease the expression of HDAC3, P300/CBP-associated factors, as well as KAT2A/GCN5 and DNMT1, while enhancing acetylation and degradation of repair proteins such as CtIP85. Additionally, SFN treatment in Caco-2 cells demethylated the NRF2 promoter region and thus activated the NRF2 signaling pathway. It was observed that DNMT expression was reduced by SFN treatment86.
In Liu and Dey’s study, a 0.12% phenethyl Isothiocyanate (PEITC)-enriched diet fed to mice lowered inflammation of the colonic mucosa and submucosa during AOM/DSS-induced colitis. Further in vitro experiments suggested the underlying mechanism was related to the inhibition of NF𝜅B1 protein by PEITC treatment. Epigenetically, the mRNA expression of NF𝜅B1 displayed an inverse correlation with tri-methylation of lysine 27 on histone 3 near the NF𝜅B1 promoter region in a time-dependent manner. PEITC increased the level of H3K27me3 with a rise in the expression on the NF𝜅B1 gene. PEITC mitigated colon carcinogenesis via modulating NF𝜅B1 signaling87.
Betalain
The betaine pigment indicaxanthin (IND) from cactus fruit exhibited anti-inflammatory and cytotoxic activity in a variety of colorectal cancer cell lines. Ragusa et al. concluded that IND affected autophagic activity by promoting the demethylation of CpG islands in promoters of ATG7 and ATG3 in Caco-2 cells. In addition, the up-regulation of the expression of LC3-II and Beclin1 by IND can further promote the formation and fusion of autophagosomes in Caco-2 cells88.
Water extract of Ilex rotunda (WIR)
A standardized water extract of Ilex rotunda (WIR) was analyzed to evaluate the potential role of a microRNA (miRNA)-dependent mechanism for the prevention of colon cancer. WIR, with its rich content of triterpenoids, restored the up-regulated levels of miR-31–5p that were triggered by AOM/DSS administration in C57BL/6 mice and inhibited ectopic expression of LATS2 and YAP genes that were regulated by miR-31–5p89.
Through epigenetic modulation, phytochemicals regulate the NRF2-ARE signaling pathway to exert antioxidant effects and mediate the NF𝜅B signaling pathway against inflammation. Table 2 lists the phytochemicals that have been shown to have potential for preventing colorectal cancer.
Table 2.
Phytochemicals | Cellular/ Animal Model | Epigenetic Mechanism | Reference |
---|---|---|---|
Curcumin | HT-29 cells | Demethylate the promoter region of DLEC1. CUR treatment lowers protein level of DNMTs and some HDACs. | 80 |
Curcumin | Male C57BL/6 mice challenged by AOM/DSS | Restore AOM-DSS-induced hypomethylation of Tnf and reverse hypomethylation of several inflammation relevant genes. | 81 |
Luteolin | HCT116 cells | Reduce methylation of Nrf2 promoter region and activate the Nrf2-ARE signaling pathway. LUT treatment declined DNMTs and HDACs. | 82 |
Resveratrol | DSS-induced colitis in Male ApcMin/+ C57BL/6 mice | Increase the level of two anti-inflmmatory miRNAs: miRNA-101b and miRNA- 455. | 84 |
Sulforaphane or its analogs | HCT116 cells and SW8409 cell | Regulate HAT/ HDAC activities and the relevant DNA repair/ damage singaling pathways. | 85 |
Sulforaphane | Caco-2 cells | Demethylate the Nrf2 promoter region and activate the Nrf2 signaling pathway. Reduce expression of DNMT. | 86 |
Phenethyl Isothiocyanate | Male C57BL/6 mice challenged by AOM/DSS; SW480 cells | Increase the methylation near the NF𝜅B1 promoter region and thus decrese the mRNA expression of NF𝜅B1. | 87 |
Indicaxanthin | Caco-2 cells | Hypomethylate the promoter regions of ATG7 and ATG3 and further promote autophagic activity. | 88 |
Water extract of Ilex rotunda | C57BL/6 mice challenged by AOM/DSS | Restore the level of miR-31–5p raised by AOM and DSS to further inhibit YAP accumulation in the nuclesu. | 89 |
Prostate cancer prevention by dietary phytochemicals
Prostate cancer (PCa) is a leading cause of male death associated with cancer in the United States90. The development from prostatic intraepithelial neoplasia to androgen-independent invasive carcinoma is a long-term process, which may take years to decades91. During tumor progression, genetic and epigenetic alterations are involved92. Two murine models have been developed to study PCa. (1) Transgenic adenocarcinoma of mouse prostate (TRAMP) mouse model is commonly used. TRAMP mice, incorporating SV40 early-region tumor antigens, mimic human prostate carcinogenesis and exhibit tumor transformation via interactions with tumor suppressor gene products93, 94. The NRF2-ARE signaling pathway is reported to be gradually downregulated during tumorigenesis in TRAMP mice95. (2) Phosphatase and tensin homolog located on chromosome 10 (PTEN) mouse model is another commonly used PCa model. PTEN is a tumor suppressor gene and one of the most frequently mutated/deleted genes in PCa96. PTEN deletion has been strongly linked to inflammation. In prostate-specific PTEN null mice, the expression of CXCL8/IL-8, a pro-inflammatory chemokine promoting tumorigenesis is increased 97. More importantly, PTEN deletion impacts the epigenome and transcriptome of prostate cells. This hypothesis was examined in a prostate-specific PTEN-KO mouse prostatic adenocarcinoma model through DNA methyl-Seq and RNA-Seq analyses. Loss of PTEN drove global changes in DNA CpG methylation and transcriptomic gene expression and was strongly associated with activation of several inflammatory and immune molecular pathways during PCa development98. These findings yield biomarkers of the critical molecular pathways that can be targeted by phytochemicals via epigenetic regulation for the prevention and treatment of PCa in human trials.
Polyphenols
Li et al. investigated the epigenetic effects of the synthetic curcumin analog FN1 in TRAMP C1 cells. FN1 was synthesized by coupling pyridyl aldehyde with tetrahydrothiopyran-4-one.99. FN1 treatment in TRAMP C1 cells demethylated the NRF2 promoter region, restored NRF2 expression, and increased the level of downstream genes, such as NQO-1, HO-1, and UGT1A1. FN1 also significantly reduced levels of DNMT1, DNMT3a, DNMT3b, and HDAC 493. The investigation of the effects of other synthetic curcumin derivatives, E10 and F10 was conducted. E10 and F10 were synthesized by coupling the substituted benzaldehyde with tetrahydropyran-4-ones and tetrahydrothiopyran-4-one, respectively99. The results showed that both compounds were more potent in increasing NRF2 expression than curcumin and SFN. F10 mitigated the summation of H3k27me3 and induced hypomethylation on the NRF2 promoter. F10 also downregulated DNMTs (DNMT1, DNMT3a, DNMT3b) and HDACs (HDAC1, HDAC4, and HDAC7). F10 can restore the NRF2 signaling pathway via demethylation, reduction of DNMTs and HDACs, and suppression of H3k27me3 accumulation 100.
Isothiocyanates
In the study by Zhang et al., Sulforaphane (SFN) treatment demethylated the first five CpGs of the NRF2 promoter in TRAMP C1 cells and activated mRNA and protein levels of NRF2 and NQO-1. SFN treatment attenuated DNMT1, DNMT3a, and HDACs 1, 4, 5, and 7 at the protein level while enhancing acetyl-histone 3 levels which bound to the NRF2 promoter101. SFN exerted its preventive effects via epigenetically modulating the NRF2 promoter region and re-activating the NRF2-ARE signaling pathway in TRAMP C1 cells.
The study using TRAMP mice by Wu et al. observed that the phenethyl Isothiocyanate (PEITC) diet reversed or attenuated the induction of cell cycle/Cdc42 signaling, inflammation, and cancer-related signaling in the prostate tissues of TRAMP mice. Pathway analysis revealed differences in signaling between wild-type and TRAMP mice, including pancreatic adenocarcinoma signaling activation in TRAMP mice. Analyzing DNA methyl sequencing data led to the observation of PEITC-activated reduction of global methylation alteration in PCa development. Integration of DNA methylation and RNA expression profiles of TRAMP and TRAMP+PEITC identified PEITC reversed the inverse correlation between RNA expression and DNA methylation in 28 genes. Among these genes, Arhgap40, Ebf4, kcnq4, and Papln were validated by qPCR 91.
In addition to decreasing global methylation, PEITC can prevent PCa cell invasion via upregulation of miRNA-194. Zhang et al. stated that PEITC treatment in LNCaP cells upregulated the expression of miRNA-194, which targeted bone morphogenetic protein 1 (BMP1) and further inhibited the expression of central oncogenic matrix metalloproteinases, MMP2 and MMP9. The axis of miRNA-194/ BMP1/ MMP2/9 unveiled the regulation of PCa cell metastasis by PEITC102.
Terpenoids
Combinatorial treatment with natural compounds including ursolic acid (UA), curcumin (CUR), and resveratrol (RES) was studied in HMVP2 cells and male FVB/N mice subcutaneously injected with HMVP2 cells. The results showed that the treatment with UA+CUR and UA+RES could inhibit prostate tumorigenesis and account for critical regulation of cancer metabolism 103. In a recent study, Wang et al. observed that UA exerted its protection against tumor development initiated by PTEN deletion at different stages of PCa. UA treatment decreased PTEN KO-triggered differentially methylated regions (DMRs) and reversed PTEN KO-induced overexpression of PCa-relevant oncogenes, Has3, Cfh, and Msx1. Correlation analysis of differentially expressed genes (DEGs) and DMRs revealed that the mRNA expression of the tumor suppressor gene (BDH2) and oncogenes (Ephas, Isg15, Nos2) were correlated with the CpG methylation status of the promoter region in the UA-treated group at the early phase. The pathway analysis indicated that UA treatment reversed PTEN KO-activated inflammatory pathways, such as NF-𝜅B signaling, IL-6 signaling, and IL-8 signaling104. Cellular and animal studies on the prevention of PCa indicate that phytochemicals provide the restoration of the NRF2-ARE signaling pathway through demethylation of the NRF2 promoter region and diminishment of DNMTs and HDACs expression.
Benzoquinone
Su et al. examined the anticancer properties of Z-Ligustilide (Lig) and supercritical CO2 extract of Radix Angelica Sinensis (RAS) in TRAMP C1 cells. RAS is a dried root of Angelica sinensis and has been served as medicinal plant and supplementary food for centuries. Lig is one of bioactive compounds from a lipophilic extract of RAS105. The finding suggested that NRF2 expression was restored via epigenetic changes. This led to increased mRNA and protein levels of NRF2 and its target genes such as HO-1, NQO1, and UGT1A1. The treatment of Lig and RAS extract demethylated the first five CpGs and significantly reduced the relative amount of methylated DNA in the NRF2 promoter region. Additionally, Lig and RAS extract treatment inhibited DNA methyltransferase activity in TRAMP C1 cells105. Table 3 summarizes the phytochemicals that affect epigenetic pathways and are used to inhibit PCa growth and contribute to cancer prevention.
Table 3.
Phytochemicals | Cellular/ Animal Model | Epigenetic Mechanism | Reference |
---|---|---|---|
Curcumin analog FN1 | TRAMP C1 cells | Demethylate the Nrf2 promoter region and decrease the level of DNMT1, DNMT3a, DNMT3b, and HDAC 4. | 93 |
Curcumin derivatives F10 | TRAMP C1 cells | Demethylate the Nrf2 promoter region, inhibition of H3k27me3 accumulation, and downregulation of DNMTs and HDACs. | 100 |
Sulforaphane | TRAMP C1 cells | Demethylate the Nrf2 promoter region, decrease the level of DNMT1, DNMT3a, and HDACs 1, 4, 5, 7, and raise the Ac-H3 level. | 101 |
Phenethyl Isothiocyanate | Male TRAMP mice | Reduce the global methylation in PCa carcinogenesis and reverse the TRAMP-induced changes in transcriptome and methylome. | 91 |
Phenethyl Isothiocyanate | LNCaP cells | Upregulate miRNA-194 and further inhibit expression of MMP2 and MMP9 to prevent metastasis. | 102 |
Ursolic acid | PTEN-KO mice | Reduce differentially methylated regions and the overexpression of PCa-associated oncogenes (Has3, Cfh, and Msx1) induced by PTEN deletion. Regulate the methylation status of promoter of tumor suppressor genes and oncogenes in the early-stage development of PCa. |
104 |
Z-Ligustilide (Lig) and Radix Angelica Sinensis (RAS) extract | TRAMP C1 cells | Hypomethylate the Nrf2 promoter region and reduce DNMT activity. | 105 |
Summary
Skin, colorectal, and prostate cancers are increasing in new cancer cases in the US. Cancer development is a long process. It encompasses an imbalance of oxidative stress and excessive inflammation that cause the dysfunction of multiple signaling pathways leading to cancer initiation and progression. Many phytochemicals in our everyday diet possess powerful antioxidant and anti-inflammatory properties that are hallmarks of cancer prevention. Recent evidence suggests that epigenetic modulation of these critical signaling pathways by phytochemicals is related to their antioxidant and anti-inflammatory effects. For example, modulation of the NRF2-ARE signaling pathway and the NF-𝜅B signaling pathway has been shown to contribute to the overall efficacy of cancer prevention by phytochemicals.
Despite a wide range of promising dietary phytochemicals that have been reported to have preventive activity in preclinical research, the translation of dietary phytochemicals to clinical use has been limited. Only a few of these agents are applied to the clinical studies. There is an urgent need to expedite the testing of these agents in clinical trials involving appropriate high-risk individuals with the proper dosing, formulation, biomarker endpoints (surrogate and tissue targeted), and integration of pharmacokinetic (PK)-pharmacodynamic (PD) inputs. Taken together, this article summarizes the latest preclinical research and hypotheses on cancer prevention by dietary phytochemicals via epigenetic modulation in cancers of the skin, colorectum, and prostate. In the general population, cancer prevention with a healthy lifestyle composed of exercise and consuming vegetables, fruits, herbs, and spices is a convenient and logical way to ingest a wide range of phytochemicals that can protect against cancer development. For higher-risk individuals, pharmacological doses of a combination of bioactive phytochemicals and/or phytochemical combinations with pharmacological agents of low toxicity would be logical to test for preventing cancer development 44, 45.
Acknowledgments:
This work was supported in part by institutional funds and by R01 AT009152 from the National Center for Complementary and Integrative Health (NCCIH), R01 CA200129 from the National Cancer Institute (NCI), and P30 ES005022 from the National Institute of Environmental Health (NIEHS).
Abbreviations
- AOM
Azoxymethane
- B[a]P
Benzo[a]pyrene
- BMP1
Bone morphogenetic protein 1
- CA
Corosolic acid
- CDDO
2-cyano 2,3-dioxoolean-1,9-dien-28-oic acid
- CpG
5’-C-phosphate-G-3’
- CRC
Colorectal cancer
- CUR
Curcumin
- DEG
Differentially expressed gene
- DMR
Differentially methylated region
- DNMT
DNA methyltransferases
- DSS
Dextran sulfate sodium
- FX
Fucoxanthin
- HAT
Histone acetyltransferase
- HDAC
Histone deacetylases
- IND
Indicaxanthin
- Lig
Z-Ligustilide
- LUT
Luteolin
- MIC-1
Moringa isothiocyanate
- miRNA
MicroRNA
- MMP
Matrix metalloproteinases
- NMSC
Non-melanoma skin cancer
- PCa
Prostate cancer
- PcG
Polycomb group proteins
- PD
Pharmacodynamics
- PEITC
Phenethyl Isothiocyanate
- PK
Pharmacokinetics
- PTEN
Phosphatase and tensin homolog located on chromosome 10
- RAS
Radix Angelica Sinensis
- SFN
Sulforaphane
- TPA
12-O-tetradecanoylphorbol-13-acetate
- TRAMP
Transgenic adenocarcinoma of mouse prostate
- UA
Ursolic acid
- UV
Ultraviolet
- WIR
Water extract of I. rotunda
Reference:
- 1.Varmus H, Unni AM, Lockwood WW. How Cancer Genomics Drives Cancer Biology: Does Synthetic Lethality Explain Mutually Exclusive Oncogenic Mutations? Cold Spring Harb Symp Quant Biol 2016;81:247–55. Epub 2017/01/27. doi: 10.1101/sqb.2016.81.030866. [DOI] [PubMed] [Google Scholar]
- 2.Fouad YA, Aanei C. Revisiting the hallmarks of cancer. Am J Cancer Res 2017;7(5):1016–36. [PMC free article] [PubMed] [Google Scholar]
- 3.Chen C, Kong AN. Dietary chemopreventive compounds and ARE/EpRE signaling. Free Radic Biol Med 2004;36(12):1505–16. Epub 2004/06/09. doi: 10.1016/j.freeradbiomed.2004.03.015. [DOI] [PubMed] [Google Scholar]
- 4.Lee JH, Khor TO, Shu L, Su ZY, Fuentes F, Kong AN. Dietary phytochemicals and cancer prevention: Nrf2 signaling, epigenetics, and cell death mechanisms in blocking cancer initiation and progression. Pharmacol Ther 2013;137(2):153–71. Epub 2012/10/09. doi: 10.1016/j.pharmthera.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ho SM, Johnson A, Tarapore P, Janakiram V, Zhang X, Leung YK. Environmental epigenetics and its implication on disease risk and health outcomes. Ilar j 2012;53(3–4):289–305. Epub 2013/06/08. doi: 10.1093/ilar.53.3-4.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Feinberg AP. The Key Role of Epigenetics in Human Disease Prevention and Mitigation. N Engl J Med 2018;378(14):1323–34. Epub 2018/04/05. doi: 10.1056/NEJMra1402513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cavalli G, Heard E. Advances in epigenetics link genetics to the environment and disease. Nature. 2019;571(7766):489–99. Epub 2019/07/26. doi: 10.1038/s41586-019-1411-0. [DOI] [PubMed] [Google Scholar]
- 8.Jha NK, Arfin S, Jha SK, Kar R, Dey A, Gundamaraju R, Ashraf GM, Gupta PK, Dhanasekaran S, Abomughaid MM, Das SS, Singh SK, Dua K, Roychoudhury S, Kumar D, Ruokolainen J, Ojha S, Kesari KK. Re-establishing the comprehension of phytomedicine and nanomedicine in inflammation-mediated cancer signaling. Semin Cancer Biol 2022;86(Pt 2):1086–104. Epub 20220223. doi: 10.1016/j.semcancer.2022.02.022. [DOI] [PubMed] [Google Scholar]
- 9.Klaunig JE. Oxidative Stress and Cancer. Curr Pharm Des 2018;24(40):4771–8. doi: 10.2174/1381612825666190215121712. [DOI] [PubMed] [Google Scholar]
- 10.Wu R, Li S, Hudlikar R, Wang L, Shannar A, Peter R, Chou PJ, Kuo HD, Liu Z, Kong AN. Redox signaling, mitochondrial metabolism, epigenetics and redox active phytochemicals. Free Radic Biol Med 2022;179:328–36. Epub 20201224. doi: 10.1016/j.freeradbiomed.2020.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Scott RE, Wille JJ, Jr., Wier ML. Mechanisms for the Initiation and Promotion of Carcinogenesis: A Review and a New Concept. Mayo Clinic Proceedings. 1984;59(2):107–17. doi: 10.1016/S0025–6196(12)60244–4. [DOI] [PubMed] [Google Scholar]
- 12.Goelz SE, Vogelstein B, Hamilton SR, Feinberg AP. Hypomethylation of DNA from benign and malignant human colon neoplasms. Science. 1985;228(4696):187–90. Epub 1985/04/12. doi: 10.1126/science.2579435. [DOI] [PubMed] [Google Scholar]
- 13.Feinberg AP, Vogelstein B. Alterations in DNA methylation in human colon neoplasia. Semin Surg Oncol 1987;3(3):149–51. Epub 1987/01/01. doi: 10.1002/ssu.2980030304. [DOI] [PubMed] [Google Scholar]
- 14.Feinberg AP, Koldobskiy MA, Göndör A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat Rev Genet 2016;17(5):284–99. Epub 2016/03/15. doi: 10.1038/nrg.2016.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bird A Perceptions of epigenetics. Nature. 2007;447(7143):396–8. doi: 10.1038/nature05913. [DOI] [PubMed] [Google Scholar]
- 16.Hudlikar R, Wang L, Wu R, Li S, Peter R, Shannar A, Chou PJ, Liu X, Liu Z, Kuo HD, Kong AN. Epigenetics/Epigenomics and Prevention of Early Stages of Cancer by Isothiocyanates. Cancer Prev Res (Phila). 2021;14(2):151–64. Epub 2020/10/16. doi: 10.1158/1940-6207.Capr-20-0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Loscalzo J, Handy DE. Epigenetic modifications: basic mechanisms and role in cardiovascular disease (2013 Grover Conference series). Pulm Circ 2014;4(2):169–74. doi: 10.1086/675979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ito S, D’Alessio AC, Taranova OV, Hong K, Sowers LC, Zhang Y. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature. 2010;466(7310):1129–33. doi: 10.1038/nature09303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Baylin SB, Jones PA. A decade of exploring the cancer epigenome - biological and translational implications. Nat Rev Cancer. 2011;11(10):726–34. Epub 20110923. doi: 10.1038/nrc3130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wu YL, Lin ZJ, Li CC, Lin X, Shan SK, Guo B, Zheng MH, Li F, Yuan LQ, Li ZH. Epigenetic regulation in metabolic diseases: mechanisms and advances in clinical study. Signal Transduct Target Ther 2023;8(1):98. Epub 20230302. doi: 10.1038/s41392-023-01333-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kasinski AL, Slack FJ. Epigenetics and genetics. MicroRNAs en route to the clinic: progress in validating and targeting microRNAs for cancer therapy. Nat Rev Cancer. 2011;11(12):849–64. Epub 20111124. doi: 10.1038/nrc3166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, Croce CM. Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci U S A 2004;101(9):2999–3004. Epub 20040218. doi: 10.1073/pnas.0307323101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Cheng Y, He C, Wang M, Ma X, Mo F, Yang S, Han J, Wei X. Targeting epigenetic regulators for cancer therapy: mechanisms and advances in clinical trials. Signal Transduct Target Ther 2019;4:62. Epub 20191217. doi: 10.1038/s41392-019-0095-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Guo Y, Yu S, Zhang C, Kong AN. Epigenetic regulation of Keap1-Nrf2 signaling. Free Radic Biol Med 2015;88(Pt B):337–49. Epub 20150625. doi: 10.1016/j.freeradbiomed.2015.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Guerriero G, Berni R, Munoz-Sanchez JA, Apone F, Abdel-Salam EM, Qahtan AA, Alatar AA, Cantini C, Cai G, Hausman JF, Siddiqui KS, Hernandez-Sotomayor SMT, Faisal M. Production of Plant Secondary Metabolites: Examples, Tips and Suggestions for Biotechnologists. Genes (Basel). 2018;9(6). Epub 20180620. doi: 10.3390/genes9060309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bhattacharya T, Dutta S, Akter R, Rahman MH, Karthika C, Nagaswarupa HP, Murthy HCA, Fratila O, Brata R, Bungau S. Role of Phytonutrients in Nutrigenetics and Nutrigenomics Perspective in Curing Breast Cancer. Biomolecules. 2021;11(8). Epub 20210809. doi: 10.3390/biom11081176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Liu RH. Health-promoting components of fruits and vegetables in the diet. Adv Nutr 2013;4(3):384S–92S. Epub 20130501. doi: 10.3945/an.112.003517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Behl T, Kumar K, Brisc C, Rus M, Nistor-Cseppento DC, Bustea C, Aron RAC, Pantis C, Zengin G, Sehgal A, Kaur R, Kumar A, Arora S, Setia D, Chandel D, Bungau S. Exploring the multifocal role of phytochemicals as immunomodulators. Biomed Pharmacother 2021;133:110959. Epub 20201113. doi: 10.1016/j.biopha.2020.110959. [DOI] [PubMed] [Google Scholar]
- 29.Behl T, Bungau S, Kumar K, Zengin G, Khan F, Kumar A, Kaur R, Venkatachalam T, Tit DM, Vesa CM, Barsan G, Mosteanu DE. Pleotropic Effects of Polyphenols in Cardiovascular System. Biomed Pharmacother 2020;130:110714. Epub 20200928. doi: 10.1016/j.biopha.2020.110714. [DOI] [PubMed] [Google Scholar]
- 30.Kapinova A, Kubatka P, Golubnitschaja O, Kello M, Zubor P, Solar P, Pec M. Dietary phytochemicals in breast cancer research: anticancer effects and potential utility for effective chemoprevention. Environ Health Prev Med 2018;23(1):36. Epub 20180809. doi: 10.1186/s12199-018-0724-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wu R, Li S, Hudlikar R, Wang L, Shannar A, Peter R, Chou PJ, Kuo HD, Liu Z, Kong AN. Redox signaling, mitochondrial metabolism, epigenetics and redox active phytochemicals. Free Radic Biol Med 2020. Epub 2020/12/29. doi: 10.1016/j.freeradbiomed.2020.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Santhiravel S, Bekhit AEA, Mendis E, Jacobs JL, Dunshea FR, Rajapakse N, Ponnampalam EN. The Impact of Plant Phytochemicals on the Gut Microbiota of Humans for a Balanced Life. Int J Mol Sci 2022;23(15). Epub 20220723. doi: 10.3390/ijms23158124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Sharma R, Thakur D. Health Promoting Phytochemicals in Vegetables: A Mini Review. International Journal of Food and Fermentation Technology. 2018;8:107–17. doi: 10.30954/2277-9396.02.2018.1. [DOI] [Google Scholar]
- 34.Pandey KB, Rizvi SI. Plant polyphenols as dietary antioxidants in human health and disease. Oxid Med Cell Longev 2009;2(5):270–8. doi: 10.4161/oxim.2.5.9498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Anthocyanins Clifford M. – Nature, occurrence and dietary burden. Journal of the Science of Food and Agriculture. 2000;80:1063–72. doi: . [DOI] [Google Scholar]
- 36.García-Lafuente A, Guillamón E, Villares A, Rostagno MA, Martínez JA. Flavonoids as anti-inflammatory agents: implications in cancer and cardiovascular disease. Inflamm Res 2009;58(9):537–52. Epub 20090421. doi: 10.1007/s00011-009-0037-3. [DOI] [PubMed] [Google Scholar]
- 37.Falleh H, Ben Jemaa M, Saada M, Ksouri R. Essential oils: A promising eco-friendly food preservative. Food Chemistry. 2020;330:127268. doi: 10.1016/j.foodchem.2020.127268. [DOI] [PubMed] [Google Scholar]
- 38.Burt S Essential oils: their antibacterial properties and potential applications in foods—a review. International Journal of Food Microbiology. 2004;94(3):223–53. doi: 10.1016/j.ijfoodmicro.2004.03.022. [DOI] [PubMed] [Google Scholar]
- 39.Zhao D-D, Jiang L-L, Li H-Y, Yan P-F, Zhang Y-L. Chemical Components and Pharmacological Activities of Terpene Natural Products from the Genus Paeonia. Molecules. 2016;21(10):1362. PubMed PMID: doi: 10.3390/molecules21101362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Santhiravel S, Bekhit AE- DA, Mendis E, Jacobs JL, Dunshea FR, Rajapakse N, Ponnampalam EN. The Impact of Plant Phytochemicals on the Gut Microbiota of Humans for a Balanced Life. International Journal of Molecular Sciences. 2022;23(15):8124. PubMed PMID: doi: 10.3390/ijms23158124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Su ZY, Zhang C, Lee JH, Shu L, Wu TY, Khor TO, Conney AH, Lu YP, Kong AN. Requirement and epigenetics reprogramming of Nrf2 in suppression of tumor promoter TPA-induced mouse skin cell transformation by sulforaphane. Cancer Prev Res (Phila). 2014;7(3):319–29. Epub 2014/01/21. doi: 10.1158/1940-6207.Capr-13-0313-t. [DOI] [PubMed] [Google Scholar]
- 42.Harbourne N, Marete E, Jacquier J-C, O’Riordan D. Stability of phytochemicals as sources of anti-inflammatory nutraceuticals in beverages — A review. Food Research International - FOOD RES INT. 2013;50. doi: 10.1016/j.foodres.2011.03.009. [DOI] [Google Scholar]
- 43.Alfurayhi R, Huang L, Brandt K. Pathways Affected by Falcarinol-Type Polyacetylenes and Implications for Their Anti-Inflammatory Function and Potential in Cancer Chemoprevention. Foods. 2023;12(6). Epub 20230311. doi: 10.3390/foods12061192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wu R, Wang L, Yin R, Hudlikar R, Li S, Kuo H- CD, Peter R, Sargsyan D, Guo Y, Liu X, Kong AN. Epigenetics/epigenomics and prevention by curcumin of early stages of inflammatory-driven colon cancer. Molecular Carcinogenesis. 2020;59(2):227–36. doi: 10.1002/mc.23146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hudlikar R, Wang L, Wu R, Li S, Peter R, Shannar A, Chou PJ, Liu X, Liu Z, Kuo HD, Kong AN. Epigenetics/epigenomics and prevention of early stages of cancer by isothiocyanates. Cancer Prev Res (Phila). 2020. Epub 2020/10/16. doi: 10.1158/1940-6207.CAPR-20-0217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Melnikova VO, Ananthaswamy HN. Cellular and molecular events leading to the development of skin cancer. Mutat Res 2005;571(1–2):91–106. doi: 10.1016/j.mrfmmm.2004.11.015. [DOI] [PubMed] [Google Scholar]
- 47.Shalhout SZ, Kaufman HL, Emerick KS, Miller DM. Immunotherapy for Nonmelanoma Skin Cancer: Facts and Hopes. Clin Cancer Res 2022;28(11):2211–20. doi: 10.1158/1078-0432.Ccr-21-2971. [DOI] [PubMed] [Google Scholar]
- 48.Ciążyńska M, Kamińska-Winciorek G, Lange D, Lewandowski B, Reich A, Sławińska M, Pabianek M, Szczepaniak K, Hankiewicz A, Ułańska M, Morawiec J, Błasińska-Morawiec M, Morawiec Z, Piekarski J, Nejc D, Brodowski R, Zaryczańska A, Sobjanek M, Nowicki RJ, Owczarek W, Słowińska M, Wróbel K, Bieniek A, Woźniacka A, Skibińska M, Narbutt J, Niemczyk W, Ciążyński K, Lesiak A. The incidence and clinical analysis of non-melanoma skin cancer. Sci Rep 2021;11(1):4337. Epub 20210222. doi: 10.1038/s41598-021-83502-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kasper M, Jaks V, Hohl D, Toftgård R. Basal cell carcinoma - molecular biology and potential new therapies. J Clin Invest 2012;122(2):455–63. Epub 20120201. doi: 10.1172/jci58779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yang Y, Wu R, Sargsyan D, Yin R, Kuo HC, Yang I, Wang L, Cheng D, Wang C, Li S, Hudlikar R, Lu Y, Kong AN. UVB drives different stages of epigenome alterations during progression of skin cancer. Cancer Lett 2019;449:20–30. Epub 20190213. doi: 10.1016/j.canlet.2019.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sarwar MS, Ramirez CN, Dina Kuo HC, Chou P, Wu R, Sargsyan D, Yang Y, Shannar A, Mary Peter R, Yin R, Wang Y, Su X, Kong AN. The environmental carcinogen benzo[a]pyrene regulates epigenetic reprogramming and metabolic rewiring in a two-stage mouse skin carcinogenesis model. Carcinogenesis. 2023;44(5):436–49. doi: 10.1093/carcin/bgad024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nandakumar V, Vaid M, Katiyar SK. (−)-Epigallocatechin-3-gallate reactivates silenced tumor suppressor genes, Cip1/p21 and p16INK4a, by reducing DNA methylation and increasing histones acetylation in human skin cancer cells. Carcinogenesis. 2011;32(4):537–44. Epub 2011/01/07. doi: 10.1093/carcin/bgq285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wei H, Tye L, Bresnick E, Birt DF. Inhibitory effect of apigenin, a plant flavonoid, on epidermal ornithine decarboxylase and skin tumor promotion in mice. Cancer Res 1990;50(3):499–502. [PubMed] [Google Scholar]
- 54.Birt DF, Mitchell D, Gold B, Pour P, Pinch HC. Inhibition of ultraviolet light induced skin carcinogenesis in SKH-1 mice by apigenin, a plant flavonoid. Anticancer Res 1997;17(1A):85–91. [PubMed] [Google Scholar]
- 55.Paredes-Gonzalez X, Fuentes F, Su ZY, Kong AN. Apigenin reactivates Nrf2 anti-oxidative stress signaling in mouse skin epidermal JB6 P + cells through epigenetics modifications. AAPS J 2014;16(4):727–35. doi: 10.1208/s12248-014-9613-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Li S, Li W, Wang C, Wu R, Yin R, Kuo HC, Wang L, Kong AN. Pelargonidin reduces the TPA induced transformation of mouse epidermal cells -potential involvement of Nrf2 promoter demethylation. Chem Biol Interact 2019;309:108701. Epub 2019/06/11. doi: 10.1016/j.cbi.2019.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mitsiogianni M, Amery T, Franco R, Zoumpourlis V, Pappa A, Panayiotidis MI. From chemo-prevention to epigenetic regulation: The role of isothiocyanates in skin cancer prevention. Pharmacol Ther 2018;190:187–201. Epub 2018/06/12. doi: 10.1016/j.pharmthera.2018.06.001. [DOI] [PubMed] [Google Scholar]
- 58.Mitsiogianni M, Koutsidis G, Mavroudis N, Trafalis DT, Botaitis S, Franco R, Zoumpourlis V, Amery T, Galanis A, Pappa A, Panayiotidis MI. The Role of Isothiocyanates as Cancer Chemo-Preventive, Chemo-Therapeutic and Anti-Melanoma Agents. Antioxidants (Basel). 2019;8(4). Epub 2019/04/21. doi: 10.3390/antiox8040106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Saha K, Hornyak TJ, Eckert RL. Epigenetic cancer prevention mechanisms in skin cancer. AAPS J 2013;15(4):1064–71. Epub 2013/08/02. doi: 10.1208/s12248-013-9513-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Penta D, Somashekar BS, Meeran SM. Epigenetics of skin cancer: Interventions by selected bioactive phytochemicals. Photodermatol Photoimmunol Photomed 2018;34(1):42–9. Epub 2017/10/05. doi: 10.1111/phpp.12353. [DOI] [PubMed] [Google Scholar]
- 61.Balasubramanian S, Adhikary G, Eckert RL. The Bmi-1 polycomb protein antagonizes the (−)-epigallocatechin-3-gallate-dependent suppression of skin cancer cell survival. Carcinogenesis. 2010;31(3):496–503. Epub 2009/12/18. doi: 10.1093/carcin/bgp314. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 62.Choudhury SR, Balasubramanian S, Chew YC, Han B, Marquez VE, Eckert RL. (−)-Epigallocatechin-3-gallate and DZNep reduce polycomb protein level via a proteasome-dependent mechanism in skin cancer cells. Carcinogenesis. 2011;32(10):1525–32. Epub 2011/07/30. doi: 10.1093/carcin/bgr171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang C, Wu R, Sargsyan D, Zheng M, Li S, Yin R, Su S, Raskin I, Kong AN. CpG methyl-seq and RNA-seq epigenomic and transcriptomic studies on the preventive effects of Moringa isothiocyanate in mouse epidermal JB6 cells induced by the tumor promoter TPA. J Nutr Biochem 2019;68:69–78. Epub 20190328. doi: 10.1016/j.jnutbio.2019.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Lora JM, Zhang DM, Liao SM, Burwell T, King AM, Barker PA, Singh L, Keaveney M, Morgenstern J, Gutierrez-Ramos JC, Coyle AJ, Fraser CC. Tumor necrosis factor-alpha triggers mucus production in airway epithelium through an IkappaB kinase beta-dependent mechanism. J Biol Chem 2005;280(43):36510–7. Epub 20050825. doi: 10.1074/jbc.M507977200. [DOI] [PubMed] [Google Scholar]
- 65.Kuo HD, Wu R, Sarwar MS, Zheng M, Wang C, Sargsyan D, Suh N, Kong AT. DNA Methylome and Transcriptome Study of Triterpenoid CDDO in TPA-Mediated Skin Carcinogenesis Model. AAPS J 2022;24(6):115. Epub 20221102. doi: 10.1208/s12248-022-00763-5. [DOI] [PubMed] [Google Scholar]
- 66.Yang Y, Yang I, Cao M, Su ZY, Wu R, Guo Y, Fang M, Kong AN. Fucoxanthin Elicits Epigenetic Modifications, Nrf2 Activation and Blocking Transformation in Mouse Skin JB6 P+ Cells. Aaps j 2018;20(2):32. Epub 2018/04/01. doi: 10.1208/s12248-018-0197-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yang Y, Yin R, Wu R, Ramirez CN, Sargsyan D, Li S, Wang L, Cheng D, Wang C, Hudlikar R, Kuo HC, Lu Y, Kong AN. DNA methylome and transcriptome alterations and cancer prevention by triterpenoid ursolic acid in UVB-induced skin tumor in mice. Mol Carcinog 2019;58(10):1738–53. Epub 2019/06/27. doi: 10.1002/mc.23046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hudlikar RR, Sargsyan D, Wu R, Su S, Zheng M, Kong AN. Triterpenoid corosolic acid modulates global CpG methylation and transcriptome of tumor promotor TPA induced mouse epidermal JB6 P+ cells. Chem Biol Interact 2020;321:109025. Epub 20200303. doi: 10.1016/j.cbi.2020.109025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tung YC, Sung PH, Chen PC, Wang HC, Lee JH, Su ZY. Chemoprevention of lotus leaf ethanolic extract through epigenetic activation of the NRF2-mediated pathway in murine skin JB6 P+ cell neoplastic transformation. J Tradit Complement Med 2023;13(4):337–44. Epub 2023/07/03. doi: 10.1016/j.jtcme.2023.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Marjaneh RM, Rahmani F, Hassanian SM, Rezaei N, Hashemzehi M, Bahrami A, Ariakia F, Fiuji H, Sahebkar A, Avan A. Phytosomal curcumin inhibits tumor growth in colitis-associated colorectal cancer. Journal of cellular physiology. 2018;233(10):6785–98. [DOI] [PubMed] [Google Scholar]
- 71.Yamauchi M, Morikawa T, Kuchiba A, Imamura Y, Qian ZR, Nishihara R, Liao X, Waldron L, Hoshida Y, Huttenhower C, Chan AT, Giovannucci E, Fuchs C, Ogino S. Assessment of colorectal cancer molecular features along bowel subsites challenges the conception of distinct dichotomy of proximal versus distal colorectum. Gut 2012;61(6):847–54. doi: 10.1136/gutjnl-2011-300865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ogino S, Chan AT, Fuchs CS, Giovannucci E. Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut 2011;60(3):397–411. doi: 10.1136/gut.2010.217182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Islam MR, Akash S, Rahman MM, Nowrin FT, Akter T, Shohag S, Rauf A, Aljohani ASM, Simal-Gandara J. Colon cancer and colorectal cancer: Prevention and treatment by potential natural products. Chemico-Biological Interactions. 2022;368:110170. doi: 10.1016/j.cbi.2022.110170. [DOI] [PubMed] [Google Scholar]
- 74.Perše M, Cerar A. Dextran sodium sulphate colitis mouse model: traps and tricks. J Biomed Biotechnol 2012;2012:718617. Epub 20120514. doi: 10.1155/2012/718617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Delker DA, McKnight SJ, 3rd, Rosenberg DW. The role of alcohol dehydrogenase in the metabolism of the colon carcinogen methylazoxymethanol. Toxicol Sci 1998;45(1):66–71. doi: 10.1006/toxs.1998.2499. [DOI] [PubMed] [Google Scholar]
- 76.Guo Y, Wu R, Gaspar JM, Sargsyan D, Su ZY, Zhang C, Gao L, Cheng D, Li W, Wang C, Yin R, Fang M, Verzi MP, Hart RP, Kong AN. DNA methylome and transcriptome alterations and cancer prevention by curcumin in colitis-accelerated colon cancer in mice. Carcinogenesis. 2018;39(5):669–80. doi: 10.1093/carcin/bgy043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hornef MW, Normark BH, Vandewalle A, Normark S. Intracellular recognition of lipopolysaccharide by toll-like receptor 4 in intestinal epithelial cells. J Exp Med 2003;198(8):1225–35. doi: 10.1084/jem.20022194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kang M, Edmundson P, Araujo-Perez F, McCoy AN, Galanko J, Keku TO. Association of plasma endotoxin, inflammatory cytokines and risk of colorectal adenomas. BMC Cancer. 2013;13:91. Epub 20130226. doi: 10.1186/1471-2407-13-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhu G, Huang Q, Huang Y, Zheng W, Hua J, Yang S, Zhuang J, Wang J, Ye J. Lipopolysaccharide increases the release of VEGF-C that enhances cell motility and promotes lymphangiogenesis and lymphatic metastasis through the TLR4- NF-κB/JNK pathways in colorectal cancer. Oncotarget 2016;7(45):73711–24. doi: 10.18632/oncotarget.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Guo Y, Shu L, Zhang C, Su Z-Y, Kong A- NT. Curcumin inhibits anchorage-independent growth of HT29 human colon cancer cells by targeting epigenetic restoration of the tumor suppressor gene DLEC1. Biochemical pharmacology. 2015;94(2):69–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Guo Y, Wu R, Gaspar JM, Sargsyan D, Su Z-Y, Zhang C, Gao L, Cheng D, Li W, Wang C. DNA methylome and transcriptome alterations and cancer prevention by curcumin in colitis-accelerated colon cancer in mice. Carcinogenesis. 2018;39(5):669–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Zuo Q, Wu R, Xiao X, Yang C, Yang Y, Wang C, Lin L, Kong AN. The dietary flavone luteolin epigenetically activates the Nrf2 pathway and blocks cell transformation in human colorectal cancer HCT116 cells. Journal of Cellular Biochemistry. 2018;119(11):9573–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cui X, Jin Y, Hofseth AB, Pena E, Habiger J, Chumanevich A, Poudyal D, Nagarkatti M, Nagarkatti PS, Singh UP. Resveratrol suppresses colitis and colon cancer associated with colitis. Cancer prevention research. 2010;3(4):549–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Altamemi I, Murphy EA, Catroppo JF, Zumbrun EE, Zhang J, McClellan JL, Singh UP, Nagarkatti PS, Nagarkatti M. Role of microRNAs in resveratrol-mediated mitigation of colitis-associated tumorigenesis in Apc(Min/+) mice. J Pharmacol Exp Ther 2014;350(1):99–109. Epub 20140509. doi: 10.1124/jpet.114.213306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Okonkwo A, Mitra J, Johnson GS, Li L, Dashwood WM, Hegde ML, Yue C, Dashwood RH, Rajendran P. Heterocyclic Analogs of Sulforaphane Trigger DNA Damage and Impede DNA Repair in Colon Cancer Cells: Interplay of HATs and HDACs. Mol Nutr Food Res 2018;62(18):e1800228. Epub 20180719. doi: 10.1002/mnfr.201800228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhou JW, Wang M, Sun NX, Qing Y, Yin TF, Li C, Wu D. Sulforaphane-induced epigenetic regulation of Nrf2 expression by DNA methyltransferase in human Caco-2 cells. Oncol Lett 2019;18(3):2639–47. Epub 20190705. doi: 10.3892/ol.2019.10569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Liu Y, Dey M. Dietary Phenethyl Isothiocyanate Protects Mice from Colitis Associated Colon Cancer. Int J Mol Sci 2017;18(9). Epub 20170906. doi: 10.3390/ijms18091908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Ragusa MA, Naselli F, Cruciata I, Volpes S, Schimmenti C, Serio G, Mauro M, Librizzi M, Luparello C, Chiarelli R, La Rosa C, Lauria A, Gentile C, Caradonna F. Indicaxanthin Induces Autophagy in Intestinal Epithelial Cancer Cells by Epigenetic Mechanisms Involving DNA Methylation. Nutrients. 2023;15(15). Epub 20230807. doi: 10.3390/nu15153495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chen G, Han Y, Feng Y, Wang A, Li X, Deng S, Zhang L, Xiao J, Li Y, Li N. Extract of Ilex rotunda Thunb alleviates experimental colitis-associated cancer via suppressing inflammation-induced miR-31–5p/YAP overexpression. Phytomedicine. 2019;62:152941. [DOI] [PubMed] [Google Scholar]
- 90.Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin 2017;67(1):7–30. Epub 20170105. doi: 10.3322/caac.21387. [DOI] [PubMed] [Google Scholar]
- 91.Wu R, Li S, Sargsyan D, Yin R, Kuo HC, Peter R, Wang L, Hudlikar R, Liu X, Kong AN. DNA methylome, transcriptome, and prostate cancer prevention by phenethyl isothiocyanate in TRAMP mice. Mol Carcinog 2021;60(6):391–402. Epub 20210413. doi: 10.1002/mc.23299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Petrylak DP. The current role of chemotherapy in metastatic hormone-refractory prostate cancer. Urology. 2005;65(5 Suppl):3–7; discussion −8. doi: 10.1016/j.urology.2005.03.053. [DOI] [PubMed] [Google Scholar]
- 93.Li W, Pung D, Su ZY, Guo Y, Zhang C, Yang AY, Zheng X, Du ZY, Zhang K, Kong AN. Epigenetics Reactivation of Nrf2 in Prostate TRAMP C1 Cells by Curcumin Analogue FN1. Chem Res Toxicol 2016;29(4):694–703. Epub 20160329. doi: 10.1021/acs.chemrestox.6b00016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Greenberg NM, DeMayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, Cunha GR, Donjacour AA, Matusik RJ, Rosen JM. Prostate cancer in a transgenic mouse. Proc Natl Acad Sci U S A 1995;92(8):3439–43. doi: 10.1073/pnas.92.8.3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Barve A, Khor TO, Reuhl K, Reddy B, Newmark H, Kong AN. Mixed tocotrienols inhibit prostate carcinogenesis in TRAMP mice. Nutr Cancer. 2010;62(6):789–94. doi: 10.1080/01635581003605896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Valkenburg KC, Williams BO. Mouse models of prostate cancer. Prostate Cancer. 2011;2011:895238. doi: 10.1155/2011/895238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Maxwell PJ, Coulter J, Walker SM, McKechnie M, Neisen J, McCabe N, Kennedy RD, Salto-Tellez M, Albanese C, Waugh DJ. Potentiation of inflammatory CXCL8 signalling sustains cell survival in PTEN-deficient prostate carcinoma. European urology. 2013;64(2):177–88. doi: 10.1016/j.eururo.2012.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Wang C, Feng Y, Zhang C, Cheng D, Wu R, Yang Y, Sargsyan D, Kumar D, Kong AN. PTEN deletion drives aberrations of DNA methylome and transcriptome in different stages of prostate cancer. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2020;34(1):1304–18. Epub 2020/01/10. doi: 10.1096/fj.201901205RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wei X, Du Z-Y, Zheng X, Cui X-X, Conney AH, Zhang K. Synthesis and evaluation of curcumin-related compounds for anticancer activity. European Journal of Medicinal Chemistry. 2012;53:235–45. doi: 10.1016/j.ejmech.2012.04.005. [DOI] [PubMed] [Google Scholar]
- 100.Li W, Su ZY, Guo Y, Zhang C, Wu R, Gao L, Zheng X, Du ZY, Zhang K, Kong AN. Curcumin Derivative Epigenetically Reactivates Nrf2 Antioxidative Stress Signaling in Mouse Prostate Cancer TRAMP C1 Cells. Chem Res Toxicol 2018;31(2):88–96. Epub 20180108. doi: 10.1021/acs.chemrestox.7b00248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Zhang C, Su ZY, Khor TO, Shu L, Kong AN. Sulforaphane enhances Nrf2 expression in prostate cancer TRAMP C1 cells through epigenetic regulation. Biochem Pharmacol 2013;85(9):1398–404. Epub 20130214. doi: 10.1016/j.bcp.2013.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Zhang C, Shu L, Kim H, Khor TO, Wu R, Li W, Kong AN. Phenethyl isothiocyanate (PEITC) suppresses prostate cancer cell invasion epigenetically through regulating microRNA-194. Mol Nutr Food Res 2016;60(6):1427–36. Epub 20160517. doi: 10.1002/mnfr.201500918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lodi A, Saha A, Lu X, Wang B, Sentandreu E, Collins M, Kolonin MG, DiGiovanni J, Tiziani S. Combinatorial treatment with natural compounds in prostate cancer inhibits prostate tumor growth and leads to key modulations of cancer cell metabolism. Npj Precis Oncol 2017;1. Epub 2017/12/05. doi: 10.1038/s41698-017-0024-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wang L, Wang C, Sarwar MS, Chou P, Wang Y, Su X, Kong AT. PTEN-knockout regulates metabolic rewiring and epigenetic reprogramming in prostate cancer and chemoprevention by triterpenoid ursolic acid. Faseb j 2022;36(11):e22626. Epub 2022/10/29. doi: 10.1096/fj.202201195R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Su ZY, Khor TO, Shu L, Lee JH, Saw CL, Wu TY, Huang Y, Suh N, Yang CS, Conney AH, Wu Q, Kong AN. Epigenetic reactivation of Nrf2 in murine prostate cancer TRAMP C1 cells by natural phytochemicals Z-ligustilide and Radix angelica sinensis via promoter CpG demethylation. Chem Res Toxicol 2013;26(3):477–85. Epub 20130308. doi: 10.1021/tx300524p. [DOI] [PubMed] [Google Scholar]