Abstract
MicroRNAs (miRNAs) have been implicated in many biological processes, cancer, and other diseases. In addition, miRNAs are dysregulated following exposure to toxic and genotoxic agents. Here we review studies evaluating modulation of miRNAs by dietary and pharmacological agents, which could potentially be exploited for inhibition of mutagenesis and carcinogenesis. This review covers natural agents, including vitamins, oligoelements, polyphenols, isoflavones, indoles, isothiocyanates, phospholipids, saponins, anthraquinones and polyunsaturated fatty acids, and synthetic agents, including thiols, nuclear receptor agonists, histone deacetylase inhibitors, antiinflammatory drugs, and selective estrogen receptor modulators. As many as 145 miRNAs, involved in the control of a variety of carcinogenesis mechanisms, were modulated by these agents, either individually or in combination. Most studies used cancer cells in vitro with the goal of modifying their phenotype by changing miRNA expression profiles. In vivo studies evaluated regulation of miRNAs by chemopreventive agents in organs of mice and rats, either untreated or exposed to carcinogens, with the objective of evaluating their safety and efficacy. The tissue specificity of miRNAs could be exploited for the chemoprevention of site-specific cancers, and the study of polymorphic miRNAs is expected to predict the individual response to chemopreventive agents as a tool for developing new prevention strategies.
Keywords: microRNA, Chemoprevention, Dietary agents, Pharmacological agents
1. Introduction
MicroRNAs (miRNAs) are small (18-25 nucleotides), noncoding, single-stranded RNAs, which negatively regulate gene expression either by translational inhibition or exonucleolytic messenger RNA (mRNA) decay [1]. These evolutionary conserved RNAs have been recognized in virtually all species, ranging from viruses to humans [2]. While the information provided by transcriptome analyses is redundant due to the fact that mRNA can be regulated posttranscriptionally, miRNAs regulate a number of genes simultaneously. The miRBase Version 16.0 has 1048 miRNA sequences annotated in the human genome, and miRNAs are believed to target about one-third of human mRNAs [3], a single miRNA targeting approximately 200 transcripts simultaneously [4].
Accordingly, miRNAs have been implicated in almost every biological process, including development, cell cycle regulation, cell growth and differentiation, stress response, and apoptosis [5]. In addition, miRNAs play a role in a variety of diseases [6] and in particular in cancer [7]. MiRNAs are known to be dysregulated as a response to toxic and genotoxic agents [8], including physical agents such as ionizing radiation [9] and UV radiation [10], chemical agents such as benzo[a]pyrene [11], hepatotoxicants [12] and drugs [13], and complex mixtures such as cigarette smoke [14,15] and environmental pollutants [16,17].
In the present review article we summarize the findings of a number of studies evaluating modulation of miRNAs by known inhibitors of mutagenesis and carcinogenesis. These inhibitors represent putative cancer chemopreventive agents, as assessed in experimental test systems and sometimes in clinical chemoprevention trials. They include both natural agents, mostly of dietary source, and synthetic agents, mostly used as pharmacological agents, which are analytically discussed in Sections 2 and 3, respectively.
Due to the large variety of mechanisms by which it is possible to inhibit mutagenesis and carcinogenesis [18-20], modulation of miRNAs as an epigenetic response to drugs [21] and dietary agents [22-26] is of particular relevance to understand their mechanism of action and to evaluate their safety and efficacy.
Table 1 summarizes the main findings relative to modulation of miRNAs by putative cancer chemopreventive agents, as inferred from both literature data available in PubMed up to December 2011 and unpublished data from our laboratory. . The investigated miRNAs are listed starting from the let-7 family and continuing with the miR series in increasing nomenclature number. Those miRNAs that are identified with the symbol § in Table 1 undergo single nucleotide polymorphisms (SNPs) in humans [27,28]. The tissue specificity indicates the cell type or organ in which each miRNA has the highest expression levels, as reported in the Mirnamap database (mirnamap.mbc.nctu.edu.tw) and literature data. The main functions regulated by each miRNA are inferred from the Mirnamap database (mirnamap.mbc.nctu.edu.tw), the Mirbase database (www.mirbase.org) and from literature data.
Table 1.
Denomination and main functions of miRNAs that were have been reported, up to December 2011, to be modulated by natural and synthetic chemopreventive agents in either in vitro or in vivo studies.
| MiRNA | Tissue specificity | Main regulated functions | Analyzed cells (in vitro) or species/organ (in vivo) | Modulating agent [Ref.] |
|---|---|---|---|---|
| let-7a § | Lung, cervix, liver | Cell proliferation, k-Ras activation, apoptosis | Human leukemia cells Human hepatocarcinoma cells Mouse lung (CS+) Mouse lung Mouse lung (CS+) Mouse liver Rat lung (CS+) Rat lung (CS+) |
↑Retinoic acid [30] ↑Ellagitannin BJA32515 [51] ↑PEITC [82, 83] ↑SAHA [UD] ↑Budesonide [82] ↓Budesonide [82] ↑PEITC [81] ↑Oltipraz+NAC [81] |
| let-7b | Lung, cervix, kidney | Cell proliferation | Human pancreatic cancer cells Human pancreatic cancer cells Human lung cancer cells Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↓Diindolylmethane [78] ↓Genistein [78] ↑SAHA [93] ↑Oltipraz [81] ↑Oltipraz+NAC [81] ↑PEITC+I3C [81] |
| let-7c | Lung, kidney | Intercellular adhesion | Human hepatocarcinoma cells Human leukemia cells Human pancreatic cancer cells Human pancreatic cancer cells Mouse lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↑EGCG [53] ↑Retinoic acid [30] ↓Genistein [78] ↓Diindolylmethane [78] ↑PEITC [82] ↑PEITC [81] ↑PEITC+I3C [81] ↑Oltipraz [81] ↑Oltipraz+NAC [81] |
| let-7d | Lung, cervix, prostate, bladder | Cell proliferation | Human leukemia cells Human pancreatic cancer cells Human pancreatic cancer cells Rat colon (AOM+) |
↑Retinoic acid [30] ↓Genistein [78] ↓Diindolylmethane [78] ↑PUFA (Fish oil) [86] |
| let-7e | Lung, cervix | Cell proliferation | Human hepatocarcinoma cells Human pancreatic cancer cells Human pancreatic cancer cells |
↑Ellagitannin BJA3121 [52] ↓Genistein [78] ↓Diindolylmethane [78] |
| let-7f | Lung, cervix, kidney, liver | Cell proliferation, k-Ras activation, angiogenesis | Rat lung (CS+) | ↑PEITC+I3C [81] |
| let-7l | Lung | Cell proliferation | Human colon carcinoma cells | ↓SAHA [90] |
| miR-9 | Brain | Apoptosis | Rat fetus central nervous system | ↓Retinoic acid [37] |
| miR-10 § | Lung, kidney, breast | Angiogenesis | Human neuroblastoma cells Human embryonic stem cells Mouse fetus brain Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↑Retinoic acid [36] ↑PUFA [87] ↓Folic acid [40] ↑5,6-Benzoflavone [81] ↑Oltipraz [81] ↑Oltipraz+NAC [81] ↑I3C [81] ↑PEITC+I3C [81] |
| miR-15 | Lymphocyte, thymus, lung, liver | Lymphocyte differentiation, apoptosis | Human breast cancer cells Human leukemia cells Human lymphoma cells Mouse liver (CS+) Rat colon (AOM+) |
↑Curcumin [58] ↑Retinoic acid [30] ↓SAHA [91] ↑Budesonide [82] ↑PUFA (Fish oil) [86] |
| miR-16 § | Lung | Apoptosis | Human hepatocarcinoma cells Human leukemia cells |
↑EGCG [53] ↑Retinoic acid [30] |
| miR-17 | Lung, kidney, bladder | Tumor suppressor gene PTEN, DICER, TGF-beta, c-Myc | Human colon cancer cells Human prostate cancer cells Human leukemia cells Human neuroblastoma cells Human lymphoma cells |
↓Resveratrol [62] ↓Resveratrol [68] ↓Retinoic acid [29] ↓Retinoic acid [35] ↓SAHA [91] |
| miR-18 | Lung, kidney, prostate | Small RNA transcription | Human hepatocarcinoma cells Human lymphoma cells |
↑EGCG [53] ↓SAHA [91] |
| miR-20 | Bladder, lung, thymus, prostate, kidney | No data available | Rat heart | ↑Resveratrol [69, 70] |
| miR-21 | Lung, kidney, bladder, liver | Tumor suppressor gene PTEN, cell proliferation | Human pancreatic cancer cells Human colon cancer cells Human colon cancer cells Human breast cancer cells Human breast cancer cells Human breast cancer cells Mouse lung (VC+) Mouse liver (CS+) Rat heart Human breast cancer cells |
↓Difluorinated curcumin [61] ↓Curcumin [60] ↓Resveratrol [62] ↑Diindolylmethane [79] ↑Retinoic acid [32] ↓Polyphenon-60 [55] ↓I3C [80] ↑Budesonide [82] ↑Resveratrol [69, 70] #Tamoxifen [99] |
| miR-22 | Muscle | Estrogen receptor alpha | Human pancreatic cancer cells Human colon cancer cells |
↑Curcumin [57] ↑SAHA [90] |
| miR-23 | Ovary, kidney, bladder | Gene transcription | Neuronal cells | ↑Retinoic acid [33] |
| miR-24 | Lung, kidney, prostate | Cell differentiation | Human embryonic stem cells | ↑PUFA [87] |
| miR-25 § | Pancreas, breast, kidney | DICER | Human colon cancer cells Human hepatocarcinoma cells Human leukemia cells |
↓Resveratrol [62] ↑EGCG [53] ↓Retinoic acid [29] |
| miR-26 | Lung, kidney, bladder, cervix, liver | TGF beta | Mouse liver Mouse lung (CS+) Mouse lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↓PEITC [82] ↑PEITC [82] ↑Budesonide [82] ↑PEITC [81] ↑5,6 benzoflavone [81] ↑Oltipraz[81] ↑NAC [81] ↑Oltipraz+NAC [81] ↑I3C [81] ↑PEITC+I3C [81] |
| miR-27 | Bladder, prostate, cervix, kidney, liver | Tumor suppressor genes, cell proliferation, stress response, protein repair | Human colon cancer cells Human breast cancer cells Human uveal melanoma cancer cells Mouse liver |
↓NO-NSAID GT-094 [97] ↓Poryphenon-60 [55] ↓Genistein [71] ↓Budesonide [82] |
| miR-29 | Kidney, lung, heart | Collagen production, inflammation, apoptosis | Human colon cancer cells Human hepatocarcinoma cells Human cardiac cells Rat heart Mouse lung (CS+) |
↓SAHA [90] ↑Ellagitannin BJA32515 [51] ↓Pioglitazone [89] ↓Pioglitazone [89] ↑PEITC [82] |
| miR-30 § | Kidney, lung | Intercellular adhesion, protein repair, NFκB activation, cell cycle, EGF activation, stem cell recruitment, multidrug resistance | Human lung cancer cells Human hepatocarcinoma cells Human hepatocarcinoma cells Mouse lung (CS+) Rat lung (CS+) |
↓SAHA [93] ↓EGCG [54] ↓Anthocyanidin [54] ↑PEITC [83] ↑PEITC+I3C [81] |
| miR-31 | Small intestine, kidney, lung | Protein synthesis and secretion, stress response | Mouse lung (VC+) Mouse lung (CS+) Mouse lung (CS+) |
↓I3C [80] ↑PEITC [82] ↑Budesonide [82] |
| miR-32 § | Apoptosis | Human leukemia cells | ↑1,25-dihydroxyvitamin D3 [43] | |
| miR-34 § | Ovary, prostate, lung | P53 | Human cancer prostate cells Human pancreatic cancer stem cells Mouse liver Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↑Selenium [49] ↑SAHA [92] ↑PEITC [82] ↑5,6 benzoflavone [81] ↑Oltipraz [81] ↑Oltipraz+NAC [81] ↑I3C [81] ↑PEITC+I3C [81] |
| miR-92 § | Lung | DICER | Human colon cancer cells Human lung cancer cells Human hepatocarcinoma cells |
↓Resveratrol [62] ↓Resveratrol [67] ↑EGCG [53] |
| miR-99 | Cervix, prostate, ovary, kidney, lung | Apoptosis | Rat lung (CS+) Rat lung (CS+) |
↑PEITC [81] ↑PEITC+I3C [81] |
| miR-100 | Liver, placenta, cervix, lung | Apoptosis | Human colon cancer cells Mouse liver |
↓SAHA 90] ↓Budesonide [82] |
| miR-106 | Thymus, kidney, bladder, lung, liver, placenta | Cell adhesion, TNF activation, stress response | Prostate cancer cells Human lung cancer cells Human colon cancer cells Mouse liver |
↓Resveratrol [68] ↓SAHA [93] ↓PUFA [88] ↓Budesonide [82] |
| miR-107 | Brain, kidney | Intracellular trafficking, apoptosis | Human leukemia cells Rat colon (AOM+) |
↑Retinoic acid [30] ↑PUFA (Fish oil) [86] |
| miR-122 § | Liver | Stress response, lipid metabolism | Rat liver (choline-def. diet) Rat liver Rat liver |
↑Folate [38, 39] ↓α-Tocopherol [48] ↑α-Tocopherol [47] |
| miR-123 | Lung | Angiogenesis, cell proliferation | Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↑NAC [81] ↑Oltipraz [81] ↑Oltipraz+NAC [81] ↑PEITC [81] ↑PEITC+I3C [81] |
| miR-124 | Brain, lung | Gene transcription, apoptosis | Human lung cancer cells Rat fetus central nervous system Rat lung (CS+) |
↑SAHA [93] ↓Retinoic acid [37] ↑PEITC+I3C [81] |
| miR-125 § | Lung, cervix, brain, ovary, prostate, bladder | Oncogene ERBB, vitamin D receptor, inflammation, gene transcription | Human melanoma cells Rat lung (CS+) Mouse lung Rat liver Rat liver Rat fetus central nervous system Mouse lung (CS+) Mouse liver Mouse liver (CS+) Mouse liver Mouse liver (CS+) Mouse lung (CS+) Rat lung (CS+) Rat lung (CS+) |
*1,25-dihydroxyvitamin D3 [45; 46] ↑I3C [81] ↑SAHA [UD] ↓α-Tocopherol [48] ↑α-Tocopherol (Vit. E) [47] ↓Retinoic acid [37] ↑PEITC [82] ↓PEITC [82] ↑PEITC [81] ↓Budesonide [82] ↑Budesonide [82] ↑PEITC [82] ↑PEITC+I3C [81] |
| miR-126 | Lung, kidney | Gene transcription | Human lung cancer cells | ↑SAHA [93] |
| miR-128 | Brain | Cell proliferation, apoptosis | Human glioma cells | ↑Ginsenoside Rh2 [84] |
| miR-129 | Brain, lung | Calmodulin transcription activation | Human lung cancer cells Human hepatocarcinoma cells |
↑SAHA [93] ↓EGCG [53] |
| miR-130 | Kidney, cervix, prostate, lung, liver, | Gene transcription, apoptosis | Mouse lung (VC+) | ↓I3C [80] |
| miR132 § | Brain | Gene transcription | Human lung cancer cells | ↑SAHA [93] |
| miR-133 | Lung, prostate | Inflammation | Mouse lung | ↓Budesonide [82] |
| miR-135 | Kidney, thyroid, lung | Ras regulation, cell adhesion | Mouse lung (CS+) | ↑PEITC [82] |
| miR-139 | Brain | Cell proliferation, cell differentiation | Human breast cancer cells | ↑Trichostatin A [94] |
| miR-140 § | Lung | P53 | Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↑NAC [81] ↑Oltipraz+NAC [81] ↑PEITC+I3C [81] |
| miR-141 | Placenta, kidney, intestine | Cell proliferation, cancer invasion | Human colon cancer cells | ↓SAHA [90] |
| miR-142 | Liver, thymus, spleen, lung | Protein repair, DNA repair, prostaglandin-mediated platelet aggregation | Mouse liver (CS+) Mouse liver |
↓PEITC [82] ↓Budesonide [82] |
| miR-145 | Prostate, cervix, ovary, bladder, lung | Protein repair, angiogenesis | Rat lung (CS+) | ↑PEITC+I3C [81] |
| miR-146 § | Lung | NFκB stress response, inflammation | Human colon fibroblasts Human neural cells Human pancreatic cancer cells Human breast cancer cells Mouse lung (VC+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↑Polyphenolic extracts [50] ↓Resveratrol [65] ↑Genistein [23] ↑Trichostatin A [94] ↓I3C [80] ↑5,6-benzoflavone [81] ↑NAC [81] ↑Oltipraz+NAC [81] ↑PEITC [81] ↑PEITC+I3C [81] |
| miR-152 | Neural tissue | Transcriptional repressor | Human neuroblastoma cells | ↑Retinoic acid [34] |
| miR-153 | Liver, brain, lung | Protein repair, protein synthesis, signal transduction | Mouse liver (CS+) | ↑PEITC [82] |
| miR-155 | Lung | TGF-beta | Human monocytic cells Human breast cancer cells Human lymphoma cells |
↓Resveratrol [63, 64] ↑Trichostatin A [94] ↓SAHA [91] |
| miR-156 | Plants (Arabidopsis, Arachis) | Anthocyanin accumulation | Arabidopsis thaliana | •Anthocyanin [77] |
| miR-181 § | Brain, thymus, kidney, lung | NFκB stress response | Human leukemia cells Mouse lung Human leukemia cells Human breast cancer cells |
↓Vitamin D3 [42] ↓PEITC [82] ↓Retinoic acid [30] #Tamoxifen [99] |
| miR-182 § | Thymus, lung | Inflammation, cell proliferation | Human breast cancer cells | ↑2,5-Hydroxyvitamin D3 [44] |
| miR-183 | Lung | Apoptosis, cell adesion | Lung cancer cells Mouse lung |
↑SAHA [93] ↑SAHA [UD] |
| miR-186 | Bladder | Apoptosis | Human lung cancer cells Human leukemia cells |
↓Curcumin [59] ↑Retinoic acid [29] |
| miR-191 | Brain, cervix, kidney, lung | Cell proliferation | Rat colon (AOM+) Rat lung (CS+) |
↑PUFA (Fish oil) [86] ↑PEITC+I3C [81] |
| miR-192 | Intestine, kidney, liver, lung | Cell proliferation, k-Ras activation | Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
↑PEITC [81] ↑PEITC+I3C [81] ↑Oltipraz+NAC [81] |
| miR-193 | Muscle, lymphocytes | Signal transduction | Human leukemia cells | ↓Retinoic acid [29] |
| miR-194 § | Intestine, kidney | Apoptosis, cell proliferation | Human lung cancer cells | ↑Resveratrol [67] |
| miR-195 | Cervix, prostate, ovary, bladder, lymphocytes | Small RNA transcription | Human leukemia cells | ↓Retinoic acid [29] |
| miR-196 § | Kidney, cervix | TGF-beta | Human colon cancer cells Human hepatocarcinoma cells Human pancreatic cancer cells |
↓Resveratrol [62] ↓EGCG [53] ↓Curcumin [57] |
| miR-197 | Brain | Apoptosis, cell proliferation | Human hepatocarcinoma cells Human hepatocarcinoma cells |
↓Ellagitannin BJA32515 [51] ↑Anthocyanidin [54] |
| miR-200 § | Kidney, lung, liver | Apoptosis, intracellular trafficking, protein repair | Human pancreatic cancer cells Human pancreatic cancer cells Human pancreatic cancer cells Human hepatocarcinoma cells Mouse lung (CS+) Mouse liver Mouse liver |
↓Difluorinated curcumin [61] ↓Genistein [78] ↓Diindolylmethane [78] ↓EGCG [53] ↑PEITC [82] ↓PEITC [82] ↓Budesonide [82] |
| miR-210 | Lung | Hipoxia-inducible factor-1 | Mouse and human lung cancer cells | ↑EGCG [56] |
| miR-215 | Intestine | Intracellular trafficking, apoptosis | Human leukemia cells Human breast cancer cells |
↑Retinoic acid [29] ↑Trichostatin A [94] |
| miR-218 § | Lung, kidney, bladder, prostate, liver | Stress response, oncogene k-Ras activation, antioxidant | Mouse lung (CS+) Mouse lung (CS+) |
↑Pioglitazone [UD] ↑Bexarotene+Pioglitazone [UD] |
| miR-221 | Prostate | SERM resistance | Human breast cancer cells Human breast cancer cells Human prostate cancer cells Human colon cancer cells |
#Tamoxifen [99; 98] #Fulvestrant [101] ↓Genistein [85] ↓SAHA [90] |
| miR-222 § | Prostate, lung, bladder | Angiogenesis, cell proliferation, SERM resistance | Human breast cancer cells Human breast cancer cells Human lymphoblastoid cells Human colon cancer cells Human prostate cancer cells Rat lung (CS+) Rat lung (CS+) Rat lung (CS+) |
#Tamoxifen [99; 98] #Fulvestrant [101] ↓Folate [41] ↓SAHA [Shin et al. 2009] ↓Genistein [85] ↑Oltipraz+NAC [81] ↑PEITC [81] ↑PEITC+I3C [81] |
| miR-223 | Spleen, lung | Protein repair, k-Ras activation | Human leukemia cells Rat lung (CS+) |
↑Retinoic acid [29; 30] ↑PEITC+I3C [81] |
| miR-290 | Lung | Stem cell marker | Mouse lung (CS+) | ↑Bexarotene+Pioglitazone [UD] |
| miR-292 | Liver | Hepatocyte growth factor-induced cell proliferation, angiogenesis | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-296 § | Muscle, prostate, lung, bladder | Thioredoxin and cysteine synthesis (antioxidants), inflammation | Mouse lung (CS+) Mouse lung (CS+) |
↑Pioglitazone [UD] ↑Bexarotene+Pioglitazone [UD] |
| miR-297 | Liver | Protein repair, cell cycle | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-299 | Liver, lung, cervix, testes | NFκB activation, stress response, peroxisome activation | Human lung cancer cells Mouse liver (CS+) |
↑Resveratrol [67] ↑PEITC [82] |
| miR-300 | Liver, | Protein repair, intracellular trafficking, cell proliferation | Mouse liver | ↓Budesonide [82] |
| miR-302 | Lung | Cell adhesion, protein repair, intracellular trafficking, cell proliferation | Mouse lung Mouse lung |
↑Myo-inositol [UD] ↓SAHA [UD] |
| miR-320 | Bladder, cervix, liver, lung | Protein repair, intracellular trafficking, cell proliferation | Mouse liver | ↓Budesonide [82] |
| miR-322 | Liver | Protein repair, cell proliferation | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-323 | Liver | Peroxisome activation, protein repair | Mouse liver | ↓PEITC [82] |
| miR-324 § | Brain, kidney, prostate | Cell proliferation | Rat colon (AOM+) | ↑PUFA (Fish oil) [86] |
| miR-331 | Liver | Stress response | Mouse liver Mouse liver |
↓PEITC [82] ↓Budesonide [82] |
| miR-335 | Lung | Insulin growth factor, cell proliferation, apoptosis | Mouse lung (CS+) Mouse lung (CS+) |
↑Pioglitazone [UD] ↑Bexarotene+Pioglitazone [UD] |
| miR-338 | Liver | Protein repair, stress response | Human lung cancer cells Mouse liver Mouse liver |
↑Resveratrol [67] ↓PEITC [82] ↓Budesonide [82] |
| miR-342 | Brain, lymphocytes | Stress response, protein repair, SERM resistance | Human breast cancer cells Human leukemia cells Human hepatocarcinoma cells |
#Tamoxifen [99;100] ↑Retinoic acid [30] ↓EGCG [53] |
| miR-345 § | Thyroid, kidney | Intracellular trafficking | Human lung cancer cells | ↓SAHA [93] |
| miR-370 | Brain | Apoptosis, inflammation | Human hepatocarcinoma cells | ↑Ellagitannin BJA3121 [52] |
| miR-373 § | Liver | Apoptosis, inflammation | Human hepatocarcinoma cells Human hepatocarcinoma cells |
↓Ellagitannin BJA32515 [51] ↑Ellagitannin BJA3121 [52] |
| miR-376 | Liver | Carbonic anhydrase (antioxidant), peroxisome biogenesis, P53, cell cycle progression, signal transduction, apoptosis, intracellular vesicle trafficking | Mouse liver (CS+) | ↑PEITC [82] |
| miR-377 | Lung | Angiogenesis | Mouse lung (VC+) | ↓I3C [80] |
| miR-382 | Lung, brain | Gene transcription | Human lung cancer cells Mouse lung (CS+) Mouse lung (CS+) |
↑SAHA [93] ↑PEITC [82] ↑Budesonide [82] |
| miR-409 § | No data available | Intracellular trafficking | Human lung cancer cells | ↑SAHA [93] |
| miR-424 | Intestine, uterus | No data available | Human colon cancer cells | ↑SAHA [90] |
| miR-452 | Prostate, cervix, kidney, lung | Stress response, cell cycle arrest in response to DNA damage | Mouse liver | ↑PEITC [82] |
| miR-463 | Lung | Cell proliferation, protein repair, stress response | Mouse lung (CS+) Mouse liver (CS+) |
↑Budesonide [82] ↑PEITC [82] |
| miR-466 § | Lung, liver | Cell proliferation,, k-Ras activation, gene transcription | Mouse lung Mouse liver Mouse liver (CS+) Mouse lung Mouse liver Mouse liver (CS+) |
↓PEITC [82] ↓PEITC [82] ↑PEITC [82] ↑SAHA [UD] ↓Budesonide [82] ↑Budesonide [82] |
| miR-467 | Liver | Cell proliferation, protein synthesis | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-470 | Liver | k-Ras activation, intracellular vesicle trafficking, xenobiotic metabolism | Mouse liver (CS+) | ↑PEITC [82] |
| miR-483 | Liver, lung | Protein repair | Mouse liver | ↑Budesonide [82] |
| miR-484 | Lung, heart | Apoptosis, cell differentiation | Mouse lung (CS+) | ↑Bexarotene+Pioglitazone [UD] |
| miR-489 | Human breast cancer cells | #Tamoxifen [99] | ||
| miR-493 | Lung | Cell differentiation, cell proliferation | Mouse lung (CS+) Mouse lung (CS+) |
↑Bexarotene [UD] ↑Bexarotene+Pioglitazone [UD] |
| miR-509 § | Testis, kidney, lung | Cell adhesion, k-Ras activation | Mouse lung | ↑Myo-inositol [UD] |
| miR-526 § | Placenta, liver | Cell proliferation, apoptosis, inflammation | Human hepatocarcinoma cells Human hepatocarcinoma cells |
↑Ellagitannin BJA3121 [52] ↓EGCG [53] |
| miR-532 | Liver | Gene transcription, apoptosis | Human hepatocarcinoma cells | ↑Anthocyanidin [54] |
| miR-539 | Liver | Protein repair, intracellular trafficking | Mouse liver | ↑Budesonide [82] |
| miR-543 | Lung | Stress response, inflammation | Mouse lung | ↓Myo-inositol [UD] |
| miR-544 | Breast | P53 | Human breast cancer cells | ↑Trichostatin A [94] |
| miR-548 § | No data available | Gene transcription | Human lung cancer cells | ↓SAHA [93] |
| miR-551 | Liver | DNA repair, inflammation, cell proliferation | Mouse liver Mouse liver |
↓PEITC [82] ↓Budesonide [82] |
| miR-582 | Prostate, bladder, kidney | Cell proliferation, apoptosis | Human lung cancer cells | ↑Resveratrol [67] |
| miR-592 | Intestine, lung | Cell adhesion, insulin growth factor, angiogenesis | Mouse lung | ↑Myo-inositol [UD] |
| miR-622 | Lung | Cell proliferation, k-Ras activation | Human bronchial cells | ↑Resveratrol [66] |
| miR-638 | Adipose tissue, intestine | Cell proliferation, apoptosis | Human lung cancer cells | ↑Bostrycin [75] |
| miR-645 | Breast | Gene transcription | Human breast cancer cells | ↓Trichostatin A [94] |
| miR-657 | Testes, brain, prostate, cervix, ovary, liver, kidney, lung | Gene transcription | Human breast cancer cells | ↓Trichostatin A [94] |
| miR-660 § | Kidney, lung | Protein repair | Human lung cancer cells | ↑SAHA [93] |
| miR-663 § | Prostate | TGF-beta | Human colon cancer cells Human leukemia cells |
↑Resveratrol [63] ↑Retinoic acid [31] |
| miR-666 | Lung | Protein repair, stress response | Mouse lung | ↓PEITC [82] |
| miR-684 | Lung | Signal transduction | Mouse lung | ↓SAHA [UD] |
| miR-687 | Liver | Tumor suppression by phosphatidylinositol catabolism, cell proliferation | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-690 | Lung | Cell proliferation, cell adhesion | Mouse liver (CS+) | ↑Budesonide [82] |
| miR-697 | Liver | Protein repair, intracellular trafficking, cell adhesion | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-706 | Lung | Intracellular trafficking, cell motility | Mouse lung | ↓PEITC [82] |
| miR-708 | Lung | Stress response, NFκB activation | Mouse lung | ↓PEITC [82] |
| miR-709 | Liver | Stress response, inflammation, lysosome activation | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-710 | Liver | Cell proliferation, collagen production, k-Ras activation | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-715 | Lung | No data available | Mouse lung (CS+) | ↑Bexarotene+Pioglitazone [UD] |
| miR-719 | Liver | Inflammation | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-742 | Liver | Protein repair, stress response | Mouse liver | ↑Budesonide [82] |
| miR-758 § | Lung | Cell proliferation, apoptosis | Human lung cancer cells | ↑Resveratrol [67] |
| miR-763 | Liver | Cell membrane integrity, peroxisome biogenesis, stress response | Mouse liver | ↓Budesonide [82] |
| miR-764 | Lung | Mitochondrial function | Mouse lung Mouse lung Mouse lung (CS+) Mouse lung (CS+) |
↓Myo-inositol [UD] ↓SAHA [UD] ↑Pioglitazone [UD] ↑Bexarotene+Pioglitazone [UD] |
| miR-804 | Liver, lung | Cell proliferation, collagen production, k-Ras activation | Mouse lung (CS+) | ↑PEITC [83] |
| miR-874 | Liver | Protein repair, intracellular vesicle trafficking, cell proliferation, P53 dependent apoptosis, inflammation, stress response | Mouse liver (CS+) | ↑PEITC [82] |
| miR-876 | Lung | No data available | Mouse lung | ↑Myo-inositol [UD] |
| miR-877 | Lung | No data available | Human lung cancer cells Human colon cancer cells |
↓SAHA [93] ↓SAHA [90] |
| miR-880 | Lung | No data available | Mouse lung | ↑Myo-inositol [UD] |
| miR-883 | Liver | Tumor suppressing activity through phosphatidylinositol catabolism, protein synthesis, intracellular vesicle trafficking, protein repair, cell proliferation | Mouse liver (CS+) Mouse liver (CS+) |
↑PEITC [82] ↑Budesonide [82] |
| miR-923 | Lung | Cell proliferation, apoptosis | Human lung cancer cells | ↑Bostrycin [75] |
| miR-936 | No data available | No data available | Human lung cancer cells | ↓SAHA [93] |
| miR-1224 | No data available | No data available | Human hepatocarcinoma cells | ↑Anthocyanidin [54] |
| miR-1296 | Prostate, ovary | Cell cycle arrest | Human prostate cancer cells Human ovarian cancer cells |
↑Genistein [74] ↑Genistein [73] |
| miR-1901 | Lung | No data available | Mouse lung | ↓Myo-inositol [UD] |
| miR-1953 | No data available | No data available | Mouse lung | ↑Myo-inositol [UD] |
(CS+), mice or rats exposed to cigarette smoke; (AOM+), rats treated with azoxymethane; (VC+), mice treated with vinyl chloride.
↑ upregulation, ↓ downregulation; *Modulation of antitumor effects; •Anthocyanins accumulation in plant is regulated by miR-156; #the reported miRNA is responsible for resistance to SERMs.
miRNA undergoing single nucleotide polymorphisms in humans.
UD, Izzotti et al., unpublished data
The fourth column in Table 1 reports either the cells that were analyzed in vitro or the species and organ analyzed in vivo. Most in vitro studies used cancer cells, mainly of human origin, in which the authors investigated the ability of putative anticancer agents to modulate the expression of miRNAs with the goal of exploring their mechanisms of action and modifying their phenotype. Apart from a couple of studies using human samples and another one using a plant, all other in vivo studies used tissues from rats or mice exposed to carcinogens, such as cigarette smoke (CS), vinyl carbamate (VC), and azoxymethane (AOM), or subjected to particular diets, such as vitamin- or choline-deficient diets. Several studies from our laboratory analyzed in parallel miRNA expression in organs of rodents, either unexposed or exposed to CS, in order to evaluate modulation by the investigated agents both of baseline expression profiles and of CS-induced dysregulation. This approach allowed us to predict both safety and efficacy of test agents at the molecular level.
The last column in Table 1 reports, for each miRNA, the results obtained, the investigated agent, and the corresponding reference. The arrows indicate whether modulation of miRNA expression occurred either in the sense of upregulation (upward arrows) or downregulation (downward arrows). The meaning of other symbols is reported in the footnote to the table.
It should be noted that some authors of the reviewed papers did not report all modulated miRNAs but made a selection of those that were evaluated to be more relevant. In general, the selection was made according to three analytical criteria, including (a) a more than two-fold variation, accompanied by a statistically significant difference; (b) inclusion in the highest or lowest quartile of distribution; and (c) confirmation of data by biological or functional analyses.
2. Modulation of miRNA expression by natural agents
2.1. Vitamins and derivatives
2.1.1. Vitamin A metabolites (retinoic acid)
All-trans-retinoic acid (RA) is a metabolite of vitamin A (all-trans-retinol) responsible for most of its biological effects. RA was tested in vitro for the ability to modulate miRNA expression in a variety of human cultured cancer cells, including acute promyelocytic leukemia (APL) cells, estrogen recepor-positive breast cancer (MCF-7) cells, embryonal carcinoma (NT2) cells, and neuroblastoma cells.
In APL cells, RA upregulated the expression of miR-186, miR-215 and miR-223, while it downregulated the expression of miR-17, miR-25, miR-193, miR-195 [29]. In another study using the same cells, RA was found to upregulate the expression of miR-15a, miR-15b, miR-16-1, let-7a-3, let-7c, let-7d, miR-107, miR-223, and miR-342, whereas miR-181b was downregulated [30]. Differentiation of APL cells by RA was reported to be mediated by miRNA modulation, mainly involving miR-663 upregulation [31].
Proliferation of MCF-7 cells was inhibited by RA via miR-21 upregulation [32]. MiR-23 was shown to play a critical role in the RA-induced neuronal differentiation of NT2 cells into neural cells [33]. In another study, differentiation of these cells was induced by RA following miR-152 upregulation [34]. In addition, RA downregulated miR-17, which in turn activated the expression of genes involved in neuroblastoma cell differentiation and apoptosis [35], and upregulated miR-10a and miR-10b, targeting the SR-family splicing factor SFRS1 [36].
RA downregulated the expression of miR-9, miR-124, and miR-125b in the central nervous system of rat fetuses, thereby increasing Bcl2- and P53- related apoptosis and inducing an abnormal development of spinal cord [37].
2.1.2. Vitamin B9 (folate)
In male Fisher rats, a diet deficient in folate, methionine and choline resulted in the formation of hepatocellular carcinoma at 54 weeks of age, in the absence of carcinogen treatment. This process was accompanied by miR-122 downregulation. Folate replenishment increased miR-122 levels and was associated with inhibition of liver tumorigenesis [38,39].
Folic acid blocked ethanol-induced teratogenesis in fetal mouse brain through miR-10a downregulation [40].
Utilizing blood samples from a population-based case-control study of head and neck squamous cell carcinoma, miR-222 was identified as being overexpressed in lymphoblastoid cells in culture obtained from subjects with a low folate intake. Folate supplementation in the culture medium restored miRNA levels, which suggests that dietary modulation of miRNA expression is reversible [41].
2.1.3. Vitamin D (calciferol) and derivatives
Vitamin D was found to modulate in vitro the expression of miRNAs in cultured human cancer cells. In particular, vitamin D3 downregulated miR-181 resulting in cell cycle arrest of human myeloid leukemia cells [42]. 1,25-Dihydroxyvitamin D3 markedly induced the expression of miR-32 in the same cells, leading to Bim targeting and inhibition of AraC-induced apoptosis [43].
In breast epithelial cells (MCF-12F), 2,5-hydroxyvitamin D3, a major vitamin D metabolite, conferred a protective role against cellular stress by modulating P53 and PCNA levels and dysregulating the expression of several miRNAs, among which miR-182 [44].
The cancer chemopreventive effects of vitamin D are mediated via binding with its receptor, whose expression is linked to miR-125b [45]. Indeed, malignant melanoma cells expressing the vitamin D receptor respond to the antiproliferative effects of vitamin D3. Endogenous vitamin D receptor-mRNA levels are inversely related with the expression of miR-125b, which is involved in the resistance against vitamin D3 antiproliferative effects in melanoma cells [46].
2.1.4. Vitamin E (tocopherol)
In Fisher 344 rats fed for 6 months diets deficient or sufficient in α-tocopherol, the major congenerer of vitamin E, vitamin E-deficiency resulted in reduced liver concentrations of miR-122a, which is involved in lipid metabolism, and miR-125b, which is involved in inflammation [47]. A review paper published by the same group reported that α-tocopherol downregulates the same miRNAs [48].
2.2. Oligoelements
Sodium selenite activated the P53 pathway and the related miRNA effector miR-34 in prostate cells. In fact, incubation of P53+/+ human prostate cancer cells with selenium triggered induction of miR-34, which was associated with a rapid transcriptional activation of P53 and upregulation of the expression of P53-targeted genes [49].
2.3. Polyphenols
2.3.1. Flavonol-rich extracts
Polyphenolics extracted from Ilex vomitoria (yaupon holly) leaves, whose main components are quercetin and kaempferol 3-rutinosides, upregulated miR-146a, a negative regulator of proinflammatory NFκB, in human colon fibroblasts (CCD-18Co), and protected these cells from inflammation [50].
2.3.2 Ellagitannins
In human hepatocellular carcinoma HepG2 cells, 1,3,4-tri-O-galloyl-6-O-caffeoyl-β-D-glucopyranose (BJA32515), a natural ellagitannin compound extracted from Balanophora japonica Makino, upregulated let-7a and miR-29a and downregulated miR-197 and miR-373. These miRNA modification resulted in inhibited cell proliferation and increased apoptosis [51]. In the same cells, 1,3-di-O-galloyl-4,6-(s)-HHDP-b-D-glucopyranose (BJA3121) dysregulated the expression of 25 miRNAs, and in particular upregulated let-7e, miR-370, miR-373, and miR-526, thus inhibiting cell proliferation [52].
2.3.3. Epigallocatechin 3-gallate and other green tea polyphenols
HepG2 cells, epigallocatechin 3-gallate (EGCG) treatment altered the expression levels of a total of 61 miRNAs, 13 of which were upregulated and 48 were downregulated. Among them, miR-16, which was confirmed to target and to inhibit the antiapoptotic protein Bcl- 2, was one of the upregulated miRNAs. This mechanism explains the proapoptotic effect exerted by EGCG. Other miRNAs changed their expression more than 2-fold as a consequence of EGCG treatment, including let-7c, miR-18, miR-25, and miR-92 (all of them upregulated), and miR-129, miR-196, miR-200, miR-342, and miR-526 (all of them downregulated) [53]. In another study using the same cells treated with EGCG, miR-30b was found to be downregulated [54].
In human breast cancer MCF-7 cells, 23 miRNAs were differentially expressed after treatment with Polyphenon-60, a green tea extract. These miRNAs included miR-21 and miR-27, which were found to be downregulated. These two miRNAs had previously been identified as being overexpressed in these cells, with miR-21 specifically implicated in the downregulation of the tumor suppressor gene tropomyosin-1 [55].
In mouse and human lung cancer cells in culture, EGCG specifically upregulated the expression of miR-210, a major miRNA modulating the hypoxya-inducible factor 1α (HIF-1α) pathway. The EGCG-induced upregulation of miR-210 stabilized HIF-1α by inhibiting its ubiquitination and subsequent proteasome degradation in lung cancer cell lines, thus leading to reduced cell proliferation rate and anchorage-independent growth [56].
2.3.4. Curcumin and analogues
Curcumin (diferuloylmethane) is a flavonoid derived from the rhizome of Curcuma longa. In vitro, curcumin altered miRNA expression in human pancreatic cancer cells (PxBC-3) by upregulating miR-22, whose predicted targets were estrogen receptor 1 and transcription factor Sp1. On the other hand, miR-196, an oncogenic miRNA involved in gastric cancer, was significantly downregulated after curcumin treatment [57].
In human breast cancer cells (MCF-7), curcumin reduced the expression of Bcl-2 by upregulating miR-15a and miR-15b [58].
In addition, alterations in miRNA expression were detected in curcumin-treated lung cancer A549 cells, including a significant downregulation of miR-186, whose targets include caspase-10. These results demonstrate that curcumin induces A549 cell apoptosis through a miRNA-mediated pathway [59].
In colorectal cancer cells (Rko and HCT116), curcumin inhibited the transcriptional regulation of miR-21 via AP-1 and suppressed cell proliferation, tumor growth, invasion and in vivo metastasis, and stabilized the expression of the tumor suppressor gene Pdcd4 in colorectal cancer cells tested in the chorioallantonic membrane invasion assay [60].
Due to the low bioavailability of curcumin in vivo, the synthetic analogue difluorinated curcumin (CDF) was evaluated in the pancreatic cancer cells AsPC-1 and MIAPaCa-2. Curcumin and its CDF analogue, either alone or in combination, attenuated the expression of miR-200 and miR-21, leading to induction of the tumor suppressor gene phosphatase and tensin homolog (PTEN), which negatively regulates the intracellular levels of phosphatidylinositol-3,4,5-trisphosphate thereby preventing cells from growing and dividing too rapidly. In the same cell lines, CDF attenuated cancer stem cell markers via changes in miR-21 and miR-200 [61].
2.3.5. Resveratrol and analogues
A series of in vitro studies were carried out with resveratrol, a stilbenoid that is found in the skin of red grapes and other fruits, and its analogues. In colon cancer cells (SW480), resveratrol downregulated several oncogenic miRNAs, including miR-17, miR-21, miR-25, miR-92a, miR-196a, thereby mediating the regulation of Dicer, PDCD4, and PTEN. Resveratrol upregulated miR-663, a tumor suppressor miRNA inhibiting TGFβ [62]. In monocytic cells, resveratrol induced a miR-663-dependent effect targeting activator protein-1 (AP-1) through the Jun signaling pathway. Interestingly, resveratrol also impaired the upregulation of oncogenic miR-155 in a miR-663-dependent manner [63]. On the whole, miR-21, miR-155 and miR-663 were recognized as the main miRNAs regulated by resveratrol [64].
Treatment of human neural cells with the resveratrol analogue CAY10512 downregulated miR-146a, whose targets include complement factor H. MiR-146 is upregulated in the brain of Alzheimer's disease patients causing repression of complement factor H, a potent anti-inflammatory mediator [65]. These findings provide evidence that miRNA regulation plays a major role in the antiinflammatory effects of resveratrol.
In human benzo[a]pyrene-transformed bronchial epithelial cells (16HBE-T), resveratrol upregulated miR-622, recognizing k-Ras as a target. MiR-622 upregulation inhibited cell proliferation, inducing G0 growth arrest and suppressing the ability of 16HBE-T cells to form colonies in vitro and to develop tumors in nude mice. k-Ras messenger RNA was predicted as a putative miR-622 target [66].
Resveratrol treatment altered miRNA expression in human non-small cell lung cancer cells (A549), with 26 of the 753 analyzed miRNAs that exhibited greater than 2-fold expression changes in resveratrol-treated cells relative to their expression levels in untreated cells. Six of the resveratrol-modulated miRNAs showed greater than 20-fold changes in expression. These included miR-92a (downregulated) and miR-194, miR-299, miR-338, miR-582, and miR-758 (all of them upregulated). Target genes of resveratrol-regulated miRNAs are related to apoptosis, cell cycle regulation, cell proliferation, and differentiation [67].
In prostate cancer cells (PCa), resveratrol downregulated 23 miRNAs and upregulated 28 miRNAs. The downregulated miRNAs included miR-17-92 and miR-106ab clusters, having well recognized oncogenic properties, while the upregulated miRNAs included several tumor suppressors, some of them targeting PTEN [68].
In an in vivo ischemia/reperfusion rat model, resveratrol pretreatment restored the expression pattern of miRNAs close to the control levels in the ischemic heart. The upregulated miRNAs included miR-20b and miR-21 (antiangiogenic), which are implicated in cardiac remodeling. These data suggest that resveratrol exerts a significant cardioprotection through miRNA modulation [69,70].
2.4. Isoflavones
2.4.1. Genistein
Genistein is an isoflavone isolated from soybean. In vitro, genistenin downregulated the expression of miR-27a and inhibited cell growth of human uveal melanoma cells (C918). The growth of these cells in vivo was significantly inhibited by genistein administration to Balb/C nu/nu mice carrying xenografts of uveal melanoma cancer cells [71].
Genistein upregulated miR-146a in human pancreatic cancer cells and inhibited their invasive potential by downregulating EGFR, NFκB, IRAK-1, and MTA-2 [72].
In ovarian cancer cells (UL-3A, UL-313), genistein modulated 53 miRNAs, which resulted in the induction of estrogen receptor expression and cell growth rate decrease [73].
In human prostate cancer cells (PC3) treated with genistein, the minichromosome maintenance gene MCM2, involved in DNA replication and commonly dysregulated in cancer cells, was downregulated through miR-1296 modulation. Genistein induced the expression of miR-1296 by up to five-fold, along with cell cycle arrest in S-phase [74]. In the same cells, genistein upregulated the tumor suppressor gene ARHI by downregulating miR-221 and 222 [75]. In human prostate cancer cell lines (PC-3, DU145, and LNCaP), genistein variously modulated miRNA expression profile [76].
2.4.2. Anthocyanin
In Arabidopsis thaliana, anthocyanin accumulation is under the regulation of miR156. At least one of the miR-156 targets, the squamosa promoter binding protein-like-9, negatively regulates anthocyanin accumulation by directly preventing expression of anthocyanin biosynthetic genes [77]. These results provide a potential target for manipulation of anthocyanin content in plants.
In hepatoma HepG2 cells, grape seed proanthocyanidin or cocoa proanthocyanidin extracts downregulated miR-30b and upregulated miR-197, miR-532, and miR-1224 [54].
2.5. Indoles
Several studies investigated the miRNA modulating activity of indole-3-carbinol (I3C), found in cruciferous vegetables, and its in vivo dimeric product 3,3'-diindolylmethane (DIM).
In human pancreatic cancer cell lines (MiaPaCa-2, Panc-1 and Aspc-1), resistant to gemcitabine, DIM and a mixture of other genistein isoflavones downregulated the expression of let-7b, let-7c, let-7d, let-7e, miR-200b, and miR-200c, thereby reversing in part the malignant phenotype and inhibiting cancer cell growth [78].
The effect DIM on miRNA expression was investigated in both estrogen-dependent MCF-7 and estrogen receptor negative, P53 mutant human breast cancer MDA-MB-468 cells. DIM dose dependently inhibited the proliferation of both cells. In addition, an in vivo xenograft model showed that DIM strongly inhibited the development of human breast tumors. DIM increased miR-21 expression causing a downregulation of Cdc25A, which resulted in inhibition of breast cancer cell proliferation. Thus, DIM was able to stop the cell cycle progression of human breast cancer cells regardless of their estrogen-dependence and P53 status [79].
I3C and DIM reversed VC-induced dysregulation of several miRNAs in the lung of female A/J mice. MiR-21, miR-31, miR-130a, miR-146b, and miR-377 were consistently upregulated, while miR-1 and miR-143 were downregulated in lung cancer as compared to normal lungs. Moreover, the upregulation of miR-21, miR-31, miR-130a, miR-146b, and miR-377 observed in VC-treated animals was abrogated by I3C treatment, suggesting that I3C could inhibit the expression of these oncogenic miRNAs. PTEN, PDCD4, and RECK were potential targets of miR-21, and I3C upregulated these tumor suppressor genes though inhibition of miR-21 [80].
In CS-exposed rats, I3C restored in lung the expression of downregulated miRNAs targeting P53 functions (miR-34b), TGF-β expression (miR-26a), ERBB2 activation (miR-125a), and angiogenesis (miR-10a) [81].
2.6. Isothiocyanates
Phenethyl isothiocyanate (PEITC), a naturally occurring phytochemical, was evaluated for the ability to modulate the expression of miRNAs, after administration with the diet, in the lung and liver of rodents, either unexposed or exposed to CS.
In the lung of CS-free mice, PEITC decreased the expression of miR-181, miR-466a, miR-666, miR-706 (2.1-fold), and miR-708. In addition, PEITC was effective in counteracting miRNA alterations induced by CS for let-7a, let-7c, miR-26, miR-29, miR-31, miR-125, miR-135, miR-200, and miR-382 [82].
In the liver of the same mice, PEITC decreased the expression of miR-26a, miR-125a, miR-142, miR-200, miR-323, miR-331, miR-338, miR-466, miR-551, and increased the expression of miR-34c, miR-299, miR-452. In addition, PEITC was effective in counteracting miRNA alterations induced by CS in mouse liver for miR-125b, miR-153, miR-292, miR-297, miR-322, miR-376b, miR-463, miR-466, miR-467, miR-470, miR-687, miR-697, miR-709, miR-710, miR-719, miR-874, and miR-883 [82].
In a separate study PEITC was able to counteract miRNA alterations induced by CS in mouse lung tissue, either normal or affected by pneumonia, but not in lung cancer tissue. The protected miRNAs were let-7a, miR-30, and miR-804 [83].
The effect of PEITC on miRNA alterations induced by CS in rat lung was evaluated by Izzotti et al. [81]. Of the five dietary agents tested, PEITC was the most effective in restoring CS-downregulated miRNAs. Major PEITC-induced miRNA targets were let-7a, let-7c, miR-26a, miR-99b, miR-123, miR-125b, miR-146, miR-192, and miR-222.
2.7. Combination of phenethyl isothiocyanate and indole-3-carbinol
Modulation of miRNAs by PEITC was increased by its combination with I3C. In fact, in the lung of CS-exposed rats treated with both PEITC and I3C, in addition to the miRNAs that were individually modulated by each compound, let-7b, let-7f, miR-30, miR-124, miR-140, miR-145, miR-191, and miR-223 were significantly upregulated. (81).
2.8. Phospholipids
Myo-inositol is widely occurring in nature and food, and is present in all living cells. Dietary myo-inositol significantly upregulated miR-302, miR-509, miR-592, miR-876, miR-880, and miR-1953, and downregulated miR-543, miR-764, and miR-1901 in mouse lung. A parallel effect on proteins targeted by these miRNAs was observed for cyclin-dependent kinase inhibitor 1A involved in the cell cycle (miR-302), Rho GTPase activating protein 1 involved in signal transduction (miR-509), epidermal growth factor receptor pathway substrate 8 involved in cell differentiation (miR-543), nuclear receptor subfamily 3 involved in inflammatory responses, cellular proliferation and differentiation (miR-543), insulin receptor involved in metabolic functions (miR-592), and serine/threonine kinase 24 involved in stress response (miR-880) [Izzotti et al., unpublished data].
2.9. Saponins
The triterpene saponin ginsenoside Rh2, extracted from the traditional medical plant ginseng, upregulated 14 miRNAs and downregulated 12 miRNAs in human glioma cells (U251, T98MG and A172). In particular, upregulation of miR-128 appears to be responsible for the antiproliferative effects of Rh2 in glioma cells [84].
2.10. Anthraquinones
The anthracenedione bostrycin, belonging to the large family of anthraquinones and isolated from marine fungi, upregulated miR-638 and miR-923 in human lung adenocarcinoma cells (A549). This effect resulted in downregulation of the PI3K/AKT pathway, thus playing a role in induction of cell cycle arrest and apoptosis in bostrycin-treated cells [85].
2.11 Polyunsaturated fatty acids
Polyunsaturated fatty acids (PUFA) contained in fish oil, administered to AOM-treated rats, inhibited colon cancer appearance and progression by increasing let-7d, mir-15, miR-107, miR-191, and miR-324 expression [86].
Sodium butyrate, a short chain fatty acid inhibiting histone deacetylase, contained in goat and buffalo cheese, regulated endodermal differentiation by upregulating miR-10 and miR-24 in cultured human embryonic stem cells [87]
In human colon cancer cells butyrate inhibited cancer cell proliferation by inhibiting miR-106 thus inducing P21 expression [88]
3. Modulation of miRNA expression by synthetic agents
3.1. Metabolic inducers (Beta-naphthoflavone)
Beta-naphthoflavone, or 5,6-benzoflavone, is a synthetic flavonoid that acts as a potent inducer of P4501A enzyme and agonist of the arylhydrocarbon receptor. This agent significantly fully counteracted CS-induced miR-10a downregulation in rat lung by increasing its expression up to 5.2 fold and restoring the same expression level detected in sham-exposed rats. In the same experimental model, Beta-naphthoflavone increased the expression of miR-26a, miR-34c, and miR-146 in CS-exposed rats as compared with rats exposed to CS, in the absence of chemopreventive agents. However, their expression was still lower than that detected in sham-exposed rats [81].
3.2. Thiols and derivatives
3.2.1. N-Acetyl-l-cysteine
The thiol N-acetyl-l-cysteine (NAC), an analogue and precursor of reduced glutathione (GSH), given with the drinking water, counteracted the CS-induced downregulation of miR-26a, miR-123, miR-140, and miR-146 in rat lung [81].
3.2.2. Oltipraz
The dithiolthione oltipraz significantly counteracted CS-induced downregulation of miRNAs by increasing the expression of let-7b, let-7c, miR-10, miR-26, miR-34, and miR-123 in rat lung [81].
3.2.3. Combination of N-acetyl-l-cysteine and oltipraz
The effect of oltipraz on miRNA expression was increased by its combination with NAC. In fact, in addition to the miRNAs modulated by the individual agents, oltipraz + NAC upregulated let-7a, miR-192, and miR-222 in the lung of CS-exposed rats [81].
3.3. Nuclear receptor agonists
3.3.1. Bexarotene
Bexarotene, also known as Targretin, is a retinoid X receptor (RXR) agonist. In mouse lung, bexarotene alone did not significantly alter miRNA expression profiles. In the same organ, bexarotene was effective in counteracting the CS-induced downregulation of pulmonary miR-493. In parallel, antibody microarray analyses indicated that two miR-493-targeted proteins were modulated by bexarotene, including keratin pan, involved in cell differentiation, and cell division cyclin 27, involved in the cell cycle [Izzotti et al., unpublished data].
3.3.2. Pioglitazone
Pioglitazone is a peroxisome proliferator-activated receptor (PPAR)-gamma agonist. In vivo pioglitazone protected the rats against myocardial ischaemia-reperfusion injury by downregulating miR-29a and miR-29c levels in the heart. The same finding was obtained in vitro in cardiac H9c2 cells. Downregulation of miR-29 by pioglitazone protected H9c2 cells from simulated ischaemia-reperfusion injury, as indicated by an increased cell survival and decreased caspase-3 activity. In contrast, overexpressing miR-29 promoted apoptosis and completely blocked the protective effect of pioglitazone. Antagomirs against miR-29a or miR-29c significantly reduced myocardial infarct size and apoptosis in hearts subjected to ischaemia-reperfusion injury. Western blot analyses demonstrated that Mcl-2, an anti-apoptotic Bcl-2 family member, was increased by miR-29 inhibition [89].
Dietary pioglitazone did not significantly affect lung miRNA profiles in mouse lung. However, this agent was effective in counteracting CS-induced alterations for miR-218, miR-296, miR-335, and miR-764. Parallel antibody microarray analyses indicated that lung proteins targeted by pioglitazone-modulated miRNAs were also modified in their expression. They included thioredoxin and Ras proteins, targets for miR-218; insulin-like growth factor, target for miR-335; and peroxisomal D3,D2-enoyl-CoA isomerase, target for miR-764 [Izzotti et al., unpublished data].
3.3.3. Combination of bexarotene and pioglitazone
Combination of bexarotene and pioglitazone effectively protected the mouse lung from CS-induced miRNA downregulation. In fact, the number of downregulated miRNAs in CS-exposed mice was 79, out of a total of 694 tested, whereas one miRNA only was downregulated in CS-exposed mice treated with bexarotene + pioglitazone. The most potently upregulated miRNAs by this combination of chemopreventive agents were miR-290, miR-484, and miR-715 [Izzotti et al., unpublished data].
3.4. Histone deacetylase inhibitors
3.4.1. Suberoylanilide hydroxamic acid (SAHA)
The histone deacetylase inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA), or Vorinostat, markedly altered the expression of 31 miRNAs in human colon carcinoma cells (HCT116) as well as downstream targets affecting cell cycle, apoptosis, and differentiation. In particular, SAHA downregulated let-7l, miR-29, miR-100, miR-141, miR-221, miR-222, and miR-877, while it upregulated miR-22 and miR-424 [90].
In human lymphoma cells (L540), SAHA downregulated the c-Myc-related miRNAs miR-17-3p, miR-17-5, and miR-18 as well as miR-15b and miR-155, which are not c-Myc-regulated [91].
SAHA was also found to restore the expression of miR-34a in human pancreatic cancer stem cells, which provides mechanistic insights and therapeutic targets for pancreatic cancers [92].
In human non-small cell lung cancer cells (A549), SAHA dysregulated the expression of 64 miRNAs having several target genes related to angiogenesis, apoptosis, chromatin modification, cell proliferation and differentiation. The miRNAs that were upregulated more than 4-fold included let-7b, miR-124, miR-126, miR-129-3p, miR-132, miR-382, miR-409-3p, and miR-660. The miRNAs that were downregulated more than 4-fold included miR-30c-1, miR-30e, miR-106a, miR-345, miR-548c-3p, miR-877, and miR-936 [93].
In vivo, the dietary administration of SAHA to mice modulated miRNA expression in the lung. In particular, SAHA upregulated let-7a, miR-125b, miR-183, and miR-466, and downregulated miR-302, miR-684, and miR-764 [Izzotti et al., unpublished data]. Interestingly, let-7 and miR-183 had also been found to be upregulated by SAHA in lung cancer cells in vitro [93]. Parallel analyses detected the modulation by SAHA of miRNA target proteins in mouse lung, including cyclin-dependent kinase inhibitor 1A (P21), a P53-dependent negative regulator of the cell cycle targeted by miR-302 and miR-106; integrin beta-1, regulating cell adhesion, targeted by miR-183; protein phosphatase 2, implicated in the negative control of cell growth and division, targeted by miR-183; importin beta-1, playing a role in signal transduction, targeted by miR-684; histone deacetylase 3, regulating epigenetic repression, targeted by miR-125 and miR-466 [Izzotti et al., unpublished data].
3.4.2. Trichostatin
The HDACi trichostatin A altered the expression profile of miRNA signatures in the apoptosis-resistant breast carcinoma cell line (MCF-7TN-R). Trichostatin A induced significant upregulation of 22 miRNAs and downregulation of 10 miRNAs. Among them, the most remarkably upregulated miRNAs were miR-139, miR-146, miR-155, miR-215,and miR-544, and the most remarkably downregulated miRNAs were miR-645 and miR-657. These results demonstrate that the anticancer activity of trichostatin A is correlated with alteration of miRNA expression profiles [94].
3.4.3. LAQ824
The HDACi LAQ824 produced a dramatic alteration in miRNA profiles in human breast cancer cells (SKBr3), 22 miRNAs being upregulated and 5 miRNAs being downregulated [95].
3.5. Anti-inflammatory agents
3.5.1. Glucocorticoids
The modulation of miRNAs by the glucocorticoid budesonide, given with the diet, was examined in the lung and liver either in CS-free mice or CS-exposed mice. In mouse lung, budesonide alone decreased the expression of one miRNA only (miR-133), while it exerted a more remarkable effect in the liver by downregulating 14 miRNAs and upregulating 3 miRNAs. The downregulated miRNAs included let-7a, miR-27a, miR-100, miR-106b, miR-125a, miR-142, miR-200b, miR-300, miR-320, miR-331, miR-338, miR-466a, miR-551, and miR-763. The upregulated miRNAs included miR-483, miR-539, miR-742. In the lung of CS-exposed mice, budesonide was effective in counteracting CS-related miRNA alterations for 5 miRNAs, including let-7a, miR-26a, miR-31, miR-382, and miR-463. In the liver of the same mice, budesonide was effective in counteracting CS-related miRNA alterations induced by CS for 15 miRNAs, including miR-15a, miR-21, miR-125b, miR-292, miR-297, miR-322, miR-466, miR-467, miR-687, miR-690, miR-697, miR-709, miR-710, miR-719, and miR-883 [82].
In humans, the expression of 227 miRNAs was examined in airway biopsies obtained from normal and mild asthmatic patients. MiRNA profiles were analyzed before and after 1 month of treatment with inhaled budesonide. No significant difference was detected in the expression of all 227 analyzed miRNAs, irrespective of treatment with budesonide. These results suggest that changes in miRNA lung expression are not involved in the anti-inflammatory action of the corticosteroid budesonide in asthmatic patients [96].
3.5.2. Nonsteroidal antiinflammatory drugs (NSAIDs)
Ethyl-2-((2,3-bis(nitroxy)propyl)disulfanyl benzoate (GT-094) is a nitric oxide donor NSAID (NO-NSAID) that is expected to undergo rapid thiol/disulfide exchange with protein sulfhydryl groups leading to NSAID (thiosalycylate) release. Using human colon cancer cells (RK0 and SW480), GT-094 was found to downregulate miR-27a, which in part may be responsible for the anticancer activity of this agent [97].
3.6. Selective estrogen receptor modulators (SERMs)
3.6.1. Tamoxifen
Mir-221 and miR-222 have consistently been implicated in the resistance to tamoxifen in breast cancer. These two miRNAs were found to be elevated in estrogen receptor alfa (ERα)-negative breast cancer cells as compared to ERα-positive cells, which suggest a role in the regulation of ERα expression [98]. Using breast cancer cells (MCF-7), either sensitive or resistant to tamoxifen, mir-221, miR-222 and miR-181 were found to have an increased expression in tamoxifen resistant cells, whereas miR-21, miR-342 and miR-489 had a decreased expression. In addition, this study demonstrated a relationship between mir-221 and miR-222 expression and HER2/neu oncoprotein overexpression in primary breast cancer cells [99]. Another study suggested that miR-342 regulates tamoxifen response in breast cancer cell lines in vitro, and clinical data indicated a relationship between reduced miR-342 expression and tamoxifen resistance [100].
3.6.2. Fulvestrant
An increased expression of mir-221 and miR-222 was found to play a role also in acquired resistance towards fulvestrant, a SERM antagonist used in hormone-sensitive breast cancers following failure of previous tamoxifen or aromatase inhibitor therapies [101].
4. Discussion
4.1.MiRNAs as targets for chemopreventive agents
The results reported in Table 1 provide evidence that many chemopreventive agents, belonging to various chemical classes and functional families, are able to modulate the expression of miRNAs in experimental test systems, either in vitro or in vivo. A total of as many as 148 miRNAs were found to be modulated in the studies reported in Table 1. Interestingly, several miRNAs were targeted by multiple chemopreventive agents. In particular, 7 of the miRNAs investigated in the cited studies, including let-7a, miR-21, miR-26, miR-34, miR-125, miR-146, and miR-200, were targeted by at least 5 chemopreventive agents each. These miRNAs play important roles in controlling several mechanisms that are involved in various stages of the carcinogenesis process, such as inflammation, stress response, cell proliferation, apoptosis, oncogene activation (k-Ras, TGF, ERBB2), modulation of oncosoppressor genes (PTEN, P53), and signal transduction pathways. It is conceivable that certain miRNAs may represent preferential targets for chemopreventives and may convenientely be used as indicators of efficacy of anticancer agents. Furthermore, it may be hypothesized that, in the future, miRNA themselves or anti-miRNA oligonucleotides may be used to suppress cancer development.
Opposite expressional directions of miRNA modulation by the same chemopreventive agents were reported in few cases, for instance with α-tocopherol, budesonide, and PEITC (see Table 1). In some cases, these findings may be related to noise in the omics studies. However, in other cases they may be due to the fact that the same miRNA was tested in different organs, which involves different pharmacokinetic and metabolic patterns. As an example, the bifunctional metabolic inducer PEITC exerted different effects in liver and lung [82].
4.2. Analiysis of miRNAs for evaluating the safety and efficacy of chemopreventive agents
The majority of the studies reported in Table 1 used cancer cell lines in vitro with the goal of evaluating the anticancer effects of test agents and other properties, such as occurrence of resistance to drugs, through modulation of miRNA expression. The main drawback of this methodology is that cancer cells are less sensitive than differentiated cells to miRNA modulation by chemopreventive agents, as assessed by comparing lung cancer tissue and the surrounding healthy tissue in mice treated with either PEITC or NAC [83].
In vivo approaches not only take into account pharmacokinetic and metabolic features of test compounds but also appear to be more appropriate to evaluate genuine cancer preventive effects of dietary and pharmacological agents. In our opinion, such a goal should be pursued by evaluating both the ability of test agents to affect baseline miRNA expression profiles, as an indicator of their safety, and their ability to inhibit miRNA alterations caused by carcinogens, as an indicator of their efficacy. Table 1 reports a number of examples of application of this kind of protocol in mice and rats, either unexposed or exposed to CS [81-83, plus unpublished data].
4.3. Rationale for designing combinations of chemopreventive agents by miRNA analysis
The therapy of the most important diseases (e.g., cancer, cardiovascular diseases, AIDS, etc.) involves the combination of different drugs. Likewise, application of a “combined chemoprevention” strategy is particularly promising. However, it is difficult to evaluate how the combined agents may interact in terms both of safety and efficacy. The rationale for designing a proper combination is to use agents having different and possibly complementary mechanisms of action. Most chemopreventive agents have pleiotropic properties and work via multiple mechanisms [18-20]. Since each miRNA targets a number of transcripts simultaneously, evaluation of miRNA expression provides a convenient tool for assessing the outcome of combinations of different agents at the molecular level.
Three combinations are reported in Table 1, including pioglitazone + bexarotene, NAC + oltipraz, and PEITC + I3C. Pioglitazone is a synthetic ligand of peroxisome proliferator-activator receptor-gamma (PPAR-gamma) and bexarotene is a synthetic agonist of retinoid X receptor (RXR), which is an obligate heterodimeric partner for other nuclear receptors, including PPAR [102]. NAC is characterized by a variety of protective properties but its primary mechanisms are nucleophilicity and antioxidant activity as a scavenger of ROS [103], whereas a major mechanism for oltipraz is the induction of electrophile detoxification enzymes [104]. PEITC is a typical phase II activity inducer [105], whereas I3C has a broad spectrum of anticancer properties, including its ability to interfere with multiple oncogenic signaling pathways that govern cell cycle progression, survival, invasion, and other aggressive phenotypes of cancer cells [106]. In addition, the data reported in Table 1 may be useful to design novel combinations of chemopreventive agents. For instance, SAHA inhibit the expression of miR-221 and miR-222, which are consistently associated with resistance to SERMs. Accordingly, a combination of SAHA and SERMs could be proposed to prevent resistance to SERMs.
4.4. Mechanisms of miRNA modulation by chemopreventive agents
For genotoxic agents, such as ionizing radiation, alteration of miRNA expression has been ascribed to the fact that P53 interacts with the Drosha/DGCR8 processing complex through an association with RNA helicase p68, which modulates the processing of pri-miRNAs to pre-miRNAs [9]. According to these data, DNA damage modulates miRNA expression via a P53-dependent mechanism [107]. The number of components of the miRNA processing machinery serving as direct transcriptional targets for P53 in response to DNA damage has been expanding by also including the endoribonuclease Dicer [108]. The central role of Dicer in the cellular response to UV induced damage is established [10]. However, in case of treatment with chemopreventive agents, it is unlikely that miRNA expression is modulated through DNA damage and P53 activation. Recent bioinformatic analyses indicate that Dicer, the enzyme involved in the cytoplasmic phase of miRNA maturation, is a preferential cytoplasmic target for mutagens. In particular, the binding affinity of 25 mutagens for each Dicer's RNase III domain was estimated by calculating the global contact-energy and the number of intermolecular contacts. The mutagens tested form stable complexes with Dicer, which are more stable than those formed by Dicer with its natural substrate, i.e., pre-miRNAs [109]. These data indicate that mutagens affect miRNA maturation by competing with pre-miRNA for Dicer binding. This is a short-term adaptive response of the cell to mutagen exposure resulting in maturation blockage for miRNAs acting as negative regulators of genes involved in stress response. However, the long-term alteration of miRNA maturation resulting from long-term exposure constitutes a stimulus towards carcinogenesis [110].
It should be noted that bioinformatic models indicate that Dicer binding by mutagens is non-covalent and involves low-energy. Accordingly, the Dicer catalytic sites are not irreversibly blocked but just change their affinity for specific substrates depending on oligonucleotidic sequences. This explains why only few selected miRNAs are affected by Dicer regulation depending on their specific structures. As an example, those miRNAs that are enriched in guanine in their terminal hairpin, such as those belonging to the let-7 family, are highly sensitive to miRNA alterations induced by mutagens.
The same bioinformatic approach revealed that also chemopreventive agents are characterized by affinity for Dicer. Indeed, it was reported that isothiocyanates and I3C show high Dicer affinity [109]. Further analyses using the same approach indicated that resveratrol, EGCG, I3C, and beta-naphthoflavone display Dicer affinity by binding the catalytic site of Dicer sub-units (Fig. 1). These findings indicate that chemopreventive agents may compete with mutagens for Dicer binding. According to this view, chemopreventive agents act through hormetic effects sharing the same molecular effect of mutagens at the Dicer epigenetic level thereby competing with them for the activation of adverse mechanisms such as the alteration of miRNA expression.
Fig. 1.
Bioinformatic analysis showing the binding sites of 4 chemopreventive agents to Dicer. The Dicer 3D structure is rotated in each panel to show the chemopreventive agent binding site. The colors identify the agent (in red) and the Dicer subunits (yellow, sub-unit A; green, sub-unit B; grey, sub-unit C; blue, sub-unit D).
4.5. Polymorphic miRNAs. A nutrigenomic/pharmacogenomic approach to cancer prevention?
A number of miRNAs targeted by chemopreventive agents, identified with the symbol § in Table 1, undergo SNPs. This feature also applies to frequently modulated miRNAs, such as let-7a, miR-34, miR-125, miR-146, and miR-200. For example, a G/U SNP at nucleotide 8 of miR-125 gene, has been reported to downregulate maturation of this miRNA [111]. The miR-125 genetic targets include the ERBB2 proto-oncogene encoding for the EGF receptor, which is highly expressed in carcinomas. MiR-125 genes, located in the 11q23-q24 region, are frequently deleted in lung cancer [112]. This miRNA was strongly downregulated by CS in rat lung [14,15]. MiR-125 was also downregulated in the airway epithelium of smoking humans [113]. Several chemopreventive agents, including NAC, oltipraz, I3C and PEITC, inhibited the CS-induced downregulation of miR-125 in rat lung [81]. Insofar the role of miRNA SNPs for predicting cancer risks has been estimated to be low [27,114]. Further studies are needed to establish the impact of miRNA polymorphisms on safety and efficacy of chemopreventive agents. It is conceivable that miRNA polymorphisms could be important for explaining the interindividual variability in the response to the protective effects of pharmacologic and dietary agents, according to a pharmacogenomic/nutrigenomic approach. Interindividual variability is one of the main factors affecting the outcome of cancer chemoprevention trials in humans.
4.6.Tissue specificity of miRNAs and chemoprevention of site-specific cancers
A further issue is the tissue specificity of miRNAs, which is reported in Table 1 next to the identification of each miRNA. This information could be useful to address the clinical use of dietary and pharmacological agents for the prevention of site-specific cancers. In fact, a given type of cancer is expected to be more effectively prevented by agents that are able to modulate miRNA profiles in the target organ. For instance, chemopreventive agents modulating miRNAs of the let-7 family, which are highly expressed in the lung and are considered to be major players in lung cancer development [115], could be proposed for the prevention of lung cancer. Those modulating miR-122 could be proposed for the prevention of liver cancer. In fact, functional and molecular studies have uncovered mechanisms that link deregulated miR-122 to pathways associated with hepatocellular carcinoma [116], to such an extent that an increase of this miRNA in serum has been proposed as a novel noninvasive biomarker for the detection of this cancer in healthy subjects [117].
5. Conclusions
A continuously expanding literature covers the issue of miRNA involvement in response to dietary and pharmacological agents. The present article reports the data relative to 31 agents, either natural or synthetic, which are known to behave as inhibitors of mutagenesis and carcinogenesis and are regarded as potential cancer chemopreventive agents. The majority of the studies reviewed, however, evaluated the effects of test agents on miRNA expression profiles in cultured cancer cell lines rather than their actual role in cancer chemoprevention. On the other hand, studies in mice and rats evaluated either the ability of test agents to alter the baseline expression of miRNAs and/or their ability to counteract miRNA alterations induced by carcinogens. In this way, it is possible to predict the in vivo effects of chemopreventive agents both in terms of safety and efficacy. In previous studies, we pursued a similar objective by evaluating transcriptome and proteome profiles in organs of rodents treated with carcinogens and/or chemopreventive agents [118,119]. However, mRNAs analysis gives redundant information, whereas proteome analysis just covers a large minority of the existing proteins.
The data generated by using animal models are likely to bear relevance to the human situation because the miRNA machinery is well conserved among species. Indeed, the miRNA alterations induced by CS in the lung of mice and rats are similar to those observed in the airway epithelium of smoking humans [120]. Accordingly, miRNA analysis in preclinical models may be useful to identify those chemopreventive agents that are worthy of being assayed in clinical trials as well as to select the identity of miRNAs to be analyzed as intermediate biomarkers. This task might be pursued in humans by minimally invasive procedures, due to the fact that miRNAs are released from target organs to the blood [121].
In conclusion, it is conceivable that miRNA analysis will become an important tool for developing new strategies for the prevention of cancer and other mutation-related diseases.
Acknowledgments
This work was supported by the U.S. National Cancer Institute (NCI, contract NO1-CN53301) and by the Associazione Italiana per la Ricerca sul Cancro (AIRC, grant No. 8909). We thank Dr. Patrizio Arrigo (National Research Council, Genoa, Italy) for providing 3D Dicer structures.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Kim VN, Nam JW. Genomics of microRNA. Trends Genet. 2006;22:165–73. doi: 10.1016/j.tig.2006.01.003. [DOI] [PubMed] [Google Scholar]
- 2.Berezikov E, Cuppen E, Plasterk RH. Approaches to microRNA discovery. Nat Genet. 2006;38:S2–7. doi: 10.1038/ng1794. [DOI] [PubMed] [Google Scholar]
- 3.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–97. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
- 4.Brennecke J, Stark A, Russell RB, Cohen SM. Principles of microRNA-target recognition. PLoS Biol. 2005;3:e85. doi: 10.1371/journal.pbio.0030085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Di Leva G, Croce CM. Roles of small RNAs in tumor formation. Trends Mol Med. 2010;16:257–67. doi: 10.1016/j.molmed.2010.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Couzin J. MicroRNAs make big impression in disease after disease. Science. 2008;319:1782–4. doi: 10.1126/science.319.5871.1782. [DOI] [PubMed] [Google Scholar]
- 7.Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 2009;10:704–14. doi: 10.1038/nrg2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wouters MD, van Gent DC, Hoeijmakers JH, Pothof J. MicroRNAs, the DNA damage response and cancer. Mutat. Res. 2011;717:54–66. doi: 10.1016/j.mrfmmm.2011.03.012. [DOI] [PubMed] [Google Scholar]
- 9.Suzuki HI, Yamagata K, Sugimoto K, Iwamoto T, Kato S, Miyazono K. Modulation of microRNA processing by p53. Nature. 2009;460:529–533. doi: 10.1038/nature08199. [DOI] [PubMed] [Google Scholar]
- 10.Pothof J, Verkaik NS, van IJcken W, Wiemer EA, Ta VT, van der Horst GT, Jaspers NG, van Gent DC, Hoeijmakers JH, Persengiev SP. MicroRNA-mediated gene silencing modulates the UV-induced DNA-damage response. EMBO J. 2009;28:2090–2099. doi: 10.1038/emboj.2009.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Duan H, Jiang Y, Zhang H, Wu Y. MiR-320 and miR-494 affect cell cycles of primary murine bronchial epithelial cells exposed to benzo[a]pyrene. Toxicol In Vitro. 2010;24:928–935. doi: 10.1016/j.tiv.2009.11.013. [DOI] [PubMed] [Google Scholar]
- 12.Elamin BK, Callegari E, Gramantieri L, Sabbioni S, Negrini M. MicroRNA response to environmental mutagens in liver. Mutat Res. 2011;717:67–76. doi: 10.1016/j.mrfmmm.2011.03.015. [DOI] [PubMed] [Google Scholar]
- 13.Lema C, Cunningham MJ. MicroRNAs and their implications in toxicological research. Toxicol. Lett. 2010;198:100–105. doi: 10.1016/j.toxlet.2010.06.019. [DOI] [PubMed] [Google Scholar]
- 14.Izzotti A, Calin G, Arrigo P, Steele VE, Croce C, De Flora S. Downregulation of microRNA expression in the lung of rats exposed to cigarette smoke. FASEB J. 2009;23:806–812. doi: 10.1096/fj.08-121384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Izzotti A, Calin GA, Steele VE, Croce CM, De Flora S. Relationship of microRNA expression in mouse lung with age and exposure to cigarette smoke and light. FASEB J. 2009;23:3243–3250. doi: 10.1096/fj.09-135251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jardim MJ. microRNAs: Implications for air pollution research. Mutat Res. 2011;1:38–45. doi: 10.1016/j.mrfmmm.2011.03.014. [DOI] [PubMed] [Google Scholar]
- 17.Sonkoly E, Pivarcsi A. MicroRNAs in inflammation and response to injuries induced by environmental pollution. Mutat. Res. 2011;717:46–53. doi: 10.1016/j.mrfmmm.2011.02.002. [DOI] [PubMed] [Google Scholar]
- 18.De Flora S, Ramel C. Mechanisms of inhibitor of mutagenesis and carcinogenesis. Classification and overview. Mutat. Res. 1988;202:285–306. doi: 10.1016/0027-5107(88)90193-5. [DOI] [PubMed] [Google Scholar]
- 19.Kelloff GJ, Boone CW, Steele VE, Crowell JA, Lubet RA, Greenwald P, Hawk ET, Fay JR, Sigman CC. Mechanistic considerations in the evaluation of chemopreventive data. IARC Sci Publ. 1996;139:203–19. [PubMed] [Google Scholar]
- 20.De Flora S, Ferguson LR. Overview of mechanisms of cancer chemopreventive agents. Mutat. Res. 2005;591:8–15. doi: 10.1016/j.mrfmmm.2005.02.029. [DOI] [PubMed] [Google Scholar]
- 21.Zhang W, Dolan ME. Emerging role of microRNAs in drug response. Curr. Opin. Mol. Ther. 2010;12:695–702. [PMC free article] [PubMed] [Google Scholar]
- 22.Davis CD, Ross SA. Evidence for dietary regulation of microRNA expression in cancer cells. Nutr. Rev. 2008;66:477–482. doi: 10.1111/j.1753-4887.2008.00080.x. [DOI] [PubMed] [Google Scholar]
- 23.Li Y, Kong D, Wang Z, Sarkar FH. Regulation of microRNAs by natural agents: an emerging field in chemoprevention and chemotherapy research. Pharm. Res. 2010;27:1027–41. doi: 10.1007/s11095-010-0105-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Link A, Balaguer F, Goel A. Cancer chemoprevention by dietary polyphenols: promising role for epigenetics. Biochem. Pharmacol. 2010;80:1771–92. doi: 10.1016/j.bcp.2010.06.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Reuter S, Gupta SC, Park B, Goel A, Aggarwal BB. Epigenetic changes induced by curcumin and other natural compounds. Genes Nutr. 2011;6:93–108. doi: 10.1007/s12263-011-0222-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Parasramka MA, Ho E, Williams DE, Dashwood RH. MicroRNAs, diet, and cancer: New mechanistic insights on the epigenetic actions of phytochemicals. Mol. Carcinog. 2012;51:213–230. doi: 10.1002/mc.20822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chin LJ, Ratner E, Leng S, Zhai R, Nallur S, Babar I, Muller RU, Straka E, Su L, Burki EA, Crowell RE, Patel R, Kulkarni T, Homer R, Zelterman D, Kidd KK, Zhu Y, Christiani DC, Belinsky SA, Slack FJ, Weidhaas JB. A SNP in a let-7 microRNA complementary site in the KRAS 3' untranslated region increases non-small cell lung cancer risk. Cancer Res. 2008;68:8535–40. doi: 10.1158/0008-5472.CAN-08-2129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Gong J, Tong Y, Zhang HM, Wang K, Hu T, Shan G, Sun* J, Guo AY. Genome-Wide Identification of SNPs in MicroRNA Genes and the SNP Effects on MicroRNA Target Binding and Biogenesis. Human Mutation. 2011;33:254–63. doi: 10.1002/humu.21641. [DOI] [PubMed] [Google Scholar]
- 29.Rossi A, D'Urso OF, Gatto G, Poltronieri P, Ferracin M, Remondelli P, Negrini M, Caporaso MG, Bonatti S, Mallardo M. Non-coding RNAs change their expression profile after Retinoid induced differentiation of the promyelocytic cell line NB4. BMC Res Notes. 2010;3:24. doi: 10.1186/1756-0500-3-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Garzon R, Pichiorri F, Palumbo T, Visentini M, Aqeilan R, Cimmino A, Wang H, Sun H, Volinia S, Alder H, Calin GA, Liu CG, Andreeff M, Croce CM. MicroRNA gene expression during retinoic acid-induced differentiation of human acute promyelocytic leukemia. Oncogene. 2007;26:4148–57. doi: 10.1038/sj.onc.1210186. [DOI] [PubMed] [Google Scholar]
- 31.Jian P, Li ZW, Fang TY, Jian W, Zhuan Z, Mei LX, Yan WS, Jian N. Retinoic acid induces HL-60 cell differentiation via the upregulation of miR-663. J Hematol Oncol. 2011;4:20. doi: 10.1186/1756-8722-4-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Terao M, Fratelli M, Kurosaki M, Zanetti A, Guarnaccia V, Paroni G, Tsykin A, Lupi M, Gianni M, Goodall GJ, Garattini E. Induction of miR-21 by retinoic acid in estrogen receptor-positive breast carcinoma cells: biological correlates and molecular targets. J. Biol. Chem. 2011;286:4027–42. doi: 10.1074/jbc.M110.184994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kawasaki H, Taira K. Functional analysis of microRNAs during the retinoic acid-induced neuronal differentiation of human NT2 cells. Nucleic Acids Res Suppl. 2003;3:243–4. doi: 10.1093/nass/3.1.243. [DOI] [PubMed] [Google Scholar]
- 34.Das S, Foley N, Bryan K, Watters KM, Bray I, Murphy DM, Buckley PG, Stallings RL. MicroRNA mediates DNA demethylation events triggered by retinoic acid during neuroblastoma cell differentiation. Cancer Res. 2010;70:7874–81. doi: 10.1158/0008-5472.CAN-10-1534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Beveridge NJ, Tooney PA, Carroll AP, Tran N, Cairns MJ. Down-regulation of miR-17 family expression in response to retinoic acid induced neuronal differentiation. Cell Signal. 2009;21:1837–45. doi: 10.1016/j.cellsig.2009.07.019. [DOI] [PubMed] [Google Scholar]
- 36.Meseguer S, Mudduluru G, Escamilla JM, Allgayer H, Barettino D. MicroRNAs-10a and -10b contribute to retinoic acid-induced differentiation of neuroblastoma cells and target the alternative splicing regulatory factor SFRS1 (SF2/ASF). J Biol Chem. 2011;286:4150–6. doi: 10.1074/jbc.M110.167817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhao JJ, Sun DG, Wang J, Liu SR, Zhang CY, Zhu MX, Ma X. Retinoic acid downregulates microRNAs to induce abnormal development of spinal cord in spina bifida rat model. Childs. Nerv. Syst. 2008;24:485–92. doi: 10.1007/s00381-007-0520-5. [DOI] [PubMed] [Google Scholar]
- 38.Kutay H, Bai S, Datta J, Motiwala T, Pogribny I, Frankel W, Jacob ST, Ghoshal K. Downregulation of miR-122 in the rodent and human hepatocellular carcinomas. J. Cell. Biochem. 2006;99:671–678. doi: 10.1002/jcb.20982. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 39.Pogribny IP, Tryndyak VP, Ross SA, Beland FA. Differential expression of microRNAs during hepatocarcinogenesis induced by methyl deficiency in rats. Nutr Rev. 2008;66(Suppl 1):S33–5. doi: 10.1111/j.1753-4887.2008.00064.x. [DOI] [PubMed] [Google Scholar]
- 40.Wang LL, Zhang Z, Li Q, Yang R, Pei X, Xu Y, Wang J, Zhou SF, Li Y. Ethanol exposure induces differential microRNA and target gene expression and teratogenic effects which can be suppressed by folic acid supplementation. Hum. Reprod. 2009;24:562–79. doi: 10.1093/humrep/den439. [DOI] [PubMed] [Google Scholar]
- 41.Marsit CJ, Eddy K, Kelsey KT. MicroRNA responses to cellular stress. Cancer Res. 2006;66:10843–10848. doi: 10.1158/0008-5472.CAN-06-1894. [DOI] [PubMed] [Google Scholar]
- 42.Wang X, Gocek E, Liu CG, Studzinski GP. MicroRNAs181 regulate the expression of p27Kip1 in human myeloid leukemia cells induced to differentiate by 1,25-dihydroxyvitamin D3. Cell Cycle. 2009;8:736–741. doi: 10.4161/cc.8.5.7870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gocek E, Wang X, Liu X, Liu CG, Studzinski GP. MicroRNA-32 upregulation by 1,25-dihydroxyvitamin D3 in human myeloid leukemia cells leads to Bim targeting and inhibition of AraC-induced apoptosis. Cancer Res. 2011;71:6230–9. doi: 10.1158/0008-5472.CAN-11-1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Peng X, Vaishnav A, Murillo G, Alimirah F, Torres KE, Mehta RG. Protection against cellular stress by 25-hydroxyvitamin D3 in breast epithelial cells. J. Cell. Biochem. 2010;110:1324–1333. doi: 10.1002/jcb.22646. [DOI] [PubMed] [Google Scholar]
- 45.Mohri T, Nakajima M, Takagi S, Komagata S, Yokoi T. MicroRNA regulates human vitamin D receptor. Int. J. Cancer. 2009;125:1328–1333. doi: 10.1002/ijc.24459. [DOI] [PubMed] [Google Scholar]
- 46.Essa S, Denzer N, Mahlknecht U, Klein R, Collnot EM, Tilgen W, Reichrath J. VDR microRNA expression and epigenetic silencing of vitamin D signaling in melanoma cells. J Steroid Biochem Mol Biol. 2010;121:110–3. doi: 10.1016/j.jsbmb.2010.02.003. [DOI] [PubMed] [Google Scholar]
- 47.Gaedicke S, Zhang X, Schmelzer C, Lou Y, Doering F, Frank J, Rimbach G. Vitamin E dependent microRNA regulation in rat liver. FEBS Lett. 2008;582:3542–3546. doi: 10.1016/j.febslet.2008.09.032. [DOI] [PubMed] [Google Scholar]
- 48.Rimbach G, Moehring J, Huebbe P, Lodge JK. Gene-regulatory activity of alpha-tocopherol. Molecules. 2010;15:1746–61. doi: 10.3390/molecules15031746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sarveswaran S, Liroff J, Zhou Z, Nikitin AY, Ghosh J. Selenite triggers rapid transcriptional activation of p53, and p53-mediated apoptosis in prostate cancer cells: Implication for the treatment of early-stage prostate cancer. Int J. Oncol. 2010;36:1419–28. doi: 10.3892/ijo_00000627. [DOI] [PubMed] [Google Scholar]
- 50.Noratto GD, Kim Y, Talcott ST, Mertens-Talcott SU. Flavonol-rich fractions of yaupon holly leaves (Ilex vomitoria, Aquifoliaceae) induce microRNA-146a and have anti-inflammatory and chemopreventive effects in intestinal myofibroblast CCD-18Co cells. Fitoterapia. 2011;82:557–69. doi: 10.1016/j.fitote.2011.01.013. [DOI] [PubMed] [Google Scholar]
- 51.Ai RT, Wu SY, Wen XY, Xu W, Lv L, Rao JJ, Wu SG. 1,3,4-tri-O-galloyl-6-O-caffeoyl-β-D-glucopyranose, a new anti-proliferative ellagitannin, regulates the expression of microRNAs in HepG(2) cancer cells. Nan Fang Yi Ke Da Xue Xue Bao. 2011;31:1641–8. [PubMed] [Google Scholar]
- 52.Wen XY, Wu SY, Li ZQ, Liu ZQ, Zhang JJ, Wang GF, Jiang ZH, Wu SG. Ellagitannin (BJA3121), an anti-proliferative natural polyphenol compound, can regulate the expression of MiRNAs in HepG2 cancer cells. Phytother. Res. 2009;23:778–84. doi: 10.1002/ptr.2616. [DOI] [PubMed] [Google Scholar]
- 53.Tsang WP, Kwok TT. Epigallocatechin gallate up-regulation of miR-16 and induction of apoptosis in human cancer cells. J. Nutr. Biochem. 2010;21:140–6. doi: 10.1016/j.jnutbio.2008.12.003. [DOI] [PubMed] [Google Scholar]
- 54.Arola-Arnal A, Bladé C. Proanthocyanidins Modulate MicroRNA Expression in Human HepG2 Cells. PLoS One. 2011;6:e25982. doi: 10.1371/journal.pone.0025982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Fix LN, Shah M, Efferth T, Farwell MA, Zhang B. MicroRNA expression profile of MCF-7 human breast cancer cells and the effect of green tea polyphenon-60. Cancer Genomics Proteomics. 2010;7:261–77. [PubMed] [Google Scholar]
- 56.Wang H, Bian S, Yang CS. Green tea polyphenol EGCG suppresses lung cancer cell growth through upregulating miR-210 expression caused by stabilizingHIF-1{alpha}. Carcinogenesis. 2011 doi: 10.1093/carcin/bgr218. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sun M, Estrov Z, Ji Y, Coombes KR, Harris DH, Kurzrock R. Curcumin (diferuloylmethane) alters the expression profiles of microRNAs in human pancreatic cancer cells. Mol. Cancer. Ther. 2008;7:464–473. doi: 10.1158/1535-7163.MCT-07-2272. [DOI] [PubMed] [Google Scholar]
- 58.Yang J, Cao Y, Sun J, Zhang Y. Curcumin reduces the expression of Bcl-2 by upregulating miR-15a and miR-16 in MCF-7 cells. Med. Oncol. 2010;27:1114–8. doi: 10.1007/s12032-009-9344-3. [DOI] [PubMed] [Google Scholar]
- 59.Zhang J, Du Y, Wu C, Ren X, Ti X, Shi J, Zhao F, Yin H. Curcumin promotes apoptosis in human lung adenocarcinoma cells through miR-186* signaling pathway. Oncol. Rep. 2010;24:1217–1223. doi: 10.3892/or_00000975. [DOI] [PubMed] [Google Scholar]
- 60.Mudduluru G, George-William JN, Muppala S, Asangani IA, Kumarswamy R, Nelson LD, Allgayer H. Curcumin regulates miR-21 expression and inhibits invasion and metastasis in colorectal cancer. Biosci Rep. 2011;31:185–97. doi: 10.1042/BSR20100065. [DOI] [PubMed] [Google Scholar]
- 61.Bao B, Ali S, Kong D, Sarkar SH, Wang Z, Banerjee S, Aboukameel A, Padhye S, Philip PA, Sarkar FH. Anti-tumor activity of a novel compound-CDF is mediated by regulating miR-21, miR-200, and PTEN in pancreatic cancer. PLoS One. 2011;6:e17850. doi: 10.1371/journal.pone.0017850. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 62.Tili E, Michaille JJ, Alder H, Volinia S, Delmas D, Latruffe N, Croce CM. Resveratrol modulates the levels of microRNAs targeting genes encoding tumor-suppressors and effectors of TGFbeta signaling pathway in SW480 cells. Biochem. Pharmacol. 2010;80:2057–2065. doi: 10.1016/j.bcp.2010.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tili E, Michaille JJ, Adair B, Alder H, Limagne E, Taccioli C, Ferracin M, Delmas D, Latruffe N, Croce CM. Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis. 2010;31:1561–1566. doi: 10.1093/carcin/bgq143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Tili E, Michaille JJ. Resveratrol, microRNAs, inflammation, and cancer. J Nucleic Acids. Epub. 2011 Aug 10; doi: 10.4061/2011/102431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lukiw WJ, Zhao Y, Cui JG. An NF-kappaB-sensitive micro RNA-146a-mediated inflammatory circuit in Alzheimer disease and in stressed human brain cells. J. Biol. Chem. 2008;283:31315–22. doi: 10.1074/jbc.M805371200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Han Z, Yang Q, Liu B, Wu J, Li Y, Yang C, Jiang Y. MicroRNA-622 functions as a tumor suppressor by targeting K-Ras and enhancing the anticarcinogenic effect of resveratrol. Carcinogenesis. 2011;7 doi: 10.1093/carcin/bgr226. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 67.Bae S, Lee EM, Cha HJ, Kim K, Yoon Y, Lee H, Kim J, Kim YJ, Lee HG, Jeung HK, Min YH, An S. Resveratrol alters microRNA expression profiles in A549 human non-small cell lung cancer cells. Mol Cells. 2011 doi: 10.1007/s10059-011-1037-z. Epub Aug 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Dhar S, Hicks C, Levenson AS. Resveratrol and prostate cancer: promising role for microRNAs. Mol Nutr Food Res. 2011;55:1219–29. doi: 10.1002/mnfr.201100141. [DOI] [PubMed] [Google Scholar]
- 69.Mukhopadhyay P, Mukherjee S, Ahsan K, Bagchi A, Pacher P, Das DK. Restoration of altered microRNA expression in the ischemic heart with resveratrol. PLoS One. 2010;5:e15705. doi: 10.1371/journal.pone.0015705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mukhopadhyay P, Das S, Gorbunov N, Ahsan MK, Otani H, Pacher P, Das DK. Modulation of miroRNA 20b with resveratrol and longevinex is linked with their potent anti-angiogenic action in the ischemic myocardium and synergestic effects of resveratrol and γ-tocotrienol. J. Cell. Mol. Med. 2011 doi: 10.1111/j.1582-4934.2011.01480.x. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sun Q, Cong R, Yan H, Gu H, Zeng Y, Liu N, Chen J, Wang B. Genistein inhibits growth of human uveal melanoma cells and affects microRNA-27a and target gene expression. Oncol. Rep. 2009;22:563–567. doi: 10.3892/or_00000472. [DOI] [PubMed] [Google Scholar]
- 72.Li Y, Sarkar FH. Down-regulation of invasion and angiogenesisrelated genes identified by cDNA microarray analysis of PC3 prostate cancer cells treated with genistein. Cancer Lett. 2002;186:157–64. doi: 10.1016/s0304-3835(02)00349-x. [DOI] [PubMed] [Google Scholar]
- 73.Parker LP, Taylor DD, Kesterson J, Metzinger DS, Gercel-Taylor C. Modulation of microRNA associated with ovarian cancer cells by genistein. Eur. J. Gynaecol. Oncol. 2009;30:616–621. [PubMed] [Google Scholar]
- 74.Majid S, Dar AA, Saini S, Chen Y, Shahryari V, Liu J, Zaman MS, Hirata H, Yamamura S, Ueno K, Tanaka Y, Dahiya R. Regulation of minichromosome maintenance gene family by microRNA-1296 and genistein in prostate cancer. Cancer Res. 2010;70:2809–2818. doi: 10.1158/0008-5472.CAN-09-4176. [DOI] [PubMed] [Google Scholar]
- 75.Chen Y, Zaman MS, Deng G, Majid S, Saini S, Liu J, Tanaka Y, Dahiya R. MicroRNAs 221/222 and genistein-mediated regulation of ARHI tumor suppressor gene in prostate cancer. Cancer Prev Res. 2011;4:76–86. doi: 10.1158/1940-6207.CAPR-10-0167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Rabiau N, Trraf HK, Adjakly M, Bosviel R, Guy L, Fontana L, Bignon YJ, Bernard-Gallon DJ. miRNAs Differentially Expressed in Prostate Cancer Cell Lines after Soy Treatment. In Vivo. 2011;25:917–21. [PubMed] [Google Scholar]
- 77.Gou JY, Felippes FF, Liu CJ, Weigel D, Wang JW. Negative regulation of anthocyanin biosynthesis in Arabidopsis by a miR156-targeted SPL transcription factor. Plant Cell. 2011;23:1512–22. doi: 10.1105/tpc.111.084525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Li Y, VandenBoom TG, 2nd, Kong D, Wang Z, Ali S, Philip PA, Sarkar FH. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009;69:6704–12. doi: 10.1158/0008-5472.CAN-09-1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Jin Y, Zou X, Feng X. 3,3'-Diindolylmethane negatively regulates Cdc25A and induces a G2/M arrest by modulation of microRNA 21 in human breast cancer cells. Anticancer Drugs. 2010;21:814–22. doi: 10.1097/CAD.0b013e32833e53ea. Retraction in: Anticancer Drugs 22 (2011) 303. [DOI] [PubMed] [Google Scholar]
- 80.Melkamu T, Zhang X, Tan J, Zeng Y, Kassie F. Alteration of microRNA expression in vinyl carbamate-induced mouse lung tumors and modulation by the chemopreventive agent indole-3-carbinol. Carcinogenesis. 2010;31:252–8. doi: 10.1093/carcin/bgp208. [DOI] [PubMed] [Google Scholar]
- 81.Izzotti A, Calin GA, Steele VE, Cartiglia C, Longobardi M, Croce CM, De Flora S. Chemoprevention of cigarette smoke-induced alterations of MicroRNA expression in rat lungs. Cancer Prev Res. 2010;3:62–72. doi: 10.1158/1940-6207.CAPR-09-0202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Izzotti A, Larghero P, Cartiglia C, Longobardi M, Pfeffer U, Steele VE, De Flora S. Modulation of microRNA expression by budesonide, phenethyl isothiocyanate and cigarette smoke in mouse liver and lung. Carcinogenesis. 2010;31:894–901. doi: 10.1093/carcin/bgq037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Izzotti A, Larghero P, Balansky R, Pfeffer U, Steele VE, De Flora S. Interplay between histopathological alterations, cigarette smoke and chemopreventive agents in defining microRNA profiles in mouse lung. Mutat Res. 2011;717:17–24. doi: 10.1016/j.mrfmmm.2010.10.003. [DOI] [PubMed] [Google Scholar]
- 84.Wu N, Wu GC, Hu R, Li M, Feng H. Ginsenoside Rh2 inhibits glioma cell proliferation by targeting microRNA-128. Acta Pharmacol. Sin. 2011;32:345–53. doi: 10.1038/aps.2010.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chen WS, Hou JN, Guo YB, Yang HL, Xie CM, Lin YC, She ZG. Bostrycin inhibits proliferation of human lung carcinoma A549 cells via downregulation of the PI3K/Akt pathway. J Exp. Clin. Cancer. Res. 2011;830:17. doi: 10.1186/1756-9966-30-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Davidson LA, Wang N, Shah MS, Lupton JR, Ivanov I, Chapkin RS. n-3 Polyunsaturated fatty acids modulate carcinogen-directed non-coding microRNA signatures in rat colon. Carcinogenesis. 2009;30:2077–84.S. doi: 10.1093/carcin/bgp245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Tzur G, Levy A, Meiri E, Barad O, Spector Y, Bentwich Z, Mizrahi L, Katzenellenbogen M, Ben-Shushan E, Reubinoff BE, Galun E. MicroRNA expression patterns and function in endodermal differentiation of human embryonic stem cells. PLoS One. 2008;3:e3726. doi: 10.1371/journal.pone.0003726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hu S, Dong TS, Dalal SR, Wu F, Bissonnette M, Kwon JH, Chang EB. The microbe-derived short chain fatty acid butyrate targets miRNA-dependent p21 gene expression in human colon cancer. PLoS One. 2011;6:e16221. doi: 10.1371/journal.pone.0016221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ye Y, Hu Z, Lin Y, Zhang C, Perez-Polo JR. Downregulation of microRNA-29 by antisense inhibitors and a PPAR-gamma agonist protects against myocardial ischaemia-reperfusion injury. Cardiovasc. Res. 2010;187:535–44. doi: 10.1093/cvr/cvq053. [DOI] [PubMed] [Google Scholar]
- 90.Shin S, Lee EM, Cha HJ, Bae S, Jung JH, Lee SM, Yoon Y, Lee H, Kim S, Kim H, Lee SJ, Park IC, Jin YW, An S. MicroRNAs that respond to histone deacetylase inhibitor SAHA and p53 in HCT116 human colon carcinoma cells. Int. J. Oncol. 2009;35:1343–1352. doi: 10.3892/ijo_00000452. [DOI] [PubMed] [Google Scholar]
- 91.Kretzner L, Scuto A, Dino PM, Kowolik CM, Wu J, Ventura P, Jove R, Forman SJ, Yen Y, Kirschbaum MH. Combining histone deacetylase inhibitor vorinostat with aurora kinase inhibitors enhances lymphoma cell killing with repression of c-Myc, hTERT, and microRNA levels. Cancer Res. 2011;71:3912–20. doi: 10.1158/0008-5472.CAN-10-2259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Nalls D, Tang SN, Rodova M, Srivastava RK, Shankar S. Targeting epigenetic regulation of miR-34a for treatment of pancreatic cancer by inhibition of pancreatic cancer stem cells. PLoS One. 2011;6:e24099. doi: 10.1371/journal.pone.0024099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Lee EM, Shin S, Cha HJ, Yoon Y, Bae S, Jung JH, Lee SM, Lee SJ, Park IC, Jin YW, An S. Suberoylanilide hydroxamic acid (SAHA) changes microRNA expression profiles in A549 human non-small cell lung cancer cells. Int. J. Mol. Med. 2009;24:45–50. doi: 10.3892/ijmm_00000204. [DOI] [PubMed] [Google Scholar]
- 94.Rhodes LV, Nitschke AM, Segar HC, Martin EC, Driver JL, Elliott S, Nam SY, Li M, Nephew KP, Burow ME, Collins-Burow BM. The histone deacetylase inhibitor trichostatin A alters microRNA expression profiles in apoptosis-resistant breast cancer cells. Oncol Rep. 2012;27:10–6. doi: 10.3892/or.2011.1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Scott GK, Mattie MD, Berger CE, Benz SC, Benz CC. Rapid alteration of microRNA levels by histone deacetylase inhibition. Cancer Res. 2006;66:1277–1281. doi: 10.1158/0008-5472.CAN-05-3632. [DOI] [PubMed] [Google Scholar]
- 96.Williams AE, Larner-Svensson H, Perry MM, Campbell GA, Herrick SE, Adcock IM, Erjefalt JS, Chung KF, Lindsay MA. MicroRNA expression profiling in mild asthmatic human airways and effect of corticosteroid therapy. PLoS One. 2009;4:e5889. doi: 10.1371/journal.pone.0005889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Pathi SS, Jutooru I, Chadalapaka G, Sreevalsan S, Anand S, Thatcher GR, Safe S. GT-094, a NO-NSAID, inhibits colon cancer cell growth by activation of a reactive oxygen species-microRNA-27a: ZBTB10-specificity protein pathway. Mol. Cancer Res. 2011;9:195–202. doi: 10.1158/1541-7786.MCR-10-0363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Zhao JJ, Lin J, Yang H, Kong W, He L, Ma X, Coppola D, Cheng JQ. MicroRNA-221/222 negatively regulates estrogen receptor alpha and is associated with tamoxifen resistance in breast cancer. J. Biol. Chem. 2008;283:31079–86. doi: 10.1074/jbc.M806041200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 99.Miller TE, Ghoshal K, Ramaswamy B, Roy S, Datta J, Shapiro CL, Jacob S, Majumder S. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J. Biol. Chem. 2008;283:29897–903. doi: 10.1074/jbc.M804612200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Cittelly DM, Das PM, Spoelstra NS, Edgerton SM, Richer JK, Thor AD, Jones FE. Downregulation of miR-342 is associated with tamoxifen resistant breast tumors. Mol. Cancer. 2010;9:317. doi: 10.1186/1476-4598-9-317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Rao X, Di Leva G, Li M, Fang F, Devlin C, Hartman-Frey C, Burow ME, Ivan M, Croce CM, Nephew KP. MicroRNA-221/222 confers breast cancer fulvestrant resistance by regulating multiple signaling pathways. Oncogene. 2011;30:1082–97. doi: 10.1038/onc.2010.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ziouzenkova O, Plutzky J. Retinoid metabolism and nuclear receptor responses: New insights into coordinated regulation of the PPAR-RXR complex. FEBS Lett. 2008;582:32–8. doi: 10.1016/j.febslet.2007.11.081. [DOI] [PubMed] [Google Scholar]
- 103.De Flora S, Izzotti A, D'Agostini F, Balansky RM. Mechanisms of N-acetylcysteine in the prevention of DNA damage and cancer, with special reference to smoking-related end-points. Carcinogenesis. 2001;22:999–1013. doi: 10.1093/carcin/22.7.999. [DOI] [PubMed] [Google Scholar]
- 104.Kensler TW, Egner PA, Dolan PM, Groopman JD, Roebuck BD. Mechanism of protection against aflatoxin tumorigenicity in rats fed 5-(2-pyrazinyl)-4-methyl-1,2-dithiol-3-thione (oltipraz) and related 1,2-dithiol-3-thiones and 1,2-dithiol-3-ones. Cancer Res. 1987;47:4271–7. [PubMed] [Google Scholar]
- 105.De Flora S, D'Agostini F, Balansky R, Camoirano A, Bennicelli C, Bagnasco M, Cartiglia C, Tampa E, Longobardi MG, Lubet RA, Izzotti A. Modulation of cigarette smoke-related end-points in mutagenesis and carcinogenesis. Mutat Res. 2003:523–524. 237–52. doi: 10.1016/s0027-5107(02)00340-8. [DOI] [PubMed] [Google Scholar]
- 106.Weng JR, Omar HA, Kulp SK, Chen CS. Pharmacological exploitation of indole-3-carbinol to develop potent antitumor agents. Mini Rev Med Chem. 2010;10:398–404. doi: 10.2174/138955710791330945. [DOI] [PubMed] [Google Scholar]
- 107.Boominathan L. The tumor suppressors p53, p63, and p73 are regulators of microRNA processing complex. PLoS One. 2010;5:e10615. doi: 10.1371/journal.pone.0010615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hu H, Gatti RA. MicroRNAs: new players in the DNA damage response. J Mol Cell Biol. 2011;3:151–158. doi: 10.1093/jmcb/mjq042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ligorio M, Izzotti A, Pulliero A, Arrigo P. Mutagens interfere with microRNA maturation by inhibiting DICER. An in silico biology analysis. Mutat Res. 2011;717:116–28. doi: 10.1016/j.mrfmmm.2011.07.020. [DOI] [PubMed] [Google Scholar]
- 110.Izzotti A, Larghero P, Longobardi M, Cartiglia C, Camoirano A, Steele VE, De Flora S. Dose-responsiveness and persistence of microRNA expression alterations induced by cigarette smoke in mouse lung. Mutat Res. 2011;717:9–16. doi: 10.1016/j.mrfmmm.2010.12.008. [DOI] [PubMed] [Google Scholar]
- 111.Duan R, Pak C, Jin P. Single nucleotide polymorphism associated with mature miR-125a alters the processing of pri-miRNA. Hum Mol Genet. 2007;16:1124–31. doi: 10.1093/hmg/ddm062. [DOI] [PubMed] [Google Scholar]
- 112.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;101:2999–3004. doi: 10.1073/pnas.0307323101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Schembri F, Sridhar S, Perdomo C, Gustafson AM, Zhang X, Ergun A, Lu J, Liu G, Zhang X, Bowers J, Vaziri C, Ott K, Sensinger K, Collins JJ, Brody JS, Getts R, Lenburg ME, Spira A. MicroRNAs as modulators of smoking-induced gene expression changes in human airway epithelium. Proc. Natl. Acad. Sci. U.S.A. 2009;106:2319–24. doi: 10.1073/pnas.0806383106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Hu Z, Chen J, Tian T, Zhou X, Gu H, Xu L, Zeng Y, Miao R, Jin G, Ma H, Chen Y, Shen H. Genetic variants of miRNA sequences and non-small cell lung cancer survival. J Clin Invest. 2008;118:2600–8. doi: 10.1172/JCI34934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Osada H, Takahashi T. Let-7 and miR-17-92: small-sized major players in lung cancer development. Cancer Sci. 2011;102:9–17. doi: 10.1111/j.1349-7006.2010.01707.x. [DOI] [PubMed] [Google Scholar]
- 116.Negrini M, Gramantieri L, Sabbioni S, Croce CM. MicroRNA involvement in hepatocellular carcinoma. Anticancer Agents Med Chem. 2011;11:500–21. doi: 10.2174/187152011796011037. [DOI] [PubMed] [Google Scholar]
- 117.Qi P, Cheng SQ, Wang H, Li N, Chen YF, Gao CF. Serum microRNAs as biomarkers for hepatocellular carcinoma in Chinese patients with chronic hepatitis B virus infection. PLoS One. 2011;6:e28486. doi: 10.1371/journal.pone.0028486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Izzotti A, Bagnasco M, Cartiglia C, Longobardi M, Camoirano A, Tampa E, Lubet RA, De Flora S. Modulation of multigene expression and proteome profiles by chemopreventive agents. Mutat Res. 2005;591:212–23. doi: 10.1016/j.mrfmmm.2005.03.032. [DOI] [PubMed] [Google Scholar]
- 119.Izzotti A, Bagnasco M, Cartiglia C, Longobardi M, Balansky RM, Merello A, Lubet RA, De Flora S. Chemoprevention of genome, transcriptome, and proteome alterations induced by cigarette smoke in rat lung. Eur J Cancer. 2005;41:1864–74. doi: 10.1016/j.ejca.2005.04.011. [DOI] [PubMed] [Google Scholar]
- 120.Perdomo C, Spira A, Schembri F. MiRNAs as regulators of the response to inhaled environmental toxins and airway carcinogenesis. Mutat Res. 2011;717:32–7. doi: 10.1016/j.mrfmmm.2011.04.005. [DOI] [PubMed] [Google Scholar]
- 121.Etheridge A, Lee I, Hood L, Galas D, Wang K. Extracellular microRNA: a new source of biomarkers. Mutat Res. 2011;717:85–90. doi: 10.1016/j.mrfmmm.2011.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]

