Open access peer-reviewed chapter - ONLINE FIRST

Transcribed-Ultra Conserved Regions (T-UCRs) a New Light on a Dark Matter

Written By

Maria Radanova

Reviewed: 25 August 2023 Published: 03 October 2023

DOI: 10.5772/intechopen.113015

Noncoding RNA - The Dark Matter of the Genome IntechOpen
Noncoding RNA - The Dark Matter of the Genome Edited by Preeti Dabas

From the Edited Volume

Noncoding RNA - The Dark Matter of the Genome [Working Title]

Dr. Preeti Dabas

Chapter metrics overview

43 Chapter Downloads

View Full Metrics

Abstract

Transcribed Ultra-Conserved Regions (T-UCRs) are a novel class of long non-coding RNAs derived from Ultra-Conserved Regions (UCRs) of DNA. The discovery of cancer-specific mutations in UCRs and their location in cancer-associated genomic regions suggests that T-UCRs also play a role in carcinogenesis. However, the mechanisms behind their actions remain unclear. Their interactions with microRNAs are not well understood and are currently a subject of debate. Like other non-coding RNAs, T-UCRs exhibit tissue- and disease-specific expression, making them promising candidates for biomarkers or therapeutic targets in cancer and other diseases. This chapter aims to review the current knowledge on the functional effects of T-UCRs in cancer and other diseases, discuss the role of T-UCRs as regulators and regulated, and present their potential as disease monitoring biomarkers.

Keywords

  • ultra-conserved regions
  • UCR
  • transcribed-ultra conserved regions
  • T-UCRs
  • non-coding RNAs

1. Introduction

The genome is a malleable substance. A large portion of it is made up of so-called “dark matter”—non-coding protein genomic sequences. Until recently, the transcripts of this part of the genome were considered unnecessary and non-functional. However, it is now known that they carry important information and play a major role in regulating the phenotype. It has been proven that the “dark matter” of the genome is transcribed into various types of RNA, most of which do not encode proteins, known as non-coding RNAs [1]. Based on transcript length, non-coding RNAs are divided into two major groups. Those longer than 200 nucleotides are referred to as long non-coding RNAs (lncRNAs), while those shorter than 200 nucleotides are known as short non-coding RNAs (sncRNAs). Among the sncRNAs, well-studied microRNAs (miRs) are involved in the process of RNA interference [2]. Transcribed Ultra-Conserved Regions (T-UCRs) belong to the lncRNA group. T-UCRs are transcribed from 481 ultra-conserved DNA sequence regions (UCRs). T-UCRs were discovered in recent years and have quickly become a new field of exploration.

The chapter aims to provide a critical review of the current scientific understanding of the mechanisms of action of T-UCRs in pathological conditions, to present possible mechanisms for regulating their expression and to summarize research that demonstrates the diagnostic, prognostic, and predictive potential of T-UCRs as biomarkers.

Advertisement

2. Discovery and classification of UCRs

UCRs were first described by Bejerano et al. and were found to be present on all chromosomes, except chromosomes 21 and Y, with an uneven distribution [3]. They are longer than 200 base pairs and are functional but do not encode proteins [4]. UCRs are often located in fragile sites and genomic regions associated with tumor-related genes, and 93% of them are transcribed in at least one normal human tissue [5]. They are characterized by absolute conservation, with no insertions or deletions, and are 100% identical in humans, rats, and mice [3].

Their initial classification of UCRs was made by Bejarano et al. with a focus on their intersection with protein-coding regions. The authors divided the 481 UCRs into three groups: 111 were classified as partly exonic (or exonic for short), 114 as possibly exonic due to inconclusive intersection with a protein-coding sequence, and the remaining 256 were classified as nonexonic [3]. The first two groups are randomly distributed along the chromosomes, while the nonexonic elements tend to cluster in introns or intergenic regions near transcription factors and developmental genes.

In 2010, Mestdagh et al. proposed a more detailed classification of UCRs. The genomic groups became five: exonic, intronic, exon-containing, intergenic, and partially exonic UCRs [6]. A large part of UCRs, more than 70% are intronic and intergenic and most of them are transcriptionally active [5, 6, 7]. The UCRs groups correspond to their transcription product T-UCRs (Figure 1).

Figure 1.

Different types of T-UCRs according to genomic localization of UCRs. (Figure created with BioRender.com.)

T-UCRs can be divided into two groups: intragenic and intergenic. The intragenic group includes T-UCRs whose UCRs are located within exons or introns of their host genes, such as exonic, intronic, or T-UCRs whose UCRs span an exon such as exon-containing, and partly exonic. Intergenic T-UCRs are those whose UCRs are located outside of current genomic annotations. This separation of T-UCRs is mainly based on the localization of their UCRs and is somewhat conditional. Moreover, many intronic UCRs have been discovered in the antisense orientation relative to their host gene. This suggests that their transcripts T-UCRs are not simply products of intronic transcription of known genes, but rather are genuine independent noncoding transcripts [3]. The T-UCRs which correspond to the sense genome sequence, referred to as “+”, and those that correspond to the antisense sequence, referred to as “+A”. The designation of T-UCRs includes this information, following the “uc.” (from ultraconserved) and the number indicating their chromosomal location (for example uc.346+ or uc.479+A).

In recent years, the number of studies on the functions of T-UCRs in the biology of different diseases has rapidly increased. These studies have been summarized and commented on in a number of review articles [4, 7, 8, 9, 10, 11, 12, 13, 14, 15]. However, the need for discussions on this topic remains at this stage because what is known about T-UCRs and UCRs is less than what is unknown. It is important to differentiate between the roles of T-UCRs and the UCRs from which they are transcribed. T-UCRs may not represent the entire RNA transcript, and their sequences often overlap with ultraconserved enhancers or are contained within other annotated genes. On the other hand, Fedorova et al. argued that ultra-conserved non-coding elements do not represent non-coding RNAs, as T-UCRs are assumed to be [16]. Although they share common functions with lncRNAs, T-UCRs also exhibit many specific ones. Additionally, they do not quite fit the view of ncRNAs, which primarily have evolutionary conservation in structure rather than in sequence [16]. To fully understand T-UCRs, it is necessary to clarify their structure, the factors that influence their expression, and their effects on both the host and neighboring genes.

Advertisement

3. Mechanisms of regulation of gene expression by T-UCRs

T-UCRs play a crucial role in regulating gene expression by influencing various processes, including pri-miR processing and maturation, mRNA degradation, transcription regulation, epigenetic modulation [7, 17].

Efforts to understand the general mechanisms of action of T-UCRs face challenges due to the fact that some of these mechanisms are also involved in regulating the expression of T-UCRs themselves. It remains unclear whether the pathogenic role of T-UCRs is more closely related to their direct participation in the regulation of the expression of disease-related genes or if the dysregulation of their own expression is the determining factor. This raises an interesting question: how can a T-UCR simultaneously act as both a regulator and a regulated entity?

3.1 Interaction between T-UCRs and mRNA

An established mechanism of action for T-UCRs includes their ability to directly bind to the 3’UTR of mRNAs, leading primarily to the degradation of the gene’s mRNA. For example, uc.206 specifically targets mRNA of the pro-apoptotic gene P53, promoting tumor cell growth in cervical cancer [18]. Additionally, uc.338 interacts directly within the 3’UTR of TIMP-1 mRNA and negatively regulates it at a post-transcriptional level. Through this mechanism, uc.338 decreases TIMP-1 but increases MMP9 expression in the TIMP-1-MMP9 signaling pathway, which may explain the tumor progression in colorectal and cervical cancers [19, 20]. HSPA12B mRNA is a direct target of uc.454. In this way, uc.454 may induce apoptosis of lung tumor cells by decreasing the expression of HSPA12B and affecting the ratio of Bcl-2/Bax [21]. Uc.454 can also inhibit migration and invasion in lung cancer by interacting with K-Ras at the posttranscriptional level, affecting the K-Ras/P63/MMP9 pathway [22]. A study by Goto et al. suggests that uc.416+A operates through a similar mechanism on the mRNA of several proteins. Uc.416+A is associated with cell growth through the downregulation of expression of inhibitors of proliferation, differentiation, and migration of tumors, such as IGFBP6, and the upregulation of proteins associated with malignant neoplasms, such as HOXB5 [23]. Mechanistically, uc.189 inhibits protein translation of EPHA2 mRNA by directly binding to its 3’UTR region and promotes the proliferation and lymph node metastasis in esophageal squamous cell carcinoma [24]. Uc.57 binds to BCL11A mRNA, which reduced tamoxifen resistance by inhibiting PI3K/AKT and MAPK signaling pathways [25]. All these studies were conducted in cancer models, where T-UCRs consistently act as negative regulators of mRNA expression. Uc.478 and uc.479 affect the expression of genes in non-cancerous conditions. Their overexpression is associated with decreased levels of GRIA3 mRNA and protein in human models of Rett syndrome [26].

Another interesting regulatory mechanism exercised by T-UCRs was presented by Zhang et al. [27]. The authors revealed a new regulatory relationship between T-UCRs and their host genes. They suggested that uc.189 positively regulates the expression of the splicing factor SRp20, but did not provide any mechanistic evidence to explain how this occurs [27].

3.2 Interaction between T-UCRs and proteins

Regarding the mechanisms of action of some T-UCRs, there is no evidence that they affect the expression of mRNAs, but functional analyzes show the levels of the corresponding proteins are significantly decreased when T-UCRs are overexpressed or increased when T-UCRs are silenced. This suggests a T-UCR-protein interaction. This is the case with uc.38 and PBX1. Zhang et al. found that uc.38 interacts with the protein and subsequently affects the expression of Bcl-2 family members, ultimately inducing apoptosis in breast cancer cells [28]. Shi et al. also revealed that T-UCRs can directly interact with proteins. In their study, uc.51 physically interacts with NONO and promotes the proliferation and migration of breast cancer cells [29].

As a type of lncRNAs, T-UCRs may have the ability to specifically bind with one or more proteins or function as scaffolds for protein complexes involved in the regulation of gene expression. Similar capabilities of T-UCRs were reported by Zambalde et al. who identified 17 proteins that interact with uc.147 either directly or indirectly [30]. For some of these interactions, Zambalde et al. suggested a role for uc.147 as a regulator of protein functions. The authors hypothesized that uc.147 could serve as a scaffold for two protein complexes: one between RHOA and CIT, and another among six proteins (WDR18, TEX10, PES1, NOP53, DKC1, and ZC3HAV1) that participate in RNA processing and regulation. Additionally, the authors found correlations between the expression of uc.147 and neighboring genes, as well as downstream UCRs uc.148 and uc.149. Zambalde et al. do not exclude the possibility that uc.147 also uses another regulatory mechanism in breast cancer—acting as a decoy for miR-190 [30].

When discussing the mechanisms of action of T-UCRs in regulation of the gene expression, it is important to mention also a less well-known lncRNA-dependent epigenetic mechanism, called by Wang et al. an “epigenetic switch point” [31]. Sun et al. discovered that uc.323 can directly interact with EZH2, which is the primary component of the histone modification complex PRC2 responsible for the N-methylation of histones. This interaction can inhibit H3K27me3 at the promoter regions of CPT1b and regulate its transcription. Thus uc.323 ameliorated cardiac hypertrophy [32]. Studies by Panatta et al. and Mancini et al. suggest that T-UCRs mediate another epigenetic mechanism. According to these authors, uc.291 modulates chromatin remodeling activity by negatively regulating ACTL6A, the catalytic subunit of the SWI/SNF chromatin remodeling complex. This releases chromatin and allows for the transcription of epidermal differentiation complex genes. In cutaneous squamous cell carcinoma, downregulation of uc.291 reduces SWI/SNF complex activity, leading to a de-differentiation phenotype [33, 34].

3.3 Interaction between T-UCRs and microRNAs

The interaction between T-UCRs and microRNAs (miRs) is another way for T-UCRs to exert their regulatory functions. This interaction can modulate the activity of miRs. The effects of this can be seen through the degradation or inactivation of T-UCRs’ targets, miRs, and primary miRs (pre-miR), or through the influence on the expression of mature miRs [9].

Table 1 presents examples of T-UCRs that act as competitive endogenous RNAs (ceRNAs) and prevent binding of miRs to their target mRNAs in T-UCRs/miR/mRNA axis.

T-UCRs ceRNAsmiRs/mRNA axisRole of T-UCRs in pathologyExpression/cancer type/disease/injuryReferences
uc.8+miR-596/MMP9Promotes invasion and metastasisIncreased in early stage of disease/BIC[35]
uc.8+ uc.201+miR-596In combination inhibit tumor suppressor miR-596[36]
uc.63+miR-130b/MMP2Involves in cell growth and cell migrationIncreased/PC[37]
uc.77miR-4676-5p/FBXW8Inhibites the proliferation tumor cells by increasing FBXW8 and CDK4 ubiquitination, which lead to negatively regulation of CDK4 and a block of the G0/G1 transitionDecreased/CRC[38]
uc.158+AmiR-193bDrives tumor growth and migration, its inhibition increased apoptosisIncreased/HCC[39]
Increased/CCA
uc.173miR-29b/CLDN1Enhances the intestinal epithelial barrier functionIntestinal mucosa[40]
pre-miR-195Stimulates intestinal epithelial regeneration, enhances growth of the intestinal mucosa in mice[41]
miR-291a-3pInhibits lead-induced neuronal apoptosisDecreased/lead-induced nerve injury[42]
uc.230miR-503/CUGBP1Promotes renewal and barrier function of intestinal epithelia, protects against cell death, enhances mucosal repairIncreased/UC[43]
uc.333miR-223/FOXO1Improves insulin resistancedecreased/NAFLD[44]
uc.339miR-339-3p
miR-663b-3p
miR-95-5p
Promotes tumor growth and migration by upregulation of Cyclin E2, a direct target of these miRsIncreased/NSCLC[45]
pri-miR-339/SLC7A11Leads to defects in the ferroptosis, which drives the metastasisIncreased/LC adenocarcinoma[46]
uc.372pri-miR-195/pri-miR-4668/AGOPromotes lipogenesis and lipid accumulation in liver by regulation of expression of FAS and ACC/SCD1 and CD36 genes by Ago2.Increased/NAFLD[47]
uc.416+AmiR-153Affects cancer growth and migrationIncreased/RC[48]

Table 1.

T-UCRs as a decoy for miRs and their role in different diseases.

Source: BlC—bladder cancer; CCA—cholangiocarcinoma; CRC—colorectal cancer; HCC—hepatocellular carcinoma; LC—lung cancer; NAFLD—nonalcoholic fatty liver disease; NSCLC—non-small cell lung cancer; PC—prostate cancer; RC—renal cancer; UC—ulcerative colitis.

Another aspect of the interaction between T-UCRs and miRs is connected to the influence of T-UCRs on the expression of mature miRs.

Liz et al. describe a possible mechanism by which T-UCRs regulate the maturation of miRs [49]. In their study, they found that uc.283+A interacts with the stem region of pri-miR-195, preventing its processing by the Drosha/DGCR8 microprocessor complex. Similarly, Zhang et al. found that another T-UCR, uc.339, prevented the Drosha/DGCR8 splicing complex from binding to pri-miR-339, inhibiting the production of mature miR-339 [46]. Uc.372 regulates the cleavage of both pri-miR-195 and pri-miR-4668 from pri-to-pre-miR by sponging them, suppressing their maturation [47]. In contrast, a new study shows that uc.160+ enhances the processing of the pri-miR-376 cluster and A-to-I editing during miR-376 maturation [50].

Is the interaction between T-UCRs and miRs direct, or is there a mediator involved? This is another question that the study by Terreri et al. addresses [36]. The authors found that the interaction between T-UCRs and miRs can be mediated by the polycomb protein Yin Yang 1 (YY1). Specifically, YY1 mediates the binding between uc.8+ and miR-596 in bladder cancer. The authors suggest that the binding of YY1 to T-UCR may alter its conformation, inhibiting the binding of miR-596 [36]. This finding reveals a new player in the interaction between T-UCRs and miRs, another factor in gene expression regulation.

In their study, Terreri et al. shed another new light on the relationship between T-UCRs and miRs. They found that silencing uc.8+ increased the expression of miR-596, making it available to bind to other T-UCRs such as uc.283+, which in turn reduced its level. Terreri et al. believed that the interaction between T-UCRs and miRs occurs in networks that modulate the availability of these non-coding RNAs. The authors referred to this as an example of inter epigenetic regulation [36].

A recent study by Corrà et al. provided further evidence of this network interaction between T-UCRs and miRs in breast cancer [51]. The authors found that silencing uc.183 and uc.96 increased the levels of pre-miR-221 and miR-221, but also that miR-221 itself reduced the expression of uc.183, uc.110, and uc.84. This raises the question of which of the two has a regulatory function, T-UCRs or miR? This question is further complicated by the fact that the study found that treatment with anti-cancer drugs affects the expression of T-UCRs, but not the expression of miR-221.

Although the number of studies investigating the mechanisms of action of T-UCRs is increasing, their primary functions remain unclear. It is important to determine whether T-UCRs exhibit most of the functions described for other classes of lncRNAs or if new, specific functions will be discovered. A key feature that distinguishes T-UCRs from other lncRNAs is their bi-directional interaction with miRs—both regulating and being regulated by them.

There are also other studies that have reported on the role of T-UCRs in a number of pathological processes, but the underlying regulatory mechanistic interactions remain unclear and warrant further investigation [52, 53, 54, 55].

Despite attempts to systematically present and summarize the mechanisms of action of T-UCRs, it cannot be categorically stated that there is a manifestation of a single mechanism during functional activity. Different manifestations of action from one T-UCR in different diseases cannot be ruled out, similar to other non-coding RNAs, although the trend in this regard with T-UCRs is significantly weaker. An idea of the diversity of mechanisms of action, their effects and the behavior of one T-UCR in different types of cancer can be given by uc.338, which is one of the most studied T-UCRs.

It is believed that uc.338 stimulates cell growth and promotes the transition from the G1 to S phase of the cell cycle, leading to increased cell migration and invasion in hepatocellular carcinoma, lung, colorectal, and cervical cancers. Uc.338 stimulates cell growth through the PI3K/AKT pathway, potentially by down-regulating p21 and up-regulating cyclin D1 [56], or in association with BMI1, by modulating the transcription of CDKN1A and partially repressing p21 [57]. Around 400 potential target genes with uc.338-binding sites were identified, many of which are involved in cell proliferation [58]. Moreover, PAI-RBP1 (plasminogen activator inhibitor-1 RNA-binding protein) was identified as a potential RNA-binding partner of uc.338 [58]. Additionally, as mentioned above, silencing uc.338 inhibits cell migration and invasion by directly regulating TIMP1 [19, 20], and after knocking down uc.338, p53 activity increased by almost 30% [58].

Advertisement

4. Mechanisms of regulation of T-UCRs expression

The expression of T-UCRs is regulated through several mechanisms, including miRs, CpG island methylation, and histone modifications. These mechanisms are all forms of epigenetic influence. In some cases, these mechanisms work together. For example, the expression of uc.160+ is simultaneously affected by miR-155 and the methylation of the host gene promoter [59, 60].

4.1 Role of microRNAs

It is crucial to further investigate the regulatory role of T-UCRs through miRs. Moreover, there is evidence indicating that miRs significantly influence the regulation of T-UCRs transcription, particularly in cancer.

MiRs as regulators of T-UCRs expression are presented mainly in cancer (Table 2). The majority of studies paid attention to the sequence complementarity between T-UCRs and miRs. This effect was first reported by Galin et al. who found that the expression levels of two T-UCRs, uc.160+ and uc.346+A, could be directly regulated in vivo by a single miRNA, miR-155, in chronic lymphocytic leukemia [5]. The role of miR-155 as a regulator of uc.160+ has also been confirmed in gastric cancer [59]. In neuroblastoma, nine T-UCRs have been suggested to be regulated by five miRNAs [61]. However, the role of miRNAs as regulators of T-UCR expression remains uncertain. This is evidenced by research conducted by the group led by Wataru Yasui, who found in 2016 that miR-153 directly suppresses uc.416+A in a model of gastric cancer [23], but in 2018 claimed that uc.416+A modulates epithelial-to-mesenchymal transition through the regulation of miR-153 in renal cell carcinoma [48]. The effect of miR-596 on uc.283 expression discussed above [36] also indicates that there is no clear, unambiguous mechanism linking miRNAs and T-UCRs.

T-UCRs targetsmiRs regulatorsCancer typeReferences
uc.160+miR-155CLL[5]
GC[59]
miR-24-1CLL[5]
uc.346+AmiR-155CLL[5]
uc.348+miR-29bCLL[5]
uc.209+miR-877-3рNeuroblastoma[61]
uc.271+miR-383
uc.312+miR-877-3р
miR-548d-5p
uc.330+miR-548d-5p
uc.371+miR-877-3р
uc.411+miR-33b-5р
uc.421+miR-877-3р
uc.435+miR-939
uc.452+miR-383
uc.416+AmiR-153GC[23]
uc.283miR-596BIC[36]

Table 2.

T-UCRs regulated through miRs in different type of cancer.

Source: BlC—bladder cancer; CLL—chronic lymphocytic leukemia; GC—gastric cancer.

4.2 Role of CpG island methylation

Another way in which T-UCRs expression is regulated is through the hypermethylation of CpG islands in their promoter regions. This effect, which results in the downregulation and inactivation of three T-UCRs (uc.160+, uc.283+A, and uc.346+), was found by Lujambio et al. [62] and conformed for uc.160+ expression by Honma et al. [60]. They also revealed that silencing of uc.160+ led to the activation of MAPK signaling via repression of PTEN expression. Hudson et al. in turn confirmed the silencing of uc.283+A due to CpG islands hypermethylation [62]. Moreover, they with Lujambio et al. reported that treatment with DNA hypomethylating agent 5-aza-2-deoxycytidine (5-Aza-dC) upregulated the T-UCRs expression in human colon cancer and prostate cancer cells [62]. Goto et al. provided further evidence for this mechanism by discovering that in gastric and prostate cancer, uc.158+A is downregulated due to DNA methylation in the promoter region upstream of its transcription gene [23]. Hudson et al. discovered that five additional T-UCRs (uc.308+A, uc.434+A, uc.241+A, uc.285+, and uc.85+) were epigenetically downregulated, and their expression could be restored by treatment with a combination of 5-Aza-dC and histone deacetylase inhibitor trichostatin A (TSA) [63]. It seems that this mechanism, of influencing the expression of T-UCRs, leads to the silencing of those T-UCRs that have tumor-suppressive functions.

4.3 Role of histone mark

According to Mestdagh et al. both intergenic and intragenic T-UCRs are marked by trimethylation of lysine 4 on histone H3 (H3K4me3), a marker for transcriptional initiation [6]. The authors compared the distribution of active H3K4me3 marks between T-UCRs, miRNAs, and protein-coding genes in order to understand the structure and function of T-UCR transcriptional units. They found that the distribution of active H3K4me3 marks in T-UCRs is different from that of protein-coding genes, but it shows a remarkable resemblance to that of miRs. As a result, the authors hypothesized that there may be commonalities in the transcriptional organization between these two classes of ncRNAs—T-UCRs and miRs, and differences between T-UCRs and protein-coding genes [6].

4.4 Role of other factors

The studies by Vannini et al. and Guo et al. present some established influences on the expression of T-UCRs, but the underlying mechanisms remain unknown [4547]. Vannini et al., observed that the tumor-suppressor TP53 directly and negatively regulates uc.339. They proposed that the deregulation of uc.339 and its manifestation as an oncogene may be caused by mutations in the TP53 gene, which frequently occurs in non-small cell lung cancer. In their study, Guo et al., discovered that the upstream transcription factor 1 (USF1) regulates the expression of uc.372 in non-alcoholic fatty liver disease. They also noted that INSM2, which shares a promoter with uc.372, is involved in this regulation. The authors found that the knockdown of INSM2 decreases the level of uc.372 and suppresses USF1’s regulation of uc.372. The regulation of T-UCRs expression by transcription factors has also been described in more recent studies on uc.147 and uc.43+ [64, 65].

Another factor affecting T-UCRs transcription is hypoxia. Hypoxia can induce T-UCRs such as uc.475+, uc.63+/+A, uc.73+A, uc.106+, and uc.134+A (also known as hypoxia-induced noncoding ultraconserved transcripts, HINCUTs) [66]. While HIF-1α-binding sites have been discovered upstream promoter regions of uc.475+ and uc.63+, the mechanism by which other HINCUTs are induced by hypoxia remains unknown [67].

Advertisement

5. The influence of genetic variants in UCRs

Despite their strong conservatism, UCRs are not resistant to variations. In 2010, Wojcik et al., claimed that UCRs are sites of very low natural variation in the general population [68]. De Grassi et al. argued that UCRs accumulate fewer mutations overall, including fewer nearly neutral mutations [69]. Depletion of prevalent neutral SNPs in UCRs was also confirmed by Silla et al. [70]. Halligan et al. argued that UCRs receive more strongly selected deleterious mutations than protein-coding genes or nearby regulatory elements [71]. Habic et al. unlike Wojcik et al., and De Grassi et al., found that the polymorphism density within UCRs is higher than the genomic average, but in support of Halligan et al. believe that SNPs in UCRs may be less stable or rarer, with deleterious consequences in populations than those outside of UCRs [72]. However, only a small portion of the established SNPs in UCRs have a proven association with human diseases. It is interesting to note that many mutations within UCRs are not fixed [16]. Habic et al. propose that not all of these regions are subject to the same level of purifying selection [72]. Silla et al. suggest that UCRs are not under selective pressure as a continuous stretch of DNA, but rather experience differential evolutionary pressure at the single nucleotide level [70]. Therefore, the question arises, can the hypothesis that UCRs are subject to stronger negative selection [73] be considered as a general principle for UCRs?

In their paper, Habic et al. [72] summarize the studies in the literature about the associations between the presence of SNPs in UCRs and the appearance of different disorders. They found 37 polymorphism/phenotype associations covering diseases such as: different types of muscular dystrophies; adolescent idiopathic scoliosis; amyotrophic lateral sclerosis; renal diseases; as well as eye-related diseases; and cancer [72]. Few studies examining the association between SNPs in UCRs and disease mention that the analyzed polymorphisms are located within conserved genomic regions. This makes it challenging to organize and categorize them systematically.

Studying SNPs in UCRs presents an opportunity to easily detect disease biomarkers. Several studies have established an association between individual SNPs in UCRs and disease risk, especially cancer risk [74, 75, 76]. Additionally, SNPs within UCRs may serve as valuable prognostic and predictive biomarkers, as demonstrated in studies by Lin et al. and Bao et al. [76, 77]. Rs8004379 SNP in uc.368+ is significantly associated with biochemical recurrence in localized prostate cancer as well as a decreased risk for prostate cancer-specific mortality in patients with advanced disease. In their analysis of the functional consequences of this SNP, Bao et al. made an interesting conclusion [77]. Given the localization of rs8004379 within an intron of the tumor suppressor NPAS3 gene, they suggested that the rs8004379 C allele may affect the expression of NPAS3 and be associated with a decreased risk of recurrence and mortality in prostate cancer patients after treatment. Rs7849 in uc.298 in a study by Lin et al. was found to be associated with an increased risk of disease recurrence in stage II patients [76]. Also, rs10211390 in uc.54 is associated with an increased risk of recurrence after 5-fluorouracil-based chemotherapy in patients with stage II and III colorectal cancer. The authors believed that individual outcomes following chemotherapy can be predicted based on the rs7849 and rs6590611 genotypes [76].

Advertisement

6. Clinical significance of T-UCRs in cancer and other diseases

In recent years, the number of studies focused on T-UCRs has notably grown. A significant part of them includes functional analyses to confirm the impact of T-UCRs in disease pathogenesis. However, these studies often do not provide details about the specific mechanisms of action and direct interactions of these T-UCRs [5, 39, 67, 78, 79, 80, 81]. Another large group of studies is descriptive, providing information about the association of T-UCRs levels with clinicopathological features of patients or reporting changes in T-UCRs expression in response to therapy. The results of this second group of studies provide an opportunity to select T-UCRs as potential diagnostic, prognostic, and predictive biomarkers. A large portion of these studies is summarized in Table 3, where information about the type of disease, the expression levels of T-UCRs, and their association with the clinical characteristics of studied patients is presented.

T-UCRs/expressionCancer type/diseaseAssociation with clinicopathological characteristicsPossible biomarkerReference
uc.8+/downregulatedBlCPathological grade pathological stage worst overall survival tumors at advanced stages—T-UCR prevalent in cytoplasmaPrognostic[35, 36]
uc.38/downregulatedBCLarger tumors tumors of more advanced stages[28]
T3 stage stage IV expression—ER, PR and HER2 receptors[82]
uc.48+/upregulatedTBDistinguish between untreated and cured patientsDiagnostic[53]
uc.51/upregulatedBCLarger tumor size metastasis[29]
uc.57/downregulatedBCTamoxifen resistancePredictive[25]
uc.63/upregulatedLuminal A BCReduced disease-free survivalPrognostic[83]
BCM1 trait/TNM classification expression of the HER2 receptor[82]
MIBCKnockdown of uc.63+ increased sensitivity to cisplatin chemotherapyPredictive[84]
PCCastration resistance, docetaxel resistance, worse prognosisSerum predictive, prognostic[37]
LCTumor stage, poor prognosisPrognostic[85]
GCAdvanced pT3/4 stages, pN1/2/3 stages, stages III/IVPrognostic[67]
uc.70/upregulatedCLLInferior clinical outcome, downregulated by CpG-ODN treatment and predict longer time to treatmentPrognostic[86]
uc.73/downregulatedCRCWorse overall survivalPrognostic[79]
uc.84/upregulatedBCLess aggressive tumors worse overall survivalPrognostic[87]
uc.98/upregulatedAS/ACSAtherosclerotic plaque instabilityDiagnostic, prognostic[54]
uc.134/downregulatedHCCWorse overall survivalPrognostic[88]
uc.147/upregulatedBCWorse overall survival worse progression-free survival in HER2+ patientsPrognostic[87]
uc.160 methylated/upregulatedCRCLymph node infiltration, lesion size[89]
Stages III/IV better overall survivalPrognostic[90]
Lower-grade gliomasBetter overall survivalPrognostic[50]
uc.189/upregulatedCSCC EACPoor prognosisPrognostic[91]
ESCCInvasion, advanced clinical stage, lymph node metastasis, poor prognosisPrognostic[92]
uc.261/upregulatedCDPositively correlated with HBI[80]
uc.268/upregulatedLuminal A BCPoor survival ratePrognostic[87]
uc.283 methylated/upregulatedCRCBetter overall survivalPrognostic[90]
uc.306/downregulatedHBV-related HCCShorter overall survivalPrognostic[93]
uc.338/upregulatedCRCDistal colon location[79]
Lymph node metastasis[19]
Larger tumor size, lymph node metastasis, poorer prognosisPrognostic[56]
NSCLCTNM stage, lymph node metastasis, distant metastasis, shorter overall survival, shorter disease-free survivalPrognostic[94]
CCWith lymph node metastasis[20]
uc.454/downregulatedNSCLCLymph node metastasis[22]

Table 3.

T-UCRs with clinical, predictive and prognostic significance.

Source: ACS—acute coronary syndrome; ALL—acute lymphoblastic leukemia; AS—atherosclerosis; BC—breast cancer; BlC—bladder cancer; CC—cervical cancer; CD—Crohn’s disease; CLL—chronic lymphocytic leukemia; CpG ODN—oligodeoxynucleotides that contain unmethylated CG dinucleotides; CRC—colorectal cancer; CSCC—cervical squamous cell carcinomas; EAC—endometrial adenocarcinomas; EC—esophagus cancer; ESCC—oesophagal squamous cell carcinomas; GC—gastric cancer; HBI—Harvey-Bradshaw Index (to evaluate the activity and severity of CD and to monitor the effect of treatment); HBV—hepatitis B-related HCC; HCC—hepatocellular carcinoma; LC—lung cancer; MIBC—muscle-invasive bladder cancer; NSCLC—non-small cell lung cancer; TB—tuberculosis; PC—prostate cancer.

Examining the functions of T-UCRs individually and searching for a single target for one T-UCR is an approach that is increasingly being replaced by more complex ones, such as constructing three-component regulatory networks—T-UCRs-miRNAs-mRNAs. Simultaneously tracking multiple T-UCR interactions within a single disease allows for the evaluation of the potential diagnostic and prognostic values of T-UCRs, as demonstrated in the study by Khalafiyan et al., where five T-UCRs including uc.8, uc.169, uc.342, uc.360, and uc.417 were found to be correlated with the overall survival of gastric cancer patients [95]. On the other hand, three-component regulatory networks provide an opportunity for a comprehensive understanding of the underlying gene expression regulation processes involved in pathogenesis.

Calin et al. first reported that dysregulation of several T-UCRs is associated with disease [5]. Later, Scaruffi et al. presented two T-UCR panels for high-risk prediction in neuroblastoma patients [61]. The first panel included 28 T-UCRs significantly associated with prognosis, while the second panel of 15 upregulated T-UCRs discriminated short- from long-survivors with high sensitivity and specificity. In 2014, Fassan et al. presented two panels of T-UCRs for biological profiling in the transformation of Barrett’s mucosa [96]. One is for distinguishing Barrett’s esophagus from the squamous epithelium and another is for evaluating progression from normal to adenocarcinoma. Zambalde et al. presented two panels of T-UCRs as diagnostic biomarkers for breast cancer and a third panel that distinguished with high sensitivity and specificity luminal A from triple-negative patients [87]. Finally, we come to studies such as that by Ciaramella et al. in which the authors propose a new biomarker panel of T-UCRs using machine learning techniques [97]. This signature panel of 13 T-UCRs is able to efficiently discriminate between normal and bladder cancer patient samples or patients with different survival extents, identifying sub-types of bladder cancer patients with different prognoses regardless of cancer grade. The implementation of Artificial Intelligence methodologies in medical applications represents a significant challenge in our time.

Advertisement

7. Future perspectives

As the number of studies on T-UCRs continues to grow, it is expected that more specific information will be uncovered about their structure, functions, and potential therapeutic applications. Understanding how T-UCRs operate in normal cells will provide insight into why and how genomic ultraconservation is maintained. A key challenge is to identify the role of T-UCRs in maintaining tissue homeostasis. By clarifying their role and mechanism of action in normal cells, it may be possible to understand the consequences of their dysregulation in pathological conditions. Increasingly, research is focusing on T-UCRs as potential molecular biomarkers or as new drug targets, particularly in cancer treatment. As biomarkers, T-UCRs are promising for both diagnosis and assessing prognosis because it is known that for some T-UCRs, the levels of expression and their localization within the cell vary at different stages of cancer. T-UCRs also hold potential as a basis for therapeutic strategies or drug combinations. The therapies are aimed at destroying cancer cells or reversing the tumor phenotype, as well as influencing mechanisms related to resistance.

Research on T-UCRs requires a complex approach due to the diverse interactions they are involved in and the intricate mechanisms regulating their expression.

Advertisement

8. Conclusions

In nearly 20 years since the first description of UCRs by Bejerano et al., scientific interest and research on them and T-UCRs have grown significantly. The interest directed towards T-UCRs is justifiably great, as their physiological significance in both normal and pathological conditions has been demonstrated. However, our understanding of their functions and the mechanisms through which they are realized still sheds little light on “dark matter”. T-UCRs, as a type of lncRNA, exhibit more characteristic features than common functions with other ncRNAs. Their interactions with miRs or proteins reveal the richness of the genetic regulatory machinery, but also provide an opportunity for its precise influence through the development of new RNA-based therapies.

Advertisement

Acknowledgments

This work was funded by the Bulgarian National Science Fund (Grand number KΠ-06-H23/6, 18.12.2018).

Advertisement

Conflict of interest

The author declares no conflict of interest.

Abbreviations

CITcitron rho-interacting serine/threonine kinase
CPT1bcarnitine palmitoyl transferase 1b
DKC1dyskerin pseudouridine synthase 1
EPHA2ephrin type-A receptor 2
EZH2enhancer of zeste homolog 2
HOXB5Homeobox B5
HSPA12Bheat shock protein family A member 12B
IGFBP1insulin-like growth factor-binding protein 1
INSM2insulinoma-associated 2 gene
MMP9matrix metalloproteinase-9
NONOnon-POU domain-containing octamer-binding protein
NOP53glioma tumor suppressor candidate region gene 2 protein
PBX1pre-B-cell leukemia homeobox 1
PES1Pescadillo Ribosomal Biogenesis factor 1
POM121transmembrane nucleoporin pseudogene
PRC2polycomb repressor complex 2
RHOARas homolog family member A
TEX10testis expressed 10
TIMP1tissue inhibitor of metalloproteinase 1
WDR18WD Repeat Domain 18
ZC3HAV1Zinc finger antiviral 1

References

  1. 1. Braicu C, Zimta AA, Harangus A, Iurca I, Irimie A, Coza O, et al. The function of non-coding RNAs in lung cancer tumorigenesis. Cancers (Basel). 2019;11(5):605. DOI: 10.3390/cancers11050605
  2. 2. Bresesti C, Vezzoli V, Cangiano B, Bonomi M. Long non-coding RNAs: Role in testicular cancers. Frontiers in Oncology. 2021;11:605606. DOI: 10.3389/fonc.2021.605606
  3. 3. Bejerano G, Pheasant M, Makunin I, Stephen S, Kent WJ, Mattick JS, et al. Ultraconserved elements in the human genome. Science. 2004;304:1321-1325. DOI: 10.1126/science.1098119
  4. 4. Gao SS, Zhang ZK, Wang XB, Ma Y, Yin GQ , Guo XB. Role of transcribed ultraconserved regions in gastric cancer and therapeutic perspectives. World Journal of Gastroenterology. 2022;28(25):2900-2909. DOI: 10.3748/wjg.v28.i25.2900
  5. 5. Calin GA, Liu CG, Ferracin M, Hyslop T, Spizzo R, Sevignani C, et al. Ultraconserved regions encoding ncRNAs are altered in human leukemias and carcinomas. Cancer Cell. 2007;12(3):215-229. DOI: 10.1016/j.ccr.2007.07.027
  6. 6. Mestdagh P, Fredlund E, Pattyn F, Rihani A, Van Maerken T, Vermeulen J, et al. An integrative genomics screen uncovers ncRNA T-UCR functions in neuroblastoma tumours. Oncogene. 2010;29(24):3583-3592. DOI: 10.1038/onc.2010.106
  7. 7. Terracciano D, Terreri S, de Nigris F, Costa V, Calin GA, Cimmino A. The role of a new class of long noncoding RNAs transcribed from ultraconserved regions in cancer. Biochimica Et Biophysica Acta. Reviews on Cancer. 2017;1868(2):449-455. DOI: 10.1016/j.bbcan.2017.09.001
  8. 8. Fabris L, Calin GA. Understanding the genomic Ultraconservations: T-UCRs and cancer. International Review of Cell and Molecular Biology. 2017;333:159-172. DOI: 10.1016/bs.ircmb.2017.04.004
  9. 9. Chava S, Challagundla KB. The transcribed-ultra conserved Regions: Novel non-coding RNA players in neuroblastoma progression. Noncoding RNA. 2019;5(2):39. DOI: 10.3390/ncrna5020039
  10. 10. Pereira Zambalde E, Mathias C, Rodrigues AC, de Souza Fonseca Ribeiro EM, Fiori Gradia D, Calin GA, et al. Highlighting transcribed ultraconserved regions in human diseases. Wiley Interdisciplinary Reviews RNA. 2020;11(2):e1567. DOI: 10.1002/wrna.1567
  11. 11. Wang Y, Zhou D, Feng Y, Chen G, Li N. T-UCRs with digestive and respiratory diseases. Bioorganic & Medicinal Chemistry Letters. 2020;30(16):127306. DOI: 10.1016/j.bmcl.2020.127306
  12. 12. Bozgeyik I. The dark matter of the human genome and its role in human cancers. Gene. 2022;811:146084. DOI: 10.1016/j.gene.2021.146084
  13. 13. Gibert MK Jr, Sarkar A, Chagari B, Roig-Laboy C, Saha S, Bednarek S, et al. Transcribed ultraconserved regions in cancer. Cells. 2022;11(10):1684. DOI: 10.3390/cells11101684
  14. 14. Wang T, Li F, Lu Z. Ultra-conserved RNA: A novel biological tool with diagnostic and therapeutic potential. Discover Oncology. 2023;14(1):41. DOI: 10.1007/s12672-023-00650-1
  15. 15. Oliveira JC. Transcribed Ultraconserved Regions: New regulators in cancer signaling and potential biomarkers. Genetics and Molecular Biology. 2023;46(1 Suppl. 2):e20220125. DOI: 10.1590/1678-4685-GMB-2022-0125
  16. 16. Fedorova L, Mulyar OA, Lim J, Fedorov A. Nucleotide composition of ultra-conserved elements shows excess of GpC and depletion of GG and CC dinucleotides. Genes (Basel). 2022;13(11):2053. DOI: 10.3390/genes13112053
  17. 17. Irimie AI, Zimta AA, Ciocan C, Mehterov N, Dudea D, Braicu C, et al. The unforeseen non-coding RNAs in head and neck cancer. Genes (Basel). 2018;9(3):134. DOI: 10.3390/genes9030134
  18. 18. Li Q , Li X, Wang C. Uc.206 regulates cell proliferation and apoptosis by targeting P53 in cervical cancer cells. Neoplasma. 2016;63(3):411-418. DOI: 10.4149/310_151017N538
  19. 19. Wang C, Wang Z, Zhou J, Liu S, Wu C, Huang C, et al. TUC.338 promotes invasion and metastasis in colorectal cancer. International Journal of Cancer. 2017;140(6):1457-1464. DOI: 10.1002/ijc.30542
  20. 20. Li Q , Shen F, Wang C. TUC338 promotes cell migration and invasion by targeting TIMP1 in cervical cancer. Oncology Letters. 2017;13(6):4526-4532. DOI: 10.3892/ol.2017.5971
  21. 21. Zhou J, Wang C, Gong W, Wu Y, Xue H, Jiang Z, et al. Uc.454 inhibited growth by targeting heat shock protein family a member 12B in non-small-cell lung cancer. Molecular Therapy - Nucleic Acids. 2018;12:174-183. DOI: 10.1016/j.omtn.2018.05.004
  22. 22. Zhou J, Wang C, Huang C, Ding Z, Shi M. TUCR.454 inhabits metastasis in lung cancer cells. Int. International Journal of Clinical and Experimental Pathology. 2018;11(3):1289-1296
  23. 23. Goto K, Ishikawa S, Honma R, Tanimoto K, Sakamoto N, Sentani K, et al. The transcribed-ultraconserved regions in prostate and gastric cancer: DNA hypermethylation and microRNA-associated regulation. Oncogene. 2016;35(27):3598-3606. DOI: 10.1038/onc.2015.445
  24. 24. Ding Z, Yan Y, Guo YL, Wang C. Esophageal carcinoma cell-excreted exosomal uc.189 promotes lymphatic metastasis. Aging (Albany NY). 2021;13(10):13846-13858. DOI: 10.18632/aging.202979
  25. 25. Zhang CH, Wang J, Zhang LX, Lu YH, Ji TH, Xu L, et al. Shikonin reduces tamoxifen resistance through long non-coding RNA uc.57. Oncotarget. 2017;8(51):88658-88669. DOI: 10.18632/oncotarget.20809
  26. 26. Siqueira E, Obiols-Guardia A, Jorge-Torres OC, Oliveira-Mateos C, Soler M, Ramesh-Kumar D, et al. Analysis of the circRNA and T-UCR populations identifies convergent pathways in mouse and human models of Rett syndrome. Molecular Therapy - Nucleic Acids. 2021;27:621-644. DOI: 10.1016/j.omtn.2021.12.030
  27. 27. Zhang M, Zhou J, Jiao L, Xu L, Hou L, Yin B, et al. Long non-coding RNA T-uc.189 modulates neural progenitor cell fate by regulating Srsf3 during mouse cerebral cortex development. Frontiers in Neuroscience. 2021;15:709684. DOI: 10.3389/fnins.2021.709684
  28. 28. Zhang LX, Xu L, Zhang CH, Lu YH, Ji TH, Ling LJ. Uc.38 induces breast cancer cell apoptosis via PBX1. American Journal of Cancer Research. 2017;7(12):2438-2451
  29. 29. Shi X, Huang X, Chen R, Li Y, Xu Y, Zhang W, et al. The transcribed ultraconserved element uc.51 promotes the proliferation and metastasis of breast cancer by stabilizing NONO. Clinical & Experimental Metastasis. 2021;38(6):551-571. DOI: 10.1007/s10585-021-10128-5
  30. 30. Zambalde EP, Bayraktar R, Schultz Jucoski T, Ivan C, Rodrigues AC, Mathias C, et al. A novel lncRNA derived from an ultraconserved region: Lnc-uc.147, a potential biomarker in luminal a breast cancer. RNA Biology. 2021;18(Suppl. 1):416-429. DOI: 10.1080/15476286.2021.1952757
  31. 31. Wang Z, Zhang XJ, Ji YX, Zhang P, Deng KQ , Gong J, et al. The long noncoding RNA Chaer defines an epigenetic checkpoint in cardiac hypertrophy. Nature Medicine. 2016;22(10):1131-1139. DOI: 10.1038/nm.4179
  32. 32. Sun Y, Fan W, Xue R, Dong B, Liang Z, Chen C, et al. Transcribed ultraconserved regions, Uc.323, ameliorates cardiac hypertrophy by regulating the transcription of CPT1b (carnitine palmitoyl transferase 1b). Hypertension. 2020;75(1):79-90. DOI: 10.1161/HYPERTENSIONAHA.119.13173
  33. 33. Panatta E, Lena AM, Mancini M, Smirnov A, Marini A, Delli Ponti R, et al. Long non-coding RNA uc.291 controls epithelial differentiation by interfering with the ACTL6A/BAF complex. EMBO Reports. 2020;21(3):e46734. DOI: 10.15252/embr.201846734
  34. 34. Mancini M, Cappello A, Pecorari R, Lena AM, Montanaro M, Fania L, et al. Involvement of transcribed lncRNA uc.291 and SWI/SNF complex in cutaneous squamous cell carcinoma. Discov. Oncologia. 2021;12(1):14. DOI: 10.1007/s12672-021-00409-6
  35. 35. Olivieri M, Ferro M, Terreri S, Durso M, Romanelli A, Avitabile C, et al. Long non-coding RNA containing ultraconserved genomic region 8 promotes bladder cancer tumorigenesis. Oncotarget. 2016;7(15):20636-20654. DOI: 10.18632/oncotarget.7833
  36. 36. Terreri S, Durso M, Colonna V, Romanelli A, Terracciano D, Ferro M, et al. New cross-talk layer between Ultraconserved non-coding RNAs, MicroRNAs and Polycomb protein YY1 in bladder cancer. Genes (Basel). 2016;7(12):127. DOI: 10.3390/genes7120127
  37. 37. Sekino Y, Sakamoto N, Goto K, Honma R, Shigematsu Y, Sentani K, et al. Transcribed ultraconserved region uc.63+ promotes resistance to docetaxel through regulation of androgen receptor signaling in prostate cancer. Oncotarget. 2017;8(55):94259-94270. DOI: 10.18632/oncotarget.21688
  38. 38. Zheng Z, Hong D, Zhang X, Chang Y, Sun N, Lin Z, et al. uc.77- downregulation promotes colorectal cancer cell proliferation by inhibiting FBXW8-mediated CDK4 protein degradation. Front. Oncologia. 2021;11:673223. DOI: 10.3389/fonc.2021.673223
  39. 39. Carotenuto P, Fassan M, Pandolfo R, Lampis A, Vicentini C, Cascione L, et al. Wnt signalling modulates transcribed-ultraconserved regions in hepatobiliary cancers. Gut. 2017;66(7):1268-1277. DOI: 10.1136/gutjnl-2016-312278
  40. 40. Wang JY, Cui YH, Xiao L, Chung HK, Zhang Y, Rao JN, et al. Regulation of intestinal epithelial barrier function by long noncoding RNA uc.173 through interaction with MicroRNA 29b. Molecular and Cellular Biology. 2018;38(13):e00010-e00018. DOI: 10.1128/MCB.00010-18
  41. 41. Xiao L, Wu J, Wang JY, Chung HK, Kalakonda S, Rao JN, et al. Long noncoding RNA uc.173 promotes renewal of the intestinal mucosa by inducing degradation of MicroRNA 195. Gastroenterology. 2018;154(3):599-611. DOI: 10.1053/j.gastro.2017.10.009
  42. 42. Nan A, Zhou X, Chen L, Liu M, Zhang N, Zhang L, et al. A transcribed ultraconserved noncoding RNA, Uc.173, is a key molecule for the inhibition of lead-induced neuronal apoptosis. Oncotarget. 2016;7(1):112-124. DOI: 10.18632/oncotarget.6590
  43. 43. Yu TX, Kalakonda S, Liu X, Han N, Chung HK, Xiao L, et al. Long noncoding RNA uc.230/CUG-binding protein 1 axis sustains intestinal epithelial homeostasis and response to tissue injury. JCI Insight. 2022;7(19):e156612. DOI: 10.1172/jci.insight.156612
  44. 44. Zhang Y, Sun J, Yao H, Lin Y, Wei J, Hu G, et al. Ultraconserved element uc.333 increases insulin sensitivity by binding to miR-223. Aging (Albany NY). 2020;12(8):6667-6679. DOI: 10.18632/aging.103020
  45. 45. Vannini I, Wise PM, Challagundla KB, Plousiou M, Raffini M, Bandini E, et al. Transcribed ultraconserved region 339 promotes carcinogenesis by modulating tumor suppressor microRNAs. Nature Communications. 2017;8(1):1801. DOI: 10.1038/s41467-017-01562-9
  46. 46. Zhang N, Huang J, Xu M, Wang Y. LncRNA T-UCR uc.339/miR-339/SLC7A11 Axis regulates the metastasis of ferroptosis-induced lung adenocarcinoma. Journal of Cancer. 2022;13(6):1945-1957. DOI: 10.7150/jca.65017
  47. 47. Guo J, Fang W, Sun L, Lu Y, Dou L, Huang X, et al. Ultraconserved element uc.372 drives hepatic lipid accumulation by suppressing miR-195/miR4668 maturation. Nature Communications. 2018;9(1):612. DOI: 10.1038/s41467-018-03072-8
  48. 48. Sekino Y, Sakamoto N, Goto K, Honma R, Shigematsu Y, Quoc TP, et al. Uc.416 + a promotes epithelial-to-mesenchymal transition through miR-153 in renal cell carcinoma. BMC Cancer. 2018;18(1):952. DOI: 10.1186/s12885-018-4863-y
  49. 49. Liz J, Portela A, Soler M, Gómez A, Ling H, Michlewski G, et al. Regulation of pri-miRNA processing by a long noncoding RNA transcribed from an ultraconserved region. Molecular Cell. 2014;55(1):138-147. DOI: 10.1016/j.molcel.2014.05.005
  50. 50. Soler M, Davalos V, Sánchez-Castillo A, Mora-Martinez C, Setién F, Siqueira E, et al. The transcribed ultraconserved region uc.160+ enhances processing and A-to-I editing of the miR-376 cluster: Hypermethylation improves glioma prognosis. Molecular Oncology. 2022;16(3):648-664. DOI: 10.1002/1878-0261.13121
  51. 51. Corrà F, Crudele F, Baldassari F, Bianchi N, Galasso M, Minotti L, et al. UC.110, and UC.84 ultra-conserved RNAs are mutually exclusive with miR-221 and are engaged in the cell cycle circuitry in breast cancer cell lines. Genes (Basel). 2021;12(12):1978. DOI: 10.3390/genes12121978
  52. 52. Zhang Q , Cheng Z, Yu Z, Zhu C, Qian L. Role of lncRNA uc.457 in the differentiation and maturation of cardiomyocytes. Molecular Medicine Reports. 2019;19(6):4927-4934. DOI: 10.3892/mmr.2019.10132
  53. 53. Li ZB, Han YS, Wei LL, Shi LY, Yi WJ, Chen J, et al. Screening and identification of plasma lncRNAs uc.48+ and NR_105053 as potential novel biomarkers for cured pulmonary tuberculosis. International Journal of Infectious Diseases. 2020;92:141-150. DOI: 10.1016/j.ijid.2020.01.005
  54. 54. Fan Z, Zhang Y, Xiao D, Ma J, Liu H, Shen L, et al. Long noncoding RNA UC.98 stabilizes atherosclerotic plaques by promoting the proliferation and adhesive capacity in murine aortic endothelial cells. Acta Biochimica et Biophysica Sinica (Shanghai). 2020;52(2):141-149. DOI: 10.1093/abbs/gmz155
  55. 55. Yu M, Guan Z, Li S, Wen X, Shi H, Qu G, et al. Gene expression profiling analysis reveals that the long non-coding RNA uc.412 is involved in mesangial cell proliferation. Molecular Medicine Reports. 2019;20(6):5297-5303. DOI: 10.3892/mmr.2019.10753
  56. 56. Zhang Y, Wang S, Qian W, Ji D, Wang Q , Zhang Z, et al. Uc.338 targets p21 and cyclin D1 via PI3K/AKT pathway activation to promote cell proliferation in colorectal cancer. Oncology Reports. 2018;40(2):1119-1128. DOI: 10.3892/or.2018.6480
  57. 57. Bo C, Li N, Li X, Liang X, An Y. Long noncoding RNA uc.338 promotes cell proliferation through association with BMI1 in hepatocellular carcinoma. Human Cell. 2016;29(4):141-147. DOI: 10.1007/s13577-016-0140-z
  58. 58. Wen HJ, Walsh MP, Yan IK, Takahashi K, Fields A, Patel T. Functional modulation of gene expression by Ultraconserved long non-coding RNA TUC338 during growth of human hepatocellular carcinoma. iScience. 2018;2:210-220. DOI: 10.1016/j.isci.2018.03.004
  59. 59. Pang L, Li Q , Zhang Y, Deng B, Wu F, Wang J, et al. Transcribed ultraconserved noncoding RNA uc.160 acts as a negative regulator in gastric cancer. American Journal of Translational Research. 2018;10(9):2822-2833
  60. 60. Honma R, Goto K, Sakamoto N, Sekino Y, Sentani K, Oue N, et al. Expression and function of uc.160+, a transcribed ultraconserved region, in gastric cancer. Gastric Cancer. 2017;20(6):960-969. DOI: 10.1007/s10120-017-0714-9
  61. 61. Scaruffi P, Stigliani S, Moretti S, Coco S, De Vecchi C, Valdora F, et al. Transcribed-ultra conserved region expression is associated with outcome in high-risk neuroblastoma. BMC Cancer. 2009;9:441. DOI: 10.1186/1471-2407-9-441
  62. 62. Lujambio A, Portela A, Liz J, Melo SA, Rossi S, Spizzo R, et al. CpG island hypermethylation-associated silencing of non-coding RNAs transcribed from ultraconserved regions in human cancer. Oncogene. 2010;29(48):6390-6401. DOI: 10.1038/onc.2010.361
  63. 63. Hudson RS, Yi M, Volfovsky N, Prueitt RL, Esposito D, Volinia S, et al. Transcription signatures encoded by ultraconserved genomic regions in human prostate cancer. Molecular Cancer. 2013;12:13. DOI: 10.1186/1476-4598-12-13
  64. 64. Rodrigues AC, Zambalde EP, Bellan DL, Trindade EDS, Ribeiro EMSF, Calin G, et al. Lnc-uc.147 is associated with disease stage of liver, gastric, and renal cancer. Biomolecules. 2023;13(2):265. DOI: 10.3390/biom13020265
  65. 65. Ding L, Gong C, Zhao J, Liu X, Li T, Rao S, et al. Noncoding transcribed ultraconserved region (T-UCR) UC.48+ is a novel regulator of high-fat diet induced myocardial ischemia/reperfusion injury. Journal of Cellular Physiology. 2019;234(6):9849-9861. DOI: 10.1002/jcp.27674
  66. 66. Ferdin J, Nishida N, Wu X, Nicoloso MS, Shah MY, Devlin C, et al. HINCUTs in cancer: Hypoxia-induced noncoding ultraconserved transcripts. Cell Death and Differentiation. 2013;20(12):1675-1687. DOI: 10.1038/cdd.2013.119
  67. 67. Sakamoto N, Sekino Y, Fukada K, Pham QT, Honma R, Taniyama D, et al. Uc.63+ contributes to gastric cancer progression through regulation of NF-kB signaling. Gastric Cancer. 2020;23(5):863-873. DOI: 10.1007/s10120-020-01070-8
  68. 68. Wojcik SE, Rossi S, Shimizu M, Nicoloso MS, Cimmino A, Alder H, et al. Non-codingRNA sequence variations in human chronic lymphocytic leukemia and colorectal cancer. Carcinogenesis. 2010;31(2):208-215. DOI: 10.1093/carcin/bgp209
  69. 69. De Grassi A, Segala C, Iannelli F, Volorio S, Bertario L, Radice P, et al. Ultradeep sequencing of a human ultraconserved region reveals somatic and constitutional genomic instability. PLoS Biology. 2010;8(1):e1000275. DOI: 10.1371/journal.pbio.1000275
  70. 70. Silla T, Kepp K, Tai ES, Goh L, Davila S, Catela Ivkovic T, et al. Allele frequencies of variants in ultra conserved elements identify selective pressure on transcription factor binding. PLoS One. 2014;9(11):e110692. DOI: 10.1371/journal.pone.0110692
  71. 71. Halligan DL, Oliver F, Guthrie J, Stemshorn KC, Harr B, Keightley PD. Positive and negative selection in murine ultraconserved noncoding elements. Molecular Biology and Evolution. 2011;28(9):2651-2660. DOI: 10.1093/molbev/msr093
  72. 72. Habic A, Mattick JS, Calin GA, Krese R, Konc J, Kunej T. Genetic variations of Ultraconserved elements in the human genome. OMICS. 2019;23(11):549-559. DOI: 10.1089/omi.2019.0156
  73. 73. Katzman S, Kern AD, Bejerano G, Fewell G, Fulton L, Wilson RK, et al. Human genome ultraconserved elements are ultraselected. Science. 2007;317(5840):915. DOI: 10.1126/science.1142430
  74. 74. Yang R, Frank B, Hemminki K, Bartram CR, Wappenschmidt B, Sutter C, et al. SNPs in ultraconserved elements and familial breast cancer risk. Carcinogenesis. 2008;29(2):351-355. DOI: 10.1093/carcin/bgm290
  75. 75. Lin M, Eng C, Hawk ET, Huang M, Greisinger AJ, Gu J, et al. Genetic variants within ultraconserved elements and susceptibility to right- and left-sided colorectal adenocarcinoma. Carcinogenesis. 2012;33(4):841-847. DOI: 10.1093/carcin/bgs096
  76. 76. Lin M, Eng C, Hawk ET, Huang M, Lin J, Gu J, et al. Identification of polymorphisms in ultraconserved elements associated with clinical outcomes in locally advanced colorectal adenocarcinoma. Cancer. 2012;118(24):6188-6198. DOI: 10.1002/cncr.27653
  77. 77. Bao BY, Lin VC, Yu CC, Yin HL, Chang TY, Lu TL, et al. Genetic variants in ultraconserved regions associate with prostate cancer recurrence and survival. Scientific Reports. 2016;6:22124. DOI: 10.1038/srep22124
  78. 78. das Chagas PF, de Sousa GR, Kodama MH, CAO DBJ, Yunes JA, Brandalise SR, et al. Ultraconserved long non-coding RNA uc.112 is highly expressed in childhood T versus B-cell acute lymphoblastic leukemia. Hematology, Transfusion and Cell Therapy. 2021;43(1):28-34. DOI: 10.1016/j.htct.2019.12.003
  79. 79. Sana J, Hankeova S, Svoboda M, Kiss I, Vyzula R, Slaby O. Expression levels of transcribed ultraconserved regions uc.73 and uc.388 are altered in colorectal cancer. Oncology. 2012;82(2):114-118. DOI: 10.1159/000336479
  80. 80. Qian XX, Cai CW, Li HY, Lai LJ, Song DJ, Qiao YQ , et al. Transcribed ultraconserved region (T-UCR) uc.261 expression is closely correlated with disease activity and intestinal permeability in Crohn's disease. Therapeutic Advances in Gastroenterology. 2019;12:1756284819880733. DOI: 10.1177/1756284819880733
  81. 81. Vannini I, Ferracin M, Fabbri F, Fabbri M. Overexpression of ultraconserved region 83- induces lung cancer tumorigenesis. PLoS One. 2022;17(1):e0261464. DOI: 10.1371/journal.pone.0261464
  82. 82. Zadrożna-Nowak A, Romanowicz H, Zadrożny M, Bryś M, Forma E, Smolarz B. Analysis of long non-coding RNA (lncRNA) uc.38 and uc.63 expression in breast carcinoma patients. Genes (Basel). 2022;13(4):608. DOI: 10.3390/genes13040608
  83. 83. Marini A, Lena AM, Panatta E, Ivan C, Han L, Liang H, et al. Ultraconserved long non-coding RNA uc.63 in breast cancer. Oncotarget. 2017;8(22):35669-35680. DOI: 10.18632/oncotarget.10572
  84. 84. Sekino Y, Sakamoto N, Ishikawa A, Honma R, Shigematsu Y, Hayashi T, et al. Transcribed ultraconserved region uc.63+ promotes resistance to cisplatin through regulation of androgen receptor signaling in bladder cancer. Oncology Reports. 2019;41(5):3111-3118. DOI: 10.3892/or.2019.7039
  85. 85. Liu X, Zhou X, Deng CJ, Zhao Y, Shen J, Wang Y, et al. Comprehensive analyses of T-UCR expression profiles and exploration of the efficacy of uc.63- and uc.280+ as biomarkers for lung cancer in Xuanwei, China. Pathology, Research and Practice. 2020;216(6):152978. DOI: 10.1016/j.prp.2020.152978
  86. 86. Bomben R, Roisman A, D'Agaro T, Castellano G, Baumann T, Delgado J, et al. Expression of the transcribed ultraconserved region 70 and the related long non-coding RNA AC092652.2-202 has prognostic value in chronic lymphocytic leukaemia. British Journal of Haematology. 2019;184(6):1045-1050. DOI: 10.1111/bjh.15237
  87. 87. Zambalde EP, Adamoski D, Gradia DF, Rabinovich I, Rodrigues AC, Ivan C, et al. Transcribed Ultraconserved Regions are associated with clinicopathological features in breast cancer. Biomolecules. 2022;12(2):214. DOI: 10.3390/biom12020214
  88. 88. Ni W, Zhang Y, Zhan Z, Ye F, Liang Y, Huang J, et al. A novel lncRNA uc.134 represses hepatocellular carcinoma progression by inhibiting CUL4A-mediated ubiquitination of LATS1. Journal of Hematology & Oncology. 2017;10(1):91. DOI: 10.1186/s13045-017-0449-4. Erratum in: J Hematol Oncol. 2023 May 8;16(1):51
  89. 89. Kottorou AE, Antonacopoulou AG, Dimitrakopoulos FD, Diamantopoulou G, Sirinian C, Kalofonou M, et al. Deregulation of methylation of transcribed-ultra conserved regions in colorectal cancer and their value for detection of adenomas and adenocarcinomas. Oncotarget. 2018;9(30):21411-21428. DOI: 10.18632/oncotarget.25115
  90. 90. Kottorou AE, Dimitrakopoulos FD, Antonacopoulou AG, Diamantopoulou G, Tsoumas D, Koutras A, et al. Differentially methylated ultra-conserved Regions Uc160 and Uc283 in adenomas and adenocarcinomas are associated with overall survival of colorectal cancer patients. Cancers (Basel). 2020;12(4):895. DOI: 10.3390/cancers12040895
  91. 91. Wang L, Wang XC, Li X, Gu Y, Zhou J, Jiang S, et al. Expression of uc.189 and its clinicopathologic significance in gynecological cancers. Oncotarget. 2017;9(7):7453-7463. DOI: 10.18632/oncotarget.23761
  92. 92. Guo Y, Wang C, Miao X, Chen S, Qian Y, Li G, et al. Upregulation of uc.189 in patients with esophageal squamous cell carcinoma and its clinicopathologic value. Pathology, Research and Practice. 2017;213(11):1400-1403. DOI: 10.1016/j.prp.2017.09.005
  93. 93. Luo HL, Chen J, Luo T, Wu FX, Liu JJ, Wang HF, et al. Downregulation of macrophage-derived T-UCR uc.306 associates with poor prognosis in hepatocellular carcinoma. Cellular Physiology and Biochemistry. 2017;42(4):1526-1539. DOI: 10.1159/000479269
  94. 94. Tian Y, Feng Y. Up-regulation of long noncoding RNA uc.338 predicts poor survival in non-small cell lung cancer. Cancer Biomarkers. 2018;22(4):781-785. DOI: 10.3233/CBM-181331
  95. 95. Khalafiyan A, Emadi-Baygi M, Wolfien M, Salehzadeh-Yazdi A, Nikpour P. Construction of a three-component regulatory network of transcribed ultraconserved regions for the identification of prognostic biomarkers in gastric cancer. Journal of Cellular Biochemistry. 2023;124(3):396-408. DOI: 10.1002/jcb.30373
  96. 96. Fassan M, Dall'Olmo L, Galasso M, Braconi C, Pizzi M, Realdon S, et al. Transcribed ultraconserved noncoding RNAs (T-UCR) are involved in Barrett's esophagus carcinogenesis. Oncotarget. 2014;5(16):7162-7171. DOI: 10.18632/oncotarget.2249
  97. 97. Ciaramella A, Di Nardo E, Terracciano D, Conte L, Febbraio F, Cimmino A. A new biomarker panel of ultraconserved long non-coding RNAs for bladder cancer prognosis by a machine learning based methodology. BMC Bioinformatics. 2023;23(Suppl. 6):569. DOI: 10.1186/s12859-023-05167-6

Written By

Maria Radanova

Reviewed: 25 August 2023 Published: 03 October 2023