Epigenetic roles of PIWI‑interacting RNAs (piRNAs) in cancer metastasis (Review)

  • Authors:
    • Jia Liu
    • Shujun Zhang
    • Binglin Cheng
  • View Affiliations

  • Published online on: September 6, 2018     https://doi.org/10.3892/or.2018.6684
  • Pages: 2423-2434
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

P‑element‑induced wimpy testis (PIWI)‑interacting RNAs (piRNAs) are epigenetic‑related short ncRNAs that participate in chromatin regulation, transposon silencing, and modification of specific gene sites. These epigenetic factors or alterations are also involved in the growth of a variety of human cancers, including lung, breast, and colon cancer. Accumulating evidence has revealed that tumor metastasis and invasion involve genetic and epigenetic factors. Cancer metastasis is characterized by epigenetic alterations including DNA methylation and histone modification. Changes in DNA methylation, H3K9me3 heterochromatin and transposable elements have been detected in several cancers. piRNAs may function in gene silencing and gene modification upstream or downstream of oncogenes in cancer cell lines or cancer tissues. In addition to piRNAs, PIWI proteins can be used as biomarkers for prognosis, diagnosis and clinical evaluation and may be factors in cancer metastasis. Here, we elucidated the possible mechanisms by which piRNAs regulate cancer metastasis, including but not restricted to influencing DNA and histone methylation and transposable elements.

Introduction

P-element-induced wimpy testis (PIWI)-interacting RNAs (piRNAs) belong to a new class of ncRNAs that have been associated with many cancers (1). piRNAs are involved in the gene regulation process in which certain nucleotides bind coding regions in gene promoters (2). piRNAs function in the epigenetic regulation of DNA methylation (3), transposable silencing and chromatin modification (4). PIWI is a type of Argonaute protein that binds to piRNAs and carries out unique functions in somatic and germ cells, and stably expressed piRNAs have also been detected in human blood (5). piRNAs may be used as biomarkers for cancer diagnosis (6). Four PIWI proteins have been discovered in humans: PIWI1, PIWI2, PIWI3, and PIWI4 (710). Furthermore, PIWI expression levels are associated with different types of cancers and clinical stages (1113). Additionally, piRNAs have been linked to proliferation, apoptosis (14), genomic instability (15), invasion, and metastasis (16) in cancer cells. The levels of PIWI and piRNAs were revealed to be significantly altered between tumor tissues and non-tumor tissues. The clinical pathological features of tumors are associated with PIWI and piRNA expression. Therefore, additional studies are needed to understand the role of piRNAs in cancer and their epigenetic mechanisms and to shed light on the potential of piRNAs for the diagnosis and prediction of clinical cancer stages.

Cancer formation involves genetic, epigenetic, and pathological mechanisms (17). Cancer is a complicated disease with distinct genetic, epigenetic and pathological features (18). Despite recent advancements in precision medicine, the assessment of pathological and clinical features remains the primary and most accurate method for diagnosing carcinoma. Preventing tumor metastasis is still a formidable problem in the world. Carcinogenesis involves genetic machineries that are present beginning in early childhood but are altered over time. Abnormal and inappropriate epigenetic alterations can regulate carcinoma development (19). Activation of oncogenes and inactivation of tumor suppressors or other cell factors and pathways cannot fully account for metastatic cancers. Under certain circumstances, epigenetic mechanisms related to piRNAs may be responsible for the downstream inactivation of genes in tumors (20). Epigenetic modifications alter gene expression rather than changing the DNA sequence (21). These alterations are based on DNA methylation and histone protein modification, and non-coding RNAs (ncRNAs) frequently participate in this process.

Epigenetic functions of PIWI and piRNAs

piRNAs play crucial roles in protecting genomic stability by inhibiting transposon activity and maintaining minimum levels of transposons in germ line, mammalian cells and other cell types (22). piRNAs reside in clusters within heterochromatin-euchromatin boundaries and exhibit repeat-rich regions with ancient fragmented transposon copies. Amplification of piRNAs occurs through the ‘ping-pang’ cycle (23). This cycle is initiated by the emergence of a primary piRNA from piRNA clusters. Primary piRNAs are antisense sequences to expressed transposons and have the ability to cleave their targets, while secondary piRNAs form the AGO3 complex. The AGO3-bound piRNAs interact with transposon targets, which include antisense transposon sequences. This interaction then produces antisense piRNAs, and the ‘ping-pang’ cycle continues.

PIWI proteins have been reported to be selectively expressed in precancerous stem cells, tumor cell lines and cells of various malignancies (24). PIWI and Aubergine (Aub) proteins accumulate in the pole plasm and transfer maternal piRNAs into germ line cells (25). It was previously believed that PIWI proteins could maintain genomic integrity in animal germ cells by silencing transposons. Without these proteins, piRNAs cannot inactivate transposons within a single generation. In mammals, the PIWI proteins MILI, MIWI2 and intracisternal A-particle (IAP) act as retrotransposons and function as transposon-inhibiting factors via transposon gene silencing (TGS) (26). MILI is active in the cytosol, and MIWI2 is active in the nucleus (27). MILI, also known as PIWIL2, and MIWI2, also known as PIWIL4, suppress transposons in the cytoplasm and nucleus. MIWI is a slicer similar to Argonaute family members, which include PIWI proteins. The function of the slicer MIWI depends on its binding motif, which has a conserved Argonaute domain sequence (27). A high degree of MIWI complementarity is required for piRNA targeting. MIWI-associated piRNAs endonucleolytically cleave their target RNAs. Therefore, silencing of transposons depends on the piRNAs involved. Using next-generation sequencing, we can analyze histone modifications and methylation across entire genomes. Moreover, we can identify ncRNAs that participate in these regulatory processes. This technology provides a link between epigenetic modifications and transcription (28).

Epigenetic functions of specific transposable elements

Transcriptional silencing, heterochromatin formation, transgene silencing, HP1α alterations, histone modifications and transposon suppression are all associated with PIWI, piRNAs, the piRNA pathway and specific transposable elements (29).

As mediators of eukaryotic evolution, transposable elements are classified as either retrotransposons or DNA transposons. Notably, retrotransposons can cause genomic variations (30), alter chromatin structures and change the expression of nearby genes by integrating into genomic locations. Retrotransposons include long terminal repeat (LTR) and non-LTR retrotransposons. LTRs are similar to retroviruses in their structure, and non-LTRs are similar to mRNAs (31).

LTRs can encode structural proteins to form virus-like particles (VLPs), which can regulate gene transcription. RNA derived from LTRs can be reverse-transcribed into cDNA and can thereby integrate into the genome (32). RNA polymerase II located at the 5′end of LTRs can transcribe LTRs. These RNA molecules are then packaged into viral particles and use the reverse transcription machinery to generate full-length DNA. The first priming event occurs between the 5′end and the 3′end of LTR, and the second priming event occurs near the 3′end of LTR leading to production of a double-stranded cDNA molecule through an additional strand transfer. Thus, the cDNA is integrated into the host genome. The main difference between retrotransposons and infectious retroviruses is the presence of the envelope (env) gene in the latter, which enables viruses to infect other cells. In contrast, exon open reading frames (ORFs) are found in retrotransposons and retroviral genomes (33).

Non-LTRs are classified as either autonomous non-LTRs, such as LINEs, or non-autonomous LTRs, such as short interspersed nuclear elements (SINEs) (34). Retrotransposition-competent LINE1 includes a 5′-untranslated region (UTR) that is rich in CpG-islands; ORF1, which binds to RNA and ORF2, which encodes proteins such as endonuclease, reverse transcriptase and cysteine-histidine-rich domains. Similar to mammalian RNAs, LINE1 RNA has a poly (A) tail at the 3′-UTR. RNA polymerase II transcribes ORF1 and ORF2 in the cytoplasm (35).

Mutations are generated during the process of evolutionary change. Proofreading polymerases repair damaged DNA sequences and eliminate potential mutations. However, some piRNAs can also mediate the activity of transposable elements (36). HP1, a chromatin-organizing protein, can affect transposon activity by regulating piRNA expression or by directly mediating the expression of transposons. We can therefore conclude that some chromatin-organizing proteins, such as HP1, act upstream or downstream of piRNAs to regulate transposons. Mutations in Aub, PIWI, and Su(var)205 are known to increase the activity of transposable elements in germline cells (37,38) (Fig. 1).

Epigenetic functions of specific piRNA pathway proteins

Uncontrolled transposons threaten genomic integrity, and these alterations can be transferred to the next generation. piRNAs bind with their partner PIWI to recognize and silence transposable elements in germ cells. Both cytoplasmic and nuclear PIWI proteins target the genome to mediate transcriptional silencing. In mice, transposon inhibition by piRNAs occurs via DNA methylation at CpG islands in the sequences of transposable elements. During this process, the piRNA pathway mediates and maintains high levels of the repressive H3K9me3 mark in LINE regions in germ cells. Furthermore, piRNAs recognize full-length elements of the actively transposing LINE family (39).

Scientists recently identified two piRNA pathway proteins that are related to transposon silencing (40). The first, CG9754, is a downstream piRNA pathway factor that participates in the nuclear PIWI-piRNA complex involved in transcriptional silencing and heterochromatin formation. CG9754 is the first protein to target RNA or DNA, in heterochromatin and transcriptional silencing (4143). PIWI is only able to silence downstream proteins if it is bound to piRNAs that are engaged with target genes (4446). Recruitment of CG9754 directly to DNA or indirectly to RNA results in potent transcriptional silencing. Thus, CG9754 is sufficient to induce transcriptional silencing by binding to RNA. In ovarian somatic cells (OSCs), CG9754 is a downstream protein of PIWI (4749). When vectors were used to delete CG9754, a decrease in the level of H3K9me3 was observed, and the insertion of transposable elements into transcriptional genomic regions was repressed. CG9754 mediates heterochromatin formation and integrates other cell factors, such as HP1α, which induces transcriptional silencing. SetDB1 is downstream of HP1α and CG9754, an important HMT effector (5052). EXD1 is the second important piRNA pathway protein. MILI slicing acts as a switch that initiates piRNA processing. Slicing activates different types of primary piRNAs in MILI and MIWI. EXD1 is a component of the PET (PIWI-EXD1-TDRD12) complex, which mediates transcriptional gene silencing (53).

piRNAs and PIWI with cancer

piRNAs exhibit a long lifetime in cells, even though they are only 24 to 30 nt long (54), indicating that they are not easily degraded and may exist in cell nuclei and cytoplasm for a longer time than other RNAs (55). piRNAs play an important role in cancer development (56). Real-time PCR and next-generation sequencing analyses have made the relationship between piRNAs and carcinogenesis increasingly clear (11). Compared with non-cancerous tissues, the expression of piR-651, piR-823, piR-4987, piR-20365, piR-20485 and piR-20583 was revealed to be altered in cancer cell lines (12,13).

piRNAs can serve as biomarkers for the prognosis, diagnosis and clinical evaluation of cancer (8). The expression level of piR-823 in gastric cancer tissues was revealed to be lower than that in non-cancerous gastric tissues (57). A transfection-mediated increase in the piR-823 level inhibited the growth of gastric cancer cells. These results were also observed in nude mice. Thus, piR-823 may be a potential marker for cancer diagnosis (58).

The expression levels of PIWIL2, PIWIL4, and piR-823 were associated with the tumor-node-metastasis (TNM) stage of renal cell carcinoma (10). Additionally, increased piR-4987 expression regulated lymph node metastasis in breast cancer.

Here, we present an example to clarify the biomarker characteristics of piRNAs. Through small RNA sequencing and real-time PCR analysis of frozen benign kidney tissues and renal cell carcinoma tissues, 26,991 piRNAs were revealed to be expressed in kidney tissues (59). In the tumor samples, 19 types of piRNAs were found to be deregulated, including 2 types that were upregulated and 17 types that were downregulated. Furthermore, differentiation was much more obvious in the metastatic renal cell carcinoma samples (16). By comparing the localized tumor samples to the metastatic samples, 46 piRNAs were found to be aberrantly expressed, 44 of which were upregulated, while only 2 were downregulated. The increased piR-32051, piR-39894, and piR-43607 are similar in length, highly homologous and derived from the same piRNA cluster on chromosome 17. Increased expression of these piRNAs is found in late-stage tumors, which means they are highly associated with renal cell carcinoma metastasis (60).

Significantly increased piR-651 levels have been observed in non-small cell lung carcinoma (NSCLC). In A549 cells, an NSCLC cell line, piR-651 increased cell viability and metastasis. The expression of piR-651 in A549 cells decreased the proportion of cells arrested in the G0/G1 phase, thereby promoting proliferation. Oncogenes and tumor suppressor genes can be detected, and piR-651 was revealed to promote cancer growth via cyclin D1 and CDK4. These results have also been verified in lung cancer tissues from patients (61).

piR-1245 has been revealed to be overexpressed in colorectal cancer, lung, breast, stomach, bladder, kidney and prostate cancer, indicating its significant role in carcinogenesis. Poor differentiation, advanced T stage, lymph node and distant metastasis were revealed to be closely related to higher expression of piR-1245 in colorectal cancer (CRC). piR-1245 also played a crucial role in clinical pathology and revealed poor overall survival (OS) in colorectal cancer patients. piR-1245 directly targeted ATF3, BTG1, DUSP1, FAS, NFKBIA, UPP1, SESN2, TP53INP1 and MDX1 to regulate tumor progression. Thus, it may be a prognostic biomarker in CRC (62).

Similar to miRNAs, which are stable in tissues, piRNAs are relatively stable and can be used to obtain reliable results in quantitative piRNA expression studies (63). piRNAs can be detected in patient plasma (64), suggesting their potential use as biomarkers to predict TNM stage and disease prognosis (65). Furthermore, piRNAs can serve as a switch allowing tumors to proliferate and metastasize (66). Additionally, piRNAs can be indicative of patient outcomes and can be helpful in selecting effective surgical methods, radiotherapy and chemotherapy to prolong patient survival time (67).

PIWI proteins have been found in human cancers, such as breast, lung, gastric, hepatocellular, colon, renal cell carcinoma, endometrial and ovarian cancer. PIWI acts via a distinct pathway to regulate carcinogenesis. PIWIL can affect transcription, causing an increase in Bcl-XL, Stat3 and cyclin D1 expression (68).

Cancer stem cells (CSCs), contain epigenetic alterations and signaling pathways characteristic of stem cells including self-renewal capacity, rapid proliferation and multiline age differentiation. PIWIs may be cancer testis antigens (CATs) and act as oncogenes or constitute markers for CSCs (69). Metastatic cancer cells appear to undergo epithelial-mesenchymal transition (EMT) and CSC-like phenotype (70). PIWIL2 expression was associated with altered expression of EMT markers.

As a member of the PIWI gene family, MILI binds to piRNAs and plays multiple roles in gene silencing (71) and chromatin remodeling (72). Transposon methylation has been observed in tumor cell lines and many types of human cancer (7375). MILI was revealed to control the activity of LINE1 by methylating its CpG island (76). The hypomethylation of LINE1 increased the risk of cancer development and may be an indicator of cancer grade and lymph node metastasis (77,78). Wang et al found that MILI affected melanoma cell metastasis and cancer-related gene expression by regulating LINE1 methylation (79). MILI is expressed in the melanoma cell line B16 but not in the highly metastatic mouse melanoma model B16BL6. Notably, knockdown of MILI in B16 cells activated MAGEA expression and increased cell migration, whereas MILI overexpression in the B16BL6 model inhibited MAGEA expression and decreased cell migration, yielding the opposite results (65,80). Depletion of MILI/MIWI2 in mice led to reduced DNA methylation of LTR-retrotransposon promoter regions (81). Thus, LINE1 methylation by MILI was revealed to controls the expression of cancer-related genes and cell migration, and MILI plays a key role in melanoma metastasis and tumor progression.

Epigenetic mechanisms of cancer metastasis related to piRNAs

The function of piRNAs in tumors is related to transposable elements and changes in chromatin structures that are caused by epigenetic alterations, such as DNA hypomethylation (60). For example, in HeLa cells, piRNAs play an important role in inhibiting transposons by interacting with the HILI protein (82). The relationship between piRNAs and epigenetics is elaborated above, and the epigenetic changes account for a large proportion in tumors. Perhaps piRNAs and transposable elements, upstream or downstream of epigenetic alterations in tumors, affect the metastasis ability of tumors. Next, we will illustrate the epigenetic mechanisms of cancer metastasis related to piRNAs.

Cancer is a genetic and epigenetic disease (83). Cancer cells have the ability to invade tissues, enter systemic circulation, and extravasate into surrounding interstitial tissues, resulting in distant capillary retention (84). The metastasis phenotype is associated with epigenetic alterations that are involved in the cancer metastasis process. Metastasis involves cellular invasion, migration and angiogenesis of the primary carcinoma. Detection of genetic or epigenetic abnormalities can be used to identify epigenetic alterations in lesions that are morphologically normal (85). Understanding the epigenetic mechanisms of tumor metastasis related to piRNAs can assist in the identification of new tumor markers and treatments (86).

Epigenetic evaluations involve examination of DNA methylation profiles, certain RNA expression profiles (87) and histone modification profiles (88). Epigenetic alterations do not involve changes in DNA sequence (89). DNA methylation and histone modification are the major types of epigenetic alteration (90). In normal tissues, DNA methylation can prevent X chromosome activation and gene mutations (91), and histone modifications can dynamically regulate gene activity.

Here, mechanisms related to epigenetic alterations are briefly summarized. First, DNA methylation not only affects the expression of individual genes but also affects DNA domains by interacting with nucleosomes, thus altering DNA packaging. DNA methylation is inevitable and occurs on the cytosine of CpG dinucleotides. In mammalian cells, methylation is conferred by four main DNA methyltransferases (DNMTs): DNMT1 (92), DNMT3A (93), DNMT3B (94,95) and DNMT3L (96). DNMT1 adds methyl groups to hemi-methylated CpG sites, DNMT3A and DNMT3B methylate novel CpG sites, and DNMT3L interacts with DNMT3A and 3B to facilitate methylation of retrotransposons. DNA demethylation and remethylation comprise a balanced process that is disrupted in cancer cells (97). Tumor progression and metastasis occur due to changes in DNA methylation. Studies investigating DNA methylation in promoter regions have been fruitful, leading to the discovery of adenomatous polyposis coli (APC), retinoic acid receptor β-2 and H-cadherin (98), which are also associated with tumor progression (99). In primary testicular tumors, scientists detected a gain of 5′end promoter CpG island methylation of the PIWIL1, PIWIL2, PIWIL4 and TDRD1 genes in association with transcriptional silencing. The DNMT3L/PIWIL2/TDRD1 complex is responsible for the loss of DNA methylation at LINE1 and IA transposons (100). The extent of DNA methylation in tumor tissues is lower than that in normal tissues, and the degree of DNA hypomethylation increases with the progression of malignancy. DNA hypomethylation is conducive to mitotic recombination, leading to chromosomal deletions and translocations, which promote chromosomal rearrangements (101). DNA methylation in malignant cells can reactivate genomic DNA repeat elements, such as long interspersed element 1 (LINE1) (77) and Alu (102). These demethylated transposons can be transcribed or transposed to other genomic regions and disrupt the genome (103). Transposable elements, which are abundant in the human genome, are highly mutagenic due to their ability to target protein-coding genes for insertion, resulting in chromosome breakage and promoting illegitimate genome rearrangement.

Another mechanism related to cancer is histone modification. There are two types of nuclear chromatin, namely, heterochromatin and euchromatin (104). In normal tissues, heterochromatin is stable during various cell cycle phases and silences during transcription (105), and the genes in euchromatin are actively transcribed. As part of an interplay with DNA methylation, facultative heterochromatin, which is associated with allelic exclusion, genomic imprinting, X chromosome stabilization (106), immunoglobulin (Igh/Igk) and T-cell receptor-α and -β (107) gene loci, is vital for normal cell lineage development and cell differentiation via somatic methylation and inactivation of germline-specific genes (108).

Histone 3 methylation and heterochromatin

Initiation, propagation and maintenance of heterochromatin are largely controlled by trimethylation of lysine 9 on histone H3 (H3K9me3) and other synergistic epigenetic modifications (109). Chromosomal regions that are abundant in repetitive DNA help H3K9me3 stabilize constitutive heterochromatin, facultative heterochromatin and intermediate or transient heterochromatin, the 3 heterochromatin subtypes. By preventing abnormal chromosome segregation, recombination and DNA replication, H3K9me3 regulates constitutive heterochromatin to stabilize genomic integrity (110).

Histone H3 determines the formation of different chromatin structures. Methylation of the N-terminal lysine of histone H3 is vital for well-documented histone modifications. H3K9me3, H3K36me3, and possibly H3K79me3 facilitate the opening of the chromatin configuration to form euchromatin, which is also associated with serine 10 phosphorylation and lysine 9 acetylation of histone H3 for active transcription of genes. H3K9me3 and H3K27me3 mainly function in the initiation, propagation and maintenance of highly compact heterochromatin to silence gene expression (111).

Histone proteins expose DNA euchromatin to facilitate the binding of transcription factors. Methylation and acetylation are the two major mechanisms by which histone function is regulated (112). Methyltransferases and demethylases modify the lysines of histone H3 to form mono-, di-, or tri-methylated lysines, which contribute to chromatin structure and gene transcription (113). Histone methyltransferases (HMTs) regulate histone proteins by transferring methyl groups from S-adenosylmethionine to lysine or arginine residues in histones. Histone acetylation is regulated by histone acetyl-transferases (HATs) and deacetylases (HDACs) (114). Not surprisingly, there are other histone modifications. The balance of these modifications and their effects on histone structure ultimately coordinate DNA exposure. Histone methylation is a key event in gene transcription, and it is plausible to speculate that this type of modification can regulate DNA replication, recombination, and damage repair (115).

H3K9me3 and transcriptional repressors

H3K9me3 recruits transcriptional repressors such as repressor element 1 silencing transcription factor (REST) and CoREST, which contain histone deacetylases (HDACs) (116,117), H3K4me3 demethylases LSD1 (118) and Rbp4 (119), to actively transcribe gene loci, leading to gene blocking and suppression of gene transcription (120). Recruitment of DNMTs as well as additional histone methylases is responsible for localized chromatin condensation when the tethering of HP1α (heterochromatin protein) and HP1ß to H3K9me2 or H3K9me3 triggers gene silencing (121). Thus, H3K9me3 acts as a natural brake to prevent unnecessary over-transcription of actively expressed genes. Attenuation of H3K9me3 by either over-transcription of demethylases or deficiency of H3K9 methyltransferases will therefore lead to sustained expression of the genes involved in either cell cycle transition or proliferation (122).

H3K9me3 and methyltransferases in cancer

Changes in chromatin structure that are caused by epigenetic alterations can contribute to cancer development. In experimental studies using mice lacking SUV39, a methyltransferase that acts on H3K9, enhanced genomic instability and incidence of B-cell lymphoma were observed (123,124). In addition, polymorphisms of SUV39 can increase lung cancer risk due to piRNA instability and decreased levels of H3K9me3. H3K9 methyltransferases include G9a for mono- and di-methylation and SUV39h1/h2 for di- and tri-methylation of H3K9. Similarly, low levels of RIZI (125), another methyltransferase of H3K9, are frequently observed in lung cancer, breast cancer, hepatocellular carcinoma, colon cancer, neuroblastoma, and melanoma (126). Methyltransferases, such as SUV39 and RZZI act as tumor suppressors, while some demethylases may have oncogenic activity. The low expression of these methyltransferases in tumor cells may be the result of increased cell proliferation, apoptosis resistance and poor differentiation (127). Global regulation of H3K9me3 has been observed in several human cancers, including colorectal, ovarian, and lung cancer, all of which are characterized by deficiency or elevated activity of H3K9 methyltransferases or changes in the expression of H3K9 demethylases (128,129).

Other epigenetic alterations in cancer

Epigenetic changes associated with piRNA expression, affect genes associated with malignant phenotypes (VE-cadherin, VEGF-C, PAX8, Keratin 7, CD13, laminin, urokinase, α3-integrin subunit and c-met) (8).

Detection of some forms of methylation in tumors can be diagnostic of cancer. 5-Hydroxymethylcytosine (5hmC) in cfDNA, which is found in blood originating from different tissues, is the basis of noninvasive prenatal diagnostic tests, organ transplant rejection diagnostics, and cancer detection. 5hmC is useful in gene regulation and cancer pathogenesis and can be used diagnostically to identify cancer types and track tumor stage. Li et al observed a progressive global loss of 5hmC in cfDNA in lung cancer, whereas disease-specific changes in the cell-free hydroxymethylome have been observed in hepatocellular carcinoma and pancreatic cancer (130).

In breast and prostate cancers (131), protease urokinase-type plasminogen activator [UPA (132)] was revealed to promote invasion and was associated with poor prognosis (133). Hypomethylation of the UPA promoter activates tumor genes and thus worsens patient outcomes. Carcinogenesis occurs prior to cancer metastases (134), and hypermethylation of tissue inhibitor of metalloproteinase-3 [TIMP3 (135)] was revealed to promote vascular growth and activate angiogenesis (136).

DNMT3Ab plays crucial role in directing EMT-associated metastasis in gastric cancer (GC). Increased DNMT3A expression was revealed to be closely related to a poor survival rate in GC, breast, lung and liver cancer. Furthermore, TNM stage and lymph node metastasis of GC cells were more closely associated with DNMT3A than with DNMT1 and DNMT3B. Increased expression of DNMT3Ab was demonstrated to promote GC cell migration and invasion as well as EMT progression. DNMT3Ab mediated the E-cadherin gene via DNA hypermethylation and histone modifications of H3K9me2 and H3K27me3. DNMT3Ab effectively regulated the expression of E-cadherin via DNA hypermethylation and histone modifications of H3K9me2 and H3K27me3. DNMT3Ab in cooperation with H3K9me2 and H3K27me3 contributed to the transcriptional regulation of E-cadherin in a Snail-dependent manner and multiple metastasis-associated genes and oncogenic signaling pathways are regulated by DNMT3Ab overexpression. Thus, it was revealed that DNMT3Ab acts as a crucial regulator of metastasis-related genes in GC (137) (Table I).

Table I.

Summary of epigenetic-related molecular biomarkers.

Table I.

Summary of epigenetic-related molecular biomarkers.

ExpressionCancerFunctionTechnology(Refs.)
DNMT1LowColon cancer (HCT116)DNMT1-associated lncRNAs contributes to aberrant DNA methylation and gene expression during colon tumorigenesisRIP-seq(92)
DNMT3AHighLung cancerRegulated by miR-708-5pBisulfate sequencing(93)
DNMT3BOverexpressionPancreatic cancer Endometrial cancer (Ishikawa cell lines)OncogeneMSP, RT-PCR, and western blotting(94,95)
DNMT3LOverexpressionEmbryonic stem cell (ESC)Differentiation delaysMSP, RT-PCR, and western blotting(96)
LINE1Higher global methylation levelsBladder cancerIncreases bladder cancer riskPre-diagnostic blood DNA(77)
Alu (with LINE1)HypomethylatedEpithelial ovarian cancerDirect cancer associationLINE1 and Alu bisulfite pyrosequencing(102)
H3K9me3 H3K27me3HighProstate cancerFacilitates PCa progressionEpidrug 5-Aza as a model(111)
HDAC inhibitors (HDACis)Cancer chemotherapeutic agentsMelanoma cell line SK-Mel-5Upregulation of activin ART-PCR and dual luciferase assay(116)
HDACsHistone deacetylase inhibitors (HDACi) and tamoxifen, and in renal cell carcinomaBreast cancer with exemestaneAnticancer therapeuticsRT-PCR(117)
LSD1HighHepatocellular carcinoma Oral cancerCSC self-renewal and tumorigenicity in HCC Notch signaling activatedRT-PCR(118)
Rbp4HighColorectal cancer Colon cancerDiagnosis serum biomarkersELISA method(119)
SUV39Highcancer cell linesHDAC/Suv39/G9a pathwayAnalysis of MICA/B surface expression by FACS(99)
RIZ1Hypermethylated and downregulatedHepatocellular carcinomaHBx repressed RIZ1 expression via DNMT1CHIP(125)
LowEndometrial cancer (EC)Potential therapeutic targetsImmunohistochemical analysis(126)
LowSiHa cervical cancer cellsStronger cell inhibition than paclitaxel aloneRT-PCR(127)
UPAHigh Lymphangioleiomyomatosis (LAM)Attenuates LAM progression and poten tially other TSC-related disordersRT-PCR(132)
TIMP3HighCervical cancerProliferation, migration, and invasion in cervical cancerRT-PCR(135)
HighOvarian cancerDiagnostic and prognostic biomarkerRT-PCR(136)

Gliomas with histone H3 lysine27-to-methionine mutations primarily occur in the central nervous system of young children, which means that there is a link between genetics and cellular context in tumorigenesis. Through single-cell RNA sequencing of 3321 cells from six primary H3K27M-glioma and matched models, Filbin et al found that H3K27M-gliomas contained cells that resembled oligodendrocyte precursor cells (OPC-like cells). OPC-like cells tend to exhibit higher proliferation and a greater tumor-propagating potential and some of these cells display PDGFRA signaling (138).

Epigenetics of ncRNAs in cancer

EMT and angiogenesis are regulated by miRNAs (139,140). miRNAs function in cell differentiation, proliferation, apoptosis and serve as tumor suppressors and tumor promoters. Additionally, miRNAs can control other genes in certain protein pathways. If the expression of certain miRNAs can be inhibited, cancer growth or cancer metastasis may be suppressed. For example, transfection of breast cancer cells with vectors inhibiting miRNA-155 was revealed to reduce the level of CXCR4. The transfected cells exhibited lower migration and invasion rates in vitro and resulted in fewer lung metastases in vivo than control cells (141).

miRNAs, short interfering RNAs (siRNAs) and piRNAs are involved in the regulation of mRNA transcripts, chromatin-mediated gene silencing, and DNA rearrangement. miRNAs accelerate de-adenylation of the poly(A) tail and downregulate the expression of some pathways, thereby downregulating the expression of hundreds of target genes. siRNAs can also control transposons. RDR2-dependent siRNAs, which are endo-siRNAs, silence transposons, retroelements and DNA methylation (142). siRNAs bind to a nascent RNA being transcribed at their target site, resulting in RNA-induced transcriptional silencing (RITS) and formation of the RNA-directed RNA polymerase complex (RDRC) at the site of intended heterochromatin formation, which in turn results in TGS. TGS occurs in the nucleus. During this process, siRNAs guide miRNAs to modify chromatin, which also influences the cell cycle (143). miR-127 and miR-136 are released near 2 CpG islands in the Rtl1 transcript and thus regulate RISC-mediated cleavage of the maternal transcript, resulting in late-fetal or neonatal lethality (144).

miRNAs are the best studied ncRNAs (145). miRNAs play a critical role in regulating the maintenance and behavior of stem cells during self-renewal and differentiation. miR-290 serves as a transcriptional repressor of DNMTs (146). DNMTs can epigenetically silence OCT4, a transcription factor in ES cells, which can renew and differentiate into other cell types (147).

In mice, the miR-290-295 miRNA cluster was revealed to act as a transcriptional repressor of the DNMTs, Dnmt3a and Dnmt3b, resulting in the appearance of long telomeres and increased telomere recombination. The expression of this cluster remained high in undifferentiated ES cells, but decreased after ES cell differentiation. This example indicates a direct or indirect function of miRNAs in regulating genes involved in self-renewal or differentiation by affecting methylation.

Discussion

piRNA expression detected in both patient lymph nodes and serum samples is related to tumor treatment failure. piRNAs can act as tumor promoters or cancer suppressors and can participate in other carcinoma cell activities. piRNAs and PIWI can serve as biomarkers for the prognosis, diagnosis and clinical evaluation of cancer, and can be helpful in selecting effective surgical methods, radiotherapy and chemotherapy to prolong patient survival time. piRNA can serve as a switch, allowing tumors to proliferate and metastasize.

In mammals, PIWI proteins function as transposon-inhibiting factors through TGS. PIWI acts via a distinct pathway to regulate carcinogenesis by affecting transcription and the expression of other carcinoma-related genes that reduce apoptosis and increase cell proliferation and transformation. Transcriptional silencing, heterochromatin formation, transgene silencing, HP1α alteration, histone modifications and transposon suppression are all associated with PIWI, piRNAs and the piRNA pathway. Transposable elements are classified as either retrotransposons or DNA transposons. Retrotransposons can be further divided into LTR and non-LTR groups. Non-LTRs are divided into the autonomous non-LTRs LINEs and non-autonomous LTRs SINEs. Chromatin-organizing proteins such as HP1, Aub and Su(var)205, act upstream or downstream of piRNAs to regulate transposons. Hypomethylation of LINE1 increases the risk of cancer development and may be an indicator of cancer grade and lymph node metastasis. LINE1 methylation by MILI was revealed to control the expression of cancer-related genes and cell migration, and MILI played a key role in melanoma metastasis and tumor progression. In the sequences of transposable elements, the piRNA pathway mediates and maintains high levels of the repressive H3K9me3 mark in LINE1 regions in germ cells. There are two identified pathway proteins that are related to transposon silencing: CG9754 and EXD1.

Perhaps piRNAs, PIWI, transposable elements and piRNA pathways, upstream or downstream of the epigenetic alterations in tumors, affect the metastasis ability of tumors. In addition, epigenetic alteration of piRNAs, cancer stem cells, CpG island methylation and EMT all participate in cancer metastasis.

Epigenetic alterations associated with cancer metastasis involve DNA methylation, histone modification and certain RNA expression profiles. First, DNA methylation affects the expression of individual genes and DNA domains. The degree of DNA hypomethylation increases as tumors progress and metastasize. DNA methylation and histone modification can activate genomic DNA repeat elements, such as LINE1 and Alu, which can be transcribed or transposed to other genomic regions and disrupt the genome, resulting in chromosome breakage and illegitimate genome rearrangement.

H3K9me3 and other synergistic epigenetic modifications control heterochromatin, but in tumors, that balance has been disrupted. Therefore, with the ability to stabilize genomic integrity by preventing abnormal chromosome segregation, recombination and DNA replication, H3K9me3 and H3K27me3 mainly function in the initiation, propagation and maintenance of highly compact heterochromatin to silence gene expression. Methylation and acetylation are the two major mechanisms that regulate histone function. HMTs regulate histone proteins. Histone acetylation is regulated by HATs and HDACs. Several HMTs exhibit tumor suppressor functions, and some demethylases exhibit oncogenic activity. H3K9me3 recruits transcriptional repressors such as REST and CoREST. The low expression of methyltransferases such as SUV39 and RZZI in tumor cells may be the result of increased cell proliferation and apoptosis resistance and poor differentiation. Global regulation of H3K9me3 has been observed in several human cancers, including colorectal, ovarian and lung cancer, all of which are characterized by deficiency or elevated activity of H3K9 methyltransferases or altered expression of H3K9 demethylases. 5hmC can be used to identify cancer type and track tumor stage. DNMT3Ab cooperated with H3K9me2 and H3K27me3 played a crucial role in directing EMT-associated metastasis in gastric cancer.

Understanding the epigenetic mechanisms of tumor metastasis related to piRNAs can assist in the identification of new tumor markers and treatments. piRNAs can be indicative of patient outcomes and can be helpful in selecting effective surgical methods, radiotherapy and chemotherapy to prolong patient survival time. piRNAs can be used as biomarkers to predict TNM stage and disease prognosis. Since preventing tumor metastasis is still a formidable problem, studies on the potential of piRNAs and epigenetic alternations for diagnosis and prediction of clinical cancer stages are greatly needed.

Acknowledgements

Not applicable.

Funding

The present review was supported by the grant 2017Y001 from the First Affiliated Hospital of Harbin Medical University Research Innovation Fund. This review was supported by the First Affiliated Hospital of Harbin Medical University and the Laboratory of Hepatosplenic Surgery Center of Heilongjiang Province. The authors are solely responsible for the content, and this manuscript does not represent the official views of the First Affiliated Hospital of Harbin Medical University.

Availability of data and materials

Not applicable.

Author's contributions

BC, SZ and JL conceived and designed the study. JL wrote the manuscript. SZ prepared the figure and the table. BC wrote the ncRNA part, reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Li C, Vagin VV, Lee S, Xu J, Ma S, Xi H, Seitz H, Horwich MD, Syrzycka M, Honda BM, et al: Collapse of germline piRNAs in the absence of Argonaute3 reveals somatic piRNAs in flies. Cell. 137:509–521. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Shrey K, Suchit A, Nishant M and Vibha R: RNA interference: Emerging diagnostics and therapeutics tool. Biochem Biophys Res Commun. 386:273–277. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Collins LJ and Penny D: The RNA infrastructure: Dark matter of the eukaryotic cell? Trends Genet. 25:120–128. 2009. View Article : Google Scholar : PubMed/NCBI

4 

Malone CD and Hannon GJ: Small RNAs as guardians of the genome. Cell. 136:656–668. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Malone CD, Brennecke J, Dus M, Stark A, McCombie WR, Sachidanandam R and Hannon GJ: Specialized piRNA pathways act in germline and somatic tissues of the Drosophila ovary. Cell. 137:522–535. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Freedman JE, Gerstein M, Mick E, Rozowsky J, Levy D, Kitchen R, Das S, Shah R, Danielson K, Beaulieu L, et al: Diverse human extracellular RNAs are widely detected in human plasma. Nat Commun. 7:111062016. View Article : Google Scholar : PubMed/NCBI

7 

Iliev R, Stanik M, Fedorko M, Poprach A, Vychytilova-Faltejskova P, Slaba K, Svoboda M, Fabian P, Pacik D, Dolezel J, et al: Decreased expression levels of PIWIL1, PIWIL2, and PIWIL4 are associated with worse survival in renal cell carcinoma patients. OncoTargets Ther. 9:217–222. 2016.

8 

Lee JH, Schutte D, Wulf G, Füzesi L, Radzun HJ, Schweyer S, Engel W and Nayernia K: Stem-cell protein Piwil2 is widely expressed in tumors and inhibits apoptosis through activation of Stat3/Bcl-XL pathway. Hum Mol Genet. 15:201–211. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Taubert H, Greither T, Kaushal D, Würl P, Bache M, Bartel F, Kehlen A, Lautenschläger C, Harris L, Kraemer K, et al: Expression of the stem cell self-renewal gene Hiwi and risk of tumour-related death in patients with soft-tissue sarcoma. Oncogene. 26:1098–1100. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Wang Y, Liu Y, Shen X, Zhang X, Chen X, Yang C and Gao H: The PIWI protein acts as a predictive marker for human gastric cancer. Int J Clin Exp Pathol. 5:315–325. 2012.PubMed/NCBI

11 

Cheng J, Guo JM, Xiao BX, Miao Y, Jiang Z, Zhou H and Li QN: piRNA, the new non-coding RNA, is aberrantly expressed in human cancer cells. Clin Chim Acta. 412:1621–1625. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Huang G, Hu H, Xue X, Shen S, Gao E, Guo G, Shen X and Zhang X: Altered expression of piRNAs and their relation with clinicopathologic features of breast cancer. Clin Transl Oncol. 15:563–568. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Law PT, Qin H, Ching AK, Lai KP, Co NN, He M, Lung RW, Chan AW, Chan TF and Wong N: Deep sequencing of small RNA transcriptome reveals novel non-coding RNAs in hepatocellular carcinoma. J Hepatol. 58:1165–1173. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Yan H, Wu QL, Sun CY, Ai LS, Deng J, Zhang L, Chen L, Chu ZB, Tang B, Wang K, et al: piRNA-823 contributes to tumorigenesis by regulating de novo DNA methylation and angiogenesis in multiple myeloma. Leukemia. 29:196–206. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Luteijn MJ and Ketting RF: PIWI-interacting RNAs: from generation to transgenerational epigenetics. Nat Rev Genet. 14:523–534. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Li Y, Wu X, Gao H, Jin JM, Li AX, Kim YS, Pal SK, Nelson RA, Lau CM, Guo C, et al: Piwi-Interacting RNAs (piRNAs) are dysregulated in renal cell carcinoma and associated with tumor metastasis and cancer-specific survival. Mol Med. 21:381–388. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Gigek CO, Chen ES, Calcagno DQ, Wisnieski F, Burbano RR and Smith MA: Epigenetic mechanisms in gastric cancer. Epigenomics. 4:279–294. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Potter VR: Initiation and promotion in cancer formation: the importance of studies on intercellular communication. Yale J Biol Med. 53:367–384. 1980.PubMed/NCBI

19 

Lujambio A, Calin GA, Villanueva A, Ropero S, Sánchez-Céspedes M, Blanco D, Montuenga LM, Rossi S, Nicoloso MS, Faller WJ, et al: A microRNA DNA methylation signature for human cancer metastasis. Proc Natl Acad Sci USA. 105:13556–13561. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Tiwari VK, McGarvey KM, Licchesi JD, Ohm JE, Herman JG, Schübeler D and Baylin SB: PcG proteins, DNA methylation, and gene repression by chromatin looping. PLoS Biol. 6:2911–2927. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Nowacka-Zawisza M and Wisnik E: DNA methylation and histone modifications as epigenetic regulation in prostate cancer (Review). Oncol Rep. 38:2587–2596. 2017. View Article : Google Scholar : PubMed/NCBI

22 

Weick EM and Miska EA: piRNAs: From biogenesis to function. Development. 141:3458–3471. 2014. View Article : Google Scholar : PubMed/NCBI

23 

Han BW and Zamore PD: piRNAs. Curr Biol. 24:R730–R733. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Aravin AA, Sachidanandam R, Girard A, Fejes-Toth K and Hannon GJ: Developmentally regulated piRNA clusters implicate MILI in transposon control. Science. 316:744–747. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Ross RJ, Weiner MM and Lin H: PIWI proteins and PIWI-interacting RNAs in the soma. Nature. 505:353–359. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Houwing S, Kamminga LM, Berezikov E, Cronembold D, Girard A, van den Elst H, Filippov DV, Blaser H, Raz E, Moens CB, et al: A role for Piwi and piRNAs in germ cell maintenance and transposon silencing in Zebrafish. Cell. 129:69–82. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Aravin AA, Sachidanandam R, Bourc'his D, Schaefer C, Pezic D, Toth KF, Bestor T and Hannon GJ: A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice. Mol Cell. 31:785–799. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Ghildiyal M and Zamore PD: Small silencing RNAs: an expanding universe. Nat Rev Genet. 10:94–108. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Peng JC and Lin H: Beyond transposons: The epigenetic and somatic functions of the Piwi-piRNA mechanism. Curr Opin Cell Biol. 25:190–194. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Feschotte C and Pritham EJ: DNA transposons and the evolution of eukaryotic genomes. Annu Rev Genet. 41:331–368. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Kubo S, Seleme MC, Soifer HS, Perez JL, Moran JV, Kazazian HH Jr and Kasahara N: L1 retrotransposition in nondividing and primary human somatic cells. Proc Natl Acad Sci USA. 103:8036–8041. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Pelisson A, Mejlumian L, Robert V, Terzian C and Bucheton A: Drosophila germline invasion by the endogenous retrovirus gypsy: Involvement of the viral env gene. Insect Biochem Mol Biol. 32:1249–1256. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Leblanc P, Desset S, Giorgi F, Taddei AR, Fausto AM, Mazzini M, Dastugue B and Vaury C: Life cycle of an endogenous retrovirus, ZAM, in Drosophila melanogaster. J Virol. 74:10658–10669. 2000. View Article : Google Scholar : PubMed/NCBI

34 

Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH, Moran JV and Kazazian HH Jr: Hot L1s account for the bulk of retrotransposition in the human population. Proc Natl Acad Sci USA. 100:5280–5285. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Alisch RS, Garcia-Perez JL, Muotri AR, Gage FH and Moran JV: Unconventional translation of mammalian LINE-1 retrotransposons. Genes Dev. 20:210–224. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Baer CF, Miyamoto MM and Denver DR: Mutation rate variation in multicellular eukaryotes: causes and consequences. Na Rev Genet. 8:619–631. 2007. View Article : Google Scholar

37 

Belinco C, Diprima SN, Wolff RE, Thorp MW, Buschette JT and Simmons MJ: Cytotype regulation in Drosophila melanogaster: synergism between telomeric and non-telomeric P elements. Genet Res. 91:383–394. 2009. View Article : Google Scholar

38 

Simmons MJ, Peterson MP, Thorp MW, Buschette JT, DiPrima SN, Harter CL and Skolnick MJ: piRNA-mediated transposon regulation and the germ-line mutation rate in Drosophila melanogaster males. Mutat Res. 773:16–21. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Pezic D, Manakov SA, Sachidanandam R and Aravin AA: piRNA pathway targets active LINE1 elements to establish the repressive H3K9me3 mark in germ cells. Genes Dev. 28:1410–1428. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Czech B, Preall JB, McGinn J and Hannon GJ: A transcriptome-wide RNAi screen in the Drosophila ovary reveals factors of the germline piRNA pathway. Mol Cell. 50:749–761. 2013. View Article : Google Scholar : PubMed/NCBI

41 

Handler D, Meixner K, Pizka M, Lauss K, Schmied C, Gruber FS and Brennecke J: The genetic makeup of the Drosophila piRNA pathway. Mol Cell. 50:762–777. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Muerdter F, Guzzardo PM, Gillis J, Luo Y, Yu Y, Chen C, Fekete R and Hannon GJ: A genome-wide RNAi screen draws a genetic framework for transposon control and primary piRNA biogenesis in Drosophila. Mol Cell. 50:736–748. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Sarot E, Payen-Groschene G, Bucheton A and Pelisson A: Evidence for a piwi-dependent RNA silencing of the gypsy endogenous retrovirus by the Drosophila melanogaster flamenco gene. Genetics. 166:1313–1321. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Le Thomas A, Rogers AK, Webster A, Marinov GK, Liao SE, Perkins EM, Hur JK, Aravin AA and Tóth KF: Piwi induces piRNA-guided transcriptional silencing and establishment of a repressive chromatin state. Genes Dev. 27:390–399. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Sienski G, Batki J, Senti KA, Dönertas D, Tirian L, Meixner K and Brennecke J: Silencio/CG9754 connects the Piwi-piRNA complex to the cellular heterochromatin machinery. Genes Dev. 29:2258–2271. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Sienski G, Donertas D and Brennecke J: Transcriptional silencing of transposons by Piwi and maelstrom and its impact on chromatin state and gene expression. Cell. 151:964–980. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Gunawardane LS, Saito K, Nishida KM, Miyoshi K, Kawamura Y, Nagami T, Siomi H and Siomi MC: A slicer-mediated mechanism for repeat-associated siRNA 5′end formation in Drosophila. Science. 315:1587–1590. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Niki Y, Yamaguchi T and Mahowald AP: Establishment of stable cell lines of Drosophila germ-line stem cells. Proc Natl Acad Sci USA. 103:16325–16330. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Saito K, Ishizu H, Komai M, Kotani H, Kawamura Y, Nishida KM, Siomi H and Siomi MC: Roles for the Yb body components Armitage and Yb in primary piRNA biogenesis in Drosophila. Genes Dev. 24:2493–2498. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Ayyanathan K, Lechner MS, Bell P, Maul GG, Schultz DC, Yamada Y, Tanaka K, Torigoe K and Rauscher FJ III: Regulated recruitment of HP1 to a euchromatic gene induces mitotically heritable, epigenetic gene silencing: A mammalian cell culture model of gene variegation. Genes Dev. 17:1855–1869. 2003. View Article : Google Scholar : PubMed/NCBI

51 

Li Y, Danzer JR, Alvarez P, Belmont AS and Wallrath LL: Effects of tethering HP1 to euchromatic regions of the Drosophila genome. Development. 130:1817–1824. 2003. View Article : Google Scholar : PubMed/NCBI

52 

Rangan P, Malone CD, Navarro C, Newbold SP, Hayes PS, Sachidanandam R, Hannon GJ and Lehmann R: piRNA production requires heterochromatin formation in Drosophila. Curr Biol. 21:1373–1379. 2011. View Article : Google Scholar : PubMed/NCBI

53 

Yang Z, Chen KM, Pandey RR, Homolka D, Reuter M, Janeiro BK, Sachidanandam R, Fauvarque MO, McCarthy AA and Pillai RS: PIWI slicing and EXD1 drive biogenesis of nuclear piRNAs from cytosolic targets of the mouse piRNA pathway. Mol Cell. 61:138–152. 2016. View Article : Google Scholar : PubMed/NCBI

54 

He W, Wang Z, Wang Q, Fan Q, Shou C, Wang J, Giercksky KE, Nesland JM and Suo Z: Expression of HIWI in human esophageal squamous cell carcinoma is significantly associated with poorer prognosis. BMC Cancer. 9:4262009. View Article : Google Scholar : PubMed/NCBI

55 

He G, Chen L, Ye Y, Xiao Y, Hua K, Jarjoura D, Nakano T, Barsky SH, Shen R and Gao JX: Piwil2 expressed in various stages of cervical neoplasia is a potential complementary marker for p16INK4a. Am J Transl Res. 2:156–169. 2010.PubMed/NCBI

56 

Rajasethupathy P, Antonov I, Sheridan R, Frey S, Sander C, Tuschl T and Kandel ER: A role for neuronal piRNAs in the epigenetic control of memory-related synaptic plasticity. Cell. 149:693–707. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Cui L, Lou Y, Zhang X, Zhou H, Deng H, Song H, Yu X, Xiao B, Wang W and Guo J: Detection of circulating tumor cells in peripheral blood from patients with gastric cancer using piRNAs as markers. Clin Biochem. 44:1050–1057. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Cheng J, Deng H, Xiao B, Zhou H, Zhou F, Shen Z and Guo J: piR-823, a novel non-coding small RNA, demonstrates in vitro and in vivo tumor suppressive activity in human gastric cancer cells. Cancer Lett. 315:12–17. 2012. View Article : Google Scholar : PubMed/NCBI

59 

Gupta K, Miller JD, Li JZ, Russell MW and Charbonneau C: Epidemiologic and socioeconomic burden of metastatic renal cell carcinoma (mRCC): A literature review. Cancer Treat Rev. 34:193–205. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Siomi MC, Sato K, Pezic D and Aravin AA: PIWI-interacting small RNAs: The vanguard of genome defence. Nat Rev Mol Cell Biol. 12:246–258. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Li D, Luo Y, Gao Y and Yang Y, Wang Y, Xu Y, Tan S, Zhang Y, Duan J and Yang Y: piR-651 promotes tumor formation in non-small cell lung carcinoma through the upregulation of cyclin D1 and CDK4. Int J Mol Med. 38:927–936. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Weng W, Liu N, Toiyama Y, Kusunoki M, Nagasaka T, Fujiwara T, Wei Q, Qin H, Lin H, Ma Y and Goel A: Novel evidence for a PIWI-interacting RNA (piRNA) as an oncogenic mediator of disease progression, and a potential prognostic biomarker in colorectal cancer. Mol Cancer. 17:162018. View Article : Google Scholar : PubMed/NCBI

63 

Wu X, Weng L, Li X, Guo C, Pal SK, Jin JM, Li Y, Nelson RA, Mu B, Onami SH, et al: Identification of a 4-microRNA signature for clear cell renal cell carcinoma metastasis and prognosis. PLoS One. 7:e356612012. View Article : Google Scholar : PubMed/NCBI

64 

Zeng Y, Qu LK, Meng L, Liu CY, Dong B, Xing XF, Wu J and Shou CC: HIWI expression profile in cancer cells and its prognostic value for patients with colorectal cancer. Chin Med J. 124:2144–2149. 2011.PubMed/NCBI

65 

Liu JJ, Shen R, Chen L, Ye Y, He G, Hua K, Jarjoura D, Nakano T, Ramesh GK, Shapiro CL, et al: Piwil2 is expressed in various stages of breast cancers and has the potential to be used as a novel biomarker. Int J Clin Exp Pathol. 3:328–337. 2010.PubMed/NCBI

66 

Grochola LF, Greither T, Taubert H, Möller P, Knippschild U, Udelnow A, Henne-Bruns D and Würl P: The stem cell-associated Hiwi gene in human adenocarcinoma of the pancreas: expression and risk of tumour-related death. Br J Cancer. 99:1083–1088. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Mei Y, Clark D and Mao L: Novel dimensions of piRNAs in cancer. Cancer Lett. 336:46–52. 2013. View Article : Google Scholar : PubMed/NCBI

68 

Tan Y, Liu L, Liao M, Zhang C, Hu S, Zou M, Gu M and Li X: Emerging roles for PIWI proteins in cancer. Acta Biochim Biophys Sin. 47:315–324. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Litwin M, Szczepanska-Buda A, Piotrowska A, Dziegiel P and Witkiewicz W: The meaning of PIWI proteins in cancer development. Oncol Lett. 13:3354–3362. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Tamura S, Isobe T, Ariyama H, Nakano M, Kikushige Y, Takaishi S, Kusaba H, Takenaka K, Ueki T, Nakamura M, et al: Ecadherin regulates proliferation of colorectal cancer stem cells through NANOG. Oncol Rep. 40:693–703. 2018.PubMed/NCBI

71 

Liu J, Carmell MA, Rivas FV, Marsden CG, Thomson JM, Song JJ, Hammond SM, Joshua-Tor L and Hannon GJ: Argonaute2 is the catalytic engine of mammalian RNAi. Science. 305:1437–1441. 2004. View Article : Google Scholar : PubMed/NCBI

72 

Yin H and Lin H: An epigenetic activation role of Piwi and a Piwi-associated piRNA in Drosophila melanogaster. Nature. 450:304–308. 2007. View Article : Google Scholar : PubMed/NCBI

73 

Chen Y, Hu W, Lu Y, Jiang S, Li C, Chen J, Tao D, Liu Y, Yang Y and Ma Y: A TALEN-based specific transcript knock-down of PIWIL2 suppresses cell growth in HepG2 tumor cell. Cell Pprolif. 47:448–456. 2014. View Article : Google Scholar

74 

Lee JH, Jung C, Javadian-Elyaderani P, Schweyer S, Schütte D, Shoukier M, Karimi-Busheri F, Weinfeld M, Rasouli-Nia A, Hengstler JG, et al: Pathways of proliferation and antiapoptosis driven in breast cancer stem cells by stem cell protein piwil2. Cancer Res. 70:4569–4579. 2010. View Article : Google Scholar : PubMed/NCBI

75 

Zhang H, Ren Y, Xu H, Pang D, Duan C and Liu C: The expression of stem cell protein Piwil2 and piR-932 in breast cancer. Surg Oncol. 22:217–223. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Lee E, Iskow R, Yang L, Gokcumen O, Haseley P, Luquette LJ III, Lohr JG, Harris CC, Ding L, Wilson RK, et al: Landscape of somatic retrotransposition in human cancers. Science. 337:967–971. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Andreotti G, Karami S, Pfeiffer RM, Hurwitz L, Liao LM, Weinstein SJ, Albanes D, Virtamo J, Silverman DT, Rothman N and Moore LE: LINE1 methylation levels associated with increased bladder cancer risk in pre-diagnostic blood DNA among US (PLCO) and European (ATBC) cohort study participants. Epigenetics. 9:404–415. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Daskalos A, Nikolaidis G, Xinarianos G, Savvari P, Cassidy A, Zakopoulou R, Kotsinas A, Gorgoulis V, Field JK and Liloglou T: Hypomethylation of retrotransposable elements correlates with genomic instability in non-small cell lung cancer. Int J Cancer. 124:81–87. 2009. View Article : Google Scholar : PubMed/NCBI

79 

Wang X, Jiang C, Fu B, Zhu R, Diao F, Xu N, Chen Z, Tao W and Li CJ: MILI, a PIWI family protein, inhibits melanoma cell migration through methylation of LINE1. Biochem Biophys Res Commun. 457:514–519. 2015. View Article : Google Scholar : PubMed/NCBI

80 

Ye Y, Yin DT, Chen L, Zhou Q, Shen R, He G, Yan Q, Tong Z, Issekutz AC, Shapiro CL, et al: Identification of Piwil2-like (PL2L) proteins that promote tumorigenesis. PLoS One. 5:e134062010. View Article : Google Scholar : PubMed/NCBI

81 

Kuramochi-Miyagawa S, Watanabe T, Gotoh K, Totoki Y, Toyoda A, Ikawa M, Asada N, Kojima K, Yamaguchi Y, Ijiri TW, et al: DNA methylation of retrotransposon genes is regulated by Piwi family members MILI and MIWI2 in murine fetal testes. Genes Dev. 22:908–917. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Lu Y, Li C, Zhang K, Sun H, Tao D, Liu Y, Zhang S and Ma Y: Identification of piRNAs in Hela cells by massive parallel sequencing. BMB Rep. 43:635–641. 2010. View Article : Google Scholar : PubMed/NCBI

83 

Virani S, Colacino JA, Kim JH and Rozek LS: Cancer epigenetics: A brief review. ILAR J. 53:359–369. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Chik F, Szyf M and Rabbani SA: Role of epigenetics in cancer initiation and progression. Adv Exp Med Biol. 720:91–104. 2011. View Article : Google Scholar : PubMed/NCBI

85 

Cavalli LR, Urban CA, Dai D, de Assis S, Tavares DC, Rone JD, Bleggi-Torres LF, Lima RS, Cavalli IJ, Issa JP, et al: Genetic and epigenetic alterations in sentinel lymph nodes metastatic lesions compared to their corresponding primary breast tumors. Cancer Genet Cytogenet. 146:33–40. 2003. View Article : Google Scholar : PubMed/NCBI

86 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

87 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

88 

Esteller M: Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet. 8:286–298. 2007. View Article : Google Scholar : PubMed/NCBI

89 

Hinoue T, Weisenberger DJ, Lange CP, Shen H, Byun HM, Van Den Berg D, Malik S, Pan F, Noushmehr H, van Dijk CM, et al: Genome-scale analysis of aberrant DNA methylation in colorectal cancer. Genome Res. 22:271–282. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Azad N, Zahnow CA, Rudin CM and Baylin SB: The future of epigenetic therapy in solid tumours-lessons from the past. Nat Rev Clin Oncol. 10:256–266. 2013. View Article : Google Scholar : PubMed/NCBI

91 

Jones PA and Taylor SM: Cellular differentiation, cytidine analogs and DNA methylation. Cell. 20:85–93. 1980. View Article : Google Scholar : PubMed/NCBI

92 

Merry CR, Forrest ME, Sabers JN, Beard L, Gao XH, Hatzoglou M, Jackson MW, Wang Z, Markowitz SD and Khalil AM: DNMT1-associated long non-coding RNAs regulate global gene expression and DNA methylation in colon cancer. Hum Mol Genet. 24:6240–6253. 2015. View Article : Google Scholar : PubMed/NCBI

93 

Liu T, Wu X, Chen T, Luo Z and Hu X: Downregulation of DNMT3A by miR-708-5p inhibits lung cancer stem cell-like phenotypes through repressing Wnt/β-catenin signaling. Clin Cancer Res. 24:1748–1760. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Wang LH, Huang J, Wu CR, Huang LY, Cui J, Xing ZZ and Zhao CY: Downregulation of miR29b targets DNMT3b to suppress cellular apoptosis and enhance proliferation in pancreatic cancer. Mol Med Rep. 17:2113–2120. 2018.PubMed/NCBI

95 

Yang L, Hou J, Cui XH, Suo LN and Lv YW: RG108 induces the apoptosis of endometrial cancer Ishikawa cell lines by inhibiting the expression of DNMT3B and demethylation of HMLH1. Eur Rev Med Pharmacol Sci. 21:5056–5064. 2017.PubMed/NCBI

96 

Heo J, Lim J, Lee S, Jeong J, Kang H, Kim Y, Kang JW, Yu HY, Jeong EM, Kim K, et al: Sirt1 regulates DNA methylation and differentiation potential of embryonic stem cells by antagonizing Dnmt3l. Cell Rep. 18:1930–1945. 2017. View Article : Google Scholar : PubMed/NCBI

97 

Guo Y, Wang M, Jia X, Zhu H, Zhi Y and Yuan L: Wnt signaling pathway upregulates DNMT1 to trigger NHERF1 promoter hypermethylation in colon cancer. Oncol Rep. 40:1165–1173. 2018.PubMed/NCBI

98 

Zochbauer-Muller S, Gazdar AF and Minna JD: Molecular pathogenesis of lung cancer. Ann Rev Physiol. 64:681–708. 2002. View Article : Google Scholar

99 

Nakajima NI, Niimi A, Isono M, Oike T, Sato H, Nakano T and Shibata A: Inhibition of the HDAC/Suv39/G9a pathway restores the expression of DNA damage-dependent major histocompatibility complex class I-related chain A and B in cancer cells. Oncol Rep. 38:693–702. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Ferreira HJ, Heyn H, del Muro Garcia X, Vidal A, Larriba S, Muñoz C, Villanueva A and Esteller M: Epigenetic loss of the PIWI/piRNA machinery in human testicular tumorigenesis. Epigenetics. 9:113–118. 2014. View Article : Google Scholar : PubMed/NCBI

101 

Sciamanna I, Vitullo P, Curatolo A and Spadafora C: A reverse transcriptase-dependent mechanism is essential for murine preimplantation development. Genes. 2:360–373. 2011. View Article : Google Scholar : PubMed/NCBI

102 

Akers SN, Moysich K, Zhang W, Lai Collamat G, Miller A, Lele S, Odunsi K and Karpf AR: LINE1 and Alu repetitive element DNA methylation in tumors and white blood cells from epithelial ovarian cancer patients. Gynecol Oncol. 132:462–467. 2014. View Article : Google Scholar : PubMed/NCBI

103 

Slotkin RK and Martienssen R: Transposable elements and the epigenetic regulation of the genome. Nat Rev Genet. 8:272–285. 2007. View Article : Google Scholar : PubMed/NCBI

104 

Shilatifard A: Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu Rev Biochem. 75:243–269. 2006. View Article : Google Scholar : PubMed/NCBI

105 

Grewal SI and Jia S: Heterochromatin revisited. Nat Rev Genet. 8:35–46. 2007. View Article : Google Scholar : PubMed/NCBI

106 

Heard E: Delving into the diversity of facultative heterochromatin: the epigenetics of the inactive X chromosome. Curr Opin Genet Dev. 15:482–489. 2005. View Article : Google Scholar : PubMed/NCBI

107 

Corcoran AE: Immunoglobulin locus silencing and allelic exclusion. Semin Immunol. 17:141–154. 2005. View Article : Google Scholar : PubMed/NCBI

108 

Skok JA, Gisler R, Novatchkova M, Farmer D, de Laat W and Busslinger M: Reversible contraction by looping of the Tcra and Tcrb loci in rearranging thymocytes. Nat Immunol. 8:378–387. 2007. View Article : Google Scholar : PubMed/NCBI

109 

Klose RJ and Zhang Y: Regulation of histone methylation by demethylimination and demethylation. Nat Rev Mol Cell Biol. 8:307–318. 2007. View Article : Google Scholar : PubMed/NCBI

110 

Feldman N, Gerson A, Fang J, Li E, Zhang Y, Shinkai Y, Cedar H and Bergman Y: G9a-mediated irreversible epigenetic inactivation of Oct-3/4 during early embryogenesis. Nat Cell Biol. 8:188–194. 2006. View Article : Google Scholar : PubMed/NCBI

111 

Lee E, Wang J, Jung Y, Cackowski FC and Taichman RS: Reduction of two histone marks, H3k9me3 and H3k27me3 by epidrug induces neuroendocrine differentiation in prostate cancer. J Cell Biochem. 119:3697–3705. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Ooi L and Wood IC: Chromatin crosstalk in development and disease: lessons from REST. Nat Rev Genet. 8:544–554. 2007. View Article : Google Scholar : PubMed/NCBI

113 

Shi Y: Histone lysine demethylases: emerging roles in development, physiology and disease. Nat Rev Genet. 8:829–833. 2007. View Article : Google Scholar : PubMed/NCBI

114 

Yang M, Gocke CB, Luo X, Borek D, Tomchick DR, Machius M, Otwinowski Z and Yu H: Structural basis for CoREST-dependent demethylation of nucleosomes by the human LSD1 histone demethylase. Mol Cell. 23:377–387. 2006. View Article : Google Scholar : PubMed/NCBI

115 

Grewal SI and Elgin SC: Transcription and RNA interference in the formation of heterochromatin. Nature. 447:399–406. 2007. View Article : Google Scholar : PubMed/NCBI

116 

Lee J, Ko J and Yi JY: Histone deacetylase inhibitor (HDACi) upregulates activin A and activates the Smad signaling pathway in melanomas. J Dermatol Sci. 90:13–20. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Terranova-Barberio M, Thomas S and Munster PN: Host histone acetylation unlocks HDAC inhibitor potential. Oncotarget. 8:106161–106162. 2017. View Article : Google Scholar : PubMed/NCBI

118 

Liu C, Liu L, Chen X, Cheng J, Zhang H, Zhang C, Shan J, Shen J and Qian C: LSD1 stimulates cancer-associated fibroblasts to drive Notch3-dependent self-renewal of liver cancer stem-like cells. Cancer Res. 78:938–949. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Fei W, Chen L, Chen J, Shi Q, Zhang L, Liu S, Li L, Zheng L and Hu X: RBP4 and THBS2 are serum biomarkers for diagnosis of colorectal cancer. Oncotarget. 8:92254–92264. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA and Shi Y: Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 119:941–953. 2004. View Article : Google Scholar : PubMed/NCBI

121 

Smith CD, Shu S, Mungall CJ and Karpen GH: The Release 5.1 annotation of Drosophila melanogaster heterochromatin. Science. 316:1586–1591. 2007. View Article : Google Scholar : PubMed/NCBI

122 

Smallwood A, Esteve PO, Pradhan S and Carey M: Functional cooperation between HP1 and DNMT1 mediates gene silencing. Genes Dev. 21:1169–1178. 2007. View Article : Google Scholar : PubMed/NCBI

123 

Peters AH, O'Carroll D, Scherthan H, Mechtler K, Sauer S, Schöfer C, Weipoltshammer K, Pagani M, Lachner M, Kohlmaier A, et al: Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell. 107:323–337. 2001. View Article : Google Scholar : PubMed/NCBI

124 

Yoon KA, Hwangbo B, Kim IJ, Park S, Kim HS, Kee HJ, Lee JE, Jang YK, Park JG and Lee JS: Novel polymorphisms in the SUV39H2 histone methyltransferase and the risk of lung cancer. Carcinogenesis. 27:2217–2222. 2006. View Article : Google Scholar : PubMed/NCBI

125 

Zhao Z, Hu Y, Shen X, Lao Y, Zhang L, Qiu X, Hu J, Gong P, Cui H, Lu S, et al: HBx represses RIZ1 expression by DNA methyltransferase 1 involvement in decreased miR-152 in hepatocellular carcinoma. Oncol Rep. 37:2811–2818. 2017. View Article : Google Scholar : PubMed/NCBI

126 

Gibbons RJ: Histone modifying and chromatin remodelling enzymes in cancer and dysplastic syndromes. Hum Mol Genet. 14:R85–R92. 2005. View Article : Google Scholar : PubMed/NCBI

127 

Lakshmikuttyamma A, Takahashi N, Pastural E, Torlakovic E, Amin HM, Garcia-Manero G, Voralia M, Czader M, DeCoteau JF and Geyer CR: RIZ1 is potential CML tumor suppressor that is down-regulated during disease progression. J Hematol Oncol. 2:282009. View Article : Google Scholar : PubMed/NCBI

128 

Espino PS, Drobic B, Dunn KL and Davie JR: Histone modifications as a platform for cancer therapy. J Cell Biochem. 94:1088–1102. 2005. View Article : Google Scholar : PubMed/NCBI

129 

Hamamoto R, Furukawa Y, Morita M, Iimura Y, Silva FP, Li M, Yagyu R and Nakamura Y: SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells. Nat Cell Biol. 6:731–740. 2004. View Article : Google Scholar : PubMed/NCBI

130 

Li W, Zhang X, Lu X, You L, Song Y, Luo Z, Zhang J, Nie J, Zheng W, Xu D, et al: 5-Hydroxymethylcytosine signatures in circulating cell-free DNA as diagnostic biomarkers for human cancers. Cell Res. 27:1243–1257. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Pulukuri SM, Estes N, Patel J and Rao JS: Demethylation-linked activation of urokinase plasminogen activator is involved in progression of prostate cancer. Cancer Res. 67:930–939. 2007. View Article : Google Scholar : PubMed/NCBI

132 

Stepanova V, Dergilev KV, Holman KR, Parfyonova YV, Tsokolaeva ZI, Teter M, Atochina-Vasserman EN, Volgina A, Zaitsev SV, Lewis SP, et al: Urokinase-type plasminogen activator (uPA) is critical for progression of tuberous sclerosis complex 2 (TSC2)-deficient tumors. J Biol Chem. 292:20528–20543. 2017. View Article : Google Scholar : PubMed/NCBI

133 

Foekens JA, Peters HA, Look MP, Portengen H, Schmitt M, Kramer MD, Brünner N, Jänicke F, Meijer-van Gelder ME, Henzen-Logmans SC, et al: The urokinase system of plasminogen activation and prognosis in 2780 breast cancer patients. Cancer Res. 60:636–643. 2000.PubMed/NCBI

134 

Miyake H, Hara I, Yamanaka K, Gohji K, Arakawa S and Kamidono S: Elevation of serum levels of urokinase-type plasminogen activator and its receptor is associated with disease progression and prognosis in patients with prostate cancer. Prostate. 39:123–129. 1999. View Article : Google Scholar : PubMed/NCBI

135 

Zhang Z, Wang J, Wang X, Song W, Shi Y and Zhang L: MicroRNA-21 promotes proliferation, migration, and invasion of cervical cancer through targeting TIMP3. Arch Gynecol Obstet. 297:433–442. 2018. View Article : Google Scholar : PubMed/NCBI

136 

Qi JH, Ebrahem Q, Moore N, Murphy G, Claesson-Welsh L, Bond M, Baker A and Anand-Apte B: A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med. 9:407–415. 2003. View Article : Google Scholar : PubMed/NCBI

137 

Cui H, Hu Y, Guo D, Zhang A, Gu Y, Zhang S, Zhao C, Gong P, Shen X, Li Y, et al: DNA methyltransferase 3A isoform b contributes to repressing E-cadherin through cooperation of DNA methylation and H3K27/H3K9 methylation in EMT-related metastasis of gastric cancer. Oncogene. 37:4358–4371. 2018. View Article : Google Scholar : PubMed/NCBI

138 

Filbin MG, Tirosh I, Hovestadt V, Shaw ML, Escalante LE, Mathewson ND, Neftel C, Frank N, Pelton K, Hebert CM, et al: Developmental and oncogenic programs in H3K27M gliomas dissected by single-cell RNA-seq. Science. 360:331–335. 2018. View Article : Google Scholar : PubMed/NCBI

139 

Gregory PA, Bracken CP, Bert AG and Goodall GJ: MicroRNAs as regulators of epithelial-mesenchymal transition. Cell Cycle. 7:3112–3118. 2008. View Article : Google Scholar : PubMed/NCBI

140 

Suarez Y and Sessa WC: MicroRNAs as novel regulators of angiogenesis. Circ Res. 104:442–454. 2009. View Article : Google Scholar : PubMed/NCBI

141 

Liang Z, Wu H, Reddy S, Zhu A, Wang S, Blevins D, Yoon Y, Zhang Y and Shim H: Blockade of invasion and metastasis of breast cancer cells via targeting CXCR4 with an artificial microRNA. Biochem Biophys Res Commun. 363:542–546. 2007. View Article : Google Scholar : PubMed/NCBI

142 

Henderson IR and Jacobsen SE: Epigenetic inheritance in plants. Nature. 447:418–424. 2007. View Article : Google Scholar : PubMed/NCBI

143 

Wassenegger M, Heimes S, Riedel L and Sanger HL: RNA-directed de novo methylation of genomic sequences in plants. Cell. 76:567–576. 1994. View Article : Google Scholar : PubMed/NCBI

144 

Seitz H, Youngson N, Lin SP, Dalbert S, Paulsen M, Bachellerie JP, Ferguson-Smith AC and Cavaillé J: Imprinted microRNA genes transcribed antisense to a reciprocally imprinted retrotransposon-like gene. Nat Genet. 34:261–262. 2003. View Article : Google Scholar : PubMed/NCBI

145 

Lin SP, Coan P, da Rocha ST, Seitz H, Cavaille J, Teng PW, Takada S and Ferguson-Smith AC: Differential regulation of imprinting in the murine embryo and placenta by the Dlk1-Dio3 imprinting control region. Development. 134:417–426. 2007. View Article : Google Scholar : PubMed/NCBI

146 

Sinkkonen L, Hugenschmidt T, Berninger P, Gaidatzis D, Mohn F, Artus-Revel CG, Zavolan M, Svoboda P and Filipowicz W: MicroRNAs control de novo DNA methylation through regulation of transcriptional repressors in mouse embryonic stem cells. Nat Struct Mol Biol. 15:259–267. 2008. View Article : Google Scholar : PubMed/NCBI

147 

Benetti R, Gonzalo S, Jaco I, Muñoz P, Gonzalez S, Schoeftner S, Murchison E, Andl T, Chen T and Klatt P: A mammalian microRNA cluster controls DNA methylation and telomere recombination via Rbl2-dependent regulation of DNA methyltransferases. Nat Struct Mol Biol. 15:268–279. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2018
Volume 40 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu J, Zhang S and Cheng B: Epigenetic roles of PIWI‑interacting RNAs (piRNAs) in cancer metastasis (Review). Oncol Rep 40: 2423-2434, 2018
APA
Liu, J., Zhang, S., & Cheng, B. (2018). Epigenetic roles of PIWI‑interacting RNAs (piRNAs) in cancer metastasis (Review). Oncology Reports, 40, 2423-2434. https://doi.org/10.3892/or.2018.6684
MLA
Liu, J., Zhang, S., Cheng, B."Epigenetic roles of PIWI‑interacting RNAs (piRNAs) in cancer metastasis (Review)". Oncology Reports 40.5 (2018): 2423-2434.
Chicago
Liu, J., Zhang, S., Cheng, B."Epigenetic roles of PIWI‑interacting RNAs (piRNAs) in cancer metastasis (Review)". Oncology Reports 40, no. 5 (2018): 2423-2434. https://doi.org/10.3892/or.2018.6684