Next Article in Journal
Anti-Fatigue Effects of Lycium barbarum Polysaccharide and Effervescent Tablets by Regulating Oxidative Stress and Energy Metabolism in Rats
Previous Article in Journal
Ayka, a Novel Curtobacterium Bacteriophage, Provides Protection against Soybean Bacterial Wilt and Tan Spot
Previous Article in Special Issue
A Regulatory Axis between Epithelial Splicing Regulatory Proteins and Estrogen Receptor α Modulates the Alternative Transcriptome of Luminal Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Integration of TE Induces Cancer Specific Alternative Splicing Events

1
Department of Integrated Biological Sciences, Pusan National University, Busan 46241, Korea
2
Institute of Systems Biology, Pusan National University, Busan 46241, Korea
3
Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan 46241, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(18), 10918; https://doi.org/10.3390/ijms231810918
Submission received: 29 August 2022 / Revised: 13 September 2022 / Accepted: 13 September 2022 / Published: 18 September 2022
(This article belongs to the Special Issue Alternative Splicing in Cancer: Role, Mechanisms and Impact)

Abstract

:
Alternative splicing of messenger RNA (mRNA) precursors contributes to genetic diversity by generating structurally and functionally distinct transcripts. In a disease state, alternative splicing promotes incidence and development of several cancer types through regulation of cancer-related biological processes. Transposable elements (TEs), having the genetic ability to jump to other regions of the genome, can bring about alternative splicing events in cancer. TEs can integrate into the genome, mostly in the intronic regions, and induce cancer-specific alternative splicing by adjusting various mechanisms, such as exonization, providing splicing donor/acceptor sites, alternative regulatory sequences or stop codons, and driving exon disruption or epigenetic regulation. Moreover, TEs can produce microRNAs (miRNAs) that control the proportion of transcripts by repressing translation or stimulating the degradation of transcripts at the post-transcriptional level. Notably, TE insertion creates a cancer-friendly environment by controlling the overall process of gene expression before and after transcription in cancer cells. This review emphasizes the correlative interaction between alternative splicing by TE integration and cancer-associated biological processes, suggesting a macroscopic mechanism controlling alternative splicing by TE insertion in cancer.

1. Introduction

The finding that more than 90% of the average pre-mRNA sequence is removed as introns in the nucleus, and only approximately 10% of the remaining pre-mRNA is combined as exonic sequences, brings forth a fundamental principle of biology, known as RNA splicing [1,2]. Alternative splicing is a regulatory process of gene expression that imparts macromolecular and cellular complexity to higher eukaryotic organisms by allowing the production of two or more variant mRNAs from a single gene. The exons of primary transcripts are spliced into structurally and functionally distinct mRNAs that have distinct arrangements by alternative splicing [3]. That is, the combination of exons is alternatively determined as the cell decides whether to eliminate a part of the pre-mRNA or include a specific part of the mature mRNA [4]. These processes are orchestrated by the spliceosome, a dynamic and powerful macromolecular machinery complex, in a synergistic and antistatic manner [5,6]. A change in alternative splicing could play a role in the occurrence of human diseases by adjusting processes including exon skipping, intron retention, and the choice of alternative splicing sites [7,8]. In particular, protein isoforms generated by dysregulated alternative splicing, known to be the hallmark of cancer, have a close relationship with cancer development and are the subject of therapeutic interventions in numerous cancers [9,10].
The insertion of TEs is expected to be the origin of alternative splicing including, exon shuffling, and constitutively spliced exons [11]. TEs are DNA sequences that can mutate the host genomes by changing their locations and facilitating chromosomal rearrangements via homologous recombination. According to the authors of [2,12] TEs, which consist of almost half of the mammalian genomes, can be classified into two major classes distinguished by their transposition mechanisms: DNA transposon and retrotransposon [13]. DNA transposons are generally extinct in higher eukaryotes and are activated through a ‘cut-and-paste’ mechanism which depends on transposase for catalyzing excision and insertion [14]. In contrast, retrotransposons change their position via a ‘copy-and-paste’ mechanism using RNA intermediates and are reverse transcribed into complementary DNA (cDNA) by reverse transcriptase while retaining the template at its original locus [15]. Retrotransposons can also be classified into two subclasses based on the presence of a long terminal repeat (LTR): LTR retrotransposons, including endogenous retrovirus (ERV) and non-LTR retrotransposons including long-interspersed elements (LINEs) and short-interspersed elements (SINEs), such as Alu and SVA elements [16,17]. Retrotransposons are subdivided into autonomous and non-autonomous retrotransposons [18]. ERVs and LINEs are autonomous retrotransposons that encode reverse transcriptase for insertion into another region of the genome [19]. In contrast, SINEs are non-autonomous retrotransposons that cannot move to other regions by themselves because of the absence of reverse transcriptase in their sequences; they require LINEs for their transposition [20].
Integration of TE into the host genome can disrupt gene function or alter gene expression by increasing alternative splicing mechanisms [21,22]. Furthermore, TEs can generate miRNAs, also called miRNAs derived from TEs (MDTE), which are 20–25 nucleotide non-coding RNA that bind to the 3′ untranslated region (UTR) of the target mRNA [23,24]. At the post-transcriptional level, miRNAs can function as vital regulatory factors by controlling the expression of specific transcripts through repressive processes, including translational suppression and initiation of mRNA degradation [25,26]. Based on the literature, it has been revealed that biological regulatory actions induced by TE insertion are significantly related to cancer development [27,28]. This review focuses on alternative splicing and TEs that have a profound effect on the onset and development of cancers through the tuning of regulatory correlations. In addition, the tumorigenic mechanisms of TE insertion and induced alternative splicing are summarized. Simultaneously, a cancer regulatory model of the interaction among TEs, alternative splicing, and miRNAs originating from TEs is presented.

2. The Intimate Connection between Alternative Splicing and Cancer

Previous research has found that 60% of alternatively spliced variants encode functionally distinct proteins [29,30]. In a typical splicing process, the removal of the intron region is mediated by major and minor spliceosomes composed of numerous splicing factors containing five uridine-rich small nuclear RNAs (snRNA U1, U2, U4, U5, and U6) and functional analogs of the major spliceosome snRNAs (U11, U12, U4atac, and U6atac) [31]. The splicing process comprises consecutive reactions that involve the assembly of spliceosome components and their interaction with cis-acting regulatory sequences [32]. To begin, snRNP U1, SF1, and U2AF bind to the intron 5′ end splicing donor site (GU), intronic branch point, and intron 3′ end splicing acceptor site (AG), respectively. Then, U2 binds to the branch point site instead of SF1, and U4/U5/U6 snRNPs form networks with U1 and U2. After the release of U1 and U4, the activated spliceosome stimulates the cleavage of the intron 5′ and 3′ ends by forming a lariat, the release of the lariat/U2/U5/U6 complex, and the joining of post-spliced exons [6,31]. However, cancers tend to choose a substitutive pathway, cancer-specific alternative splicing, that generates several mRNA transcripts from the same gene locus with potentially different genetic functions, known as isoforms [33]. The differentially expressed isoforms in cancer, known as cancer-specific transcripts, are caused by alterations in internal or external factors. Subsequently, various biological processes that promote cancer development have changed (Figure 1) [34].

2.1. Cancer Promoting Mutations in Alternative Splicing Causing Factors

Fine-tuning between cis-acting elements and trans-acting factors coordinates alternative splicing networks [35]. Cancers are significantly affected by abnormal splicing events caused by somatic mutations in cis-acting splicing sequences and irregular alteration of trans-acting elements, including different activities of regulatory splicing factors and mutations in the core components, for example, RNA-binding proteins (RBPs), of the splicing machinery [36]. Changes in core splicing factors are related to cancer pathologies through dysregulation of cancer signaling transduction [37]. Numerous studies have identified that mutations in SRSF1, SRSF2, SF3B1, and U2AF1, which are essential components of the major spliceosome, are associated with the development of several types of cancers. SRSF1, which is upregulated in breast cancer, induces SRSF1-regulated alternative splicing events, such as exon inclusion and skipping, by binding to the 5′ or 3′ splice site. Overexpression of the exon-9-included CASC4 variant by SRSF1-regulated alternative splicing, which is considered a potential target for therapeutic development, increased proliferation, and decreased apoptosis [38]. Another example is the mutation of the splicing factor SRSF2, especially correlated with blood cancer types, which accelerates differential splicing of hn-RNP proteins in the SRSF2P95H mutant cell line [39,40] or alteration of RNA binding affinities [41]. Cancer-associated mutations in SF3B1 induce aberrant splicing of specific genes, such as DVL2, a regulator of Notch signaling, or disrupt interactions with other collaborating proteins, DDX42 and DDX46 [42,43]. In addition, mutation of U2AF1 in the S34 mutation hotspot region adjusted the progress of noncanonical translation by causing alternative splicing [44].
Furthermore, other types of RBPs contribute to dysregulated alternative splicing in cancer. For example, RNA-binding motif proteins adjust alternative splicing in cancer cells. Breast cancer-specific expression of SRPK1 accumulates phosphorylated RBM4 in the cytoplasm and then increases RBM4-regulated splicing transcripts of IR-B and MCL-1S [45]. RBM5, which is downregulated in bladder cancer, inhibits apoptosis [46], and RBM10 suppresses endometrial cancer proliferation by causing VEGFA alternative splicing [47]. Likewise, RBM6 represses the growth and progression of tumors and laryngocarcinoma by decreasing the expression of EGFR, extracellular signal-regulated kinase (ERK), and phosphorylated (p)-ERK [48]. Taken together, mutations accumulated in alternative splicing-causing factors lead to functional loss or change in the basal splicing process, thus contributing to the generation of cancer-specific splicing variants.

2.2. Cancer-Specific Transcripts Generated from Several Mechanisms of Alternative Splicing

Alternative splicing events affect the genetic flexibility and adaptability of cell biology in healthy cell metabolism, whereas in cancer, alterations in cancer cell processes occur via adjustment of apoptosis, invasion, proliferation, angiogenesis, and dysregulated metabolism. Through earlier computational analyses and microarray experiments, it has been revealed that alternatively spliced isoforms under oncogenic circumstances have a close relationship with cancer mechanisms involved in onset and development [49,50,51,52]. A list of cancer-specific transcripts that are abnormally expressed by alternative splicing and continuous regulatory processes is listed in Table 1. These previous findings have shown that the expression of cancer-specific transcripts, which encode major factors modulating important biological processes, is increased or decreased by alternative splicing mechanisms such as exon skipping, alternative 5′ or 3′ SS usage, mutually exclusive exons, intron retention, and usage of alternative first or last exons. The altered expression of abnormal transcripts promotes cancer progression by disrupting the normal regulatory system via multiple pathways (Figure 1).

3. Regulation of Cancer Related-Biological Processes by TE Induced Alternative Splicing

Inserted TEs modulate numerous biological processes including development [95], adaptive evolution [96], and disease progression, including cancer [97]. After integration of the genome, some activated TEs can modify the expression or transcriptional responsiveness of specific genes by stimulating alternative splicing through certain processes, such as: exonization; exon disruption; providing splicing donor/acceptor sites; alternative regulatory regions; premature stop codons; and inducing epigenetic alterations at the transcriptional level (Figure 2). Previous studies have found that disease biological pathways are affected by disease-specific transcript variants triggered by TE insertion [98,99]. In particular, overexpression of TEs is closely correlated with the onset and development of cancer by causing abnormal alterations in cancer progression.

3.1. Exonization and Exon Disruption

In a process called exonization, TEs can integrate into genomic regions and offer recognition by the splicing machinery as a newly recruited exon [100]. Approximately 4% of human genes contain TE motifs in their coding regions, indicating that exons may have been derived from the exonization of TEs [101,102,103,104,105,106]. Some studies have identified that exonized LINEs in the human genome provide an additional domain and produce abnormal transcripts through diverse alternative splicing mechanisms in cancers. For example, overexpression of the cancer-specific cadherin-12 (CDH12) variant, a subtype of the N-cadherin family, can be generated by somatic LINE-1 insertion and induces migration and invasion of colon cancer cells by targeting the transcription factor, Snail [99,107]. Other researchers have also found that overexpressed LINE-1 MYC products are characterized in breast cancer patients [108,109]. Furthermore, Alu inserted into the coding region can act as an alternative exon in lung cancer-related genes. One study showed that the expression of the canonical ADAR2 transcript, which functions as a tumor suppressor gene, was downregulated and other aberrant transcripts were inversely upregulated. The TE-derived isoforms among these aberrant transcripts are formed by the inclusion of an alternative exon 5a, which introduces a 120-nucleotide coding Alu-repeat sequence [110,111]. According to other studies, Alu can also integrate into MYH11, which is important for cell migration and adhesion by encoding smooth muscle myosin and make longer isoforms by adding additional exons. This insertion led to a frameshift mutation and over-production of a truncated protein [112,113].
In contrast, some proportion of inserted TEs into the coding sequence result in complete loss-of-function mutations and drastic changes in the encoded proteins by prompting gene disruption under oncogenic circumstances [114]. As a representative example, mutations in BRCA2, mainly observed in breast and ovarian cancers, could be induced by the insertion of Alu elements. The Alu element, integrated into the coding region of BRCA2, resulted in the elimination of the targeted exon 3 from the equivalent mRNA molecule by target site duplication, containing a specific 9 bp long segment as a recognition site for the transposition machinery [115]. Additionally, other studies have shown that TRPC6, which is important for cell proliferation and migration, is strongly expressed in breast cancer epithelial cells. The disrupted transcript produced by the LTR insertion is overexpressed in breast cancer [116,117]. In a systemic analysis, two genes, SHSC1 and KLK2, were abnormally spliced through an internal exon skipping mechanism after LINE-1 integration [112]. In particular, some TEs activate gene disruption, which promotes oncogenic processes via integration with tumor suppressor genes. The tumor suppressor gene, LRP1B, which suppresses cancer cell growth and invasion, was downregulated in colon cancer patients and 19 retrotransposon insertions were observed [118]. The data show that these genetic alterations are caused by exonization or genetic disruption, which adjusts the proportion of variants in a biological direction favorable to the occurrence and growth of cancer.

3.2. Providing Splicing Donor/Acceptor Site and Stop Codon

TEs are often composed of many splice-donor and acceptor sites, which lead to irregular splicing processes by interacting with splicing factors and RBPs. RBPs serve site preferences, which prompt them to reach specific regions of TEs [119,120]. TEs change the expression of cancer-related gene variants through the suggestion of alternative 5′ or 3′ SSs. In particular, inserted Alu retroelements, which contain multiple sites with sequences similar to those of SSs, could be considered a real exon by offering pseudo-SSs [101,102,121,122]. One study identified that PDZK1, which plays a crucial role in ion-channel organization, upregulates gene expression by providing alternative 5′ sites via the inserted Alu [123,124]. Other studies have also found that Alu offers 5′ alternative sites to the tumorigenic gene, HINFP, which activates cyclin E/CDK2 in the cell cycle and regulates DNA damage-induced cell cycle checkpoints [124,125,126].
The insertion of TE inside the intronic and coding regions of a premature mRNA can introduce an irregular stop codon or polyadenylation signal, resulting in truncated transcripts. Human antigen R (HuR) or fused in sarcoma (FUS) proteins, characterized by binding preference to U-rich motifs, alternatively bind to inserted TE regions and induce the nonsense-mediated decay process [127]. One research team confirmed that a short variant isoform of CHM was generated from the insertion of LTR12C as a carrier of an early stop codon and was highly expressed in colon cancer cell lines and tumor samples [128]. Furthermore, LINE-1 elements retain a polyadenylation signal within their own sequences, and AATAAA sequences are usually generated in the A-rich tail region of SINEs and LINEs [129]. The LINE-1 insertion into the last exon of APC, a tumor suppressor gene, led to disruption through the proposal of a polyadenylation site and was associated with the development of sporadic colorectal tumors [130]. Another study has also shown that germ line L1 insertions into MCC as an upstream inhibitor of the Wnt/β-catenin pathway can repress the expression of MCC and overexpress the β-catenin protein. This study suggests a functional link between L1 insertions and HCC-predisposing mutations [131]. Specific variants created by alternative SSs and stop codons are not only used as potential cancer diagnostic biomarkers but also for therapeutic applications.

3.3. Providing Alternative Regulatory Sequences Such as Enhancer, Repressor and Promoter

TEs, especially major families of retrotransposons, including LINE and SINE, are involved in the adjustment of upstream open reading frame-related genetic expression by operating cis-acting elements, such as promoters, enhancers, and repressors, to control gene expression [132,133]. TE insertion can boost the upregulation of the cis-open reading frame, causing the stimulation of oncogenic traits for cancer development. For example, MAD1L1, a cell cycle regulator, has an LTR sequence-derived promoter as one of two promoters. Isoforms induced by the LTR promoter were abundant in various tumors compared to the universally expressed form and increased cancer cell proliferation [134]. Similarly, carbonic anhydrase, which is relevant to ion, fluid, and acid-base balance, CA1, is over-expressed in colon cancer by the LTR-derived primary promoter [134,135]. In addition, an alternative promoter generated by LINE-1 insertion elevates the expression of DBC-1, which acts as an interface between apoptosis and colon cancer progression by controlling wnt/β-catenin-mediated expression of MACC1 [99,136]. Additionally, the LINE-1 integrated region into SYT1 acts as an alternative promoter and upregulates SYT1 expression, which over-activates the regulatory mechanism in membrane interactions in lung cancer [137].
On the contrary, TEs can contribute to cancer development by promoting expression of the truncated isoform after insertion, thereby downregulating the original function of the corresponding gene. For example, ARID3A has a tumor suppressive function that inhibits somatic cell reprogramming according to loss-of-function analysis [138]. According to previous research, the Alu sequence inserted into the regulatory region of ARID3A sequences increased the production of truncated proteins, which are unable to repress dedifferentiation in lung cancer cells [139]. Furthermore, some studies have indicated that TE-derived promoters activate tyrosine kinase receptors as oncogenes in colon cancer. The proto-oncogene MET1 is controlled by an alternative LINE-1 promoter within the canonical intron 2 by increasing the abnormal isoform translated into a truncated protein [140,141]. In another case, the LTR-derived alternative promoter accelerated ERBB4 expression, which alters cell proliferation and differentiation, migration, and apoptosis, resulting in an increase in the truncated isoform [99,142]. TE-derived regulatory regions have changed the overall expression level of transcripts by upregulating or downregulating cis-acting genes and are involved in diverse cancer-specific biological changes.

3.4. Epigenetic Alteration

TEs, mostly silenced in normal situations, lose their repressed markers, such as DNA methylation and suppressive histone modifications, by epigenetic dysregulation in cancer cells [143]. Many tumors have over-expressed the DNA demethylating enzymes TET2 and TET3, which result from higher ERV expression. That is, the demethylation of specific genes might be directly related to the transcription level of TEs, including ERV [144]. In renal cell cancer, DNA hypomethylation also activated TEs, ERVs expression, and immune signaling [145]. Another study also discovered that hypomethylation of retrotransposons can lead to their activation and translocation to other regions of the genome and stimulate an increase in genomic instability in T-cell lymphoma [146]. Likewise, several studies have focused on the activation of LINE-1 elements by demethylation of their own sequences linked to the development of cancers [147,148,149], such as prostate carcinoma and hepatocellular carcinoma [150,151,152]. In accordance with transcriptome analysis for chronic lymphocytic leukemia, the results suggest that TEs are globally hypomethylated compared to normal tissues [153]. In melanoma, LINE-1 hypomethylation was also closely correlated with the shortened period of relapse and survival time of patients and was connected with the metastatic conversion of primary cancer [154].
Additionally, when TEs are inserted into a tumor suppressor gene, they may cause cancer by providing new methylation or histone modifications to regulatory sequences [143]. Numerous studies have shown that inserted TEs can spread repressive epigenetic markers to regions adjacent to genomic sequences. Based on previous studies, TEs, especially highly repetitive Alu elements, are regarded as methylation centers in the genome [155,156]. Through epigenetic pattern analysis, one study revealed that Alu, integrated into intron 1, might offer additional methylation to MLH1 in correlation with trans-acting elements. Expression of MLH1, which is closely associated with mismatch repair, might be downregulated by hypermethylated Alu elements and is predicted to be closely associated with cancer development [157]. As carriers of epigenetic markers, TEs can integrate into the genomic region and generate tumorigenic status by changing the location and proportion of epigenetic markers in the regulatory region.

4. MDTEs as Regulatory Elements Linking Alternative Splicing by TE Integration and Cancer Organically

4.1. MDTEs’ Regulatory Processes Related to Cancer Progression and Cancer Therapy

Inserted TEs occasionally produce miRNAs from their sequences [24]. The TE sequence is transcribed as a primary miRNA and processed into precursor miRNAs through cleavage by proteins such as DGCR and Drosha. Precursor miRNAs move to the cytoplasm through the nuclear transport protein exportin-5 and are cleaved by proteins including TRBP and Dicer to form a miRNA duplex. One of the two complementary strands creates an RNA-induced silencing complex with functional proteins and binds to mRNA for mRNA degradation or translational repression. Through epigenetic regulation, MDTEs act as regulators that control the expression of several alternative transcripts (Figure 3) [158,159]. The MDTEs adjust the portion of oncogenic transcripts not only through basic inhibitory mechanisms but also through interactive mechanisms by interacting with other genetic elements, such as long non-coding RNA, transcription factors, and circular RNA [160,161,162,163]. Overexpression or knockdown of genes required for oncogenic processes under the adjustment of MDTEs had an effect on cancer onset and progression [164,165]. In brief, TE insertion affects pre-transcriptional regulation by directly participating in the alternative splicing process and has a big impact on fine-tuning the expression of transcripts at the post-translational level by creating a regulatory element, called miRNA, from their sequences.
Numerous studies have indicated that miRNAs are closely related to the expression of cancer-related genes as significant regulatory factors through the repression or activation of their target genes, contributing to the onset and development of human cancer types [166,167,168]. Oncogenic miRNAs in previous studies have confirmed that a substantial proportion of miRNAs known to affect the development of cancer are derived from TEs. Despite the importance of biological correlations, few studies on the interactive relationship between MDTE and cancer progression have been conducted.
According to previous research, TEs are important drug targets in the field of disease treatment because they extend from disease-causing factors [169,170,171]. Particularly in cancer, TEs have been applied as major drug targets for cancer treatment for the design of anticancer drugs. In prostate cancer, reverse transcriptase (RT), the enzyme which is encoded as region of the open reading frame 2 of LINE-1, is used as a target for anti-prostate cancer drugs. Inhibitors of reverse transcriptase (RT) repressed the proliferation of cancer cells and tumor progression and inversely promoted differentiation in animal models [172,173]. TEs were also used as vital coordinators related to cancer therapy by generating miRNAs from their sequences. Exosomal MDTEs can modulate cancer cell resistance, leading to tumor recurrence by regulating the chemosensitivity of cancer cells, which are promoted by altered cellular signaling pathways in chemotherapy [174]. As a representative example, exosomal miR-151a produced by LINE controls drug resistance in glioblastoma and pancreatic adenocarcinoma. Exosomal miR-151a sensitized temozolomide (TMZ)-resistant glioblastoma cells to TMZ by interacting with X-ray repair cross-complementing 4 (XRCC4) [175]. In pancreatic ductal adenocarcinoma, macrophage-derived exosomal miR-151a decreases the sensitivity of cancer cells to gemcitabine, dramatically [176]. Although there are still several technical limitations in the clinical trial of cancer using TEs and MDTEs, they are crucially considered as a future drug target.

4.2. Cancer Controlling MDTEs in the Top 5 Mortality Cancers

According to the global cancer report of 2020, lung cancer is the leading cause of cancer death (18% of the total cancer-related deaths), followed by colon (9.4%), liver (8.3%), stomach (7.7%), and breast (6.9%) cancers for both sexes combined [177]. Table 2 indicates the MDTEs dysregulated in five major mortality-causing cancers in the previously published literature over the last five years (2018–2022). The information on the origin of the specific TE sequence of each miRNA was organized with regard to the information of the human genome 38 registered in the UCSC genome browser. MDTEs can change cancer pathways by regulating the expression of their direct target genes on the basis of interactions with various genetic factors, leading to cancer induction or progression.

4.2.1. Lung Cancer

Lung cancer is the second most frequently diagnosed cancer with the highest death rate in 2020 [177]. MDTEs have been considered more sensitive potential prognostic biomarkers and therapeutic indicators in lung cancer than other tumor markers. Overexpressed hsa-miR-421 and hsa-miR-1290, both derived from LINE (L1) and DNA transposons, respectively, were linked to a poor prognosis and development of lung cancer, including advanced tumor stage, enlarged tumor size, lymph node involvement, and distant metastasis [178,179]. Increased or decreased miRNAs can alter the biological mechanisms of lung cancer into a favorable environment for cancer development by modulating the expression of their target genes. For instance, upregulated miR-4317 functions as a potential suppressor of lung cancer by directly binding fibroblast growth factor 9 and cyclin D2, resulting in the inhibition of proliferation, colony formation, migration, and invasion [180]. Similarly, miR-1246 prevented cell invasion and epithelial mesenchymal transition by interacting with C-X-C chemokine receptor type 4 and blocking the JAK/STAT and PI3K/AKT signal pathways in lung cancer cells [181].
In-depth studies have shown that MDTEs are involved in substantive cancer treatment. For example, miR-181b and miR-885-3p are closely related to chemoresistance by targeting BCL2 and Aurora A, respectively [182,183]. In some cases, miRNA regulation in lung cancer has a molecular connection with non-coding RNA or circular RNA. According to one study, MALAT1, an upregulated long non-coding RNA, promotes proliferation, apoptosis, migration, and invasion in non-small cell lung cancer by downregulating miR-374b-5p and inversely upregulating SRSF7 [184]. Another study also revealed a correlative axis among circular RNA, target mRNA, and miRNAs. Additionally, Circ-ZKSCAN1 increased FAM83A expression and restrained MAPK signaling by targeting carcinogenic miR-330-5p as a sponge to aid non-small cell lung cancer progression [185].

4.2.2. Colon Cancer

Colon cancer is a secondary cause of death and the third most frequently diagnosed- cancer type worldwide in both sexes [177]. Biological processes regulated by miRNAs are connected with colon cancer incidence and development. Cell growth and proliferation were stimulated by downregulation of miR-583-3p and miR-1273g-3p, inducing overexpression of their target genes PSME3 and MAGEA3/6 in colon cancer cells [186,187]. Some studies have found that exosome-transmitted miRNA-335-5p and miR-340-5p, directly controlled by LncRNA LINC00662, promote colorectal cancer invasion and metastasis through facilitating epithelial-mesenchymal transition and activating the ERK signaling pathway [188,189]. Moreover, angiogenesis in colon cancer has been over-activated by miR-181a, broadly known as an oncogenic miRNA that stimulates over-activation of VEGF signaling in various cancer types [165,190]. In addition, miR-552 serves as an indicator of poor prognosis in cancer patients and is a potential diagnostic target by regulating the expression of PTEN [191].
Some miRNAs derived from LINE act as broad tumor suppressor miRNAs related to tumor hallmarks such as proliferation, growth, apoptosis, and migration. Downregulated expression of miR-708 in various cancers, including colorectal cancer tissues and cell lines, activates proliferation and metastasis and inhibits apoptosis via the targeting of ZEB1 through the Akt/mTOR signaling pathway [192]. Additionally, miR-28-5p was identified as a component of the combined regulatory axis UCA1/miR-28-5p/HOXB3, which controls tumor size and stage, cell growth, and migration in colon cancer [193].

4.2.3. Liver Cancer

Liver cancer is the third leading cause of cancer-associated deaths worldwide. Liver cancer has a poor prognosis for late diagnosis at advanced and metastatic stages without representative prior symptoms and sufficient therapeutic approaches [194]. Studies of diagnostic biomarkers are important for the early diagnosis of liver cancer. One study showed that upregulated exosomal miR-224 derived from the DNA transposon is a diagnostic and prognostic biomarker of hepatocellular carcinoma, resulting in a lower survival rate and increased proliferation and invasion [195]. Moreover, overexpression of miR-493-5p and miR-608 suppresses the proliferation and invasion of liver cancer cells by regulating the expression of their target genes, VAMP2 and the BET family protein BRD4, respectively [196,197]. Some miRNAs originating from SINE negatively control the Warburg effect, a distinctive metabolic phenomenon that favorably utilizes glucose through aerobic glycolysis by silencing their target genes [198]. miR-342-3p, a tumor suppressor miRNA, inhibits cancer cell proliferation by inactivating the IGF-1R-mediated PI3K/AKT/GLUT1 signaling pathway. Suppression of IGF-1R weakens glycolysis by decreasing glucose uptake, lactate generation, ATP production, and extracellular acidification rate, inversely increasing the oxygen consumption rate in hepatoma cells, causing activation of proliferation [199]. Another study demonstrated that forced expression of miR-885-5p enhanced aerobic glycolysis by reducing glucose uptake and lactate production through inhibition of hexokinase 2, which catalyzes the first step of glycolysis [200].
Liver cancer-controlling miRNAs also have a close correlation with long non-coding RNAs and form a regulatory axis with other factors, including transcription factors and target genes of miRNA, based on the competing endogenous RNA hypothesis that lncRNAs might act as a molecular sponge for miRNA. One study has identified that lncRNA H19 and miR-326 are expressed inversely in hepatocellular carcinoma and control the expression of TWIST1, a downstream target of miR-326, tempting changes in cancer cell growth, migration, and invasion [201]. Another study confirmed a negative correlation between lncRNA MIR31HG and miR-575. MIR31HG suppresses proliferation and invasion of liver cancer cells by inhibiting miR-575, an upstream regulator of the tumorigenicity 7-like (ST7L) gene [202].

4.2.4. Stomach Cancer

Stomach cancer, the most common malignant tumor, ranks fifth in incidence and fourth in mortality in global cancer statistics for 2020 [177]. The onset of stomach cancer is often caused by the abnormal expression of specific genes. Several studies have confirmed that changes in the expression of cancer-related genes can be regulated by MDTEs. Consequently, MDTEs can control cancer biological pathways and manage cancer progression. Both miR-585 and miR-1269, are derived from the LTR element and inversely regulate the proliferation of stomach cancer cells. The tumor suppressor miRNA miR-585 binds to MAPK1 and prevents its expression, leading to suppression of tumor proliferation and migration [203]. On the other hand, oncogenic miRNA miR-1269 promotes proliferation and cell cycle G1-S transition by activating the AKT signaling pathway while suppressing apoptosis by targeting RASSF9 via the Bax/Bcl-2 signaling pathway [204]. Similarly, upregulated LINE derived-miR-552 also functions as an oncogenic miRNA, as an accelerator of gastric cancer progression, increased metastasis, and worsens therapeutic outcomes by targeting forkhead box O1 (FOXO1) and modulating the PI3K/AKT pathway [205]. Cancer cells induce polymorphisms in major oncogenes to circumvent this regulatory mechanism of MDTE. A recent study found that MUC4, a regulator of cell apoptosis and tumorigenesis, is aberrantly expressed in numerous cancer types. The evaluated expression of the rs2641726 C allele of MUC4 was significantly concerned with cancer incidence by providing a binding site to attenuate its interaction with miR-581 [206].
In addition, long non-coding RNAs and circular RNAs also contribute to the correlation between MDTEs and their target genes in gastric cancer. One study has verified that the novel abundantly expressed lncRNA RP11-290F20.3, named GC-related lncRNA1 (GCRL1), could change gastric cell proliferation and metastasis both in vitro and in vivo by sponging the tumor suppressor miRNA miRNA-885-3p and stimulating overexpression of the target gene cyclin-dependent kinase 4 (CDK4) [207]. In addition, long non-coding RNAs such as TRPM2-AS and LINC00324 act as miRNA sponges for MDTEs such as miR-612 and miR-3200-5p and attenuate tumorigenesis by increasing the expression of their target genes [208,209]. Furthermore, circular RNAs, Circ_0008287, and Circ-LDLRAD3, boost immune escape mechanisms or cancer cell viability criteria such as cell growth, migration, and invasion by regulating MDTEs, miR-548c-3p, and miR-224-5p, its target genes axis in stomach cancer [210,211].

4.2.5. Breast Cancer

Breast cancer is the most common cancer diagnosed in women and the most prevalent cancer in 2020 in both sexes [177]. Improved survival outcomes of breast cancer are associated with understanding the molecular processes driving breast cancer development, including the interaction of miRNA and target mRNA. For example, miR-421 is a valuable diagnostic biomarker that adjusts breast cell proliferation through targeting RDCD4 [212,213]. According to other studies, as a tumor suppressor miRNA, miR-326, and miR-340-5p derived from DNA transposon controlled vital tumor pathways, ErbB/PI3K and Wnt/beta-catenin signaling pathways [214,215].
In particular, a few miRNAs facilitate tumorigenesis of triple-negative breast cancer, which is a subset categorized by the negative expression of human epidermal growth factor receptor 2, estrogen, and progesterone receptors, and is also considered one of the highest-risk and poorest prognostic subtypes of breast cancer [216,217]. LINE-originated miRNAs, miR-582-5p, and miR374-5p, stimulate cancer invasion and metastasis by antagonizing their target genes, CMTM8 and ARRB1 [218,219]. Moreover, upexpressed miR-224-5p, derived from the DNA transposon in triple-negative breast cancer cells, enhances cell proliferation, migration, and invasion by inhibiting CASP9 [220].
Table 2. The list of miRNAs dysregulated in five major mortality-causing cancers in the last 5 years.
Table 2. The list of miRNAs dysregulated in five major mortality-causing cancers in the last 5 years.
Cancer TypeMiRNASubclassSuperfamilyTarget GeneReference
Lung cancerhsa-miR-421LINEL2-[178]
hsa-miR-4317SINEMIRFGF9, CCND2[180]
hsa-miR-330-5pSINEMIRRASSF1A[221]
FAM83A[185]
hsa-miR-374b-5pLINEL2SRSF7[184]
hsa-miR-544aDNA transposonhAT-CharlieFBXW7[222]
hsa-miR-1183LINEL2PDPK1[223]
hsa-miR-181aLINERTE-BovBGAS7[224]
hsa-miR-181bLINERTE-BovBBcl-2[182]
hsa-miR-340DNA transposonTcMar-Mariner-[225]
hsa-miR-340-5pDNA transposonTcMar-MarinerKPNA4[226]
hsa-miR-885-3pSINEMIRAurora A[183]
hsa-miR-378a-3pSINEMIRCDK4/CDK6[227]
hsa-miR-1246LTRERVL-MaLRCXCR4[181]
hsa-miR-1290DNA transposonTcMar-Tigger-[179]
hsa-miR-326DNA transposonhAT-Tip100hsa_circ_0003998[228]
Sp1[229]
hsa-miR-608LINEL2MIF[230]
Colon cancerhsa-miR-585-3pLTRERVL-MaLRPSME3[186]
hsa-miR-335-5pSINEMIRRASA1[188]
hsa-miR-181aLINERTE-BovBSRCIN1[190]
hsa-miR-708LINEL2ZEB1[192]
hsa-miR-1273SINEAluMAGEA3/6[187]
circPIP5K1A[231]
hsa-miR-340-5pDNA transposonTcMar-MarinerCLDN8, IL22[189]
hsa-miR-552LINEL1PTEN, p53[191]
hsa-miR-28-5pLINEL2HOXB3[193]
hsa-miR-374b-5pLINEL2LRH-1[232]
Liver cancerhsa-miR-23cSINEMIRERBB2IP[233]
hsa-miR-575SINEMIRST7L[202]
hsa-miR-608LINEL2BRD4[197]
hsa-miR-326DNA transposonhAT-Tip100TWIST1[201]
hsa-miR-645DNA transposonhAT-CharlieSOX30[234]
hsa-miR-493-5pLINEL2VAMP2[196]
hsa-miR-224DNA transposonDNA transposonGNMT[195]
hsa-miR-342SINEtRNA-RTEIGF-1R[199]
MCT1[235]
hsa-miR-378aSINEMIRVEGFR, PDGFRβ, c-Raf[236]
hsa-miR-885-5pSINEMIRHK2[200]
hsa-miR-421LINEL2MAPK14[237]
Stomach cancerhsa-miR-575SINEMIRPTEN[238]
hsa-miR-581DNA transposonhAT-CharlieMUC4[206]
hsa-miR-552LINEL1FOXO1[205]
hsa-miR-885-3pSINEMIRCDK4[207]
hsa-miR-421LINEL2Hsacirc0001546[212,239]
hsa-miR-181aLINERTE-BovBcaprin-1[240]
hsa-miR-612SINEMIRIGF2BP1, FOXM1[208]
hsa-miR-224-5pDNA transposonDNA transposoncirc-LDLRAD3[211]
hsa-miR-3200-5pLTRERVLBCAT1[209]
hsa-miR-1269LTRERVLRASSF9[204]
hsa-miR-585LTRERVL-MaLRMAPK1[203]
hsa-miR-4317SINEMIRZNF322[241]
hsa-miR-548c-3pDNA transposonTcMar-MarinerCLIC1[210]
Breast cancerhsa-miR-130a-3pLINERTE-BovBFOSL1, RAB5B[242]
hsa-miR-582-5pLINECR1CMTM8[218]
hsa-miR-224-5pDNA transposonDNA transposonCASP9[220]
Smad4[243]
hsa-miR-1246LTRERVL-MaLR-[244]
hsa-miR-326DNA transposonhAT-Tip100EGFR, ErbB2, ErbB3, AKT1, AKT2, AKT3[214]
hsa-miR-708LINEL2-[245]
hsa-miR-374a-5pLINEL2ARRB1[219]
hsa-miR-335-5pSINEMIREphA4[246]
hsa-miR-181aLINERTE-BovBAK024094[161]
hsa-miR-340-5pDNA transposonTcMar-MarinerLGR5[215]
hsa-miR-421LINEL2PDCD4[213]

4.3. Cancer Regulatory MDTEs in Other Cancer Types

In addition, it has been confirmed that MDTEs can function as vital controlling factors in other cancer types (Table 3). In renal cancer, DNA transposon-derived hsa-miR-224 in collaboration with miR-193a-3p promotes cell proliferation and migration by targeting al-pha-2,3-sialyltransferase IV and activating the PI3K/AKT pathway [247]. Similarly, miR-340 prevents the proliferation, migration, and invasion of squamous cell carcinoma cells by directly targeting the Ras homolog gene family member A [248]. In bladder cancer, miR-374 and miR-612 function as tumor suppressor miRNAs through interaction with ZEB2 and malic enzyme 1 [249,250]. Upregulated miR-582-5p directly targets AKT3 and affects cell proliferation and apoptosis in endometrial carcinoma [251]. LINE-originated miRNAs, miR-1271, 887-3p, and miR-552, regulate the progression of pancreatic and ovarian cancers by inhibiting each target gene [252,253,254]. Some MDTEs have the same effect on cancer as their regulators. In cancer studies, miR-224 and miR-374b were shown to be oncogenic miRNAs and tumor suppressor miRNAs, respectively, in cervical cancer by binding to pentraxin 3 and junctional adhesion molecule-2, respectively [255,256].
In addition, MDTEs have a significant effect on chemotherapy as well as their role as regulators of cancer progression. For example, the expression of the circulating miRNA, miR-1290, was downregulated in the plasma of oral squamous cell carcinoma patients compared to that in healthy volunteers. According to clinicopathological and Cox regression analyses, oral cancer patients with lower expression of miR-1290 showed poor pathological response to preoperative chemoradiotherapy and a lower five year overall survival rate. As a valuable biomarker, circulating miR-1290 can predict the clinical response to chemoradiotherapy and the overall survival rate in patients with oral squamous cell carcinoma [257]. Poor survival rates by increasing chemoresistance were caused by LTR-derived miR-1246, overexpressed in oral cancer patient tissues. Moreover, miR-1246 represses CCNG2 expression, leading to cancer cell stemness progression, which can represent relapse and metastasis [258]. Thus, MDTEs can be applied as therapeutic and diagnostic biomarkers, as well as expression regulators of oncogenes promoting cancer development and tumor suppressor genes inhibiting the generation of tumors. However, their biological and clinical value in patients with cancer has not yet been fully explored, and few research papers illuminating the relationship between MDTE and cancer have been conducted.

5. Conclusions

This review focuses on the biological consequences of TEs as a key source of alternative splicing and as a vital transcriptional regulator of various oncogenic processes associated with the onset and development of cancer. Under normal conditions, RNA processing fidelity is preserved by basal splicing, which maintains normal physiological homeostasis. Conversely, with the incidence of cancer, disruption of regulatory homeostasis by TE insertion can produce cancer-specific transcripts and contribute to the development of cancer by altering the expression of cancer progression-related genes. Moreover, miRNAs derived from TE can regulate a portion of cancer-specific transcripts at the post-transcriptional level. That is, integration of TE acts as a critical regulator of many aberrant tumorigenic processes implicated in cancer pathogenesis, including the cell cycle, apoptosis, EMT, metabolic deregulation, and angiogenesis (Figure 4). Despite this scientifically revealed organic relationship, there have been few studies on the correlation between alternative splicing by inserted TEs and cancer-related MDTEs by analyzing in vivo data originating from samples of cancer patients. Hence, in-depth research and systematic analyses of these interactions are necessary to provide therapeutic insights into cancer treatment and a better understanding of oncogenic regulatory mechanisms from a macroscopic point of view.

Author Contributions

Conceptualization, H.-S.K. and W.R.K.; writing—original draft, W.R.K.; review and editing, E.G.P., Y.J.L., W.H.B. and D.H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Abascal, F.; Tress, M.L.; Valencia, A. Alternative splicing and co-option of TEs: The case of TMPO/LAP2α and ZNF451 in mammals. Bioinformatics 2015, 31, 2257–2261. [Google Scholar] [CrossRef] [PubMed]
  2. International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef] [PubMed]
  3. Hallegger, M.; Llorian, M.; Smith, C.W. Alternative splicing: Global insights. FEBS J. 2010, 277, 856–866. [Google Scholar] [CrossRef] [PubMed]
  4. Ben-Dov, C.; Hartmann, B.; Lundgren, J.; Valcárcel, J. Genome-wide analysis of alternative pre-mRNA splicing. J. Biol. Chem. 2008, 283, 1229–1233. [Google Scholar] [CrossRef]
  5. Effenberger, K.A.; Perriman, R.J.; Bray, W.M.; Lokey, R.S.; Ares, M., Jr.; Jurica, M.S. A high-throughput splicing assay identifies new classes of inhibitors of human and yeast spliceosomes. J. Biomol. Screen. 2013, 18, 1110–1120. [Google Scholar] [CrossRef] [PubMed]
  6. Wahl, M.C.; Will, C.L.; Lührmann, R. The spliceosome: Design principles of a dynamic RNP machine. Cell 2009, 136, 701–718. [Google Scholar] [CrossRef]
  7. Matlin, A.J.; Clark, F.; Smith, C.W. Understanding alternative splicing: Towards a cellular code. Nat. Rev. Mol. Cell Biol. 2005, 6, 386–398. [Google Scholar] [CrossRef]
  8. Faustino, N.A.; Cooper, T.A. Pre-mRNA splicing and human disease. Genes Dev. 2003, 17, 419–437. [Google Scholar] [CrossRef]
  9. Venables, J.P.; Klinck, R.; Bramard, A.; Inkel, L.; Dufresne-Martin, G.; Koh, C.; Gervais-Bird, J.; Lapointe, E.; Froehlich, U.; Durand, M. Identification of alternative splicing markers for breast cancer. Cancer Res. 2008, 68, 9525–9531. [Google Scholar] [CrossRef]
  10. Kahles, A.; Lehmann, K.-V.; Toussaint, N.C.; Hüser, M.; Stark, S.G.; Sachsenberg, T.; Stegle, O.; Kohlbacher, O.; Sander, C.; Caesar-Johnson, S.J. Comprehensive analysis of alternative splicing across tumors from 8,705 patients. Cancer Cell 2018, 34, 211–224.e6. [Google Scholar] [CrossRef] [Green Version]
  11. Kim, E.; Goren, A.; Ast, G. Alternative splicing: Current perspectives. Bioessays 2008, 30, 38–47. [Google Scholar] [CrossRef] [PubMed]
  12. Herron, P. Mobile DNA II. Heredity 2004, 92, 476. [Google Scholar] [CrossRef]
  13. Wells, J.N.; Feschotte, C. A field guide to eukaryotic TEs. Annu. Rev. Genet. 2020, 54, 539. [Google Scholar] [CrossRef] [PubMed]
  14. Ramakrishnan, M.; Satish, L.; Sharma, A.; Vinod, K.K.; Emamverdian, A.; Zhou, M.; Wei, Q. Transposable elements in plants: Recent advancements, tools and prospects. Plant Mol. Biol. Rep. 2022, 1–18. [Google Scholar] [CrossRef]
  15. Mercan, L.; Bülbül, C.E.; Bilgi, F.; Marakli, S. Determination of plant-specific retrotransposons in chicken. Turk. J. Agric. For. 2022, 46, 67–73. [Google Scholar]
  16. de Assis, R.; Baba, V.Y.; Cintra, L.A.; Gonçalves, L.S.A.; Rodrigues, R.; Vanzela, A.L.L. Genome relationships and LTR-retrotransposon diversity in three cultivated Capsicum L. (Solanaceae) species. BMC Genom. 2020, 21, 237. [Google Scholar] [CrossRef]
  17. Liu, G.; Jiang, H.; Sun, W.; Zhang, J.; Chen, D.; Murchie, A.I. The function of twister ribozyme variants in non-LTR retrotransposition in Schistosoma mansoni. Nucleic Acids Res. 2021, 49, 10573–10588. [Google Scholar] [CrossRef]
  18. Wicker, T.; Stritt, C.; Sotiropoulos, A.G.; Poretti, M.; Pozniak, C.; Walkowiak, S.; Gundlach, H.; Stein, N. TE Populations Shed Light on the Evolutionary History of Wheat and the Complex Co-Evolution of Autonomous and Non-Autonomous Retrotransposons. Adv. Genet. 2022, 3, 2100022. [Google Scholar] [CrossRef]
  19. Kubota, S.; Ishikawa, T.; Kawata, K.; Hattori, T.; Nishida, T. Retrotransposons manipulating mammalian skeletal development in chondrocytes. Int. J. Mol. Sci. 2020, 21, 1564. [Google Scholar] [CrossRef]
  20. Maquat, L.E. Short interspersed nuclear element (SINE)-mediated post-transcriptional effects on human and mouse gene expression: SINE-UP for active duty. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2020, 375, 20190344. [Google Scholar] [CrossRef]
  21. Maxwell, P.H. Consequences of ongoing retrotransposition in mammalian genomes. Adv. Genom. Genet. 2014, 4, 129–142. [Google Scholar] [CrossRef] [Green Version]
  22. Ullastres, A.; Merenciano, M.; González, J. Regulatory regions in natural TE insertions drive interindividual differences in response to immune challenges in Drosophila. Genome Biol. 2021, 22, 265. [Google Scholar] [CrossRef] [PubMed]
  23. Szatkowska, M.; Krupa, R. Regulation of DNA damage response and homologous recombination repair by miRNA in human cells exposed to ionizing radiation. Cancers 2020, 12, 1838. [Google Scholar] [CrossRef]
  24. Campo, S.; Sánchez-Sanuy, F.; Camargo-Ramírez, R.; Gómez-Ariza, J.; Baldrich, P.; Campos-Soriano, L.; Soto-Suárez, M.; San Segundo, B. A novel TE-derived miRNA participates in plant immunity to rice blast disease. Plant Biotechnol. J. 2021, 19, 1798–1811. [Google Scholar] [CrossRef] [PubMed]
  25. Krützfeldt, J.; Rösch, N.; Hausser, J.; Manoharan, M.; Zavolan, M.; Stoffel, M. MiRNA-194 is a target of transcription factor 1 (Tcf1, HNF1α) in adult liver and controls expression of frizzled-6. Hepatology 2012, 55, 98–107. [Google Scholar] [CrossRef]
  26. Liz, J.; Esteller, M. lncRNAs and miRNAs with a role in cancer development. Biochim. Biophys. Acta Gene Regul. Mech. 2016, 1859, 169–176. [Google Scholar] [CrossRef]
  27. Grundy, E.E.; Diab, N.; Chiappinelli, K.B. TE regulation and expression in cancer. FEBS J. 2022, 289, 1160–1179. [Google Scholar] [CrossRef]
  28. Beckmann, P.J.; Largaespada, D.A. Transposon insertion mutagenesis in mice for modeling human cancers: Critical insights gained and new opportunities. Int. J. Mol. Sci. 2020, 21, 1172. [Google Scholar] [CrossRef]
  29. Pan, Q.; Shai, O.; Lee, L.J.; Frey, B.J.; Blencowe, B.J. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat. Genet. 2008, 40, 1413–1415. [Google Scholar] [CrossRef]
  30. Leoni, G.; Le Pera, L.; Ferrè, F.; Raimondo, D.; Tramontano, A. Coding potential of the products of alternative splicing in human. Genome Biol. 2011, 12, R9. [Google Scholar] [CrossRef]
  31. Papasaikas, P.; Valcárcel, J. The spliceosome: The ultimate RNA chaperone and sculptor. Trends Biochem. Sci. 2016, 41, 33–45. [Google Scholar] [CrossRef]
  32. Kastner, B.; Will, C.L.; Stark, H.; Lührmann, R. Structural insights into nuclear pre-mRNA splicing in higher eukaryotes. Cold Spring Harb. Perspect. Biol. 2019, 11, a032417. [Google Scholar] [CrossRef] [PubMed]
  33. Furlanis, E.; Traunmüller, L.; Fucile, G.; Scheiffele, P. Landscape of ribosome-engaged transcript isoforms reveals extensive neuronal-cell-class-specific alternative splicing programs. Nat. Neurosci. 2019, 22, 1709–1717. [Google Scholar] [CrossRef] [PubMed]
  34. Bonnal, S.C.; López-Oreja, I.; Valcárcel, J. Roles and mechanisms of alternative splicing in cancer—Implications for care. Nat. Rev. Clin. Oncol. 2020, 17, 457–474. [Google Scholar] [CrossRef] [PubMed]
  35. Baralle, F.E.; Giudice, J. Alternative splicing as a regulator of development and tissue identity. Nat. Rev. Mol. Cell Biol. 2017, 18, 437–451. [Google Scholar] [CrossRef]
  36. Zhang, J.; Manley, J.L. Misregulation of pre-mRNA alternative splicing in cancer. Cancer Discov. 2013, 3, 1228–1237. [Google Scholar] [CrossRef]
  37. Visconte, V.; Nakashima, M.O.; Rogers, H.J. Mutations in splicing factor genes in myeloid malignancies: Significance and impact on clinical features. Cancers 2019, 11, 1844. [Google Scholar] [CrossRef]
  38. Anczuków, O.; Akerman, M.; Cléry, A.; Wu, J.; Shen, C.; Shirole, N.H.; Raimer, A.; Sun, S.; Jensen, M.A.; Hua, Y. SRSF1-regulated alternative splicing in breast cancer. Mol. Cell 2015, 60, 105–117. [Google Scholar] [CrossRef]
  39. Liang, Y.; Tebaldi, T.; Rejeski, K.; Joshi, P.; Stefani, G.; Taylor, A.; Song, Y.; Vasic, R.; Maziarz, J.; Balasubramanian, K. SRSF2 mutations drive oncogenesis by activating a global program of aberrant alternative splicing in hematopoietic cells. Leukemia 2018, 32, 2659–2671. [Google Scholar] [CrossRef]
  40. Jafari, P.A.; Ayatollahi, H.; Sadeghi, R.; Sheikhi, M.; Asghari, A. Prognostic significance of SRSF2 mutations in myelodysplastic syndromes and chronic myelomonocytic leukemia: A meta-analysis. Hematology 2018, 23, 778–784. [Google Scholar] [CrossRef]
  41. Zhang, J.; Lieu, Y.K.; Ali, A.M.; Penson, A.; Reggio, K.S.; Rabadan, R.; Raza, A.; Mukherjee, S.; Manley, J.L. Disease-associated mutation in SRSF2 misregulates splicing by altering RNA-binding affinities. Proc. Natl. Acad. Sci. USA 2015, 112, E4726–E4734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhao, B.; Hu, X.; Zhou, Y.; Shi, Y.; Qian, R.; Wan, Y. Characterization of the aberrant splicing of DVL2 induced by cancer-associated SF3B1 mutation. Biochem. Biophys. Res. Commun. 2021, 546, 21–28. [Google Scholar] [CrossRef] [PubMed]
  43. Zhao, B.; Li, Z.; Qian, R.; Liu, G.; Fan, M.; Liang, Z.; Hu, X.; Wan, Y. Cancer-associated mutations in SF3B1 disrupt the interaction between SF3B1 and DDX42. J. Biochem. 2022, 172, 117–126. [Google Scholar] [CrossRef] [PubMed]
  44. Palangat, M.; Anastasakis, D.G.; Fei, D.L.; Lindblad, K.E.; Bradley, R.; Hourigan, C.S.; Hafner, M.; Larson, D.R. The splicing factor U2AF1 contributes to cancer progression through a noncanonical role in translation regulation. Genes Dev. 2019, 33, 482–497. [Google Scholar] [CrossRef]
  45. Lin, J.-C.; Lin, C.-Y.; Tarn, W.-Y.; Li, F.-Y. Elevated SRPK1 lessens apoptosis in breast cancer cells through RBM4-regulated splicing events. RNA 2014, 20, 1621–1631. [Google Scholar] [CrossRef]
  46. Zhang, Y.-P.; Liu, K.-L.; Wang, Y.-X.; Yang, Z.; Han, Z.-W.; Lu, B.-S.; Qi, J.-C.; Yin, Y.-W.; Teng, Z.-H.; Chang, X.-L. Down-regulated RBM5 inhibits bladder cancer cell apoptosis by initiating an miR-432-5p/β-catenin feedback loop. FASEB J. 2019, 33, 10973–10985. [Google Scholar] [CrossRef]
  47. Dou, X.Q.; Chen, X.J.; Wen, M.X.; Zhang, S.Z.; Zhou, Q.; Zhang, S.Q. Alternative splicing of VEGFA is regulated by RBM10 in endometrial cancer. Kaohsiung J. Med. Sci. 2020, 36, 13–19. [Google Scholar] [CrossRef]
  48. Wang, Q.; Wang, F.; Zhong, W.; Ling, H.; Wang, J.; Cui, J.; Xie, T.; Wen, S.; Chen, J. RNA-binding protein RBM6 as a tumor suppressor gene represses the growth and progression in laryngocarcinoma. Gene 2019, 697, 26–34. [Google Scholar] [CrossRef]
  49. French, P.J.; Peeters, J.; Horsman, S.; Duijm, E.; Siccama, I.; Van Den Bent, M.J.; Luider, T.M.; Kros, J.M.; van der Spek, P.; Sillevis Smitt, P.A. Identification of differentially regulated splice variants and novel exons in glial brain tumors using exon expression arrays. Cancer Res. 2007, 67, 5635–5642. [Google Scholar] [CrossRef]
  50. Milani, L.; Fredriksson, M.; Syvanen, A.-C. Detection of alternatively spliced transcripts in leukemia cell lines by minisequencing on microarrays. Clin. Chem. 2006, 52, 202–211. [Google Scholar] [CrossRef]
  51. Li, H.-R.; Wang-Rodriguez, J.; Nair, T.M.; Yeakley, J.M.; Kwon, Y.-S.; Bibikova, M.; Zheng, C.; Zhou, L.; Zhang, K.; Downs, T. Two-dimensional transcriptome profiling: Identification of mRNA isoform signatures in prostate cancer from archived paraffin-embedded cancer specimens. Cancer Res. 2006, 66, 4079–4088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Roy, M.; Xu, Q.; Lee, C. Evidence that public database records for many cancer-associated genes reflect a splice form found in tumors and lack normal splice forms. Nucleic Acids Res. 2005, 33, 5026–5033. [Google Scholar] [CrossRef] [PubMed]
  53. Boise, L.H.; González-García, M.; Postema, C.E.; Ding, L.; Lindsten, T.; Turka, L.A.; Mao, X.; Nuñez, G.; Thompson, C.B. bcl-x, a bcl-2-related gene that functions as a dominant regulator of apoptotic cell death. Cell 1993, 74, 597–608. [Google Scholar] [CrossRef]
  54. Gayle, S.S.; Sahni, J.M.; Webb, B.M.; Weber-Bonk, K.L.; Shively, M.S.; Spina, R.; Bar, E.E.; Summers, M.K.; Keri, R.A. Targeting BCL-xL improves the efficacy of bromodomain and extra-terminal protein inhibitors in triple-negative breast cancer by eliciting the death of senescent cells. J. Biol. Chem. 2019, 294, 875–886. [Google Scholar] [CrossRef] [PubMed]
  55. Takehara, T.; Liu, X.; Fujimoto, J.; Friedman, S.L.; Takahashi, H. Expression and role of Bcl-xL in human hepatocellular carcinomas. Hepatology 2001, 34, 55–61. [Google Scholar] [CrossRef]
  56. Prinos, P.; Garneau, D.; Lucier, J.-F.; Gendron, D.; Couture, S.; Boivin, M.; Brosseau, J.-P.; Lapointe, E.; Thibault, P.; Durand, M. Alternative splicing of SYK regulates mitosis and cell survival. Nat. Struct. Mol. Biol. 2011, 18, 673–679. [Google Scholar] [CrossRef]
  57. Du, Z.; Fan, M.; Kim, J.-G.; Eckerle, D.; Lothstein, L.; Wei, L.; Pfeffer, L.M. Interferon-resistant Daudi cell line with a Stat2 defect is resistant to apoptosis induced by chemotherapeutic agents. J. Biol. Chem. 2009, 284, 27808–27815. [Google Scholar] [CrossRef]
  58. Schittenhelm, M.M.; Walter, B.; Tsintari, V.; Federmann, B.; Saipi, M.B.; Akmut, F.; Illing, B.; Mau-Holzmann, U.; Fend, F.; Lopez, C.D. Alternative splicing of the tumor suppressor ASPP2 results in a stress-inducible, oncogenic isoform prevalent in acute leukemia. EBioMedicine 2019, 42, 340–351. [Google Scholar] [CrossRef]
  59. Liu, J.H.; Wei, S.; Lamy, T.; Li, Y.; Epling-Burnette, P.; Djeu, J.Y.; Loughran, T.P., Jr. Blockade of Fas-dependent apoptosis by soluble Fas in LGL leukemia. Blood Am. J. Hematol. 2002, 100, 1449–1453. [Google Scholar] [CrossRef]
  60. Inaba, H.; Komada, Y.; Li, Q.S.; Zhang, X.L.; Tanaka, S.; Azuma, E.; Yamamoto, H.; Sakurai, M. mRNA expression of variant Fas molecules in acute leukemia cells. Am. J. Hematol. 1999, 62, 150–158. [Google Scholar] [CrossRef]
  61. Kamihira, S.; Yamada, Y.; Tomonaga, M.; Sugahara, K.; Tsuruda, K. Discrepant expression of membrane and soluble isoforms of Fas (CD95/APO-1) in adult T-cell leukaemia: Soluble Fas isoform is an independent risk factor for prognosis. Br. J. Haematol. 1999, 107, 851–860. [Google Scholar] [CrossRef] [PubMed]
  62. Fukumoto, T.; Iwasaki, T.; Okada, T.; Hashimoto, T.; Moon, Y.; Sakaguchi, M.; Fukami, Y.; Nishigori, C.; Oka, M. High expression of Mcl-1L via the MEK-ERK-phospho-STAT 3 (Ser727) pathway protects melanocytes and melanoma from UVB-induced apoptosis. Genes Cells 2016, 21, 185–199. [Google Scholar] [CrossRef] [PubMed]
  63. Bria, E.; Di Modugno, F.; Sperduti, I.; Iapicca, P.; Visca, P.; Alessandrini, G.; Antoniani, B.; Pilotto, S.; Ludovini, V.; Vannucci, J. Prognostic impact of alternative splicing-derived hMENA isoforms in resected, node-negative, non-small-cell lung cancer. Oncotarget 2014, 5, 11054. [Google Scholar] [CrossRef]
  64. Philippar, U.; Roussos, E.T.; Oser, M.; Yamaguchi, H.; Kim, H.-D.; Giampieri, S.; Wang, Y.; Goswami, S.; Wyckoff, J.B.; Lauffenburger, D.A. A Mena invasion isoform potentiates EGF-induced carcinoma cell invasion and metastasis. Dev. Cell 2008, 15, 813–828. [Google Scholar] [CrossRef] [PubMed]
  65. Oudin, M.J.; Hughes, S.K.; Rohani, N.; Moufarrej, M.N.; Jones, J.G.; Condeelis, J.S.; Lauffenburger, D.A.; Gertler, F.B. Characterization of the expression of the pro-metastatic MenaINV isoform during breast tumor progression. Clin. Exp. Metastasis 2016, 33, 249–261. [Google Scholar] [CrossRef]
  66. Di Modugno, F.; Iapicca, P.; Boudreau, A.; Mottolese, M.; Terrenato, I.; Perracchio, L.; Carstens, R.P.; Santoni, A.; Bissell, M.J.; Nisticò, P. Splicing program of human MENA produces a previously undescribed isoform associated with invasive, mesenchymal-like breast tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 19280–19285. [Google Scholar] [CrossRef]
  67. Narla, G.; DiFeo, A.; Yao, S.; Banno, A.; Hod, E.; Reeves, H.L.; Qiao, R.F.; Camacho-Vanegas, O.; Levine, A.; Kirschenbaum, A. Targeted inhibition of the KLF6 splice variant, KLF6 SV1, suppresses prostate cancer cell growth and spread. Cancer Res. 2005, 65, 5761–5768. [Google Scholar] [CrossRef]
  68. Hatami, R.; Sieuwerts, A.M.; Izadmehr, S.; Yao, Z.; Qiao, R.F.; Papa, L.; Look, M.P.; Smid, M.; Ohlssen, J.; Levine, A.C. KLF6-SV1 drives breast cancer metastasis and is associated with poor survival. Sci. Transl. Med. 2013, 5, 169ra12. [Google Scholar] [CrossRef]
  69. Prochazka, L.; Tesarik, R.; Turanek, J. Regulation of alternative splicing of CD44 in cancer. Cell. Signal. 2014, 26, 2234–2239. [Google Scholar] [CrossRef]
  70. Wielenga, V.J.; Heider, K.-H.; Johan, G.; Offerhaus, A.; Adolf, G.R.; van den Berg, F.M.; Ponta, H.; Herrlich, P.; Pals, S.T. Expression of CD44 variant proteins in human colorectal cancer is related to tumor progression. Cancer Res. 1993, 53, 4754–4756. [Google Scholar]
  71. Ni, J.; Cozzi, P.J.; Hao, J.L.; Beretov, J.; Chang, L.; Duan, W.; Shigdar, S.; Delprado, W.J.; Graham, P.H.; Bucci, J. CD44 variant 6 is associated with prostate cancer metastasis and chemo-/radioresistance. Prostate 2014, 74, 602–617. [Google Scholar] [CrossRef] [PubMed]
  72. Zhang, Y.; Zhao, Y.; Jiang, G.; Zhang, X.; Zhao, H.; Wu, J.; Xu, K.; Wang, E. Impact of p120-catenin isoforms 1A and 3A on epithelial mesenchymal transition of lung cancer cells expressing E-cadherin in different subcellular locations. PLoS ONE 2014, 9, e88064. [Google Scholar] [CrossRef] [PubMed]
  73. Matsuda, Y.; Hagio, M.; Seya, T.; Ishiwata, T. Fibroblast Growth Factor Receptor 2 IIIc as a Therapeutic Target for Colorectal Cancer CellsFGFR2IIIc in Colorectal Cancer. Mol. Cancer Ther. 2012, 11, 2010–2020. [Google Scholar] [CrossRef] [PubMed]
  74. Kawase, R.; Ishiwata, T.; Matsuda, Y.; Onda, M.; Kudo, M.; Takeshita, T.; Naito, Z. Expression of fibroblast growth factor receptor 2 IIIc in human uterine cervical intraepithelial neoplasia and cervical cancer. Int. J. Oncol. 2010, 36, 331–340. [Google Scholar]
  75. Ranieri, D.; Rosato, B.; Nanni, M.; Magenta, A.; Belleudi, F.; Torrisi, M.R. Expression of the FGFR2 mesenchymal splicing variant in epithelial cells drives epithelial-mesenchymal transition. Oncotarget 2016, 7, 5440. [Google Scholar] [CrossRef]
  76. Ghigna, C.; Giordano, S.; Shen, H.; Benvenuto, F.; Castiglioni, F.; Comoglio, P.M.; Green, M.R.; Riva, S.; Biamonti, G. Cell motility is controlled by SF2/ASF through alternative splicing of the Ron protooncogene. Mol. Cell 2005, 20, 881–890. [Google Scholar] [CrossRef]
  77. Collesi, C.; Santoro, M.M.; Gaudino, G.; Comoglio, P.M. A splicing variant of the RON transcript induces constitutive tyrosine kinase activity and an invasive phenotype. Mol. Cell. Biol. 1996, 16, 5518–5526. [Google Scholar] [CrossRef]
  78. Ben-Hur, V.; Denichenko, P.; Siegfried, Z.; Maimon, A.; Krainer, A.; Davidson, B.; Karni, R. S6K1 alternative splicing modulates its oncogenic activity and regulates mTORC1. Cell Rep. 2013, 3, 103–115. [Google Scholar] [CrossRef]
  79. Mei, H.; Wang, Y.; Fan, J.; Lin, Z. Alternative splicing of S6K1 promotes non-small cell lung cancer survival. Tumor Biol. 2016, 37, 13369–13376. [Google Scholar] [CrossRef]
  80. Bechara, E.G.; Sebestyén, E.; Bernardis, I.; Eyras, E.; Valcárcel, J. RBM5, 6, and 10 differentially regulate NUMB alternative splicing to control cancer cell proliferation. Mol. Cell 2013, 52, 720–733. [Google Scholar] [CrossRef]
  81. Haupt, Y.; Maya, R.; Kazaz, A.; Oren, M. Mdm2 promotes the rapid degradation of p53. Nature 1997, 387, 296–299. [Google Scholar] [CrossRef] [PubMed]
  82. Kubbutat, M.H.; Jones, S.N.; Vousden, K.H. Regulation of p53 stability by Mdm2. Nature 1997, 387, 299–303. [Google Scholar] [CrossRef] [PubMed]
  83. Sigalas, I.; Calvert, A.H.; Anderson, J.J.; Neal, D.E.; Lunec, J. Alternatively spliced mdm2 transcripts with loss of p53 binding domain sequences: Transforming ability and frequent detection in human cancer. Nat. Med. 1996, 2, 912–917. [Google Scholar] [CrossRef] [PubMed]
  84. Biselli-Chicote, P.; Oliveira, A.; Pavarino, E.; Goloni-Bertollo, E. VEGF gene alternative splicing: Pro-and anti-angiogenic isoforms in cancer. J. Cancer Res. Clin. Oncol. 2012, 138, 363–370. [Google Scholar] [CrossRef]
  85. Abou Faycal, C.; Gazzeri, S.; Eymin, B. A VEGF-A/SOX2/SRSF2 network controls VEGFR1 pre-mRNA alternative splicing in lung carcinoma cells. Sci. Rep. 2019, 9, 336. [Google Scholar] [CrossRef]
  86. Albuquerque, R.J.; Hayashi, T.; Cho, W.G.; Kleinman, M.E.; Dridi, S.; Takeda, A.; Baffi, J.Z.; Yamada, K.; Kaneko, H.; Green, M.G. Alternatively spliced vascular endothelial growth factor receptor-2 is an essential endogenous inhibitor of lymphatic vessel growth. Nat. Med. 2009, 15, 1023–1030. [Google Scholar] [CrossRef]
  87. Clower, C.V.; Chatterjee, D.; Wang, Z.; Cantley, L.C.; Vander Heiden, M.G.; Krainer, A.R. The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform expression and cell metabolism. Proc. Natl. Acad. Sci. USA 2010, 107, 1894–1899. [Google Scholar] [CrossRef]
  88. Christofk, H.R.; Vander Heiden, M.G.; Harris, M.H.; Ramanathan, A.; Gerszten, R.E.; Wei, R.; Fleming, M.D.; Schreiber, S.L.; Cantley, L.C. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008, 452, 230–233. [Google Scholar] [CrossRef]
  89. Poulikakos, P.I.; Persaud, Y.; Janakiraman, M.; Kong, X.; Ng, C.; Moriceau, G.; Shi, H.; Atefi, M.; Titz, B.; Gabay, M.T. RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF (V600E). Nature 2011, 480, 387–390. [Google Scholar] [CrossRef]
  90. Takeuchi, A.; Yokoyama, S.; Nakamori, M.; Nakamura, M.; Ojima, T.; Yamaguchi, S.; Mitani, Y.; Shively, J.E.; Yamaue, H. Loss of CEACAM1 is associated with poor prognosis and peritoneal dissemination of patients with gastric cancer. Sci. Rep. 2019, 9, 12702. [Google Scholar] [CrossRef]
  91. Chen, L.; Chen, Z.; Baker, K.; Halvorsen, E.M.; da Cunha, A.P.; Flak, M.B.; Gerber, G.; Huang, Y.-H.; Hosomi, S.; Arthur, J.C. The short isoform of the CEACAM1 receptor in intestinal T cells regulates mucosal immunity and homeostasis via Tfh cell induction. Immunity 2012, 37, 930–946. [Google Scholar] [CrossRef]
  92. Helfrich, I.; Singer, B.B. Size matters: The functional role of the CEACAM1 isoform signature and its impact for NK cell-mediated killing in melanoma. Cancers 2019, 11, 356. [Google Scholar] [CrossRef] [PubMed]
  93. Comstock, C.E.; Augello, M.A.; Benito, R.P.; Karch, J.; Tran, T.H.; Utama, F.E.; Tindall, E.A.; Wang, Y.; Burd, C.J.; Groh, E.M. Cyclin D1 Splice Variants: Polymorphism, Risk, and Isoform-Specific Regulation in Prostate CancerCyclin D1b Regulation in Prostate Cancer. Clin. Cancer Res. 2009, 15, 5338–5349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Zhu, J.; Sen, S.; Wei, C.; Frazier, M.L. Cyclin D1b represses breast cancer cell growth by antagonizing the action of cyclin D1a on estrogen receptor α-mediated transcription. Int. J. Oncol. 2010, 36, 39–48. [Google Scholar] [PubMed]
  95. Garcia-Perez, J.L.; Widmann, T.J.; Adams, I.R. The impact of TEs on mammalian development. Development 2016, 143, 4101–4114. [Google Scholar] [CrossRef]
  96. Schrader, L.; Schmitz, J. The impact of TEs in adaptive evolution. Mol. Ecol. 2019, 28, 1537–1549. [Google Scholar] [CrossRef]
  97. Bernard, A.; Boidot, R.; Végran, F. Alternative Splicing in Cancer and Immune Cells. Cancers 2022, 14, 1726. [Google Scholar] [CrossRef]
  98. Kaer, K.; Speek, M. Retroelements in human disease. Gene 2013, 518, 231–241. [Google Scholar] [CrossRef]
  99. Lee, E.; Iskow, R.; Yang, L.; Gokcumen, O.; Haseley, P.; Luquette, L.J., III; Lohr, J.G.; Harris, C.C.; Ding, L.; Wilson, R.K. Landscape of somatic retrotransposition in human cancers. Science 2012, 337, 967–971. [Google Scholar] [CrossRef]
  100. Krasileva, K.V. The role of TEs and DNA damage repair mechanisms in gene duplications and gene fusions in plant genomes. Curr. Opin. Plant Biol. 2019, 48, 18–25. [Google Scholar] [CrossRef]
  101. Makałowski, W.; Mitchell, G.A.; Labuda, D. Alu sequences in the coding regions of mRNA: A source of protein variability. Trends Genet. 1994, 10, 188–193. [Google Scholar] [CrossRef]
  102. Sorek, R.; Ast, G.; Graur, D. Alu-containing exons are alternatively spliced. Genome Res. 2002, 12, 1060–1067. [Google Scholar] [CrossRef] [PubMed]
  103. Nekrutenko, A.; Li, W.-H. TEs are found in a large number of human protein-coding genes. Trends Genet. 2001, 17, 619–621. [Google Scholar] [CrossRef]
  104. Singer, S.S.; Männel, D.N.; Hehlgans, T.; Brosius, J.; Schmitz, J. From “junk” to gene: Curriculum vitae of a primate receptor isoform gene. J. Mol. Biol. 2004, 341, 883–886. [Google Scholar] [CrossRef]
  105. Gotea, V.; Makałowski, W. Do TEs really contribute to proteomes? Trends Genet. 2006, 22, 260–267. [Google Scholar] [CrossRef]
  106. Zhang, X.H.-F.; Chasin, L.A. Comparison of multiple vertebrate genomes reveals the birth and evolution of human exons. Proc. Natl. Acad. Sci. USA 2006, 103, 13427–13432. [Google Scholar] [CrossRef]
  107. Ma, J.; Zhao, J.; Lu, J.; Wang, P.; Feng, H.; Zong, Y.; Ou, B.; Zheng, M.; Lu, A. Cadherin-12 enhances proliferation in colorectal cancer cells and increases progression by promoting EMT. Tumor Biol. 2016, 37, 9077–9088. [Google Scholar] [CrossRef]
  108. Xu, J.; Chen, Y.; Olopade, O.I. MYC and breast cancer. Genes Cancer 2010, 1, 629–640. [Google Scholar] [CrossRef]
  109. Morse, B.; Rotherg, P.G.; South, V.J.; Spandorfer, J.M.; Astrin, S.M. Insertional mutagenesis of the myc locus by a LINE-1 sequence in a human breast carcinoma. Nature 1988, 333, 87–90. [Google Scholar] [CrossRef]
  110. Li, Z.; Tian, Y.; Tian, N.; Zhao, X.; Du, C.; Han, L.; Zhang, H. Aberrant alternative splicing pattern of ADAR2 downregulates adenosine-to-inosine editing in glioma. Oncol. Rep. 2015, 33, 2845–2852. [Google Scholar] [CrossRef]
  111. Wang, X.; Ren, X.; Liu, W.; Chen, X.; Wei, J.; Gong, Z.; Yan, Y.; Xu, Z. Role of downregulated ADARB1 in lung squamous cell carcinoma. Mol. Med. Rep. 2020, 21, 1517–1526. [Google Scholar] [CrossRef] [PubMed]
  112. Clayton, E.A.; Rishishwar, L.; Huang, T.-C.; Gulati, S.; Ban, D.; McDonald, J.F.; Jordan, I.K. An atlas of TE-derived alternative splicing in cancer. Philos. Trans. R. Soc. B 2020, 375, 20190342. [Google Scholar] [CrossRef] [PubMed]
  113. Ma, Q.; Xu, Y.; Liao, H.; Cai, Y.; Xu, L.; Xiao, D.; Liu, C.; Pu, W.; Zhong, X.; Guo, X. Identification and validation of key genes associated with non-small-cell lung cancer. J. Cell. Physiol. 2019, 234, 22742–22752. [Google Scholar] [CrossRef] [PubMed]
  114. Dubin, M.J.; Scheid, O.M.; Becker, C. Transposons: A blessing curse. Curr. Opin. Plant Biol. 2018, 42, 23–29. [Google Scholar] [CrossRef] [PubMed]
  115. Teugels, E.; De Brakeleer, S.; Goelen, G.; Lissens, W.; Sermijn, E.; De Grève, J. De novo Alu element insertions targeted to a sequence common to the BRCA1 and BRCA2 genes. Hum. Mutat. 2005, 26, 284. [Google Scholar] [CrossRef]
  116. Guilbert, A.; Dhennin-Duthille, I.; Hiani, Y.E.; Haren, N.; Khorsi, H.; Sevestre, H.; Ahidouch, A.; Ouadid-Ahidouch, H. Expression of TRPC6 channels in human epithelial breast cancer cells. BMC Cancer 2008, 8, 125. [Google Scholar] [CrossRef]
  117. Zhang, Y.; Romanish, M.T.; Mager, D.L. Distributions of TEs reveal hazardous zones in mammalian introns. PLoS Comput. Biol. 2011, 7, e1002046. [Google Scholar] [CrossRef]
  118. Cajuso, T.; Sulo, P.; Tanskanen, T.; Katainen, R.; Taira, A.; Hänninen, U.A.; Kondelin, J.; Forsström, L.; Välimäki, N.; Aavikko, M. Retrotransposon insertions can initiate colorectal cancer and are associated with poor survival. Nat. Commun. 2019, 10, 4022. [Google Scholar] [CrossRef]
  119. Becklin, K.L.; Smeester, B.A.; Moriarity, B.S. Cancer gene discovery utilizing sleeping beauty transposon mutagenesis. In Cancer Driver Genes; Humana Press: New York, NY, USA, 2019; pp. 161–170. [Google Scholar]
  120. Burns, K.H. TEs in cancer. Nat. Rev. Cancer 2017, 17, 415–424. [Google Scholar] [CrossRef]
  121. Lev-Maor, G.; Sorek, R.; Shomron, N.; Ast, G. The birth of an alternatively spliced exon: 3’splice-site selection in Alu exons. Science 2003, 300, 1288–1291. [Google Scholar] [CrossRef]
  122. Krull, M.; Brosius, J.R.; Schmitz, J.R. Alu-SINE exonization: En route to protein-coding function. Mol. Biol. Evol. 2005, 22, 1702–1711. [Google Scholar] [CrossRef] [PubMed]
  123. Kocher, O.; Pal, R.; Roberts, M.; Cirovic, C.; Gilchrist, A. Targeted disruption of the PDZK1 gene by homologous recombination. Mol. Cell. Biol. 2003, 23, 1175–1180. [Google Scholar] [CrossRef] [PubMed]
  124. Rebollo, R.; Farivar, S.; Mager, D.L. C-GATE-catalogue of genes affected by TEs. Mob. DNA 2012, 3, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Su, C.; Gao, G.; Schneider, S.; Helt, C.; Weiss, C.; O’Reilly, M.A.; Bohmann, D.; Zhao, J. DNA damage induces downregulation of histone gene expression through the G1 checkpoint pathway. EMBO J. 2004, 23, 1133–1143. [Google Scholar] [CrossRef]
  126. Xie, R.; Medina, R.; Zhang, Y.; Hussain, S.; Colby, J.; Ghule, P.; Sundararajan, S.; Keeler, M.; Liu, L.-J.; Van der Deen, M. The histone gene activator HINFP is a nonredundant cyclin E/CDK2 effector during early embryonic cell cycles. Proc. Natl. Acad. Sci. USA 2009, 106, 12359–12364. [Google Scholar] [CrossRef]
  127. Lucas, B.A.; Lavi, E.; Shiue, L.; Cho, H.; Katzman, S.; Miyoshi, K.; Siomi, M.C.; Carmel, L.; Ares, M., Jr.; Maquat, L.E. Evidence for convergent evolution of SINE-directed Staufen-mediated mRNA decay. Proc. Natl. Acad. Sci. USA 2018, 115, 968–973. [Google Scholar] [CrossRef]
  128. Jung, Y.-D.; Huh, J.-W.; Kim, D.-S.; Kim, Y.-J.; Ahn, K.; Ha, H.-S.; Lee, J.-R.; Yi, J.M.; Moon, J.-W.; Kim, T.-O. Quantitative analysis of transcript variants of CHM gene containing LTR12C element in humans. Gene 2011, 489, 1–5. [Google Scholar] [CrossRef]
  129. Lee, J.Y.; Ji, Z.; Tian, B. Phylogenetic analysis of mRNA polyadenylation sites reveals a role of TEs in evolution of the 3′-end of genes. Nucleic Acids Res. 2008, 36, 5581–5590. [Google Scholar] [CrossRef]
  130. Miki, Y.; Nishisho, I.; Horii, A.; Miyoshi, Y.; Utsunomiya, J.; Kinzler, K.W.; Vogelstein, B.; Nakamura, Y. Disruption of the APC gene by a retrotransposal insertion of L1 sequence in a colon cancer. Cancer Res. 1992, 52, 643–645. [Google Scholar]
  131. Shukla, R.; Upton, K.R.; Muñoz-Lopez, M.; Gerhardt, D.J.; Fisher, M.E.; Nguyen, T.; Brennan, P.M.; Baillie, J.K.; Collino, A.; Ghisletti, S. Endogenous retrotransposition activates oncogenic pathways in hepatocellular carcinoma. Cell 2013, 153, 101–111. [Google Scholar] [CrossRef]
  132. Savage, A.L.; Schumann, G.G.; Breen, G.; Bubb, V.J.; Al-Chalabi, A.; Quinn, J.P. Retrotransposons in the development and progression of amyotrophic lateral sclerosis. J. Neurol. Neurosurg. Psychiatry 2019, 90, 284–293. [Google Scholar] [CrossRef] [PubMed]
  133. Drongitis, D.; Aniello, F.; Fucci, L.; Donizetti, A. Roles of TEs in the different layers of gene expression regulation. Int. J. Mol. Sci. 2019, 20, 5755. [Google Scholar] [CrossRef] [PubMed]
  134. van de Lagemaat, L.N.; Landry, J.-R.; Mager, D.L.; Medstrand, P. TEs in mammals promote regulatory variation and diversification of genes with specialized functions. Trends Genet. 2003, 19, 530–536. [Google Scholar] [CrossRef] [PubMed]
  135. Mori, M.; Staniunas, R.J.; Barnard, G.F.; Jessup, J.M.; Steele, G.D., Jr.; Chen, L.B. The significance of carbonic anhydrase expression in human colorectal cancer. Gastroenterology 1993, 105, 820–826. [Google Scholar] [CrossRef]
  136. Kim, H.J.; Moon, S.J.; Kim, S.-H.; Heo, K.; Kim, J.H. DBC1 regulates Wnt/β-catenin-mediated expression of MACC1, a key regulator of cancer progression, in colon cancer. Cell Death Dis. 2018, 9, 831. [Google Scholar] [CrossRef]
  137. Jun, H.J.; Johnson, H.; Bronson, R.T.; de Feraudy, S.; White, F.; Charest, A. The Oncogenic Lung Cancer Fusion Kinase CD74-ROS Activates a Novel Invasiveness Pathway through E-Syt1 PhosphorylationOncogenic Properties of ROS Fusion Kinases. Cancer Res. 2012, 72, 3764–3774. [Google Scholar] [CrossRef]
  138. Popowski, M.; Templeton, T.D.; Lee, B.-K.; Rhee, C.; Li, H.; Miner, C.; Dekker, J.D.; Orlanski, S.; Bergman, Y.; Iyer, V.R. Bright/Arid3A acts as a barrier to somatic cell reprogramming through direct regulation of Oct4, Sox2, and Nanog. Stem Cell Rep. 2014, 2, 26–35. [Google Scholar] [CrossRef]
  139. Jang, H.S.; Shah, N.M.; Du, A.Y.; Dailey, Z.Z.; Pehrsson, E.C.; Godoy, P.M.; Zhang, D.; Li, D.; Xing, X.; Kim, S. TEs drive widespread expression of oncogenes in human cancers. Nat. Genet. 2019, 51, 611–617. [Google Scholar] [CrossRef]
  140. Babaian, A.; Mager, D.L. Endogenous retroviral promoter exaptation in human cancer. Mob. DNA 2016, 7, 24. [Google Scholar] [CrossRef]
  141. Hur, K.; Cejas, P.; Feliu, J.; Moreno-Rubio, J.; Burgos, E.; Boland, C.R.; Goel, A. Hypomethylation of long interspersed nuclear element-1 (LINE-1) leads to activation of proto-oncogenes in human colorectal cancer metastasis. Gut 2014, 63, 635–646. [Google Scholar] [CrossRef]
  142. Williams, C.S.; Bernard, J.K.; Beckler, M.D.; Almohazey, D.; Washington, M.K.; Smith, J.J.; Frey, M.R. ERBB4 is over-expressed in human colon cancer and enhances cellular transformation. Carcinogenesis 2015, 36, 710–718. [Google Scholar] [CrossRef] [PubMed]
  143. Sharma, S.; Kelly, T.K.; Jones, P.A. Epigenetics in cancer. Carcinogenesis 2010, 31, 27–36. [Google Scholar] [CrossRef] [PubMed]
  144. Mendis, S.R.; Topham, J.T.; Titmuss, E.; Williamson, L.; Pleasance, E.D.; Culibrk, L.; Karasinska, J.; Liu, S.L.; Lee, M.; Aird, J. Comprehensive transcriptome analysis reveals link between epigenetic dysregulation, ERV expression and immunogenicity in metastatic colorectal carcinoma. Am. J. Clin. Oncol. 2019, 37, 3535. [Google Scholar] [CrossRef]
  145. De Cubas, A.A.; Dunker, W.; Zaninovich, A.; Hongo, R.A.; Bhatia, A.; Panda, A.; Beckermann, K.E.; Bhanot, G.; Ganesan, S.; Karijolich, J. DNA hypomethylation promotes TE expression and activation of immune signaling in renal cell cancer. JCI Insight 2020, 5, e137569. [Google Scholar] [CrossRef] [PubMed]
  146. Howard, G.; Eiges, R.; Gaudet, F.; Jaenisch, R.; Eden, A. Activation and transposition of endogenous retroviral elements in hypomethylation induced tumors in mice. Oncogene 2008, 27, 404–408. [Google Scholar] [CrossRef]
  147. Ross, J.P.; Rand, K.N.; Molloy, P.L. Hypomethylation of repeated DNA sequences in cancer. Epigenomics 2010, 2, 245–269. [Google Scholar] [CrossRef]
  148. Watanabe, Y.; Maekawa, M. Methylation of DNA in cancer. Adv. Clin. Chem. 2010, 52, 145–167. [Google Scholar]
  149. Kulis, M.; Queirós, A.C.; Beekman, R.; Martín-Subero, J.I. Intragenic DNA methylation in transcriptional regulation, normal differentiation and cancer. Biochim. Biophys. Acta Gene Regul. Mech. 2013, 1829, 1161–1174. [Google Scholar] [CrossRef]
  150. Florl, A.; Steinhoff, C.; Müller, M.; Seifert, H.; Hader, C.; Engers, R.; Ackermann, R.; Schulz, W. Coordinate hypermethylation at specific genes in prostate carcinoma precedes LINE-1 hypomethylation. Br. J. Cancer 2004, 91, 985–994. [Google Scholar] [CrossRef]
  151. Santourlidis, S.; Florl, A.; Ackermann, R.; Wirtz, H.C.; Schulz, W.A. High frequency of alterations in DNA methylation in adenocarcinoma of the prostate. Prostate 1999, 39, 166–174. [Google Scholar] [CrossRef]
  152. Kim, M.-J.; White-Cross, J.A.; Shen, L.; Issa, J.-P.J.; Rashid, A. Hypomethylation of long interspersed nuclear element-1 in hepatocellular carcinomas. Mod. Pathol. 2009, 22, 442–449. [Google Scholar] [CrossRef] [PubMed]
  153. Ferreira, P.G.; Jares, P.; Rico, D.; Gómez-López, G.; Martínez-Trillos, A.; Villamor, N.; Ecker, S.; González-Pérez, A.; Knowles, D.G.; Monlong, J. Transcriptome characterization by RNA sequencing identifies a major molecular and clinical subdivision in chronic lymphocytic leukemia. Genome Res. 2014, 24, 212–226. [Google Scholar] [CrossRef] [PubMed]
  154. Ecsedi, S.I.; Hernandez-Vargas, H.; Lima, S.C.; Herceg, Z.; Adany, R.; Balazs, M. Transposable hypomethylation is associated with metastatic capacity of primary melanomas. Int. J. Clin. Exp. Pathol. 2013, 6, 2943. [Google Scholar] [PubMed]
  155. Xiang, S.; Liu, Z.; Zhang, B.; Zhou, J.; Zhu, B.-D.; Ji, J.; Deng, D. Methylation status of individual CpG sites within Alu elements in the human genome and Alu hypomethylation in gastric carcinomas. BMC Cancer 2010, 10, 1–11. [Google Scholar] [CrossRef]
  156. Turker, M.S.; Bestor, T.H. Formation of methylation patterns in the mammalian genome. Mutat. Res. Rev. Mutat. Res. 1997, 386, 119–130. [Google Scholar] [CrossRef]
  157. Wang, X.; Fan, J.; Liu, D.; Fu, S.; Ingvarsson, S.; Chen, H. Spreading of Alu methylation to the promoter of the MLH1 gene in gastrointestinal cancer. PLoS ONE 2011, 6, e25913. [Google Scholar] [CrossRef] [PubMed]
  158. Shirley, K.; Reichard, K.; Grover, N. Small Non-coding RNA, miRNA in Gene Regulation. In Fundamentals of RNA Structure and Function; Springer: Cham, Switzerland, 2022; pp. 167–190. [Google Scholar]
  159. Ghosh, A.; Platt, R.N., II; Vandewege, M.W.; Tabassum, R.; Hsu, C.-Y.; Isberg, S.R.; Peterson, D.G.; Finger, J.W., Jr.; Kieran, T.J.; Glenn, T.C. Identification and characterization of miRNAs (miRNAs) and their TE origins in the saltwater crocodile, Crocodylus porosus. Anal. Biochem. 2020, 602, 113781. [Google Scholar] [CrossRef] [PubMed]
  160. Sun, S.-N.; Hu, S.; Shang, Y.-P.; Li, L.-Y.; Zhou, H.; Chen, J.-S.; Yang, J.-F.; Li, J.; Huang, Q.; Shen, C.-P. Relevance function of miRNA-708 in the pathogenesis of cancer. Cell. Signal. 2019, 63, 109390. [Google Scholar] [CrossRef]
  161. Zheng, T.; Li, Y.; Li, W. LncRNA AK024094 aggravates the progression of breast cancer through regulating miRNA-181a. Eur. Rev. Med. Pharmacol. Sci. 2020, 4, 1913–1921. [Google Scholar]
  162. Fang, C.; Huang, X.; Dai, J.; He, W.; Xu, L.; Sun, F. The circular RNA circFARSA sponges miRNA-330-5p in tumor cells with bladder cancer phenotype. BMC Cancer 2022, 22, 373. [Google Scholar] [CrossRef]
  163. Lee, H.-E.; Park, S.-J.; Huh, J.-W.; Imai, H.; Kim, H.-S. The enhancer activity of long interspersed nuclear element derived miRNA 625 induced by NF-κB. Sci. Rep. 2021, 11, 3139. [Google Scholar] [CrossRef] [PubMed]
  164. Lee, H.-E.; Huh, J.-W.; Kim, H.-S. Bioinformatics analysis of evolution and human disease related TE-derived miRNAs. Life 2020, 10, 95. [Google Scholar] [CrossRef] [PubMed]
  165. Rezaei, T.; Amini, M.; Hashemi, Z.S.; Mansoori, B.; Rezaei, S.; Karami, H.; Mosafer, J.; Mokhtarzadeh, A.; Baradaran, B. miRNA-181 serves as a dual-role regulator in the development of human cancers. Free Radic. Biol. Med. 2020, 152, 432–454. [Google Scholar] [CrossRef] [PubMed]
  166. Makondi, P.T.; Wei, P.-L.; Huang, C.-Y.; Chang, Y.-J. Development of novel predictive miRNA/target gene pathways for colorectal cancer distance metastasis to the liver using a bioinformatic approach. PLoS ONE 2019, 14, e0211968. [Google Scholar] [CrossRef]
  167. Wu, S.-G.; Chang, T.-H.; Liu, Y.-N.; Shih, J.-Y. MiRNA in lung cancer metastasis. Cancers 2019, 11, 265. [Google Scholar] [CrossRef]
  168. Liu, B.; Shyr, Y.; Cai, J.; Liu, Q. Interplay between miRNAs and host genes and their role in cancer. Brief. Funct. Genom. 2019, 18, 255–266. [Google Scholar] [CrossRef] [Green Version]
  169. Cappucci, U.; Torromino, G.; Casale, A.M.; Camon, J.; Capitano, F.; Berloco, M.; Mele, A.; Pimpinelli, S.; Rinaldi, A.; Piacentini, L. Stress-induced strain and brain region-specific activation of LINE-1 transposons in adult mice. Stress 2018, 21, 575–579. [Google Scholar] [CrossRef]
  170. Curtin, F.; Bernard, C.; Levet, S.; Perron, H.; Porchet, H.; Médina, J.; Malpass, S.; Lloyd, D.; Simpson, R.; RAINBOW-T1D Investigators. A new therapeutic approach for type 1 diabetes: Rationale for GNbAC1, an anti-HERV-W-Env monoclonal antibody. Diabetes Obes. Metab. 2018, 20, 2075–2084. [Google Scholar] [CrossRef]
  171. Weber, B.; Kimhi, S.; Howard, G.; Eden, A.; Lyko, F. Demethylation of a LINE-1 antisense promoter in the cMet locus impairs Met signalling through induction of illegitimate transcription. Oncogene 2010, 29, 5775–5784. [Google Scholar] [CrossRef]
  172. Houede, N.; Piazza, P.V.; Pourquier, P. LINE-1 as a therapeutic target for castration-resistant prostate cancer. Front. Biosci. 2018, 23, 1292–1309. [Google Scholar]
  173. Sciamanna, I.; De Luca, C.; Spadafora, C. The reverse transcriptase encoded by LINE-1 retrotransposons in the genesis, progression, and therapy of cancer. Front. Chem. 2016, 4, 6. [Google Scholar] [CrossRef] [PubMed]
  174. Kosaka, N.; Takeshita, F.; Yoshioka, Y.; Hagiwara, K.; Katsuda, T.; Ono, M.; Ochiya, T. Exosomal tumor-suppressive microRNAs as novel cancer therapy: “exocure” is anoTher. choice for cancer treatment. Adv. Drug Deliv. Rev. 2013, 65, 376–382. [Google Scholar] [CrossRef] [PubMed]
  175. Zeng, A.; Wei, Z.; Yan, W.; Yin, J.; Huang, X.; Zhou, X.; Li, R.; Shen, F.; Wu, W.; Wang, X. Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma. Cancer Lett. 2018, 436, 10–21. [Google Scholar] [CrossRef] [PubMed]
  176. Binenbaum, Y.; Fridman, E.; Yaari, Z.; Milman, N.; Schroeder, A.; Ben David, G.; Shlomi, T.; Gil, Z. Transfer of miRNA in Macrophage-Derived Exosomes Induces Drug Resistance in Pancreatic AdenocarcinomaExosomes Induce Gemcitabine Resistance in Pancreatic Cancer. Cancer Res. 2018, 78, 5287–5299. [Google Scholar] [CrossRef]
  177. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  178. Li, Y.; Cui, X.; Li, Y.; Zhang, T.; Li, S. Upregulated expression of miR-421 is associated with poor prognosis in non-small-cell lung cancer. Cancer Manag. Res. 2018, 10, 2627. [Google Scholar] [CrossRef]
  179. Wu, Y.; Wei, J.; Zhang, W.; Xie, M.; Wang, X.; Xu, J. Serum exosomal miR-1290 is a potential biomarker for lung adenocarcinoma. OncoTargets Ther. 2020, 13, 7809. [Google Scholar] [CrossRef]
  180. He, X.; Chen, S.-Y.; Yang, Z.; Zhang, J.; Wang, W.; Liu, M.-Y.; Niu, Y.; Wei, X.-M.; Li, H.-M.; Hu, W.-N. miR-4317 suppresses non-small cell lung cancer (NSCLC) by targeting fibroblast growth factor 9 (FGF9) and cyclin D2 (CCND2). J. Exp. Clin. Cancer Res. 2018, 37, 1–16. [Google Scholar] [CrossRef]
  181. Xu, X.; Cao, L.; Zhang, Y.; Lian, H.; Sun, Z.; Cui, Y. MiRNA-1246 inhibits cell invasion and epithelial mesenchymal transition process by targeting CXCR4 in lung cancer cells. Cancer Biomark. 2018, 21, 251–260. [Google Scholar] [CrossRef]
  182. Liu, H.-N.; Qie, P.; Yang, G.; Song, Y.-B. miR-181b inhibits chemoresistance in cisplatin-resistant H446 small cell lung cancer cells by targeting Bcl-2. Arch. Med. Sci. 2018, 14, 745–751. [Google Scholar] [CrossRef]
  183. Cao, J.; Geng, J.; Chu, X.; Wang, R.; Huang, G.; Chen, L. miRNA-885-3p inhibits docetaxel chemoresistance in lung adenocarcinoma by downregulating Aurora A. Oncol. Rep. 2019, 41, 1218–1230. [Google Scholar] [CrossRef]
  184. Song, J.; Su, Z.; Shen, Q. Long non-coding RNA MALAT1 regulates proliferation, apoptosis, migration and invasion via miR-374b-5p/SRSF7 axis in non-small cell lung cancer. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1853–1862. [Google Scholar] [PubMed]
  185. Wang, Y.; Xu, R.; Zhang, D.; Lu, T.; Yu, W.; Wo, Y.; Liu, A.; Sui, T.; Cui, J.; Qin, Y. Circ-ZKSCAN1 regulates FAM83A expression and inactivates MAPK signaling by targeting miR-330-5p to promote non-small cell lung cancer progression. Transl. Lung Cancer Res. 2019, 8, 862. [Google Scholar] [CrossRef] [PubMed]
  186. Liu, C.; Yang, J.; Wu, H.; Li, J. Downregulated miR-585-3p promotes cell growth and proliferation in colon cancer by upregulating PSME3. OncoTargets Ther. 2019, 12, 6525. [Google Scholar] [CrossRef] [PubMed]
  187. Wu, F.; Liu, F.; Dong, L.; Yang, H.; He, X.; Li, L.; Zhao, L.; Jin, S.; Li, G. miR-1273g silences MAGEA3/6 to inhibit human colorectal cancer cell growth via activation of AMPK signaling. Cancer Lett. 2018, 435, 1–9. [Google Scholar] [CrossRef]
  188. Sun, X.; Lin, F.; Sun, W.; Zhu, W.; Fang, D.; Luo, L.; Li, S.; Zhang, W.; Jiang, L. Exosome-transmitted miRNA-335-5p promotes colorectal cancer invasion and metastasis by facilitating EMT via targeting RASA1. Mol. Ther. Nucleic Acids 2021, 24, 164–174. [Google Scholar] [CrossRef]
  189. Cheng, B.; Rong, A.; Zhou, Q.; Li, W. LncRNA LINC00662 promotes colon cancer tumor growth and metastasis by competitively binding with miR-340-5p to regulate CLDN8/IL22 co-expression and activating ERK signaling pathway. J. Exp. Clin. Cancer Res. 2020, 39, 5. [Google Scholar] [CrossRef]
  190. Sun, W.; Wang, X.; Li, J.; You, C.; Lu, P.; Feng, H.; Kong, Y.; Zhang, H.; Liu, Y.; Jiao, R. MiRNA-181a promotes angiogenesis in colorectal cancer by targeting SRCIN1 to promote the SRC/VEGF signaling pathway. Cell Death Dis. 2018, 9, 1–13. [Google Scholar] [CrossRef]
  191. Im, J.; Nam, S.K.; Lee, H.S. MiRNA-552 expression in colorectal cancer and its clinicopathological significance. J. Pathol. Transl. Med. 2021, 55, 125–131. [Google Scholar] [CrossRef]
  192. Sun, S.; Hang, T.; Zhang, B.; Zhu, L.; Wu, Y.; Lv, X.; Huang, Q.; Yao, H. miRNA-708 functions as a tumor suppressor in colorectal cancer by targeting ZEB1 through Akt/mTOR signaling pathway. Am. J. Transl. Res. 2019, 11, 5338. [Google Scholar]
  193. Cui, M.; Chen, M.; Shen, Z.; Wang, R.; Fang, X.; Song, B. LncRNA-UCA1 modulates progression of colon cancer through regulating the miR-28-5p/HOXB3 axis. J. Cell. Biochem. 2019, 120, 6926–6936. [Google Scholar] [CrossRef] [PubMed]
  194. Ahmadian, E.; Janas, D.; Eftekhari, A.; Zare, N. Application of carbon nanotubes in sensing/monitoring of pancreas and liver cancer. Chemosphere 2022, 302, 134826. [Google Scholar] [CrossRef] [PubMed]
  195. Cui, Y.; Xu, H.-F.; Liu, M.-Y.; Xu, Y.-J.; He, J.-C.; Zhou, Y.; Cang, S.-D. Mechanism of exosomal miRNA-224 in development of hepatocellular carcinoma and its diagnostic and prognostic value. World J. Gastroenterol. 2019, 25, 1890. [Google Scholar] [CrossRef] [PubMed]
  196. Wang, G.; Fang, X.; Han, M.; Wang, X.; Huang, Q. MiRNA-493-5p promotes apoptosis and suppresses proliferation and invasion in liver cancer cells by targeting VAMP2. Int. J. Mol. Med. 2018, 41, 1740–1748. [Google Scholar] [PubMed]
  197. He, L.; Meng, D.; Zhang, S.-H.; Zhang, Y.; Deng, Z.; Kong, L.-B. miRNA-608 inhibits human hepatocellular carcinoma cell proliferation via targeting the BET family protein BRD4. Biochem. Biophys. Res. Commun. 2018, 501, 1060–1067. [Google Scholar] [CrossRef]
  198. Tyagi, K.; Mandal, S.; Roy, A. Recent advancements in therapeutic targeting of the Warburg effect in refractory ovarian cancer: A promise towards disease remission. Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188563. [Google Scholar] [CrossRef]
  199. Liu, W.; Kang, L.; Han, J.; Wang, Y.; Shen, C.; Yan, Z.; Tai, Y.; Zhao, C. miR-342-3p suppresses hepatocellular carcinoma proliferation through inhibition of IGF-1R-mediated Warburg effect. OncoTargets Ther. 2018, 11, 1643. [Google Scholar] [CrossRef]
  200. Xu, F.; Yan, J.-J.; Gan, Y.; Chang, Y.; Wang, H.-L.; He, X.-X.; Zhao, Q. miR-885-5p negatively regulates warburg effect by silencing hexokinase 2 in liver cancer. Mol. Ther. Nucleic Acids 2019, 18, 308–319. [Google Scholar] [CrossRef] [Green Version]
  201. Wei, L.Q.; Li, L.; Lu, C.; Liu, J.; Chen, Y.; Wu, H. Involvement of H19/miR-326 axis in hepatocellular carcinoma development through modulating TWIST1. J. Cell. Physiol. 2019, 234, 5153–5162. [Google Scholar] [CrossRef]
  202. Yan, S.; Tang, Z.; Chen, K.; Liu, Y.; Yu, G.; Chen, Q.; Dang, H.; Chen, F.; Ling, J.; Zhu, L. Long non-coding RNA MIR31HG inhibits hepatocellular carcinoma proliferation and metastasis by sponging miRNA-575 to modulate ST7L expression. J. Exp. Clin. Cancer Res. 2018, 37, 214. [Google Scholar] [CrossRef]
  203. Hu, L.; Wu, H.; Wan, X.; Liu, L.; He, Y.; Zhu, L.; Liu, S.; Yao, H.; Zhu, Z. MiRNA-585 suppresses tumor proliferation and migration in gastric cancer by directly targeting MAPK1. Biochem. Biophys. Res. Commun. 2018, 499, 52–58. [Google Scholar] [CrossRef] [PubMed]
  204. Liu, W.-L.; Wang, H.-X.; Shi, C.-X.; Shi, F.-Y.; Zhao, L.-Y.; Zhao, W.; Wang, G.-H. MiRNA-1269 promotes cell proliferation via the AKT signaling pathway by targeting RASSF9 in human gastric cancer. Cancer Cell Int. 2019, 19, 308. [Google Scholar] [CrossRef] [PubMed]
  205. Zhao, Y.; Zhang, J.; Yang, W.; Yang, Z.; Zhou, K. MiRNA-552 accelerates the progression of gastric cancer by targeting FOXO1 and regulating PI3K/AKT pathway. J. Oncol. 2021, 2021, 9966744. [Google Scholar] [CrossRef] [PubMed]
  206. Nabatchian, F.; Naiini, M.R.; Moradi, A.; Tabatabaeian, H.; Hoghoughi, N.; Azadeh, M.; Ghaedi, K. miR-581-related single nucleotide polymorphism, rs2641726, located in MUC4 gene, is associated with gastric cancer incidence. Indian J. Clin. Biochem. 2019, 34, 347–351. [Google Scholar] [CrossRef] [PubMed]
  207. Lin, Z.; Zhou, Z.; Guo, H.; He, Y.; Pang, X.; Zhang, X.; Liu, Y.; Ao, X.; Li, P.; Wang, J. Long non-coding RNA gastric cancer-related lncRNA1 mediates gastric malignancy through miRNA-885-3p and cyclin-dependent kinase 4. Cell Death Dis. 2018, 9, 1–16. [Google Scholar] [CrossRef]
  208. Xiao, J.; Lin, L.; Luo, D.; Shi, L.; Chen, W.; Fan, H.; Li, Z.; Ma, X.; Ni, P.; Yang, L. Long non-coding RNA TRPM2-AS acts as a miRNA sponge of miR-612 to promote gastric cancer progression and radioresistance. Oncogenesis 2020, 9, 1–15. [Google Scholar] [CrossRef]
  209. Wang, S.; Cheng, Y.; Yang, P.; Qin, G. Silencing of long non-coding RNA LINC00324 interacts with miRNA-3200-5p to attenuate the tumorigenesis of gastric cancer via regulating BCAT1. Gastroenterol. Res. Pract. 2020, 2020, 4159298. [Google Scholar] [CrossRef]
  210. Li, B.; Liang, L.; Chen, Y.; Liu, J.; Wang, Z.; Mao, Y.; Zhao, K.; Chen, J. Circ_0008287 promotes immune escape of gastric cancer cells through impairing miRNA-548c-3p-dependent inhibition of CLIC1. Int. Immunopharmacol. 2022, 111, 108918. [Google Scholar] [CrossRef]
  211. Wang, Y.; Yin, H.; Chen, X. Circ-LDLRAD3 enhances cell growth, migration, and invasion and inhibits apoptosis by regulating MiR-224-5p/NRP2 axis in gastric cancer. Dig. Dis. Sci. 2021, 66, 3862–3871. [Google Scholar] [CrossRef]
  212. Miao, Y.; Zhang, Y.; Wang, L.; Yin, L. Identifying the diagnostic value of miRNA-421 in gastric cancer patients: A meta-analysis. bioRxiv 2018, 468983. [Google Scholar] [CrossRef]
  213. Wang, Y.; Liu, Z.; Shen, J. MiRNA-421-targeted PDCD4 regulates breast cancer cell proliferation. Int. J. Mol. Med. 2019, 43, 267–275. [Google Scholar] [PubMed]
  214. Ghaemi, Z.; Soltani, B.M.; Mowla, S.J. MiRNA-326 functions as a tumor suppressor in breast cancer by targeting ErbB/PI3K signaling pathway. Front. Oncol. 2019, 9, 653. [Google Scholar] [CrossRef] [PubMed]
  215. Shi, S.; Chen, X.; Liu, H.; Yu, K.; Bao, Y.; Chai, J.; Gao, H.; Zou, L. LGR5 acts as a target of miR-340-5p in the suppression of cell progression and drug resistance in breast cancer via Wnt/β-catenin pathway. Gene 2019, 683, 47–53. [Google Scholar] [CrossRef] [PubMed]
  216. Bianchini, G.; De Angelis, C.; Licata, L.; Gianni, L. Treatment landscape of triple-negative breast cancer—Expanded options, evolving needs. Nat. Rev. Clin. Oncol. 2022, 19, 91–113. [Google Scholar] [CrossRef] [PubMed]
  217. Shi, J.; Liu, F.; Song, Y. Progress: Targeted therapy, immunotherapy, and new chemotherapy strategies in advanced triple-negative breast cancer. Cancer Manag. Res. 2020, 12, 9375. [Google Scholar] [CrossRef]
  218. Zeng, X.; Ma, X.; Guo, H.; Wei, L.; Zhang, Y.; Sun, C.; Han, N.; Sun, S.; Zhang, N. MiRNA-582-5p promotes triple-negative breast cancer invasion and metastasis by antagonizing CMTM8. Bioengineered 2021, 12, 10126–10135. [Google Scholar] [CrossRef]
  219. Son, D.; Kim, Y.; Lim, S.; Kang, H.-G.; Kim, D.-H.; Park, J.W.; Cheong, W.; Kong, H.K.; Han, W.; Park, W.-Y. miR-374a-5p promotes tumor progression by targeting ARRB1 in triple negative breast cancer. Cancer Lett. 2019, 454, 224–233. [Google Scholar] [CrossRef]
  220. Zhang, L.; Zhang, X.; Wang, X.; He, M.; Qiao, S. MiRNA-224 promotes tumorigenesis through downregulation of caspase-9 in triple-negative breast cancer. Dis. Markers 2019, 2019, 7378967. [Google Scholar]
  221. Chen, T.; Yang, Z.; Liu, C.; Wang, L.; Yang, J.; Chen, L.; Li, W. Circ_0078767 suppresses non-small-cell lung cancer by protecting RASSF1A expression via sponging miR-330-3p. Cell Prolif. 2019, 52, e12548. [Google Scholar] [CrossRef] [Green Version]
  222. Liu, X.; Ma, J.; Xu, F.; Li, L. TINCR suppresses proliferation and invasion through regulating miR-544a/FBXW7 axis in lung cancer. Biomed. PharmacoTher. 2018, 99, 9–17. [Google Scholar] [CrossRef]
  223. Zhou, Y.; Zheng, X.; Xu, B.; Chen, L.; Wang, Q.; Deng, H.; Jiang, J. Circular RNA hsa_circ_0004015 regulates the proliferation, invasion, and TKI drug resistance of non-small cell lung cancer by miR-1183/PDPK1 signaling pathway. Biochem. Biophys. Res. Commun. 2019, 508, 527–535. [Google Scholar] [CrossRef] [PubMed]
  224. Ping, W.; Gao, Y.; Fan, X.; Li, W.; Deng, Y.; Fu, X. MiR-181a contributes gefitinib resistance in non-small cell lung cancer cells by targeting GAS7. Biochem. Biophys. Res. Commun. 2018, 495, 2482–2489. [Google Scholar] [CrossRef] [PubMed]
  225. Zhang, Z.; Wang, Y.; Zhang, W.; Li, J.; Liu, W.; Lu, W. Long non-coding RNA SNHG14 exerts oncogenic functions in non-small cell lung cancer through acting as an miR-340 sponge. Biosci. Rep. 2019, 39, BSR20180941. [Google Scholar] [CrossRef] [PubMed]
  226. Li, X.; Yu, M.; Yang, C. YY1-mediated overexpression of long non-coding RNA MCM3AP-AS1 accelerates angiogenesis and progression in lung cancer by targeting miR-340-5p/KPNA4 axis. J. Cell. Biochem. 2020, 121, 2258–2267. [Google Scholar] [CrossRef] [PubMed]
  227. Wang, M.; Sun, X.; Yang, Y.; Jiao, W. Long non-coding RNA OIP5-AS1 promotes proliferation of lung cancer cells and leads to poor prognosis by targeting miR-378a-3p. Thorac. Cancer 2018, 9, 939–949. [Google Scholar] [CrossRef]
  228. Yu, W.; Jiang, H.; Zhang, H.; Li, J. Hsa_circ_0003998 promotes cell proliferation and invasion by targeting miR-326 in non-small cell lung cancer. OncoTargets Ther. 2018, 11, 5569. [Google Scholar] [CrossRef]
  229. Wang, R.; Xu, J.; Xu, J.; Zhu, W.; Qiu, T.; Li, J.; Zhang, M.; Wang, Q.; Xu, T.; Guo, R. MiR-326/Sp1/KLF3: A novel regulatory axis in lung cancer progression. Cell Prolif. 2019, 52, e12551. [Google Scholar] [CrossRef]
  230. Yu, H.; Wang, X.; Han, X.; Cao, B. MiR-608 exerts tumor suppressive function in lung adenocarcinoma by directly targeting MIF. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 4908–4916. [Google Scholar]
  231. Zhang, Q.; Zhang, C.; Ma, J.-X.; Ren, H.; Sun, Y.; Xu, J.-Z. Circular RNA PIP5K1A promotes colon cancer development through inhibiting miR-1273a. World J. Gastroenterol. 2019, 25, 5300. [Google Scholar] [CrossRef]
  232. Qu, R.; Hao, S.; Jin, X.; Shi, G.; Yu, Q.; Tong, X.; Guo, D. MiRNA-374b reduces the proliferation and invasion of colon cancer cells by regulation of LRH-1/Wnt signaling. Gene 2018, 642, 354–361. [Google Scholar] [CrossRef]
  233. Zhang, L.; Wang, Y.; Wang, L.; Yin, G.; Li, W.; Xian, Y.; Yang, W.; Liu, Q. miR-23c suppresses tumor growth of human hepatocellular carcinoma by attenuating ERBB2IP. Biomed. PharmacoTher. 2018, 107, 424–432. [Google Scholar] [CrossRef]
  234. Tao, J.; Liu, Z.; Wang, Y.; Wang, L.; Yin, G.; Yang, W.; Tu, K.; Liu, Q. MiRNA-645 represses hepatocellular carcinoma progression by inhibiting SOX30-mediated p53 transcriptional activation. Int. J. Biol. Macromol. 2019, 121, 214–222. [Google Scholar] [CrossRef] [PubMed]
  235. Komoll, R.-M.; Hu, Q.; Olarewaju, O.; von Döhlen, L.; Yuan, Q.; Xie, Y.; Tsay, H.-C.; Daon, J.; Qin, R.; Manns, M.P. MiRNA-342-3p is a potent tumour suppressor in hepatocellular carcinoma. J. Hepatol. 2021, 74, 122–134. [Google Scholar] [CrossRef] [PubMed]
  236. Fu, H.; Zhang, J.; Pan, T.; Ai, S.; Tang, L.; Wang, F. miR-378a enhances the sensitivity of liver cancer to sorafenib by targeting VEGFR, PDGFRβ and c-Raf. Mol. Med. Rep. 2018, 17, 4581–4588. [Google Scholar] [CrossRef] [PubMed]
  237. Xu, L.; Feng, X.; Hao, X.; Wang, P.; Zhang, Y.; Zheng, X.; Li, L.; Ren, S.; Zhang, M.; Xu, M. CircSETD3 (Hsa_circ_0000567) acts as a sponge for miRNA-421 inhibiting hepatocellular carcinoma growth. J. Exp. Clin. Cancer Res. 2019, 38, 98. [Google Scholar] [CrossRef]
  238. Wang, Y.-N.; Xu, F.; Zhang, P.; Wang, P.; Wei, Y.-N.; Wu, C.; Cheng, S.-J. MiRNA-575 regulates development of gastric cancer by targeting PTEN. Biomed. PharmacoTher. 2019, 113, 108716. [Google Scholar] [CrossRef]
  239. Wu, Q.; Wang, H.; Liu, L.; Zhu, K.; Yu, W.; Guo, J. Hsa_circ_0001546 acts as a miRNA-421 sponge to inhibit the chemoresistance of gastric cancer cells via ATM/Chk2/p53-dependent pathway. Biochem. Biophys. Res. Commun. 2020, 521, 303–309. [Google Scholar] [CrossRef]
  240. Lu, Q.; Chen, Y.; Sun, D.; Wang, S.; Ding, K.; Liu, M.; Zhang, Y.; Miao, Y.; Liu, H.; Zhou, F. MiRNA-181a functions as an oncogene in gastric cancer by targeting caprin-1. Front. Pharmacol. 2019, 9, 1565. [Google Scholar] [CrossRef]
  241. Hu, X.; Zhang, M.; Miao, J.; Wang, X.; Huang, C. miRNA-4317 suppresses human gastric cancer cell proliferation by targeting ZNF322. Cell Biol. Int. 2018, 42, 923–930. [Google Scholar] [CrossRef]
  242. Kong, X.; Zhang, J.; Li, J.; Shao, J.; Fang, L. MiR-130a-3p inhibits migration and invasion by regulating RAB5B in human breast cancer stem cell-like cells. Biochem. Biophys. Res. Commun. 2018, 501, 486–493. [Google Scholar] [CrossRef]
  243. Cheng, Y.; Li, Z.; Xie, J.; Wang, P.; Zhu, J.; Li, Y.; Wang, Y. MiRNA-224-5p inhibits autophagy in breast cancer cells via targeting Smad4. Biochem. Biophys. Res. Commun. 2018, 506, 793–798. [Google Scholar] [CrossRef] [PubMed]
  244. Zhai, L.-Y.; Li, M.-X.; Pan, W.-L.; Chen, Y.; Li, M.-M.; Pang, J.-X.; Zheng, L.; Chen, J.-X.; Duan, W.-J. In situ detection of plasma exosomal miRNA-1246 for breast cancer diagnostics by a Au nanoflare probe. ACS Appl. Mater. Interfaces 2018, 10, 39478–39486. [Google Scholar] [CrossRef] [PubMed]
  245. Ramchandani, D.; Lee, S.K.; Yomtoubian, S.; Han, M.S.; Tung, C.-H.; Mittal, V. Nanoparticle Delivery of miR-708 Mimetic Impairs Breast Cancer MetastasismiR-708 Mimetic in TNBC Therapy. Mol. Cancer Ther. 2019, 18, 579–591. [Google Scholar] [CrossRef] [PubMed]
  246. Dong, Y.; Liu, Y.; Jiang, A.; Li, R.; Yin, M.; Wang, Y. MiRNA-335 suppresses the proliferation, migration, and invasion of breast cancer cells by targeting EphA4. Mol. Cell. Biochem. 2018, 439, 95–104. [Google Scholar] [CrossRef] [PubMed]
  247. Pan, Y.; Hu, J.; Ma, J.; Qi, X.; Zhou, H.; Miao, X.; Zheng, W.; Jia, L. MiR-193a-3p and miR-224 mediate renal cell carcinoma progression by targeting alpha-2, 3-sialyltransferase IV and the phosphatidylinositol 3 kinase/Akt pathway. Mol. Carcinog. 2018, 57, 1067–1077. [Google Scholar] [CrossRef] [PubMed]
  248. Wang, H.; Guo, W.; Jian, Q.; Xue, K.; Huang, M.; Chi, S.; Li, C.; Li, C. MiRNA-340 inhibits squamous cell carcinoma cell proliferation, migration and invasion by downregulating RhoA. J. Dermatol. Sci. 2018, 92, 197–206. [Google Scholar] [CrossRef]
  249. Wang, S.; Zhang, G.; Zheng, W.; Xue, Q.; Wei, D.; Zheng, Y.; Yuan, J. MiR-454-3p and miR-374b-5p suppress migration and invasion of bladder cancer cells through targetting ZEB2. Biosci. Rep. 2018, 38, BSR20181436. [Google Scholar] [CrossRef]
  250. Liu, M.; Chen, Y.; Huang, B.; Mao, S.; Cai, K.; Wang, L.; Yao, X. Tumor-suppressing effects of miRNA-612 in bladder cancer cells by targeting malic enzyme 1 expression. Int. J. Oncol. 2018, 52, 1923–1933. [Google Scholar]
  251. Li, L.; Ma, L. Upregulation of miR-582-5p regulates cell proliferation and apoptosis by targeting AKT3 in human endometrial carcinoma. Saudi J. Biol. Sci. 2018, 25, 965–970. [Google Scholar] [CrossRef]
  252. Li, N.; Wang, C.; Zhang, P.; You, S. Emodin inhibits pancreatic cancer EMT and invasion by up-regulating miRNA-1271. Mol. Med. Rep. 2018, 18, 3366–3374. [Google Scholar]
  253. Xu, X.; Zheng, S. MiR-887-3p negatively regulates STARD13 and promotes pancreatic cancer progression. Cancer Manag. Res. 2020, 12, 6137. [Google Scholar] [CrossRef] [PubMed]
  254. Zhao, W.; Han, T.; Li, B.; Ma, Q.; Yang, P.; Li, H. miR-552 promotes ovarian cancer progression by regulating PTEN pathway. J. Ovarian Res. 2019, 12, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Yu, L.M.; Wang, W.W.; Qi, R.; Leng, T.G.; Zhang, X.L. MiRNA-224 inhibition prevents progression of cervical carcinoma by targeting PTX3. J. Cell. Biochem. 2018, 119, 10278–10290. [Google Scholar] [CrossRef] [PubMed]
  256. Li, G.C.; Cao, X.Y.; Li, Y.N.; Qiu, Y.Y.; Li, Y.N.; Liu, X.J.; Sun, X.X. MiRNA-374b inhibits cervical cancer cell proliferation and induces apoptosis through the p38/ERK signaling pathway by binding to JAM-2. J. Cell. Physiol. 2018, 233, 7379–7390. [Google Scholar] [CrossRef]
  257. Nakashima, H.; Yoshida, R.; Hirosue, A.; Kawahara, K.; Sakata, J.; Arita, H.; Yamamoto, T.; Toya, R.; Murakami, R.; Hiraki, A. Circulating miRNA-1290 as a potential biomarker for response to chemoradiotherapy and prognosis of patients with advanced oral squamous cell carcinoma: A single-center retrospective study. Tumor Biol. 2019, 41, 1010428319826853. [Google Scholar] [CrossRef]
  258. Lin, S.-S.; Peng, C.-Y.; Liao, Y.-W.; Chou, M.-Y.; Hsieh, P.-L.; Yu, C.-C. miR-1246 targets CCNG2 to enhance cancer stemness and chemoresistance in oral carcinomas. Cancers 2018, 10, 272. [Google Scholar] [CrossRef]
  259. Zhu, G.; Zhou, L.; Liu, H.; Shan, Y.; Zhang, X. MiRNA-224 promotes pancreatic cancer cell proliferation and migration by targeting the TXNIP-mediated HIF1α pathway. Cell. Physiol. Biochem. 2018, 48, 1735–1746. [Google Scholar] [CrossRef]
Figure 1. Effects of alternative splicing on biological processes related to cancer progression through the generation of cancer-specific transcripts. Alternative splicing events in cancer are activated by mutations in cis-acting elements and aberrant expression levels of trans-acting factors. The initiation of alternative splicing is stimulated by aggregation of splicing-mediated factors to precursor mRNA. This procedure is mediated by diverse regulatory processes including exon skipping, alternative 5′ or 3′ usage, mutually exclusive exons, intron retention, and use of alternative first or last exons. The production of cancer-specific transcripts created by alternative splicing is implicated in cancer biological processes; apoptosis, cell cycle, metabolism, invasion, proliferation, and angiogenesis. Light yellow, light blue and light orange rectangles indicated basal exons. Yellow rectangle indicated newly integrated exon. Pre-mRNA—precursor mRNA; SS—splicing site; SR protein—Serine/arginine—rich protein; ESE—exonic splicing enhancers; ESS—exonic splicing silencers; ISE—intronic splicing enhancers; ISS—intronic splicing silencers; hn-RNP—heterogeneous nuclear ribonucleoproteins.
Figure 1. Effects of alternative splicing on biological processes related to cancer progression through the generation of cancer-specific transcripts. Alternative splicing events in cancer are activated by mutations in cis-acting elements and aberrant expression levels of trans-acting factors. The initiation of alternative splicing is stimulated by aggregation of splicing-mediated factors to precursor mRNA. This procedure is mediated by diverse regulatory processes including exon skipping, alternative 5′ or 3′ usage, mutually exclusive exons, intron retention, and use of alternative first or last exons. The production of cancer-specific transcripts created by alternative splicing is implicated in cancer biological processes; apoptosis, cell cycle, metabolism, invasion, proliferation, and angiogenesis. Light yellow, light blue and light orange rectangles indicated basal exons. Yellow rectangle indicated newly integrated exon. Pre-mRNA—precursor mRNA; SS—splicing site; SR protein—Serine/arginine—rich protein; ESE—exonic splicing enhancers; ESS—exonic splicing silencers; ISE—intronic splicing enhancers; ISS—intronic splicing silencers; hn-RNP—heterogeneous nuclear ribonucleoproteins.
Ijms 23 10918 g001
Figure 2. The induction mechanisms of alternative splicing by integrated TEs (A) basal splicing process without TE integration. Insertion of TE is interrelated with the progression of alternative splicing through induction mechanisms; (B) exonization, (C) exon disruption, (D) providing splicing donor/acceptor sites, (E) providing stop codons, (F) alternative enhancer or repressor, (G) alternative promoter, and (H) epigenetic alteration. Yellow, light purple and orange rectangles indicated basal exons.
Figure 2. The induction mechanisms of alternative splicing by integrated TEs (A) basal splicing process without TE integration. Insertion of TE is interrelated with the progression of alternative splicing through induction mechanisms; (B) exonization, (C) exon disruption, (D) providing splicing donor/acceptor sites, (E) providing stop codons, (F) alternative enhancer or repressor, (G) alternative promoter, and (H) epigenetic alteration. Yellow, light purple and orange rectangles indicated basal exons.
Ijms 23 10918 g002
Figure 3. General regulatory process of miRNAs derived from TEs to control the expression of oncogenic transcripts at the post-transcriptional level. RISC-RNA-induced silencing complex. Yellow, light purple and orange rectangles indicated basal exons.
Figure 3. General regulatory process of miRNAs derived from TEs to control the expression of oncogenic transcripts at the post-transcriptional level. RISC-RNA-induced silencing complex. Yellow, light purple and orange rectangles indicated basal exons.
Ijms 23 10918 g003
Figure 4. Impact of cancer-specific alternative splicing promoted by TE integration. Integrated TE is a critical regulator of many aberrant cancer-related biological processes implicated in cancer pathogenesis. (A) Fundamental splicing process in normal cell biology. (B) Macroscopic regulatory mechanism of cancer-specific transcript expression under the cancer environment. EMT: Epithelial-Mesenchymal Transition. Yellow, light purple and orange rectangles indicated basal exons.
Figure 4. Impact of cancer-specific alternative splicing promoted by TE integration. Integrated TE is a critical regulator of many aberrant cancer-related biological processes implicated in cancer pathogenesis. (A) Fundamental splicing process in normal cell biology. (B) Macroscopic regulatory mechanism of cancer-specific transcript expression under the cancer environment. EMT: Epithelial-Mesenchymal Transition. Yellow, light purple and orange rectangles indicated basal exons.
Ijms 23 10918 g004
Table 1. Differential expression of cancer-specific isoforms produced by alternative splicing events that adjust cancer-related biological processes.
Table 1. Differential expression of cancer-specific isoforms produced by alternative splicing events that adjust cancer-related biological processes.
Cancer Related
Biological Process
Type of Alternative SplicingGeneProteinIsoformRegulatory ProcessRef
ApoptosisAlternative 5′ SS usageBCL2L1Bcl-2-Like Protein 1BCl-XLThrough the alternative use of two competing 5′ SSs in exon 2, produced BCL-XL which has an antiapoptotic effect and functions as a dominant regulator.[53,54,55]
Exon skippingSYKSpleen Associated Tyrosine KinaseSYK(S)Switching SYK(L) to SYK(S) generated by exon 9 skipping induces apoptosis in ovarian cancer.[56]
Intron retentionSTAT2Signal Transducer and Activator of Transcription 2STAT2 + I19STAT2 + I19, splice variant containing intron 19 which has a stop codon before the Src homology 2 domain, leads to disruption of STAT dimerization and suppresses IFN-induced apoptosis in IFN-resistant cells.[57]
Exon skippingASPP2Apoptosis-Stimulating of P53 Protein 2ASPP2KASPP2K, which has a truncated C-terminal domain losing the p53 binding regions by exon skipping, possesses dominant-negative activity, impairing the induction of p53 dependent apoptosis and promoting cancer aggressiveness.[58]
Exon skippingFASFas Cell Surface Death ReceptorsFASAn alternatively spliced isoform, soluble Fas (sFAS), generated by the skipping of exon 6 that encodes the transmembrane domain, cannot localize to the plasma membrane. As a result, upregulated sFAS inhibits the extrinsic pathway of apoptosis in various cancer types.[59,60,61]
Exon inclusionMCL1Myeloid Leukemia Cell Differentiation Protein Mcl-1MCL1-LMelanocytes upregulate MCL-1L, a splicing variant of MCL1 by exon 2 inclusion, in response to UVB radiation to protect themselves against apoptosis, whereas melanoma cells elevating MCL1-L expression without UV exposure are resistant to apoptosis.[62]
Invasion
(EMT)
Exon skippingENAH (MENA)ENAH Actin
Regulator
MENA v6ENAH (known as Mena), controls actin nucleation as well as cell morphology and motility. Expression of the exon skipped splicing isoform, Mena11a, has been correlated with epithelial markers and decreased invasion. Inversely, increased expression of MenaINV by exon inclusion has been associated with mesenchymal markers and increased invasion and metastasis.[63,64,65,66]
Exon inclusionMENA INV
Alternative 5′ SS usageKLF6Kruppel Like
Factor 6
KLF6-SV1KLF6-SV1 uses an alternative 5′SS, causing frameshift, and produces a protein isoform that contains 21 novel amino acids but lacking all three of the zinc finger domains. Upregulated expression of KLF6-SV1 increases cell survival, migration, and invasion in various cancer cells.[67,68]
Exon inclusionCD44CD44 AntigenCD44v8-10Expression of CD44v8-10, an alternative isoform including the variable portion of exon 8 to 10, induces a higher metastatic potential of cancer cells than the standard form of CD44 in breast cancer cell lines.[69]
CD44v6CD44 variant including variable exon 6 (CD44v6) has been identified that promotes the development of metastasis by involving epithelial-mesenchymal transition in cancers.[70,71]
Alternative 5′ SS usageCTNND1p120-cateninp120-1Ap120-catenin (p120ctn) isoforms produced by alternative 5′SS usage, p120-1A, and -3A, induced the EMT of tumor cells. Especially, in non-small cell lung cancer (NSCLC), both p120-1A and - 3A inhibited EMT and decreased cell invasiveness in cells with membrane E-cadherin. In cells with cytoplasmic E-cadherin, p120-1A stimulated EMT and cell invasiveness, while p120-3A prevented EMT and decreased cell invasiveness.[72]
p120-3A
Mutually exclusive exonFGFRFibroblast Growth Factor ReceptorFGFR lllcIncreased level of FGFR-IIIc by mutually exclusive exon9 has been detected in a variety of tumors and correlated with tumor progression, such as increased grading and invasiveness, by promoting cancer cells to acquire mesenchymal characteristics.[73,74,75]
Exon skippingRONMacrophage Stimulating 1 ReceptorΔRONThe skipping of Exon 11(ΔRON) brought about the deletion of an extra cellular domain that affects the proteolytic maturation of protein and increases cancer invasiveness through sustaining constitutively active status.[76,77]
ProliferationExon inclusionRPS6KB1Ribosomal Protein
S6 Kinase B1
RPS6KB1-2RPS6KB1-2 made by inclusion of three cassette exons 6a, 6b, and 6c, caused the shorter isoform to lack a portion of the kinase domain. RPS6KB1-2 has contributed to cell proliferation and tumor growth via mTORC1 and 4E-BP1 phosphorylation.[78,79]
Exon inclusionNUMBNUMB Endocytic Adaptor ProteinNUMB-PRR(L)In lung cancer cells, RBM10 mutations identified that disrupt splicing regulation of NUMB (Exon 9 inclusion) which is a key target of RBM5, 6, and 10 in the control of cell proliferation, to correlate with cell growth.[80]
Exon inclusionSYKSpleen Associated Tyrosine KinaseSYK(L)SYK(L), which includes exon 9 compared to the shorter isoform (SYK(S)), stimulates cell survival and tumor malignancy in many cancers by driving expression of epidermal growth factor.[56]
Exon skippingMDM2E3 Ubiquitin-Protein Ligase Mdm2MDM2-ANormal type of MDM2 could bind to p53 and facilitate proteasomal degradation of p53 as an ubiquitin ligase. Four of the splice isoforms (MDM2-A, -B, -C, and -D) by exon skipping in human cancers lack part of the p53-binding domain. Spliced isoforms could not bind to p53, enhancing degradation of p53 and cell proliferation.[81,82,83]
MDM2-B
MDM2-C
MDM2-D
AngiogenesisAlternative 3′ SS usageVEGFVascular Endothelial Growth FactorVEGFxxxVEGFxxx isoforms, produced by alternative 3′SS usage, were overexpressed in many cancers, and resulted in proangiogenic effects.[84]
Alternative 3′ SS usageVEGFVascular Endothelial Growth FactorVEGF 165sVEGFR1-113, a truncated version of VEGFR1, lacks its transmembrane and tyrosine kinase domains due to intron 13 retention.VEGF165 is a pro-angiogenic factor made by 3′ SS usage. One study identified that sVEGFR1-113 is considered to be a natural antagonist of VEGFA and upregulated under the mechanism associating the VEGF165/SOX2/SRSF2 network in anti-angiogenic therapied squamous lung carcinoma cells.[85,86]
Intron retentionVEGFRVascular Endothelial Growth Factor Receptor 1sVEGFR1-113
Dysregulated
metabolism
Mutually exclusive exonPKMPyruvate Kinase M2PKM2PKM2 had mutually exclusive exons containing Exon 10 not Exon 9 and is ubiquitously expressed in tumors. Substituting PKM2 with PKM1 in the tumor decreases lactate production and increases oxidative phosphorylation. Therefore, tumor growth is repressed.[87,88]
Immune
response
Exon skippingBRAFB-Raf Proto-Oncogene, Serine/Threonine KinaseBRAF(V600E)BRAF(V600E) transcripts by exon skipping (exon 4–8) brought about in-frame deletion of the N-terminal RAS-binding domain, resulting in melanoma cell resistance which is insensitive to inhibitors such as drug (PLX4032).[89]
Exon skippingCEACAM1CEA Cell Adhesion Molecule 1CEACAM1(S)The short isoform of CEACAM1, CEACAM1 (S) upregulated in many cancer types. This variant enlarged secretory IgA production by B cells and was associated with poor prognosis and peritoneal dissemination in gastric cancer.[90,91,92]
Cell cycleIntron retentionCCND1CyclinD1CyclinD1bThe formation of the cyclin D1b variant was linked with intron 4 retention, also concerned with cell cycle progression and proliferation in various cancers, competing with the same target, CDK4 with Cyclin D1a.[93,94]
Table 3. The list of miRNAs dysregulated in various cancer types in the last 5 years.
Table 3. The list of miRNAs dysregulated in various cancer types in the last 5 years.
Cancer TypeMiRNASubclassSuperfamilyTarget GeneReference
Renal cancerhsa-miR-224-3pDNA transposonDNA transposonST3GalIV[247]
Skin cancerhsa-miR-340-5pDNA transposonTcMar-MarinerRhoA[248]
Bladder cancerhsa-miR-374b-5pLINEL2ZEB2[249]
hsa-miR-330-5pSINEMIRcircFARSA[162]
hsa-miR-612SINEMIRME1[250]
Cervical cancerhsa-miR-224-5pDNA transposonDNA transposonPTX3[255]
hsa-miR-374b-5pLINEL2JAM2[256]
Endometrial cancerhsa-miR-582-5pLINECR1AKT3[251]
Oral cancerhsa-miR-1290DNA transposonTcMar-Tigger-[257]
hsa-miR-1246LTRERVL-MaLRCCNG2[258]
Pancreatic cancerhsa-miR-1271LINEL2E-cadherin, ZEB1, TWIST1[252]
hsa-miR-224-5pDNA transposonDNA transposonTXNIP[259]
hsa-miR-887-3pLINEL2STARD13[253]
Ovarian cancerhsa-miR-552LINEL1PTEN[254]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, W.R.; Park, E.G.; Lee, Y.J.; Bae, W.H.; Lee, D.H.; Kim, H.-S. Integration of TE Induces Cancer Specific Alternative Splicing Events. Int. J. Mol. Sci. 2022, 23, 10918. https://doi.org/10.3390/ijms231810918

AMA Style

Kim WR, Park EG, Lee YJ, Bae WH, Lee DH, Kim H-S. Integration of TE Induces Cancer Specific Alternative Splicing Events. International Journal of Molecular Sciences. 2022; 23(18):10918. https://doi.org/10.3390/ijms231810918

Chicago/Turabian Style

Kim, Woo Ryung, Eun Gyung Park, Yun Ju Lee, Woo Hyeon Bae, Du Hyeong Lee, and Heui-Soo Kim. 2022. "Integration of TE Induces Cancer Specific Alternative Splicing Events" International Journal of Molecular Sciences 23, no. 18: 10918. https://doi.org/10.3390/ijms231810918

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop