Skip to main content
Current Genomics logoLink to Current Genomics
. 2013 Nov;14(7):441–452. doi: 10.2174/13892029113146660011

Tumor Protein p63/microRNA Network in Epithelial Cancer Cells

Edward A Ratovitski 1,*
PMCID: PMC3867720  PMID: 24396276

Abstract

Non-coding microRNAs are involved in multiple regulatory mechanisms underlying response of cancer cells to stress leading to apoptosis, cell cycle arrest and autophagy. Many molecular layers are implicated in such cellular response including epigenetic regulation of transcription, RNA processing, metabolism, signaling. The molecular interrelationship between tumor protein (TP)-p53 family members and specific microRNAs is a key functional network supporting tumor cell response to chemotherapy and potentially playing a decisive role in chemoresistance of human epithelial cancers. TP63 was shown to modulate the expression of numerous microRNAs involved in regulation of epithelial cell proliferation, differentiation, senescence, “stemness” and skin maintenance, epithelial/ mesenchymal transition, and tumorigenesis in several types of epithelial cancers (e.g. squamous cell carcinoma, ovarian carcinoma, prostate carcinoma, gastric cancer, bladder cancer, and breast tumors), as well as in chemoresistance of cancer cells. TP63/microRNA network was shown to be involved in cell cycle arrest, apoptosis, autophagy, metabolism and epigenetic transcriptional regulation, thereby providing the groundwork for novel chemotherapeutic venues.

Keywords: Apoptosis, Autophagy, Cell cycle arrest, Cell metabolism, Chemoresistance, Epigenetics, microRNA, TP63.

1. INTRODUCTION

Multiple molecular mechanisms are implicated in regulation of gene expression in human cells in various physiologic and pathophysiologic conditions [1]. They include but not limited to epigenetic alterations of DNA methylation, histone methylation or demethylation, histone acetylation or deacetylation, formation of multiple complexes between distinct chromatin components and transcription factors, RNA processing and RNA translation, post-translational modifications of nascent proteins [1, 2]. Finally, a modulation of gene expression by non-coding small interfering microRNAs is also implicated in epigenetic control of gene expression [2-5].

MicroRNAs (miRs) are small 18-24-nucleotide non-coding RNAs, which act through the RNA interference pathway; repress target gene expression largely by modulating translation and mRNA stability [6]. MicroRNAs are sequentially processed from longer precursor molecules that are encoded by the microRNA genes [6]. Primary microRNA transcript (pri-miRNA) is processed in the nucleus by the RNA-induced silencing complex (RISC) to generate mature microRNA [6]. The pri-miRNAs contain one or more ~ 7 base pair stem-loop structures. The ribonuclease DROSHA excises the stem-loop structure to form the precursor microRNA (or pre-miRNA) [7]. After export into the cytoplasm, the pre-miRNA is cleaved by the ribonuclease DICER to generate a short RNA duplex [6-8]. Mature microRNAs bind to RISC and to target mRNAs by base pairing [usually within the 3′-untranslated region (UTR)] subsequently causing an inhibition of protein translation and/or degradation of the mRNA [9, 10]. Levels of the target proteins are consequently reduced, whereas mRNA levels may or may not be decreased [10]. One microRNA could potentially modulate several mRNAs and a few microRNAs might regulate the expression of the same mRNA target [9-11].

MicroRNA expression is deregulated in a wide range of human diseases including cancer [12-14]. Altered expression of microRNA genes has been found in a variety of tumor types and specific microRNAs have shown the oncogenic, tumor-suppressive or apoptotic properties [15, 16]. Certain microRNAs were shown to mediate epigenetic regulation of gene transcription and metabolism, the induction of cell death, cell cycle arrest, autophagy and senescence and contribute to epithelial stem cell maturation [16-19]. On one hand, microRNAs were recently shown to directly bind to gene promoter and gene terminus sequences, thereby modulating specific gene expression at the transcription level [20-23]. On the other hand, transcriptional networks that are often deregulated in cancer cells may lead to altered transcription of specific microRNA genes [24-26]. For example, miR-34 was shown to be regulated by the tumor protein (TP)-p53 transcription factor, “the guardian of the genome”, which regulates the cellular response to stress and cancer-initiating events such as DNA damage [24, 26].

Non-coding regulatory microRNAs may also have therapeutic applications by which cancer-causing microRNAs could be modulated to restore the normal cellular function (27, 28). The modified cholesterol-conjugated antisense RNAs designated “antagomirs” were shown to effectively inhibit microRNA function in vivo in the adult mouse [29]. The competitive microRNA inhibitors (“microRNA sponges”) were reported to de-repress microRNA targets as strongly as chemically modified antisense oligonucleotides [30].

Accumulating evidence supports that microRNAs, whose transcription regulated by TP53 family factors (TP53, TP63, and TP73), could contribute in multiple signaling pathways involved in cell cycle arrest, apoptosis, autophagy, metabolism and epigenetic transcriptional regulation, thereby potentially underlying the mechanisms leading to epithelial cell maintenance, and tumor development and chemoresistance [4, 19, 24, 26, 31-46]. As a member of the TP53 family, TP63 transcription factor is likely to play its decisive role in transcriptional and post-transcriptional regulation of microRNAs in epithelial cancers, epithelial differentiation and epithelial/mesenchymal transition (EMT) [31-38, 41, 42, 45, 46]. Complex gene expression machinery complicates to fully recognize the role for TP63 in modulation of microRNA expression [47]. Due to distinct promoters and multiple splicing events, TP63 encodes six protein isoforms; three of them contain the long transactivation (TA-) domain (TAp63 isoforms α, β and γ), whereas other three are lacking this TA domain (∆Np63 isoforms α, β and γ), as reviewed in [47]. Emerging data strongly suggests that TAp63 isoforms function in similar manner as TP53 or TP73 by inducing cell death and tumor suppression, while ∆Np63 isoforms are frequently acting in an opposite manner by promoting the oncogenic function and modulating the cell death contributing to tumor cell chemoresistance [47].

Number of reports showed that upon ATM-dependent phosphorylation of ∆Np63α, this transcription factor is able to regulate expression of downstream gene targets implicated in control of cell death (e.g. cell cycle arrest, apoptosis, and autophagy). Moreover, the phosphorylated (p)-∆Np63α was shown to induced or reduce the transcription and processing of microRNAs, which subsequently contributed to modulation of targets involved in cell death and survival [48-53]. This review is designed to give a first glimpse on potential roles of TP63-regulated microRNAs in a few key cellular processes that contribute to tumor cell proliferation, cell death, cell metabolism and epigenetic regulation of gene expression.

2. TP63 TRANSCRIPTIONALLY REGULATE VARIOUS microRNAs THAT MODULATE NUMEROUS TARGETS IN EPITHELIAL CANCER CELLS

TP53 family members were shown acting as candidate drivers of microRNA overexpression [43]. Expression of both TP73 and TP63 is significantly correlated with expression of microRNAs whose promoters contain TP53 family binding sites in head/neck and ovarian carcinomas [33, 43]. Validated data showed that TP53 family binding sites modulate promoter activity of the miR-200 family and miR-429, which are known regulators of cancer stem cells and epithelial/mesenchymal transitions, as well as promoters for miR-181a-5p, miR-374a-5p, miR-519a-3p miR-630 and miR-885-3p, which were reported to play a regulatory role in cell cycle arrest, apoptosis, and autophagy [32, 33, 36, 38, 41, 45, 46, 52]. Moreover, miR-200 family, miR-1246 and miR-155 were shown to be direct targets for TP53 and TP63, respectively, while miR-193a-5p is regulated negatively by TP63 and positively by TP73 at the transcriptional level [31, 33, 36, 39, 40]. ΔNp63α was shown to inhibit miR-138, -181a, -181b, and -130b expression by binding directly to TP63-responsive elements located in close proximity to the genomic loci of these microRNAs in primary keratinocytes [41]. TP63 was shown to maintain cell cycle progression by directly repressing miR-34a and miR-34c in primary keratinocytes and in embryonic skin [46]. TP63 directly binds to TP53-consensus sites in both miR-34a and miR-34c regulatory regions resulting in reduction of their transcription, which leads to a restored cell cycle progression and expression of cyclin D1 (CCND1) and cyclin-dependent kinase (CDK)-4 [46]. ΔNp63α was found to promote miR-205 transcription and to subsequently control EMT via modulation of ZEB1/2 levels in human bladder cancer cells [45]. Multiple TP63-regulated microRNAs (miR-17, miR-20b, miR-30a, miR-106a, miR-143 and miR-455-3p) were involved in epidermal differentiation [44].

Using the microRNA expression chip arrays, subsequently validated by quantitative PCR expression analysis several reports showed altered expression of miR-485-5p, miR-297, miR-185-5p miR-194-5p, miR-574a-3p, miR-720, miR-98-5p, miR-29c-3p, miR-101-3p, miR-22-3p, miR-34c-3p, miR-206, miR-429, miR-339-3p, miR-203a, miR-25-3p, miR-155-5p, miR-148a-3p, miR-125b, miR-181a-5p, miR-374a-5p, miR-519a-3p, miR-630, miR-885-3p and miR-1246 in human epithelial cancers including squamous cell carcinoma, ovarian carcinoma, prostate carcinoma, gastric cancer, bladder cancer, and breast tumors [31-46, 49-54]. Accumulating evidence shows that a number of above-listed microRNAs were found to modulate the expression of critical proteins involved in tumor cell response to DNA damage, tumorigenesis, tumor cell death, and tumor metastasis, such as EGFR, SIRT1, ZNF652, CARM1, TP63, SKP2, STMN1, MSI2, ROCK1, NOTCH1, ZEB1/2, BMI, CCND1, CDK4, ULK1, ATG2, ATG4, ATG5, ATG7, ATG9, ATG10, ATG12, ATG16, BECN1, RAB5A, and MAPK8/9 [31-46, 49-54].

Using the luciferase activity assays mediated by the target mRNA 3’-untranslated regions (UTR), we and others further found that several direct targets of tested microRNAs, including ataxia telangiectasia mutated kinase (ATM), autophagy-related 5 and 10 (ATG5 and ATG10), caspase/ apoptosis-related cysteine peptidases 2 and 3 (CASP2 and 3); cyclin-dependent kinase 1 and 2 (CDK1 and 2); cyclin-dependent kinase inhibitor 1C and 2B (CDKN1C and 2B), and nuclear factor Y beta (NFYB), as reviewed in [51, 53, and Table].

The selected microRNA mimics inhibited the luciferase activity fused to the specific mRNA 3’-UTR by ~32-56% compared to scrambled microRNA, while their protein targets were found downregulated subsequently affecting the tumor cell response to chemotherapeutic platinum agents [49-53]. In order to classify the effect of TP63/microRNA circuitry, we divided the microRNA-regulated products on the following categories: epigenetic regulation of transcription, cell metabolism, autophagy, cell cycle arrest and apoptosis.

3. TP63-DEPENDENT microRNAs AND EPIGENETIC REGULATION OF TRANSCRIPTION

While the binding of transcription factors and other chromatin accessory components to the corresponding promoter element is essential for transcriptional function, the final outcome (activation or repression) is very likely to be determined by numerous chromatin accessory/remodeling proteins [1, 3, 55-58]. Transcriptional activating mechanisms include: demethylation of promoter DNA sequences, acetylation or demethylation of histones altering histone interactions with other chromatin proteins and nucleosome properties subsequently affecting the chromatin remodeling. However, the transcription repression mechanisms include: methylation of promoter DNA sequences and methylation or deacetylation of histone molecules forming nucleosome structures around promoter sequences [55-61].

Previous studies shed a light on the role for TP63 (e.g. ∆Np63α) in many of these epigenetic regulatory layers. Recent reports showed that ∆Np63α represses anti-proliferative genes via accumulation of acetylated H2A.Z, while physical association of HDAC1 and HDAC2 with TP63 mediates transcriptional repression of BCL2 and PUMA and tumor maintenance in squamous cell carcinoma [62, 63]. Moreover, the ATM-dependent p-∆Np63α is capable to regulate a plethora of various microRNAs that showed a potential to affect the expression levels of many components of epigenetic regulatory machinery (Table, Part A).

Table 1.

TP63-regulated microRNAs in Epithelial Cancers

A. Epigenetic Regulation
MicroRNA (hsa-miR) Protein Target Tissues/cells References
92b-3p KAT2B, HDAC9 SCC Ratovitski, in preparation
185-5p ATF6, DNMT1, SREBF2 SCC This review
194-5p KAT6B, SIRT1, ATM SCC This review
196-3p MBD1 SCC This review
297 SIRT3, DNMT3A, SKP2, ATM SCC [52], Ratovitski, in preparation
382-3p NFYB SCC This review
485-5p KDM4C SCC Ratovitski, in preparation
610 ATF5 SCC This review
630 EZH2, ZBTB2, KAT3B SCC [52]
637 ATF3 SCC This review
885-3p CARM1 SCC [52]
920 KAT6B, NFYB SCC This review
181a-5p HDAC4, SIRT1, KAT2B, ATM, TP63 SCC [52]
374a-5p SP1, NFYB, CRTC2, KAT2B, ATM, TP63 SCC [52]
519a-3p KDM2A, BHLHE41, ATM, TP63 SCC [52]
29c-3p SIRT1, HDAC4, KDM2A, DNMT3B SCC [74, 75], Ratovitski, in preparation
22-3p KAT6B, SIRT1, KDM3A, MECP2 SCC This review
34c-3p DNMT1 SCC Ratovitski, in preparation
339-3p DNMT3B SCC This review
203a NFYA, CITED2, KAT6B, TP63, ATM SCC [38, 41, 52], This review
206 CITED, KAT6A SCC This review
25-3p HDAC9 SCC Ratovitski, in preparation
155-5p SP3, KDM2A SCC This review
148a-3p DNMT1, DNMT3B SCC, breast and gastric cancers [76, 77, 145], This review
101a-3p EZH2, DNMT3A SCC [80, 81], This review
429 CITED2, E2F3, NFYA SCC This review
455-3p KAT2B SCC This review
27a-3p KDM3A, HDAC9, TP53 SCC Ratovitski, in preparation
183-5p KDM3A SCC This review
362-3p SIN3A, E2F1 SCC This review
603 ATM, TP63 SCC This review
B. Cell Metabolism
MicroRNA (hsa-miR) Protein Target Tissues/cells References
92b-3p ATOX1 SCC This review
297 ATP7A SCC This review
382-3p ETNK SCC This review
485-5p ETNK, H6PD SCC This review
885-3p AKT1 SCC [50]
29c-3p CPS1, AKT2 SCC This review
203a ATP7B, CPS1, FADS1 SCC This review
18a-5p CPS1, CPS2 (CAD) SCC This review
146b-3p CPS1 SCC This review
101a-3p COX2, AKT1 Gastric cancer, Prostate cancer [93]
185-5p SREBF, FADS1, HMGCR Prostate cancer [94]
34a IMPDH Lung Cancer [90]
C. Cell Cycle Arrest and Apoptosis
MicroRNA (hsa-miR) Protein Target Tissues/cells References
92b-3p CDKN1C SCC This review
185-5p CASP2, CASP14, PARP11 SCC This review
194-5p CASP7 SCC This review
485-5p PARP8, DFFA SCC This review
760 BMF SCC This review
885-3p CASP3 SCC [50]
519a-3p CASP2, CDKN2B SCC [49, 53]
34c-3p BMF SCC This review
98-5p CASP3 SCC [53]
25-3p CDKN1C SCC [53]
155-5p APAF1 SCC This review
29c-3p BMF SCC This review
18a-5p CASP7 SCC This review
214-3p DFFA, BCL2, BCL2L2 SCC, cervical cancer [53, 120], This review
659-3p CHEK1 SCC This review
7a-5p CASP3, XIAP SCC, cervical cancer [109], This review
221-3p CDKN1B SCC This review
429 CASP2, CDKN2B, CDK2, BCL2 SCC [53]
29c-3p CDK2 SCC [53]
382-3p CDK1 SCC [53]
D. Autophagy
MicroRNA (hsa-miR) Protein Target Tissues/cells References
181a-5p ATG5 SCC [52, 53]
374a-5p ATG4A, ATG5, UVRAG SCC [52, 53]
519a-3p ATG10, ATG16L1, UVRAG SCC [52, 53]
22-3p ATG2B SCC This review
34c-3p ATG4C, DRAM1 SCC This review
339-3p GABARAPL1 SCC This review
203a ATG2B, GABARAPL1 SCC [53]
155-5p GABARAPL1 SCC This review
485-3p MAP1LC3B SCC This review
214-3p SQSTM1, ATG12 SCC This review
183-5p ATG12 SCC [53]
98-5p ATG10 SCC [53]
27-3p ATG10 SCC [53]
603 ATG10 SCC [53]
630 UVRAG, ATG2B, ATG4C, ATG12 SCC [52, 53], This review
297 ATG5 SCC [53]
101a-3p ATG4D, RAB5A HCC, breast cancer [127]
885-3p ULK2, ATG16 SCC [51]
93a SQSTM1 [53]
194-3p GABARAPL1 SCC This review

Using a set of bioinformatics tools, p-∆Np63α- dependent microRNAs were predicted to modulate the protein targets (Table, part A) involved in DNA methylation (DNMT1, DNMT3A, DNMT3B, MBD1 and MECP2), histone acetylation (KAT2B, KAT3B and KAT6B), histone deacetylation (HDAC4, HDAC9, SIRT1, SIRT3 and SIRT5), histone demethylation (KDM2A, KDM3A and KDM4C), polycomb repressive complex (EZH2, SIN3A), transcription factors (ATF3, ATF5, ATF6, E2F1, E2F3, SREBF2, SP1, SP3, NFYA, NFYB and TP53), co-activators (CITED2 and CRTC2), co-repressors (BHLHE41 and ZBTB2) and RNA splicing (SRSF2), as reviewed in [52]. Intriguingly, many protein targets listed above are likely to be affected by several microRNAs, while certain microRNAs could potentially modulate several targets (Table, Category A). Since expression of specific microRNAs is upregulated or downregulated through the cisplatin-/p-∆Np63α- dependent mechanism, the final outcome on specific epigenetic regulators is not easy to predict. For example, specific microRNAs (miR-185-5p or miR-297) that are upregulated in cisplatin-treated squamous cell carcinoma cells (SCC-11) appeared to decrease the protein levels for DNMT1 or DNMT3A, while downregulated microRNAs (148a-3p and 101-3p) could potentially lead to increase of these proteins [53, Ratovitski, in preparation]. Similarly, miR-92-3p could decrease the HDAC9, while miR-25-3p and 27a-3p would increase this protein level in SCC-11 cells exposed to cisplatin treatment [53, Ratovitski, in preparation].

The repression of gene transcription could occur directly by hypermethylation of CpG dinucleotides within the promoter DNA regions by DNA methyltransferases (DNMT1, 3A and 3B), or indirectly through interaction with methyl-CpG-binding proteins (e.g. MBD1 or MECP2) leading ultimately to inability of transcription factors to bind their recognition sites [1, 2, 64]. DNMT1 preserves the methylation DNA patterns throughout each cell division, while DNMT3A and 3B transfer a methyl group to unmethylated DNA sequences [64]. There are other enzymes involved in adding or removing methyl or acetyl groups to histones: histone methyltransferases (EZH1 and 2), histone demethylases (KDM1-KDM8), histone acetyltransferases (KAT2A-KAT8), and histone deacetylases (HDAC1-9, SIN3 complex), as reviewed in [65-67].

Although methylation of the promoter DNA leads to transcriptional repression, the methylated histones can either activate or repress gene transcription [65-67]. For example, the trimethylation of H3 lysine 4 or H3 lysine 36 leads to gene activation, whereas the trimethylation of H3 lysine 27, or di- and tri-methylation of H3 lysine 9, or trimethylation of H4 lysine 20 would lead to gene repression [65-67]. Similarly, histone demethylases would affect the gene transcription in an opposite fashion [65-67]. On the other hand, histone acetylation catalyzed by histone acetyltransferases is linked to transcriptional activation, while deacetylation of histones by HDACs is often leading to a transcriptional repression [62]. An increased DNA methylation can silence tumor suppressor and pro-apoptotic genes, whereas the demethylation of DNA can induce the oncogenic and anti-apoptotic genes, thereby such epigenetic mechanisms, which control transcription of genes involved in cell differentiation, proliferation, survival and apoptosis are often deregulated in cancer cells leading to malignant phenotypes [1, 2, 65-67].

Finally, the intricate network of epigenetic regulation of gene expression has been further enriched by the non-coding microRNAs, whose ability to modulate the level of target proteins via binding to their respective mRNA 3’-UTR sequences was complemented by the direct effect of microRNAs on the epigenetic machinery [20-23, 68-71].

The expression of microRNA, which is altered in almost all human cancers, is affected by the same epigenetic mechanisms as mRNA transcription [66]. The ability of microRNAs to regulate the components of the epigenetic machinery, targeting molecules involved in the DNA methylation, histone acetylation, and modulation of transcription factors is also started to emerge creating a controlled feedback mechanism [2, 4, 72-74]. For example, the introduction of miR-148a and miR-34b/c in cancer cells inhibited their motility, reduced tumor growth, and impaired metastasis formation in tumorgraft models, and led to a down-regulation of the microRNA oncogenic targets, such as c-MYB, c-MYC, E2F3, CDK6, HDAC, and TGIF2 [72-74]. miR-29 family was reported to directly target DNMT3A, -3B, and indirectly DNMT1 through regulation of the transactivator Sp1 [74, 75]. miR-148a was shown to directly target DNMT3B by binding a recognition site located in the coding region [76, 77]. miR-140, miR-148a, miR-152 and miR-301 were shown to modulate DNMT1, while miR-101a-3p was found to regulate the expression of EZH2, catalytic subunit of the polycomb repressive complex 2, which mediates epigenetic gene silencing by trimethylating histone H3 lysine 27, and miR-449a was found to modulate HDAC1 inducing cell cycle arrest, apoptosis and a senescent phenotype in many cancer cells [82]. Taken together, these data strongly support the notion that alterations of microRNA landscape in cancers cells are very likely to affect the epigenetic regulation of genes involved in cell death and survival, and thereby could be useful biomarkers and targets for the future development of anti-cancer biotherapies.

4. TP63-DEPENDENT microRNAs AND CELL METABOLISM

Under many environmental stresses that induce DNA damage, including chemotherapy, tumor cells respond by changing their metabolism, which allows sustaining tumor growth under the fluctuations in energy availability. Mounting evidence shows the interplay between microRNAs and oncogenes/tumor suppressors, via key metabolic enzyme effecters, which could facilitate the Warburg Effect (anaerobic production of energy) in cancer cells [83-85]. Changing levels of glucose modulate miR-451 expression, thereby affecting cell proliferation but enhancing migration and survival [86]. miR-451 was shown to repress CAB39, the binding partner of LKB1 leading to a subsequent regulation of the LKB1/AMPK pathway [86]. miR-33a/b plays a crucial role in controlling cholesterol and lipid metabolism by targeting phosphoenol-pyruvate carboxykinase (PCK1), glucose-6-phosphatase (G6PC), carnitine palmitoyltransferase 1A (CPT1A), and AMP-activated protein kinase (AMPK α 1) via the sterol-regulatory element-binding transcription factors (SREBF) [87]. Additionally, miR-103 and miR-107 were reported to regulate insulin and glucose homeostasis through a modulation of pantothenate kinase, while miR-34a affects hepatic lipid homeostasis [88].

TP53 transcription factor that controls cell cycle arrest, cell death, autophagy, and glucose metabolism was shown to trigger a metabolic switch to the Warburg effect found in the most cancer cells [89]. Recent studies show that TP53-inducible miR-34a modulates the expression of glucose metabolic enzymes (e.g. (hexokinase 1 and 2, glucose-6-phosphate isomerase, and pyruvate dehydrogenase kinase 1), as well as a nucleotide biosynthesis by repression of inosine 5'-monophosphate dehydrogenase, a rate-limiting enzyme for de novo purine biosynthesis, needed for a sustained tumor cell proliferation [90, 91]. Moreover, phosphatidylinositol 3-kinase that regulates the levels of phosphorylated phosphatidylinositol at the plasma membrane, and plays a key role in cancer cell metabolism is targeted by miR-123a, miR-136, miR-320, miR-422, and miR-506 [92]. Additionally, miR-152, miR-148a, miR-148b, miR-299-5p, miR-19a/b, miR-122a, miR-421, and miR-494 regulate the citrate synthase gene, which encodes a major enzyme in tricarboxylic acid (Krebs) cycle [85, 92]. miR-101a-3p was found to play a critical role in the regulation of cyclooxygenase-2 (COX-2) expression gastric cancer specimens and cell lines leading to a decreased cell proliferation and increased apoptosis in vitro and in vivo [93]. miR-185-5p and miR-342 were shown to inhibit SREBF-1 and 2 expression and downregulate their targets, fatty acid synthase 1 and 3-hydroxy-3-methylglutaryl CoA reductase in prostate cancer, as well as inhibited tumorigenicity, cell growth, migration and invasion in prostate cancer cell culture and xenograft models [94].

The cisplatin-/p-∆Np63α- dependent microRNAs could potentially affect the critical metabolic pathways through a modulation of specific protein levels (e.g. ATOX1, ATP7A, ATP7B, ETNK, H6PD, CPS1, CPS2/CAD, FADS1, AKT1 and AKT2), as shown in Table (Category B). Future studies are underway to provide a proof of concept for this notion. Previous reports showed that TP63-dependent microRNAs (e.g. miR-885-3p) modulated the 3’-UTR activity driven by AKT1 [50], while p-∆Np63α was found to transcriptionally regulate many metabolic enzymes by interacting with SREBF1 [95]. Interestingly, CPS1 expression is likely to be increased in SCC-11 cells upon cisplatin exposure since four microRNAs (miR-29c-3p, miR-203a, miR-18a-5p and miR-146b-3p) are downregulated under these experimental conditions [53]. Interestingly that a few proteins involved in intracellular copper binding, transport and regulation (ATOX1, ATP7A and ATP7B) are also shown to bind cisplatin, thereby are likely to play a role in platinum chemoresistance [96]. p-∆Np63α-dependent microRNAs are likely to modulate the metabolic enzymes implicated in carbohydrate (H6PD), lipid (ETNK, FADS1), amino acid (CPS1) and pyrimidine (CPS2/CAD) metabolism (Table). Finally, miR-101a by modulating the activation of serine/threonine kinases, AKT1 and AKT2, was shown to suppress apoptosis by phosphorylation of components of the apoptotic machinery [97, 98] subsequently linking the tumor cell metabolism to the apoptotic response or lack thereof. These data support further studies, which are needed to establish a mechanistic link between TP63-dependent regulation of metabolic enzymes through transcription and microRNA modulation. Overall this microRNA network could potentially contribute to dramatic alterations of metabolism and biosynthesis of many metabolic compounds leading to energy imbalance and deregulated tumor cell proliferation, as reviewed in [95].

5. TP63-DEPENDENT microRNAs AND CELL CYCLE ARREST AND APOPTOSIS

As well known, cell cycle regulation occurs through modulation of activity of CDK by cyclins (positive regulation) leading to cell proliferation and CDK inhibitors (negative regulation) inducing a temporary cell cycle arrest in G1 phase, or a permanent cell cycle arrest, if induced by damaged DNA, often resulting in a cell death [99, 100]. MiR-449a, -b, and -c are potent inducers of cell death, cell cycle arrest, and/or cell differentiation, as well as miR-34 regulated by TP53, while also induced by the cell cycle regulatory transcription factor E2F1 [71]. These microRNAs were shown to downregulate histone acetyltransferases and activate TP53, while modulating CDK and their association partners provide an asymmetric feedback loop to balance E2F and p53 activities [101, 102]. A few reports showed that CDK inhibitors could also be targets for a microRNA regulation (e.g. miR-22, miR-296, miR-423 and miR-519a-3p for CDKN1A; miR-221 and miR-222 for CDKN1A, CDKN1B and CDKN1C), as shown elsewhere [101, 102]. The cell cycle-regulating microRNAs are incorporated into a large regulatory network to control the self-renewal of stem cells by inducing or inhibiting differentiation and function of cell cycle-regulating microRNAs in cancer [103].

Alternatively to cell cycle arrest, the caspase-dependent and -independent pathways could also lead to the cell death through apoptosis when cells face irreversible stress [104-106]. Members of the BCL2 family play crucial roles in regulating intrinsic apoptotic pathway, while in the extrinsic apoptotic pathway CASP8 and CASP10 are activated upon stimuli [105, 106]. Both apoptotic pathways converge on the level of CASP3 activation, which in turn cleaves various intracellular substrates and cause the specific morphological changes [104-106]. During the caspase-dependent apoptosis, the caspase cascade pathways are being initiated, which include initiator caspases (e.g., CASP2, CASP8, CASP9, and CASP10), and effector (executioner) caspases (e.g., CASP3, CASP6, CASP7). The latter are being activated by the former and subsequently cleave other protein substrates within the cell, to trigger the apoptotic process [106].

MicroRNAs were shown to modulate the function of various pro- and anti- apoptotic proteins under cisplatin pressure [107, 108]. For example, miR-885-3p (or miR-708), Let7a-1 (or miR-24), and miR-155-5p were shown to decrease CASP3, XIAP, and APAF1 levels, respectively [50, 109-112]. Multiple microRNAs designated as ‘apoptomirs’ (e.g. miR-15a, miR-16, miR-125b, miR-153, miR-519a-3p, miR-205, miR-210, miR-214, miR-429, miR-503, miR-26a, miR-29b, miR-193a-3p and miR-133a) were reported to reduce the expression of BCL2 family members through their respective 3-'UTR region binding sites in various cancer cells [113-122]. Intriguingly, downregulation of PDGFR-α/β by siRNA or miR-34a/c strongly augmented the response to TNF-related apoptosis inducing ligand (TRAIL) while reducing migratory and invasive capacity of non-small cell lung carcinomas [123]. The TRAIL-mediated apoptosis pathway was shown to be sensitive to miR-29, miR-130a, miR-133b, miR-185-5p, miR-221, and miR-222 [124, 125]. Whereas, miR-483-3p displayed a tumor suppressive function in SCC by targeting CDC25A resulting in cell cycle arrest and API5, BIRC5, and RAN leading to pro-apoptotic pathway, which significantly hampered tumor growth of SCC tumorgrafts [126].

Exposure of SCC cells to cisplatin was also leads to a modulation of certain proteins involved in cell cycle arrest or apoptosis ultimately resulting in cell death phenotype. Specific microRNAs, whose transcription is upregulated by p-∆Np63α, were reported to decrease the levels of CASP2, CASP3, CASP7 and CASP14, PARP8 and PARP11, BMF, DFFA and CDKN1C, while downregulated microRNAs were shown to increase of CASP2, CASP3 and CASP7, BMF, CDKN1B, CDKN1C and CDKN2B, APAF1, DFFA and CHEK1, as shown in Table (Category C). Taken together these data strongly support the notion that some of the TP63-regulated microRNAs are likely to contribute to the tumor cell survival and response to chemotherapy alone on in combination with the specific microRNA mimics or inhibitors [50, 53].

6. TP63-DEPENDENT microRNAs AND AUTOPHAGY

Emerging evidence shows that the specific microRNAs are involved in the regulation of autophagy and play a role in modulating the cross talk between autophagy and apoptosis contributing to cancer and tumor cell response to chemotherapy [19, 127-132]. Autophagy is an intrinsic tightly controlled catabolic cellular process in which proteins and organelles are eliminated through delivery to lysosomes, while preserving the cell function and survival [133, 134]. Deregulation of autophagy under stress leading to a malfunction of the autophagic regulatory mechanisms contributes to cancer and tumor cell response to chemotherapy [134-138].

MicroRNAs were shown to modulate numerous signaling intermediates of autophagic pathway (e.g. miR-519a-3p for ATG10, and ATG16L1; miR-101a-3p for ATG4D and RAB5A; miR-17, miR-20, miR-93a and miR-106 for SQSTM1; miR-204 for MAP1LC3; miR-885-3p for ULK2 and ATG16; miR-630 for ATG12 and UVRAG; miR-30a for BECN1; miR-181a-5p for ATG5; miR-630 for ATG12; miR-376b for ATG4C and BECN1; miR-375 for ATG7; miR-374a-5p for ATG4A, ATG5 and UVRAG; miR-34a for ATG9; and miR-130a for ATG2), as reviewed in [19, 127-132].

By inhibiting BECN1, miR-30a leads to the suppression of autophagic phenotype in cancer cells, thereby contributing to cancer progression [128]. ATG4-ATG8 conjunction is a crucial step in the autophagosome biogenesis pathway, thereby underscoring the importance of miR-101a-dependent regulation [129]. SQSTM1 (p62), a multiple domain protein that acts as a signaling hub, was identified as a key target for multiple microRNAs [130]. SQSTM1 can interfere with autophagy via binding to the autophagic regulator ATG8/MAP1LC3. Thus, elimination of SQSTM1 through microRNA modulation may potentially inhibit the proliferation of these tumor cells [130]. MiR-376b was reported to control starvation and mTOR inhibition-related autophagy by targeting ATG4C and BECN1 [131].

Intriguingly, several confirmed targets of microRNAs are also important mediators in the cross regulation between autophagy and apoptosis [19, 127, 132]. For example, the physical interaction between BECN1 and proteins in the anti-apoptotic family (BCL2, MCL1, BCL2L1) is pivotal for the interplay between the autophagic and apoptotic pathways [128, 139-141]. Normally, BECN1 and anti-apoptotic BCL2 proteins can bind to each other to maintain cellular homeostasis [139]. However, upon stress, BECN1 and BCL2 proteins disassociate, thereby promoting autophagy and inhibiting apoptosis, respectively. ATG5, in addition to the promotion of autophagy, enhances susceptibility towards apoptotic stimuli [142]. Enforced expression of ATG5 renders tumor cells sensitive to chemotherapy, whereas silencing the ATG5 with siRNA resulted in partial resistance to anticancer drugs. This tumor cell response was associated with calpain-mediated ATG5 cleavage resulting in cytochrome c release and caspase activation suggesting a molecular link between autophagy and apoptosis [143].

Exposure of SCC-11 cells to cisplatin treatment leads to the p-∆Np63α- dependent modulation of numerous microRNAs potentially implicated in regulation of autophagic signaling intermediates [50, 51, 53]. Although the most of the autophagic proteins appeared to be induced since their corresponding microRNAs are downregulated in SCC-11 cells upon cisplatin exposure, a few proteins are likely to be reduced by upregulated microRNAs (miR-194-3p, miR-297 and miR-630), as reviewed in [50, 51, 53]. However, all protein targets (ATG2B, ATG4A, ATG4C, ATG5, ATG10, ATG12, ATG16L1, DRAM1, GABARAPL1, MAP1LC3B, SQSTM1 and UVRAG1) seem to be affected by both downregulated and upregulated microRNAs (Table, Category D), and as reviewed in [50, 51, 53]. Intriguingly, the specific microRNAs were further shown to modulate resistant phenotype of SCC cells in vitro, thereby providing groundwork for novel chemotherapeutic venues for head and neck cancer [50, 51, 53].

7. CONCLUSIONS

Accumulating evidence shows that microRNAs, whose transcription is regulated by many transcriptional factors, including TP53 members (TP53, TP63 and TP73) contribute to multiple mechanisms implicated in control of tumor cell homeostasis, proliferation and survival under chemotherapeutic pressure [31, 43]. The diverse actions of these microRNAs affected by TAp63 isoforms and ∆Np63 isoforms complicate the tumor cell response even more. While the former are generally act as pro-apoptotic and tumor suppressive agents, the latter function more as an anti-apoptotic and oncogenic factors. However, p-∆Np63α occupies a more intermediate niche, since the microRNAs regulated by this transcriptional factor are capable to function in a pro-apoptotic and cell cycle arrest manner, as well as modulate the survival pathway of autophagy, therefore supporting the sensitive response to chemotherapeutic agents (e.g. platinum drugs), as reviewed in [49-53]. Intriguingly, many microRNAs, whose transcription was shown (miR-181a-5p, miR-374a-5p, miR-519a-3p, miR-203a) or predicted to be regulated by TP63 (miR-297 and miR-603), are, in fact, shown [41, 52], or predicted to maintain the feedback control of the TP63 protein levels, as well as likely to modulate the TP63 phosphorylation status via ATM inhibition (Table, Category A).

Over recent years, microRNAs have emerged as major players in the complex networks of gene regulation and have been implicated in various aspects of human disease and were designated as one of the key hallmarks of cancer [144]. In addition to oncogenes and tumor suppressor genes, microRNAs and their regulatory networks should be taken into account to understand the complex molecular mechanisms underlying malignant transformation and acquired chemoresistance to anti-cancer drugs. MicroRNAs are important regulators of numerous aspects of metabolic homeostasis, physiology and disease. On one hand, microRNAs were shown or predicted to regulate the transcription and protein levels of numerous transcription factors and epigenetic/chromatin accessory components or signaling proteins involved in regulation of metabolic enzymes and critical regulators of cell cycle arrest, apoptosis or autophagy [50, 51, 53]. On the other hand, microRNAs could regulate the production of certain metabolites by directly affecting the levels of metabolic enzymes [83, 95]. Therapeutic use of microRNA mimic or inhibitors to suppress certain stages or steps of tumor cell metabolism is likely to lead to new anti-cancer biotherapeutic strategies [28, 83].

ACKNOWLEDGEMENTS

I thank Dr. Yiping Huang (Johns Hopkins University School of Medicine) for many years of collaboration and friendship, her dedication and support. This study was supported in part by the Flight Attendant Research Institutions grant (#082469).

CONFLICT OF INTERESTS

The author(s) confirm that this article content has no conflicts of interest.

ABBREVIATIONS

ATM

 = Ataxia telangiectasia mutated

CDK

 = Cyclin-dependent kinase

DNMT

 = DNA methyltransferase

microRNA

 = miR

p

 = Phosphorylated

RISC

 = RNA-induced silencing complex

SCC

 = Squamous cell carcinoma

SREBF

 = Sterol-regulatory element-binding transcription factor

TA

 = Transactivation

TNF

 = Tumor necrosis factor

TP

 = Tumor protein

TRAIL

 = Tumor necrosis factor related apoptosis inducing ligand

XIAP

 = X-linked inhibitor of apoptosis protein

UTR

 = Untranslated region

REFERENCES

  • 1.Tsai HC, Baylin SB. Cancer epigenetics: linking basic biology to clinical medicine. Cell Res. 2011;21:502–517. doi: 10.1038/cr.2011.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Iorio MV, Piovan C, Croce CM. Interplay between microRNAs and the epigenetic machinery: an intricate network. Biochim. Biophys. Acta. 2010;1799:694–701. doi: 10.1016/j.bbagrm.2010.05.005. [DOI] [PubMed] [Google Scholar]
  • 3.Wiklunde D, Kjems J, Clark SJ. Epigenetic architecture and miRNA: reciprocal regulators. Epigenomics. 2010;2:823–840. doi: 10.2217/epi.10.51. [DOI] [PubMed] [Google Scholar]
  • 4.Esteller M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011;12:861–874. doi: 10.1038/nrg3074. [DOI] [PubMed] [Google Scholar]
  • 5.Lovat F, Valeri N, Croce CM. MicroRNAs in the pathogenesis of cancer. Semin. Oncol. 2011;38:724–733. doi: 10.1053/j.seminoncol.2011.08.006. [DOI] [PubMed] [Google Scholar]
  • 6.Pratt AJ, MacRae IJ. The RNA-induced silencing complex: a versatile gene-silencing machine. J. Biol. Chem. 2009;284:17897–17901. doi: 10.1074/jbc.R900012200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.MacFarlane LA, Murphy PR. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genomics. 2010;11:537–561. doi: 10.2174/138920210793175895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Pasquinelli AE. MicroRNAs and their targets: recognition, regula-tion and an emerging reciprocal relationship. Nat. Rev. Genet. 2012;13:271–282. doi: 10.1038/nrg3162. [DOI] [PubMed] [Google Scholar]
  • 9.Guo H, Ingolia NT, Weissman JS, Bartel DP. Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 2010;466:835–840. doi: 10.1038/nature09267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Pillai RS, Bhattacharyya SN, Filipowicz W. Repression of protein synthesis by miRNAs: how many mechanisms?. Trends Cell Biol. 2007;17:118–126. doi: 10.1016/j.tcb.2006.12.007. [DOI] [PubMed] [Google Scholar]
  • 11.Lim LP, Lau NC, Garrett-Engele P, Grimson A, Schelter JM, Castle J, Bartel DP, Linsley PS, Johnson JM. Microarray analy-sis shows that some microRNAs downregulate large numbers of target mRNAs. Nature. 2005;433:769–773. doi: 10.1038/nature03315. [DOI] [PubMed] [Google Scholar]
  • 12.Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat. Rev. Cancer. 2006;6:857–866. doi: 10.1038/nrc1997. [DOI] [PubMed] [Google Scholar]
  • 13.Lu J, Getz G, Miska eA, Alvarez-Saavedra e, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, Downing JR, Jacks T, Horvitz HR, Golub TR. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834–838. doi: 10.1038/nature03702. [DOI] [PubMed] [Google Scholar]
  • 14.Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio M, Roldo C, Ferracin M, Prueitt RL, Yanaihara N, Lanza G, Scarpa A, Vecchione A, Negrini M, Harris CC, Croce CM. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci U S A 15. 2006;103:2257–2261. doi: 10.1073/pnas.0510565103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Krutovskikh VA, Herceg Z. Oncogenic microRNAs (OncomiRs) as a new class of cancer biomarkers. Bioessays. 2010;32:894–904. doi: 10.1002/bies.201000040. [DOI] [PubMed] [Google Scholar]
  • 16.Miska eA. How microRNAs control cell division, differentiation and death. Curr. Opin. Genet. Dev. 2005;15:563–568. doi: 10.1016/j.gde.2005.08.005. [DOI] [PubMed] [Google Scholar]
  • 17.Lee YS, Kim HK, Chung S, Kim KS, Dutta A. Depletion of human miR-125b reveals that it is critical for the proliferation of differentiated cells but not for the down-regulation of putative targets during differentiation. J. Biol. Chem. 2005;280:6635–16641. doi: 10.1074/jbc.M412247200. [DOI] [PubMed] [Google Scholar]
  • 18.Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat. Genet. 2007;39:673–677. doi: 10.1038/ng2003. [DOI] [PubMed] [Google Scholar]
  • 19.Xu J, Wang Y, Tan X, Jing H. MicroRNAs in autophagy and their emerging roles in crosstalk with apoptosis. Autophagy. 2012;8:873–882. doi: 10.4161/auto.19629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kim DH, Saetrom P, Snøve O, Rossi JJ. MicroRNA-directed transcriptional gene silencing in mammalian cells. Proc. Natl. Acad. Sci. U.S.A. 2008;105:16230–16235. doi: 10.1073/pnas.0808830105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Place RF, Li LC, Pookot D, Noonane J, Dahiya R. MicroRNA-373 induces expression of genes with complementary promoter sequences. Proc. Natl. Acad. Sci. U.S.A. 2008;105:1608–1613. doi: 10.1073/pnas.0707594105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Suzuki K, Kelleher AD. Transcriptional regulation by promoter targeted RNAs. Curr. Top. Med. Chem. 2009;9:1079–1087. doi: 10.2174/156802609789630875. [DOI] [PubMed] [Google Scholar]
  • 23.Younger ST, Corey DR. Transcriptional regulation by miRNA mimics that target sequences downstream of gene termini. Mol. Biosyst. 2011;7:2383–2388. doi: 10.1039/c1mb05090g. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chang TC, Wentzele A, Kent OA, Ramachandran K, Mullendore M, Lee KH, Feldmann G, Yamakuchi M, Ferlito M, Lowenstein CJ, Arking DE, Beer MA, Maitra A, Mendell JT. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol. Cell. 2007;26:745–752. doi: 10.1016/j.molcel.2007.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sylvestre Y, De Guire V, Queridoe Mukhopadhyay UK, Bourdeau V, Major F, Ferbeyre G, Chartrand P. An E2F/miR-20a autoregulatory feedback loop. J. Biol. Chem. 2007;282:2135–2143. doi: 10.1074/jbc.M608939200. [DOI] [PubMed] [Google Scholar]
  • 26.He L, He X, Lim LP, de Stanchinae Xuan Z, Liang Y, Xue W, Zender L, Magnus J, Ridzon D, Jackson AL, Linsley PS, Chen C, Lowe SW, Cleary MA, Hannon GJ. A microRNA component of the p53 tumour suppressor network. Nature. 2007;447:1130–1134. doi: 10.1038/nature05939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Iguchi H, Kosaka N, Ochiya T. Versatile applications of microRNA in anti-cancer drug discovery: from therapeutics to bio-markers. Curr. Drug Discov. Technol. 2010;7:95–105. doi: 10.2174/157016310793180648. [DOI] [PubMed] [Google Scholar]
  • 28.Galasso M, Elena Sana M, Volinia S. Non-coding RNAs: a key to future personalized molecular therapy?. Genome Med. 2010;2:12. doi: 10.1186/gm133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, Stoffel M. Silencing of microRNAs in vivo with ‘antagomirs’. Nature. 2005;438:685–689. doi: 10.1038/nature04303. [DOI] [PubMed] [Google Scholar]
  • 30.Ebert MS, Neilson JR, Sharp PA. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nat. Methods. 2007;4:721–726. doi: 10.1038/nmeth1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ory B, Ellisen LW. A microRNA-dependent circuit controlling p63/p73 homeostasis: p53 family cross-talk meets therapeutic op-portunity. Oncotarget. 2011;2:259–264. doi: 10.18632/oncotarget.244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Yi R, Poy MN, Stoffel M. Fuchse A skin microRNA pro-motes differentiation by repressing 'stemness'. Nature. 2008;452:225–229. doi: 10.1038/nature06642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Knoufe C, Garg K, Arroyo JD, Correa Y, Sarkar D, Parkin RK, Wurz K, O'Briant KC, Godwin AK, Urban ND, Ruzzo WL, Gentleman R, Drescher CW, Swishere M, Tewari M. An integrative genomic approach identifies p73 and p63 as activators of miR-200 microRNA family transcription. Nucleic Acids Res. 2012;40:499–510. doi: 10.1093/nar/gkr731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Melar-New M, Laimins LA. Human papillomaviruses modulate expression of microRNA 203 upon epithelial differentiation to control levels of p63 proteins. J. Virol. 2010;84:5212–5221. doi: 10.1128/JVI.00078-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chikh A, Matin RN, Senatore V, Hufbauer M, Lavery D, Raimondi C, Ostano P, Mello-Grand M, Ghimenti C, Bahta A, Khalaf S, Akgu¨l B, Braun KM, Chiorino G, Philpott MP, Harwood CA, Bergamaschi D. iASPP/p63 autoregulatory feedback loop is required for the homeostasis of stratified epithelia. EMBO J. 2011;30:4261–4273. doi: 10.1038/emboj.2011.302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tucci P, Agostini M, Grespi F, Markerte K, Terrinoni A, Vousden KH, Muller PA, Dötsch V, Kehrloesser S, Sayan BS, Giaccone G, Lowe SW, Takahashi N, Vandenabeele P, Knight R, Levine AJ, Melino G. Loss of p63 and its microRNA-205 target results in enhanced cell migration and metastasis in prostate cancer. Proc. Natl. Acad. Sci. U. S. A. 2012;109:15312–15317. doi: 10.1073/pnas.1110977109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.McKenna DJ, McDade SS, Patel D, McCance DJ. MicroRNA 203 expression in keratinocytes is dependent on regula-tion of p53 levels by E6. J. Virol. 2010;84:10644–10652. doi: 10.1128/JVI.00703-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lena AM, Shalom-Feuerstein R, Rivetti di Val Cervo P, Aberdam D, Knight RA, Melino G, Candi e. miR-203 re-presses 'stemness' by repressing DeltaNp63. Cell Death Differ. 2008;15:1187–1195. doi: 10.1038/cdd.2008.69. [DOI] [PubMed] [Google Scholar]
  • 39.Liao JM, Zhou X, Zhang Y, Lu H. MiR-1246: a new link of the p53 family with cancer and Down syndrome. Cell Cycle. 2012;11:2624–2630. doi: 10.4161/cc.20809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Neilsen PM, Noll JE, Mattiske S, Bracken CP, Gregory PA, Schulz RB, Lim SP, Kumar R, Suetani RJ, Goodall GJ, Callen DF. Mutant p53 drives invasion in breast tumors through up-regulation of miR-155. Oncogene. 2013;32:2992–3000. doi: 10.1038/onc.2012.305. [DOI] [PubMed] [Google Scholar]
  • 41.Rivetti di Val Cervo P, Lena AM, Nicoloso M, Rossi S, Mancini M, Zhou H, Saintigny G, Dellambrae Odorisio T, Mahé C, Calin GA, Candie Melino G. p63-microRNA feedback in keratinocyte senescence. Proc. Natl. Acad. Sci. U. S. A. 2012;109:1133–1138. doi: 10.1073/pnas.1112257109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jackson SJ, Zhang Z, Feng D, Flagg M, O'Loughline Wang D, Stokes N, Fuchse Yi R. Rapid and widespread suppression of self-renewal by microRNA-203 during epidermal differentiation. Development. 2013;140:1882–1891. doi: 10.1242/dev.089649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Bailey SG, Sanchez-Elsner T, Stephanou A, Cragg MS, Townsend PA. Regulating the genome surveillance system: miRNAs and the p53 super family. Apoptosis. 2010;15:541–552. doi: 10.1007/s10495-010-0456-1. [DOI] [PubMed] [Google Scholar]
  • 44.Wu N, Sulpicee Obeid P, Benzina S, Kermarrec F, Combe S, Gidrol X. The miR-17 family links p63 protein to MAPK signaling to promote the onset of human keratinocyte differentiation. PLoS One. 2012;7:e45761. doi: 10.1371/journal.pone.0045761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Tran MN, Choi W, Wszolek MF, Navai N, Lee IL, Nitti G, Wen S, Florese R, Siefker-Radtke A, Czerniak B, Dinney C, Barton M, McConkey DJ. The p63 protein isoform ?Np63a inhibits epithelial-mesenchymal transition in human bladder cancer cells: role of MIR-205. J. Biol. Chem. 2013;288:3275–3288. doi: 10.1074/jbc.M112.408104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Antonini D, Russo MT, De Rosa L, Gorrese M, Del Vecchio L, Missero C. Transcriptional repression of miR-34 family contributes to p63-mediated cell cycle progression in epidermal cells. J. Invest. Dermatol. 2010;130:1249–1257. doi: 10.1038/jid.2009.438. [DOI] [PubMed] [Google Scholar]
  • 47.Bergholz J, Xiao ZX. Role of p63 in Development, Tumorigene-sis and Cancer Progression. Cancer Microenviron. 2012;5:311–322. doi: 10.1007/s12307-012-0116-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Huang Y, Sen T, Nagpal J, Upadhyay S, Trink B, Ratovitski e, Sidransky D. ATM kinase is a master switch for the DeltaNp63alpha phosphorylation/degradation in human head and neck squamous cell carcinoma cells upon DNA damage. Cell Cycle. 2008;7:2846–2855. doi: 10.4161/cc.7.18.6627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Huang Y, Chuang A, Hao H, Talbot C, Sen T, Trink B, Sidransky D, Ratovitski e. Phospho-?Np63a is a key regulator of the cisplatin-induced microRNAome in cancer cells. Cell Death Differ. 2011;18:1220–1230. doi: 10.1038/cdd.2010.188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Huang Y, Chuang AY, Ratovitski eA. Phospho-?Np63a/miR-885-3p axis in tumor cell life and cell death upon cisplatin exposure. Cell Cycle. 2011;10:3938–3947. doi: 10.4161/cc.10.22.18107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Huang Y, Guerrero-Preston R, Ratovitski eA. Phospho-?Np63a-dependent regulation of autophagic signaling through transcription and micro-RNA modulation. Cell Cycle. 2012;11:1247–1259. doi: 10.4161/cc.11.6.19670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Huang YP, Kesselman D, Kizub D, Guerrero-Preston R, Ratovitski eA. Tumor protein p63/ microRNA feedback regulation in squamous cell carcinomas upon cisplatin exposure. Cell Cycle. 2013;12:684–697. doi: 10.4161/cc.23598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ratovitski eA. Phospho-?Np63a-dependent microRNAs modulate chemoresistance of squamous cell carcinoma cells to cisplatin: At the crossroads of cell life and death. FEBS Lett. 2013;587:2536–2541. doi: 10.1016/j.febslet.2013.06.020. [DOI] [PubMed] [Google Scholar]
  • 54.Zheng B, Liang L, Wang C, Huang S, Cao X, Zha R, Liu L, Jia D, Tian Q, Wu J, Ye Y, Wang Q, Long Z, Zhou Y, Du C, He X, Shi Y. MicroRNA-148a suppresses tumor cell invasion and metastasis by downregulating ROCK1 in gastric cancer. Clin. Cancer Res. 2011;17:7574–7583. doi: 10.1158/1078-0432.CCR-11-1714. [DOI] [PubMed] [Google Scholar]
  • 55.Gonzalez S, Pisano DG, Serrano M. Mechanistic principles of chromatin remodeling guided by siRNAs and miRNAs. Cell Cycle. 2008;7:2601–2608. doi: 10.4161/cc.7.16.6541. [DOI] [PubMed] [Google Scholar]
  • 56.Cho H, Orphanides G, Sun X, Yang X, Ogryzko V, Leese Nakatani Y, Reinberg D. A human RNA polymerase II complex containing factors that modify chromatin structure. Mol. Cell. Biol. 1998;18:5355–5363. doi: 10.1128/mcb.18.9.5355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zhang Y, Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes and Dev. 2001;15:2343–2360. doi: 10.1101/gad.927301. [DOI] [PubMed] [Google Scholar]
  • 58.Chen K, Rajewsky N. The evolution of gene regulation by tran-scription factors and micro-RNAs. Nat. Rev. Genet. 2007;8:93–103. doi: 10.1038/nrg1990. [DOI] [PubMed] [Google Scholar]
  • 59.Cloos PA, Christensen J, Agger K, Helin K. Erasing the methyl mark: histone demethylases at the center of cellular differentiation and disease. Genes and Dev. 2008;22:1115–1140. doi: 10.1101/gad.1652908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Müller J, Verrijzer P. Biochemical mechanisms of gene regulation by polycomb group protein complexes. Curr. Opin. Genet. Dev. 2009;19:150–158. doi: 10.1016/j.gde.2009.03.001. [DOI] [PubMed] [Google Scholar]
  • 61.Zeng X, Chen S, Huang H. Phosphorylation of EZH2 by CDK1 and CDK2: a possible regulatory mechanism of transmis-sion of the H3K27me3 epigenetic mark through cell divisions. Cell Cycle. 2011;10:579–583. doi: 10.4161/cc.10.4.14722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ramsey MR, He L, Forster N, Ory B, Ellisen LW. Physical association of HDAC1 and HDAC2 with p63 mediates transcriptional repression and tumor maintenance in squamous cell carcinoma. Cancer Res. 2011;71:4373–4379. doi: 10.1158/0008-5472.CAN-11-0046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gallant-Behm CL, Ramsey MR, Bensard CL, Nojek I, Tran J, Liu M, Ellisen LW, Espinosa JM. ?Np63a represses anti-proliferative genes via H2A. deposition. Genes and Dev. 2012;26:2325–2236. doi: 10.1101/gad.198069.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hatziapostolou M, Iliopoulos D. Epigenetic aberrations during oncogenesis. Cell. Mol. Life Sci. 2011;68:1681–702. doi: 10.1007/s00018-010-0624-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Berger SL. Histone modifications in transcriptional regulation. Curr. Opin. Genet. Dev. 2002;12:142–148. doi: 10.1016/s0959-437x(02)00279-4. [DOI] [PubMed] [Google Scholar]
  • 66.Margueron R, Trojer P, Reinberg D. The key to development: interpreting the histone code? Curr. Opin. Genet. Dev. 2005;15:163–176. doi: 10.1016/j.gde.2005.01.005. [DOI] [PubMed] [Google Scholar]
  • 67.Nightingale KP, O'Neill LP, Turner BM. Histone modifications: signalling receptors and potential elements of a heritable epigenetic code. Curr. Opin. Genet. Dev. 2006;16:125–136. doi: 10.1016/j.gde.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 68.Guil S, Esteller M. DNA methylomes, histone codes and miRNAs: tying it all together. Int. J. Biochem. Cell Biol. 2009;41:87–95. doi: 10.1016/j.biocel.2008.09.005. [DOI] [PubMed] [Google Scholar]
  • 69.Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150:12–27. doi: 10.1016/j.cell.2012.06.013. [DOI] [PubMed] [Google Scholar]
  • 70.Sandoval J, Esteller M. Cancer epigenomics: beyond genomics. Curr. Opin. Genet. Dev. 2012;22:50–55. doi: 10.1016/j.gde.2012.02.008. [DOI] [PubMed] [Google Scholar]
  • 71.Lopez-Serra P, Esteller M. DNA methylation-associated silencing of tumor-suppressor microRNAs in cancer. Oncogene. 2012;31:1609–1622. doi: 10.1038/onc.2011.354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Roccaro AM, Sacco A, Jia X, Azab AK, Maiso P, Ngo HT, Azab F, Runnels J, Quang P, Ghobrial IM. mi-croRNA-dependent modulation of histone acetylation in Waldenstrom macroglobulinemia. Blood. 2010;116:1506–1514. doi: 10.1182/blood-2010-01-265686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lujambio A, Calin GA, Villanueva A, Ropero S, Sánchez-Céspedes M, Blanco D, Montuenga LM, Rossi S, Nicoloso MS, Faller WJ, Gallagher WM, Eccles SA, Croce CM, Esteller M. A microRNA DNA methylation signature for human cancer metastasis. Proc. Natl. Acad. Sci. U. S. A. 2008;105:13556–13561. doi: 10.1073/pnas.0803055105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Calle-garie Liu S, Alder H, Costinean S, Fernandez-Cymering C, Volinia S, Guler G, Morrison CD, Chan KK, Marcucci G, Calin GA, Huebner K, Croce CM. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc. Natl. Acad. Sci. U. S. A. 2007;104:15805–15810. doi: 10.1073/pnas.0707628104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Garzon R, Liu S, Fabbri M, Liu Z, Heaphy CE, Callegarie Schwind S, Pang J, Yu J, Muthusamy N, Have-lange V, Volinia S, Blum W, Rush LJ, Perrotti D, Andreeff M, Bloomfield CD, Byrd JC, Chan K, Wu LC, Croce CM, Marcucci G. MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1. Blood. 2009;113:6411–6418. doi: 10.1182/blood-2008-07-170589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Duursma AM, Kedde M, Schrier M, le Sage C, Agami R. miR-148 targets human DNMT3b protein coding region. RNA. 2008;14:872–877. doi: 10.1261/rna.972008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Xu Q, Jiang Y, Yin Y, Li Q, He J, Jing Y, Qi YT, Xu Q, Li W, Lu B, Peiper SS, Jiang BH, Liu LZ. A regu-latory circuit of miR-148a/152 and DNMT1 in modulating cell transformation and tumor angiogenesis through IGF-IR and IRS1. J. Mol. Cell Biol. 2013;5:3–13. doi: 10.1093/jmcb/mjs049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Takata A, Otsuka M, Yoshikawa T, Kishikawa T, Hikiba Y, Obi S, Goto T, Kang YJ, Maeda S, Yoshida H, Omata M, Asahara H, Koike K. MiRNA-140 acts as a liver tumor suppressor by controlling NF-?B activity via directly targeting Dnmt1 expres-sion. Hepatology. 2013;57:162–170. doi: 10.1002/hep.26011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wang YS, Chou WW, Chen KC, Cheng HY, Lin RT, Juo SH. MicroRNA-152 mediates DNMT1-regulated DNA me-thylation in the estrogen receptor a gene. PLoS One. 2012;7:e30635. doi: 10.1371/journal.pone.0030635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B, Laxman B, Cao X, Jing X, Ramnarayanan K, Brenner JC, Yu J, Kim JH, Han B, Tan P, Kumar-Sinha C, Lonigro RJ, Palanisamy N, Maher CA, Chinnaiyan AM. Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science. 2008;322:1695–1699. doi: 10.1126/science.1165395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhang JG, Guo JF, Liu DL, Liu Q, Wang JJ. MicroRNA-101 exerts tumor-suppressive functions in non-small cell lung cancer through directly targeting enhancer of zeste homolog 2. J. Thorac. Oncol. 2011;6:671–678. doi: 10.1097/JTO.0b013e318208eb35. [DOI] [PubMed] [Google Scholar]
  • 82.Noonane J, Place RF, Pookot D, Basak S, Whitson JM, Hirata H, Giardina C, Dahiya R. miR-449a targets HDAC-1 and induces growth arrest in prostate cancer. Oncogene. 2009;28:1714–1724. doi: 10.1038/onc.2009.19. [DOI] [PubMed] [Google Scholar]
  • 83.Chen B, Li H, Zeng X, Yang P, Liu X, Zhao X, Liang S. Roles of microRNA on cancer cell metabolism. J. Transl. Med. 2012;10:228. doi: 10.1186/1479-5876-10-228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Biggar KK, Storey KB. The emerging roles of microRNAs in the molecular responses of metabolic rate depression. J. Mol. Cell Biol. 2011;3:167–175. doi: 10.1093/jmcb/mjq045. [DOI] [PubMed] [Google Scholar]
  • 85.Tibiche C Wange. MicroRNA regulatory patterns on the human metabolic network. Open Syst. Biol. J. 2008;1:1–8. [Google Scholar]
  • 86.Godlewski J, Nowicki MO, Bronisz A, Nuovo G, Palatini J, De Lay M, Van Brocklyn J, Ostrowski MC, Chioccae A, Lawler SE. MicroRNA-451 regulates LKB1/AMPK signaling and allows adaptation to metabolic stress in glioma cells. Mol. Cell. 2010;37:620–632. doi: 10.1016/j.molcel.2010.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dávalos A, Goedeke L, Smibert P, Ramírez CM, Warrier NP, Andreo U, Cirera-Salinas D, Rayner K, Suresh U, Pastor-Pareja JC, Espluguese Fishere A, Penalva LO, Moore KJ, Suárez Y, Laie C, Fernández-Hernando C. miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc. Natl. Acad. Sci. U. S. A. 2011;108:9232–9237. doi: 10.1073/pnas.1102281108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wilfred BR, Wang WX, Nelson PT. Energizing miRNA research: a review of the role of miRNAs in lipid metabolism, with a prediction that miR-103/107 regulates human metabolic pathways. Mol. Genet. Metab. 2007;91:209–217. doi: 10.1016/j.ymgme.2007.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Maddocks OD, Vousden KH. Metabolic regulation by p53. J. Mol. Med. (Berl) 2011;89:237–245. doi: 10.1007/s00109-011-0735-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kim HR, Roe JS, Lee JE, Hwang IY, Choe J, Youn HD. A p53-inducible microRNA-34a downregulates Ras signaling by targeting IMPDH. Biochem. Biophys. Res. Commun. 2012;418:682–688. doi: 10.1016/j.bbrc.2012.01.077. [DOI] [PubMed] [Google Scholar]
  • 91.Kim HR, Roe JS, Lee JE, Choe J, Youn HD. p53 regulates glucose metabolism by miR-34a. Biochem. Biophys. Res. Commun. 2013;437:225–231. doi: 10.1016/j.bbrc.2013.06.043. [DOI] [PubMed] [Google Scholar]
  • 92.Jeon TI, Osborne TF. SREBPs: metabolic integrators in physiology and metabolism. Trends Endocrinol. Metab. 2012;23:65–72. doi: 10.1016/j.tem.2011.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.He XP, Shao Y, Li XL, Xu W, Chen GS, Sun HH, Xu HC, Xu X, Tang D, Zheng XF, Xue YP, Huang GC, Sun WH. Downregulation of miR-101 in gastric cancer correlates with cyclooxygenase-2 overexpression and tumor growth. FEBS J. 2012;279:4201–4212. doi: 10.1111/febs.12013. [DOI] [PubMed] [Google Scholar]
  • 94.Li X, Chen YT, Josson S, Mukhopadhyay NK, Kim J, Freeman MR, Huang WC. MicroRNA-185 and 342 Inhibit Tumorigenicity and Induce Apoptosis through Blockade of the SREBP Metabolic Pathway in Prostate Cancer Cells. Plo. One;2013, 8:e70987. doi: 10.1371/journal.pone.0070987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Huang YP, Bell LN, Okamura J, Kim MS, Mohney RP, Guerrero-Preston R, Ratovitskie A. Phospho-?Np63?/SREBF1 interaction: Bridging Cell Metabolism and Cisplatin Resistance. Cell Cycle. 2012;11:3810–3827. doi: 10.4161/cc.22022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mangala LS, Zuzel V, Schmandt R, Leshanee S, Halder JB, Armaiz-Pena GN, Spannuth WA, Tanaka T, Shahzad MM, Lin YG, Nick AM, Danes CG, Lee JW, Jennings NB, Vivas-Mejia PE, Wolf JK, Coleman RL, Siddik ZH, Lopez-Berestein G, Lutsenko S, Sood AK. Therapeutic Targeting of ATP7B in Ovarian Carcinoma. Clin. Cancer Res. 2009;15:3770–3780. doi: 10.1158/1078-0432.CCR-08-2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Sachdeva M, Wu H, Ru P, Hwang L, Trieu V, Mo YY. MicroRNA-101-mediated Akt activation and estrogen-independent growth. Oncogene. 2011;30:822–831. doi: 10.1038/onc.2010.463. [DOI] [PubMed] [Google Scholar]
  • 98.Su H, Yang JR, Xu T, Huang J, Xu L, Yuan Y, Zhuang SM. MicroRNA-101, downregulated in hepatocellular carcinoma, promotes apoptosis and suppresses tumorigenicity. Cancer Res. 2009;69:1135–1142. doi: 10.1158/0008-5472.CAN-08-2886. [DOI] [PubMed] [Google Scholar]
  • 99.Canavese M, Santo L, Raje N. Cyclin dependent kinases in cancer: potential for therapeutic intervention. Cancer Biol. Ther. 2012;13:451–457. doi: 10.4161/cbt.19589. [DOI] [PubMed] [Google Scholar]
  • 100.Medema RH, Macurek L. Checkpoint control and cancer. Oncogene. 2012;31:2601–2613. doi: 10.1038/onc.2011.451. [DOI] [PubMed] [Google Scholar]
  • 101.Lizé M, Klimke A, Dobbelstein M. MicroRNA-449 in cell fate determination. Cell Cycle. 2011;10:2874–2882. doi: 10.4161/cc.10.17.17181. [DOI] [PubMed] [Google Scholar]
  • 102.Kim YK, Yu J, Han TS, Park SY, Namkoong B, Kim DH, Hur K, Yoo MW, Lee HJ, Yang HK, Kim VN. Functional links between clustered microRNAs: suppression of cell-cycle inhibitors by microRNA clusters in gastric cancer. Nucl. Acid Res. 2009;37:1672–1681. doi: 10.1093/nar/gkp002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang Y, Blelloch R. Cell cycle regulation by microRNAs in stem cells. Results Probl. Cell Differ. 2011;53:459–472. doi: 10.1007/978-3-642-19065-0_19. [DOI] [PubMed] [Google Scholar]
  • 104.Walensky L. BCL-2 in the crosshairs: tipping the balance of life and death. Cell Death Differ. 2006;13:1339–1350. doi: 10.1038/sj.cdd.4401992. [DOI] [PubMed] [Google Scholar]
  • 105.Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat. Rev. Mol. Cell Biol. 2008;9:47–59. doi: 10.1038/nrm2308. [DOI] [PubMed] [Google Scholar]
  • 106.Kumar S. Caspase function in programmed cell death. Cell Death Differ. 2007;14:32–43. doi: 10.1038/sj.cdd.4402060. [DOI] [PubMed] [Google Scholar]
  • 107.Melino G, Knight R. MicroRNAs meet cell death. Cell Death Differ. 2010;17:189–190. doi: 10.1038/cdd.2009.122. [DOI] [PubMed] [Google Scholar]
  • 108.Galluzzi L, Morsellie Vitale I, Kepp O, Senovilla L, Criollo A, Servant N, Paccard C, Hupé P, Robert T, Ripoche H, Lazar V, Harel-Bellan A, Dessen P, Barillote Kroemer G. MiR-181a and miR-630 Regulate Cisplatin-Induced Cancer Cell Death. Cancer Res. 2010;70:1793–1804. doi: 10.1158/0008-5472.CAN-09-3112. [DOI] [PubMed] [Google Scholar]
  • 109.Liu S, Zhang P, Chen Z, Liu M, Li X, Tang H. MicroRNA-7 downregulates XIAP expression to suppress cell growth and promote apoptosis in cervical cancer cells. FEBS Lett. 2013;587:2247–2253. doi: 10.1016/j.febslet.2013.05.054. [DOI] [PubMed] [Google Scholar]
  • 110.Song T, Zhang X, Zhang L, Dong J, Cai W, Gao J, Hong B. miR-708 promotes the development of bladder carcinoma via direct repression of Caspase-2. J. Cancer Res. Clin. Oncol. 2013;139:1189–1198. doi: 10.1007/s00432-013-1392-6. [DOI] [PubMed] [Google Scholar]
  • 111.Zang YS, Zhong YF, Fang Z, Li B, An J. MiR-155 inhibits the sensitivity of lung cancer cells to cisplatin via negative regulation of Apaf-1 expression. Cancer Gene. Ther. 2012;19:773–778. doi: 10.1038/cgt.2012.60. [DOI] [PubMed] [Google Scholar]
  • 112.Xie Y, Tobin LA, Camps J, Wangsa D, Yang J, Rao M, Witaspe Awad KS, Yoo N, Ried T, Kwong KF. MicroRNA-24 regulates XIAP to reduce the apoptosis threshold in cancer cells. Onc. gene;2013, 32:2442–2451. doi: 10.1038/onc.2012.258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, Wojcik SE, Aqeilan RI, Zupo S, Dono M, Rassenti L, Alder H, Volinia S, Liu CG, Kipps TJ, Negrini M, Croce C. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. U. S. A. 2005;102:13944–13949. doi: 10.1073/pnas.0506654102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Xu J, Liao X, Wong C. Downregulations of B-cell lymphoma 2 and myeloid cell leukemia sequence 1 by microRNA 153 induce apoptosis in a glioblastoma cell line DBTRG-05MG. Int. J. Cancer. 2010;126:1029–1035. doi: 10.1002/ijc.24823. [DOI] [PubMed] [Google Scholar]
  • 115.Qiu T, Zhou L, Wang T, Xu J, Wang J, Chen W, Zhou X, Huang Z, Zhu W, Shu Y, Liu P. miR-503 regulates the resistance of non-small cell lung cancer cells to cisplatin by targeting Bcl-2. Int. J. Mol. Med. 2013;32:593–598. doi: 10.3892/ijmm.2013.1439. [DOI] [PubMed] [Google Scholar]
  • 116.Kwon JE, Kim BY, Kwak SY, Bae IH, Han YH. Ionizing radiation-inducible microRNA miR-193a-3p induces apoptosis by directly targeting Mcl-1. Apoptosis. 2013;18:896–909. doi: 10.1007/s10495-013-0841-7. [DOI] [PubMed] [Google Scholar]
  • 117.Ji F, Zhang H, Wang Y, Li M, Xu W, Kang Y, Wang Z, Wang Z, Cheng P, Tong D, Li C, Tang H. MicroRNA-133a, downregulated in osteosarcoma, suppresses proliferation and promotes apoptosis by targeting Bcl-xL and Mcl-1. Bone. 2013;56:220–226. doi: 10.1016/j.bone.2013.05.020. [DOI] [PubMed] [Google Scholar]
  • 118.Gao J, Li L, Wu M, Liu M, Xie X, Guo J, Tang H, Xie X. MiR-26a inhibits proliferation and migration of breast cancer through repression of MCL-1. PLos One. 2013;8:e65138. doi: 10.1371/journal.pone.0065138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Verdoodt B, Neid M, Vogt M, Kuhn V, Liffers ST, Palisaar RJ, Noldus J, Tannapfel A. Mirmohammadsadegh, A. MicroRN -205;a novel regulator of the anti-apoptotic protein Bcl2, is downregulated in prostate cancer. Int. J. Oncol. 2013, 43, 307-314. cell survival and enhances cisplatin-induced cytotoxicity via down-regulation of Bcl2l2 in cervical cancer cells. FEBS Lett. 2013, 587:488–495. doi: 10.3892/ijo.2013.1915. [DOI] [PubMed] [Google Scholar]
  • 120.Chio CC, Lin JW, Cheng HA, Chiu WT, Wang YH, Wang JJ, Hsing CH, Chen RM. MicroRNA-210 targets an-tiapoptotic Bcl-2 expression and mediates hypoxia-induced apoptosis of neuroblastoma cells. Arch. Toxicol. 2013;87:459–468. doi: 10.1007/s00204-012-0965-5. [DOI] [PubMed] [Google Scholar]
  • 121.Gong J, Zhang JP, Li B, Zeng C, You K, Chen MX, Yuan Y, Zhuang SM. MicroRNA-125b promotes apoptosis by regulating the expression of Mcl-1, Bcl-w and IL-6R. Oncogene. 2013;32:3071–3079. doi: 10.1038/onc.2012.318. [DOI] [PubMed] [Google Scholar]
  • 122.Garofalo M, Jeon YJ, Nuovo GJ, Middleton J, Secchiero P, Joshi P, Alder H, Nazaryan N, Di Leva G, Romano G, Crawford M, Nana-Sinkam P, Croce CM. MiR-34a/c-Dependent PDGFR-a/ß Downregulation Inhibits Tumorigenesis and Enhances TRAIL-Induced Apoptosis in Lung Cancer. Plos One. 2013;8:e67581. doi: 10.1371/journal.pone.0067581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lu T, Shao N, Ji C. Targeting microRNAs to modulate TRAIL-induced apoptosis of cancer cells. Cancer Gene Ther. 2013;20:33–37. doi: 10.1038/cgt.2012.81. [DOI] [PubMed] [Google Scholar]
  • 124.Patron JP, Fendler A, Bild M, Jung U, Müller H, Arntzen M , Piso C, Stephan C, Thiede B, Mollenkopf HJ, Jung K, Kaufmann SH, Schreiber J. MiR-133b targets antiapoptotic genes and enhances death receptor-induced apoptosis. PloS One. 2012;7:e35345. doi: 10.1371/journal.pone.0035345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Bertero T, Bourget-Ponzio I, Puissant A, Loubat A, Mari B, Meneguzzi G, Auberger P, Barbry P, Ponzio G, Rezzonico R. Tumor suppressor function of miR-483-3p on squamous cell carcinomas due to its pro-apoptotic properties. Cell Cycle. 2013;12:2183–2193. doi: 10.4161/cc.25330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Frankel LB, Lund AH. MicroRNA regulation of autophagy. Carcinogenesis. 2012;3:2018–2025. doi: 10.1093/carcin/bgs266. [DOI] [PubMed] [Google Scholar]
  • 127.Zhu H, Wu H, Liu X, Li B, Chen Y, Ren X, Liu CG, Yang JM. Regulation of autophagy by a beclin 1-targeted mi-croRNA miR-30a in cancer cells. Autophagy. 2009;5:816–823. doi: 10.4161/auto.9064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Frankel LB, Wen J, Lees M, Høyer-Hansen M, Farkas T, Krogh A, Jäättelä M, Lund AH. microRNA-101 is a potent in-hibitor of autophagy. EMBO J. 2011;30:4628–4641. doi: 10.1038/emboj.2011.331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, Bray K, Reddy A, Bhanot G, Gelinas C, Di-paola RS, Karantza-Wadsworth V, White e. Autophagy suppresses tumorigenesis through elimination of p62. Cell. 2009;137:1062–1075. doi: 10.1016/j.cell.2009.03.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Korkmaz G, le Sage C, Tekirdag KA, Agami R, Gozuacik D. miR-376b controls starvation and mTOR inhibition-related autophagy by targeting ATG4C and BECN1. Autophagy. 2012;8:165–176. doi: 10.4161/auto.8.2.18351. [DOI] [PubMed] [Google Scholar]
  • 131.Fu LL, Wen X, Bao JK, Liu B. MicroRNA-modulated autophagic signaling networks in cancer. Int. J. Biochem. Cell Biol. 2012;44:733–736. doi: 10.1016/j.biocel.2012.02.004. [DOI] [PubMed] [Google Scholar]
  • 132.Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132:27–42. doi: 10.1016/j.cell.2007.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Kroemer G, Mariño G, Levine B. Autophagy and the integrated stress response. Mol. Cell. 2010;40:280–293. doi: 10.1016/j.molcel.2010.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Moscat J, Diaz-Meco MT. p62 at the crossroads of autophagy, apoptosis, and cancer. Cell. 2009;137:1001–1004. doi: 10.1016/j.cell.2009.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Jegga AG, Schneider L, Ouyang X, Zhang J. Systems biology of the autophagy-lysosomal pathway. Autophagy. 2011;7:477–489. doi: 10.4161/auto.7.5.14811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Xu Y, An Y, Wang Y, Zhang C, Zhang H, Huang C, Jiang H, Wang X, Li X. miR-101 inhibits autophagy and en-hances cisplatin-induced apoptosis in hepatocellular carcinoma cells. Oncol. Rep. 2013;29:2019–2024. doi: 10.3892/or.2013.2338. [DOI] [PubMed] [Google Scholar]
  • 137.Frankel LB, Wen J, Lees M, Høyer-Hansen M, Farkas T, Krogh A, Jäättelä M, Lund AH. microRNA-101 is a potent in-hibitor of autophagy. EMBO J. 2011;30:4628–4641. doi: 10.1038/emboj.2011.331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Maiuri MC, Le Toumelin G, Criollo A, Rain JC, Gautier F, Juin P, Tasdemire Pierron G, Troulinaki K, Taver-narakis N, Hickman JA, Geneste O, Kroemer G. Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J. 2007;26:2527–2539. doi: 10.1038/sj.emboj.7601689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Germain M, Nguyen AP, Le Grand JN, Arbour N, Vanderluit JL, Park DS, Opferman JT, Slack RS. MCL-1 is a stress sensor that regulates autophagy in a developmentally regulated manner. EMBO J. 2011;30:395–407. doi: 10.1038/emboj.2010.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Luo S, Rubinsztein DC. Apoptosis blocks Beclin 1-dependent autophagosome synthesis: an effect rescued by Bcl-xL. Cell Death Differ. 2010;17:268–277. doi: 10.1038/cdd.2009.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Yousefi S, Simon HU. Apoptosis regulation by autophagy gene 5. Crit. Rev. Oncol. Hematol. 2007 63 241-244. critrevonc. 2007;06:2005. doi: 10.1016/j.critrevonc.2007.06.005. [DOI] [PubMed] [Google Scholar]
  • 142.Yousefi S, Perozzo R, Schmid I, Ziemiecki A, Schaffner T, Scapozza L, Brunner T, Simon HU. Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat. Cell Biol. 2006;8:1124–1132. doi: 10.1038/ncb1482. [DOI] [PubMed] [Google Scholar]
  • 143.Hanahan D, Weinberg RA. Hallmarks of cancer: the next genera-tion. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 144.Zhu A, Xia J, Zuo J, Jin S, Zhou H, Yao L, Huang H, Han Z. MicroRNA-148a is silenced by hypermethylation and interacts with DNA methyltransferase 1 in gastric cancer. Med. Oncol. 2012;29:2701–2709. doi: 10.1007/s12032-011-0134-3. [DOI] [PubMed] [Google Scholar]

Articles from Current Genomics are provided here courtesy of Bentham Science Publishers

RESOURCES