Skip to main content

Current understanding of functional peptides encoded by lncRNA in cancer

Abstract

Dysregulated gene expression and imbalance of transcriptional regulation are typical features of cancer. RNA always plays a key role in these processes. Human transcripts contain many RNAs without long open reading frames (ORF, > 100 aa) and that are more than 200 bp in length. They are usually regarded as long non-coding RNA (lncRNA) which play an important role in cancer regulation, including chromatin remodeling, transcriptional regulation, translational regulation and as miRNA sponges. With the advancement of ribosome profiling and sequencing technologies, increasing research evidence revealed that some ORFs in lncRNA can also encode peptides and participate in the regulation of multiple organ tumors, which undoubtedly opens a new chapter in the field of lncRNA and oncology research. In this review, we discuss the biological function of lncRNA in tumors, the current methods to evaluate their coding potential and the role of functional small peptides encoded by lncRNA in cancers. Investigating the small peptides encoded by lncRNA and understanding the regulatory mechanisms of these functional peptides may contribute to a deeper understanding of cancer and the development of new targeted anticancer therapies.

Introduction

Cancer threatens the health of people all over the world and is the cause of a large number of deaths each year [1]. Disorders of gene expression and imbalance in transcription are typical indicators of cancer. In fact, both coding and non-coding RNAs play a key role during these processes [2]. With the development of high-throughput sequencing technology, a large number of ncRNAs have been identified as key regulators in a variety of pathophysiological conditions including cancer [3,4,5]. Long non-coding RNAs (lncRNAs), by definition, refer to RNA with transcripts longer than 200 nucleotides (nt), which have no long open reading frames (ORFs, > 100 amino acids) and lack the ability to code proteins. Therefore, lncRNA were originally regarded as trash produced by the transcription process. Upon further research, lncRNAs have been proven to drive many important cancer phenotypes by interacting with other cellular macromolecules, including DNA, RNA and protein [5]. They can regulate cancer processes through chromatin remodeling [6], transcription [7] or translation [8, 9] regulation, RNA editing [10], RNA degradation [11], RNA splicing [12] and miRNA sponge [13]. Like mRNAs, lncRNAs are transcribed, spliced, capped, and polyadenylated. For a long time, it was overlooked whether the short open reading frames (sORF) in lncRNAs could be translated. Recently, with the advancement of ribosome profiling and sequencing technologies, it has been proved that the sORF in lncRNAs can encode peptides (less than 100 amino acids) or small proteins that perform some important biological functions [14,15,16]. According to current knowledge, the sORF, with canonical start and stop codon, go through the translation process in a way similar to that of mRNA. lncRNAs that are translated preferentially localize in the cytoplasm and their translation efficiency is similar to that of mRNAs [17].

Studies have confirmed that peptides can act as key regulators of many basic cellular processes such as development, differentiation, proliferation, splicing regulation, apoptosis and cell metabolism [18,19,20,21,22,23]. In fact, both lncRNA and polypeptides encoded by lncRNA can mediate a variety of biological functions, especially in the regulation of cancer progression. lncRNA AGPG and lncRNA AFAP1-AS1 can promote tumor progression by regulating PFKFB3-mediated glycolysis reprogramming or epigenetic inhibition of p21 expression, respectively [24, 25]. HOXB-AS3 is a peptide of 53 amino acids encoded by lncRNA HOXB-AS3 [20]. The expression of HOXB-AS3 peptide is decreased in cancer tissues. The HOXB-AS3 peptide rather than lncRNA can inhibit the growth of colorectal cancer (CRC). ASRPS is a peptide of 60 amino acids encoded by LINC00908, which is down-regulated in triple-negative breast cancer (TNBC), and can inhibit angiogenesis and exert an anti-tumor effect in TNBC [26]. The deciphering of the function of lncRNA-encoded peptides/proteins has just begun.

In this review, we describe the biological function of lncRNA in cancer, introduce the coding ability of lncRNA and the current methods to evaluate the coding potential. Finally, we discuss the most updated findings on the role of small peptides encoded by lncRNA in cancer.

Biological function of lncRNA in cancer

LncRNA has been recognized as a key regulator in cancer [27]. Previous studies have shown that lncRNA affects some important biological functions in the tumor microenvironment including cell proliferation, migration and invasion, apoptosis and autophagy, epithelial-mesenchymal transformation (EMT), cancer stemness, etc. (Fig. 1) via mechanisms involving chromatin remodeling, RNA editing, RNA splicing, transcription and translation regulation [28,29,30,31,32,33].

Fig. 1
figure 1

LncRNAs influence key processes in cancer. LncRNAs themselves can regulate the progression of cancer by affecting cell proliferation, migration and invasion, autophagy and apoptosis, EMT and cancer stemness. They can also encode peptides to participate in cancer regulation

Cell proliferation, migration and invasion

Cell proliferation, migration and invasion play an important role in the progression of tumors. Through proliferation, migration and invasion, the tumor cells gradually spread to the adjacent tissue and become malignant cancers. lncRNAs are critically involved in this process. XIST (X inactivation specific transcript) is a newly discovered carcinogenic lncRNA, which promotes the malignant phenotype of many cancers [34]. Competitive endogenous RNA (ceRNA) form a network in which different RNAs, both coding and noncoding, influence each other's expression by binding to microRNA response elements (MREs). In thyroid carcinoma, XIST acts as a ceRNA sponging miR-34a and competes with MET for miR-34a binding. MET is a receptor tyrosine kinase that promotes cell proliferation through PI3K/AKT signaling and miR-34a is considered to be a tumor suppressor targeting MET, thus XIST promotes the proliferation of thyroid cancer by down-regulating miR-34a and increasing MET [35]. In pancreatic cancer (PC), ZEB1 (the key regulator of EMT and cell invasion) is the downstream target of MiR-429. Its overexpression can accelerate the migration, invasion and EMT of PC cells. XIST, as the ceRNA of miRNA-429, upregulates the expression of ZEB1, leading to PC cell migration, invasion and EMT enhancement [36]. In non-small cell lung cancer (NSCLC), XIST acts as the ceRNA of miR-744, inhibits the feedback loop of miR-744/RING1 and activates the Wnt/ β-catenin signaling pathway, which results in enhanced proliferation of NSCLC cells [37]. It has also been reported that XIST can affect tumor progression by regulating cell proliferation, migration, and invasion in nasopharyngeal carcinoma [38], glioma [39], oral squamous cell carcinoma [40], colorectal cancer [41], and hepatocellular carcinoma [42]. ABHD11-AS1 is a lncRNA that is highly expressed in various cancers including papillary thyroid carcinoma (PTC). MiR-199a-5p, as a tumor suppressor, inhibits the carcinogenic function of its downstream target SLC1A5. ABHD11-AS1, as the ceRNA of miR-199a-5p, blocks the tumor inhibitory function of miR-199a-5p, thereby enhancing the proliferation, migration and invasion of PTC cells [43]. It has also been confirmed that ABHD11-AS1 can influence tumor progression by regulating cell proliferation, migration, and invasion in colorectal cancer [44], pancreatic cancer [45], and epithelial ovarian cancer [46]. In short, the majority of cancer-related lncRNAs can affect cell proliferation, migration and invasion, thus regulating tumor progression. Some representative lncRNAs are listed in Table 1.

Table 1 Cancer-related lncRNA affecting cell proliferation, migration and invasion

Apoptosis and autophagy

Apoptosis (or type I cell death) is an orderly cellular process in living organisms, which features a variety of morphological changes including cell shrinkage, nuclear fragmentation and chromatin condensation. In this manner, all cellular components are eventually degraded and digested by other living cells [100]. Autophagy is characterized by the presence of autophagic vacuoles (autophagosomes) which are finally delivered to lysosomes for degradation. It often causes damaged cells or excess aging proteins/organelles to be swallowed and degraded, making the cells more conducive for survival [101]. However, in some circumstances, it also activates an alternative cell death pathway (or type II cell death) [102]. Many lncRNAs affect tumor progression by regulating these two processes. In gastric cancer (GC), lncRNA HAGLROS inhibits autophagic cell death by competitively sponging miR-100-5p to increase mTOR expression. At the same time, HAGLROS interacts and activates the mTORC1 signaling pathway, which serves as a negative regulator of autophagy, thus promoting the proliferation of GC cells [103]. LncRNA DANCR is transcriptionally activated by KLF5, a gene highly expressed in GC. Knockdown of KLF5 inhibits the DANCR/miR-194/AKT2 axis to enhance autophagy and decrease cancer cell viability [104]. LncRNA SNHG11 accelerates the progression of GC by activating oncogenic autophagy, which is dependent on the induction of ATG12 through the miR-483-3p/miR-1276 [105]. In glioblastoma, lncRNA AC003092.1 acts as a ceRNA to suppress miR-195 and promote the expression of Konitz protease inhibitor (TFPI-2). It enhances temozolomide (TMZ) chemosensitivity through TFPI-2-induced cell apoptosis [106]. It has also been reported that lncRNA CASC9 promotes cancer progression by enhancing cell proliferation and inhibiting autophagy dependent cell apoptosis by activating the AKT/mTOR signaling pathway in oral squamous cell carcinoma [107]. LncRNA NBR2 can repress autophagy-induced cell proliferation and down-regulate ERK and JNK signals in hepatocellular carcinoma (HCC), thereby inhibiting the malignant progression of HCC [108].

Epithelial-mesenchymal transformation (EMT)

EMT refers to the transition of cells from an epithelial state to a mesenchymal state and is associated with various tumor processes such as tumorigenesis, metastasis, invasion, and malignant progression [109,110,111]. EMT can be divided into partial, incomplete or mixed EMT states based on the characteristics between the epithelial state and the complete mesenchymal state of the cells [112]. Some lncRNAs may play a role in the progression of cancer by affecting EMT. In NSCLC, lncRNA linc00673 has been shown to facilitate tumor progression through EMT by acting as a ceRNA to sponge miR-150-5p, leading to increased expression of ZEB1 which is a proven regulatory factor in the promotion of EMT, thus enhancing the proliferation, migration, invasion and EMT of NSCLC [113]. In HCC, MiR-15b is a carcinogenic gene that promotes cancer migration, invasion, EMT and angiogenesis. Programmed cell death 4 (PDCD4) is a tumor suppressor gene that suppresses tumor migration, invasion, EMT and angiogenesis. Interestingly, IncRNA miR503HG acts as a ceRNA to sponge miR-15b leading to increased PDCD4 expression, which in turn inhibits HCC migration, invasion, EMT and angiogenesis [114]. LncRNA AB073614 is significantly up-regulated in CRC, resulting in enhanced activation of JAK/STAT3 signaling, a pathway that promotes cancer metastasis and EMT and accelerates tumor progression [115]. In lung cancer, lncRNA-LINP1 can down-regulate transforming growth factor β (TGF- β), a key regulator of EMT in tumor progression, and inhibit tumor EMT through the TGF- β / SMAD pathway [116].

Cancer stemness

Some cancer cells in the tumor cell population have the ability to self-renew, differentiate and proliferate. They resist anticancer treatments and maintain survival of cancer cells. These cells are called cancer stem cells (CSCs) [117]. Undoubtedly, CSCs have become a stumbling block in the treatment of tumors and investigation of them is deemed to hold great potential in cancer treatment. In recent years, some lncRNAs have been reported to be involved in the regulation of cancer stemness. In CRC, lncRNA H19 is expressed by cancer associated fibroblasts (CAFs) and delivered by CAF derived exosome. It acts as a ceRNA sponge of miR-141 (CRC cell stemness inhibitor) to activate β-catenin signaling and promote the stemness of CRC cells [118]. In GC, lncRNA MACC1-AS1 is induced by transforming growth factor β1 (TGF-β1) secreted by mesenchymal stem cells (MSCs), which suppresses the expression of miR-145-5p (the stemness & chemoresistance inhibitor) to accelerate FAO-dependent (fatty acid oxidation) stemness and chemotherapeutic resistance of GC cells [29]. In breast cancer (BC), lncRNA FGF13-AS1 can regulate RNA binding proteins and insulin-like growth factor 2 mRNA binding proteins (IGF2BPs) to shorten the half-life of c-Myc (Myc) mRNA. Myc (c-Myc) is a recognized carcinogenic transcription factor, which regulates cancer cell growth, proliferation, apoptosis and stemness [119]. LncRNA FGF13-AS1 suppresses the stemness of BC cells by accelerating Myc metabolism. In general, lncRNA can regulate cancer stemness, which is an important biological function of cancer-related lncRNA.

Tumor immunity

Recent studies have found that lncRNA not only regulates the progression of many cancers, but also participates in the regulation of immune process, which plays an important role in innate immunity and acquired immunity [209]. LncRNAs may affect cancer progression through immune regulation [210]. Based on this, ImmLnc, which is a tool designed to identify immunomodulatory related lncRNA, has been developed by researchers [211].

Ability of lncRNA to encode small peptides

As early as 20 years ago, a study revealed that lncRNA possesses sORF that can encode small peptides with biological functions in soybean [120]. After further research, it has been confirmed that these short ORFs from lncRNA can be captured by ribosomes and then translated into corresponding peptides with biological functions. In Drosophila, a ncRNA called polished rice (pri) actually has several short sORFs that encode peptides of 11 or 32 amino acids in length and participate in epithelial morphogenesis [121]. In human, as early as 2015, lncRNA CRNDE has been found to encode an endogenous CRNDEP peptide, which was located in the nucleus and may participate in oxygen metabolism and regulate cell proliferation [172]. With the in-depth study of peptides/proteins encoded by lncRNA in recent years, some lncRNAs with the ability to encode small peptides have been found. In addition, they have some biological functions, especially in tumor regulation. Interestingly, these lncRNAs can function both through their RNAs and the encoded peptides, such as lncRNA HOXB-AS3. Research has shown that lncRNA HOXB-AS3 can bind ErbB3-binding protein 1 (EBP1) and affect the EBP1-NPM1 complex formation to regulate rRNA transcription [123]. At the same time, it can also encode a 53aa peptide that plays oncogenic or tumor suppressive roles in different cancers, which will be discussed later. Overall, peptides encoded by lncRNAs can participate in the regulation of various cancer processes. This is usually achieved by binding of small peptides to their downstream targets, which are directly or indirectly involved in the occurrence and development of cancer. The binding of peptides with their targets will change the balance of tumor processes, stabilizing oncogenic signals or inhibiting tumor suppressive pathways to promote cancer progression, or upregulating tumor suppressor signals or inhibiting oncogenic pathways to inhibit cancer progression (Fig. 1).

In fact, some lncRNAs’ ORF can be translated and these small peptides are further processed into small antigenic peptides on MHC class I proteins, activating CD8 immune cells to inhibit tumor growth [212]. The peptides encoded by lncRNA can also mediate antigen presentation, CD4 + T cell response, interleukin production, etc. in the immune process [213]. This is an important functional discovery of lncRNA-derived peptides, which will drive the research of tumor immunotherapy and cancer vaccines to a new level.

Prediction of lncRNA coding potential

With the advancement of high-throughput sequencing technology, RNA-seq can be used to identify protein-coding RNA and non-coding RNA [124]. Methods built on this have been developed to predict the coding potential of lncRNAs, using computer-based, high-throughput sequencing technology and experimental methods that comprehensively evaluate the sequence characteristics and ORF of lncRNA, and finally predict the coding potential of lncRNA and identify candidate small peptides (Table 2).

Table 2 Tools for predicting lncRNA coding potential

Methods based on sequence

Advances in sequencing technologies have led to more precise assessment of sequence features of lncRNA, including sequence homology, conservation, nucleotide composition, secondary structure, Open Reading Frame (ORF), etc. Relying on a variety of sequencing results, the coding potential of lncRNA can be predicted by methods with high precision based on sequence features. These methods include COME, CPAT and PORTRAIT. Coding Potential Computation Tool (COME) not only has high prediction accuracy, but also characterizes multiple coding parameters of lncRNA and is applicable to a variety of organisms [127]. CPAT is a tool that uses a logistic regression model to predict the coding performance of lncRNAs, and the model is based on the comprehensive evaluation of four sequence features: ORF size, ORF coverage, Fickett TESTCODE statistic and hexamer usage bias [128]. PORTRAIT is a software originally proposed to screen ncRNAs in transcriptomes from not so well-characterized organisms. Based on the support vector machine (SVM) algorithm, protein-coding or non-coding genes can be classified with high specificity. Thus it can be used to evaluate the coding potential of ncRNAs [129].

In addition, the coding potential of lncRNAs can be evaluated based on the sequence features of their ORFs. Research has found that lncRNA contain several ORFs shorter than mRNAs (sORFs), in which the longer ORFs located in the cytoplasm may be more easily captured by ribosomes, thereby facilitating translation [17, 140, 141]. We can distinguish whether the translated ORF comes from lncRNA or from mRNA via marker mapping and mutation experiments [142, 143]. Therefore, the identification of sORFs is an important indicator when evaluating the coding capacity of lncRNA. There are two important characteristics in the ORF sequence: one is the length and the other is the integrity [130, 144]. Moreover, ORFs that can be translated should have sequence homology and should be conserved. By identifying sORFs (10–100 amino acids) with coding potential in a given nucleotide sequence, a tool to identify sORFs in lncRNA was developed: sORFfinder [134]. Through this package, the ORFs from lncRNA can be calculated quickly. This kind of software is developing rapidly, and the latest version is csORF-finder designed by NUAA, which makes the computing performance more powerful [145]. Additionally, GWIPS-Viz [135, 136] is a database for the identification of sORFs based on ribosome occupancy analysis, which can identify ORFs with high coding potential in lncRNA (lncRNA has a large number of identical and conservative fragments protected by ribosomes). A peptide encoded by lncRNA has been predicted in this manner and has proven to be biologically functional [26].

Computer-based machine learning methods

In the past decade, machine learning technology has been widely used in biological applications including genome annotation [146, 147], protein binding prediction [148, 149], identification of key transcriptional drivers of cancer [150, 151], prediction of metabolic function of complex microbial communities and characterization of transcriptional regulatory networks [152,153,154,155]. In recent years, with the development of bioinformatics, machine learning technology has been applied to the prediction of gene expression by building models [156], the study of gene splicing [157], and the identification of long non-coding RNA [158,159,160,161]. Even more recently, machine learning methods have been applied to the identification of transcript coding potential. After the discovery that some non-coding RNAs have the ability to encode peptides, the machine learning technology was brought to the prediction of the coding ability of ncRNA. MiPepid [137], a tool dedicated to the identification of micropeptides, is based on machine learning techniques. MiPepid has been trained using an existing database and logistic regression with 4-mer features to achieve a high degree of accuracy while running fast. DeepCPP [139], a deep neural network for RNA coding potential prediction, is based on nucleotide bias information and minimum distribution similarity features. Its advantage is the improved ability to identify sORFs RNA (lncRNA, etc.). NAMS webserver [138] is a web server that predicts the coding potential and functional annotation of plant transcripts. Based on its computing power, it can also be used to evaluate the coding potential of lncRNA genes.

Experimental approach

As more and more functional small peptides encoded by lncRNA are discovered, researchers have also designed experimental methods for verification. Ribosomal sequencing is a method used to evaluate the coding potential of ncRNAs. By sequencing ribosome-protected RNA fragments, the ribosome enrichment information of lncRNA sequences can be used to predict whether the lncRNA has the potential to encode peptides [17]. Many lncRNA encoded peptides have been discovered in this manner [20, 162]. After the initial identification, a vector expressing the FLAG-labeled ORF can be transfected into cells. If the ORF is translatable, it will drive the FLAG tag to be translated together, and immunofluorescence can be used to determine whether the corresponding protein is produced followed by other methodologies such as Western Blot, etc. [163,164,165,166]. Next, the start codon can be mutated, and once again used to detect whether there is a FLAG band at the corresponding molecular weight by Western blotting, in order to determine whether the translation occurs normally [167]. At the same time, the endogenously expressed lncRNA encoded peptide can be detected with antibodies raised against amino acid sequences in the peptide by western blotting and/or IP/MS. In addition, pull-down experiments such as co-immunoprecipitation (CO-IP) can also be used to discover proteins that interact with the functional peptide [168], which can further be detected by MS.

Functions of peptides encoded by LncRNA in cancer

Some lncRNAs possess small open reading frames (sORFs), which can exert biological functions by encoding functional small peptides. Several studies have found that small peptides encoded by lncRNA play a key role in the regulation of various cancer processes [169]. Some small peptides can cause worsening of the cancer phenotype and manifest as oncogenic peptides, while others can inhibit tumor proliferation, metastasis and invasion, manifested as tumor suppressor peptides. RNA-binding regulatory peptide (RBRP) is a functional peptide of 71aa encoded by lncRNA LINC00266-1. Studies have shown that RBRP is a regulatory subunit of RNA m6A reader IGF2BP1 complex by binding directly to the GxxG motif in the KH3–4 di-domain of IGF2BP1, which is indispensable for m6A recognition and interaction. The increased expression of RBRP in tumors promotes the recognition and binding of m6A reader IGF2BP1 to the targeted RNA in order to enhance the mRNA stability of proto-oncogenes (such as c-Myc, etc.), which induces tumorigenesis [170]. PACMP is a 44aa micropeptide encoded by lncRNA CTD-2256P15.2 with multiple functions. It not only suppresses CtIP-KLHL15-dependent CtIP ubiquitination, but also promotes PARP1-induced PAR polymerization by combining with DNA damage-mediated polychains. Both PARP1 and CtIP are important targets in cancer, so PACMP has the potential to become a high-value anti-cancer target [122]. The forced expression of TUBL, a 87aa peptide encoded by lncRNA TINCR, promotes cell cycle progression in normal human epidermal keratinocytes. Mice lacking this protein exhibit decreased cell cycle progression in skin-keratinocytes, delayed wound healing, and the protein may promote the proliferation of cancer cells [171]. A similar functional peptide encoded by lncRNA that is involved in cell proliferation is CRNDEP (84aa) encoded by lncRNA CRNDE [172]. A number of functional small peptides encoded by lncRNAs play crucial roles in different types of cancers (Table 3 and Fig. 2).

Table 3 Peptides encoded by lncRNA in cancer
Fig. 2
figure 2

Functions of peptides encoded by lncRNA in cancer. LncRNA can encode peptides to promote tumor development in glioma, lung cancer, HCC, OSCC, ESCC, breast cancer and melanoma or inhibit tumor development in HNSCC, TNBC and CRC

Hepatocellular carcinoma (HCC)

Liver cancer ranks fourth among all cancer-related deaths worldwide, and a large number of people die from hepatocellular carcinoma (HCC) each year [195]. It has been confirmed that a variety of small proteins encoded by lncRNA can affect the occurrence and progression of HCC. SMIM30 is a 59aa peptide encoded by LINC00998. SMIM30 promotes the carcinogenesis of HCC by activating the MAPK signaling pathway. The underlying mechanism involves the binding of the peptide to non-receptor tyrosine kinase Sploy, which drives its membrane anchoring and phosphorylation, and then activates the mitogen-activated protein kinase (MAPK) pathway thus promoting the proliferation and migration of HCC cells [181]. KRASIM is a 99aa peptide encoded by lncRNA NCBP2-AS2. Overexpression of KRASIM can reduce the level of proto-oncogene KRAS protein, which then inhibits the ERK signaling pathway in HCC cells causing a reduction in the growth and proliferation of HCC cells. In the cytoplasm of human HCC cells, KRASIM interacts and colocalizes with KRAS protein [182]. The lncRNA HBVPTPAP encodes a peptide of 145aa called HBVPTPAP. HBVPTPAP is mainly located in the cytoplasm and can induce mitochondrial apoptosis by activating the JAK/STAT signaling pathway, thus inducing apoptosis of HCC cells. This regulation was through the interaction between the HBVPTPAP polypeptide and PILRA [183]. PINT87aa is a functional small peptide of 87aa encoded by LINC-PINT. PINT87aa can induce senescence of hepatoma cells by blocking PHB2 transcription through the direct binding of PINT87aa and FOXM1 DNA binding domain [184]. A study by Guo et al. has shown that lncRNA ZFAS1 has several small open reading frames (smORF), one of which has been proven to be up-regulated in HCC tumor tissue, but is scarcely expressed in normal liver tissue, which implies that smORF may be related to the occurrence and development of hepatocellular carcinoma. However, the detailed mechanism needs to be further elucidated [185].

Colorectal cancer (CRC)

More than 1.2 million patients are diagnosed with colorectal cancer (CRC) each year, and more than 600,000 people die from the disease [196]. In CRC, functional small peptides encoded by a variety of lncRNAs regulate tumor progression. As mentioned above, HOXB-AS3 peptide promotes the proliferation of oral squamous cell carcinoma cells. The oncogenic role of LncRNA HOXB-AS3 has also been implicated in different cancer types. On the contrary, in CRC, lncRNA HOXB-AS3 inhibits the reprogramming of glucose metabolism through its encoded 53aa peptide. This antagonizes the regulation of splicing of pyruvate kinase M (PKM) mediated by hnRNP A1, and ultimately inhibits the growth of CRC. This process is achieved by the competitive binding of HOXB-AS3 peptide to the ariginine residues of hnRNP A1 which blocks the binding of its own arginine residue to exon 9 of PKM [20]. Further investigation is pending into whether the function of lncRNA HOXB-AS3 and its peptide differs depending on cancer type. ASAP is a 94aa peptide encoded by LINC00467, which is associated with ATP synthesis. Clinically, ASAP is associated with the malignant phenotype in patients, whereby high expression of ASAP indicates poor survival in patients with CRC. As for the mechanism, ASAP peptide promotes the proliferation of CRC cells by promoting ATP synthesis, thereby increasing the activity of ATP synthase and the rate of oxygen consumption by mitochondria, while the deletion of ASAP can inhibit the growth of colon tumors in vitro [174]. LINC00675 encodes a small protein of 79aa, FORCP, endogenously expressed mainly in the cytoplasm and can promote tumor cell apoptosis in response to stress in the endoplasmic reticulum. Functionally, FORCP can also inhibit cancer cell proliferation and clone formation to further inhibit tumor progression [176]. SRSP is a functional small peptideof 130aa that is encoded by LOC90024, which can promote CRC tumorigenesis and progression. Long SP4 isomer (L-SP4 protein) is a carcinogenic protein, while short SP4 isomer (S-SP4 peptide) is a non-carcinogenic protein. SRSP peptide increases the probability of SRSF3 binding to exon 3 of transcription factor SP4, thus inducing the formation of carcinogenic protein L-SP4, which leads to tumorigenesis [177]. Pep-AP is a short peptide encoded by lnc-AP, which can reverse the resistance of colon cancer cells and make them more sensitive to oxaliplatin [173]. Kikuchi et al. reported that PVT1 is a carcinogenic peptide encoded by lncRNA PVT1, located downstream of transcription factor MYC gene and is abnormally overexpressed in various cancers. In CRC, PVT1 carcinogenic peptide can be detected in CD8 T cells and peripheral blood mononuclear cells of patients [175].

Breast cancer & triple-negative breast cancer (TNBC)

According to the global cancer statistics in 2020, breast cancer ranked first in the incidence of malignant tumors in the world, and it was the fifth leading cause of death among malignant tumors [197]. It has been reported that lncRNA CASIMO1 encodes a 10 kDa microprotein CASIMO1 in breast cancer. The overexpression of CASIMO1 causes binding to the oncogene SQLE, promoting its accumulation at the protein level and subsequently accelerates the proliferation of breast cancer. Mutation of lncRNA CASIMO1 translation promoter or knockout of lncRNA CASIMO1 causes a loss of its carcinogenic effect [190]. Triple-negative breast cancer (TNBC) accounts for 15% of breast cancers with a high degree of malignancy and usually poor prognosis. In TNBC, the three main breast cancer tumor markers: estrogen receptor (ER), progesterone receptor (PR), and Her2 are all negative [198]. ASRPS and CIP2A-BP are two small proteins with tumor suppressive roles in TNBC. ASRPS is a functional small peptide of 60aa encoded by LINC00908. ASRPS can inhibit tumor progression and the mechanism is that the ASRPS peptide binds STAT3 directly through the coiled coil domain (CCD), and then down-regulates the phosphorylation of STAT3, resulting in a decrease in the expression of VEGF [26]. A 5.5 kDa peptide encoded by LINC00665 has been designated as CIP2A-BP. CIP2A-BP can inhibit the progression of TNBC. It replaces the B56γ subunit after binding to the tumor gene CIP2A, and then stimulates PP2A activity, thus reducing the expression of MMP-2, MMP-9, and Snail by inhibiting the PI3K/AKT/NFκB signaling pathway [179].

Upper gastrointestinal cancer

The oral cavity is the starting point of the upper digestive tract and has important physiological functions. Oral cancer is becoming a global public health problem. At present, about 377,000 people suffer from oral squamous cell carcinoma and 177,000 people die each year [197]. The tumor promoting role of lncRNA HOXB-AS3 has been well documented in various types of cancers [99, 199, 200]. HOXB-AS3 peptide is a small 53aa peptide encoded by lncRNA HOXB-AS3. In oral squamous cell carcinoma (OSCC), HOXB-AS3 and its encoded peptides can promote the proliferation of cancer cells, which is achieved by the direct binding of HOXB-AS3 peptide to IGF2BP2 to maintain the stability of mRNA stability of the oncogene c-Myc [188]. Esophageal squamous cell carcinoma (ESCC) is threatening more than 400,000 people worldwide, and men are more vulnerable than women [197]. A study by Wu et a.l has shown that LINC00278, a Y-linked lncRNA, down-regulated in male ESCC, encoded a micropeptide called YY1BM. YY1BM binds to YY1 to suppress the expression of eEF2K simulated by YY1 and the androgen receptor (AR), which promotes apoptosis and inhibits the proliferation of ESCC. However, YY1BM is down-regulated in ESCC, which in turn promotes cancer progression [191].

Other types of cancer

Lung cancer is especially important in the eyes of the public, since it causes more than 1.76 million death per year [201]. Studies with lung cancer have found that lncRNA UBAP1-AST6 encodes a peptide of 12.8 kDa, called UBAP1-AST6. It can promote cell proliferation and clone formation, but the specific mechanism is not clear. This peptide may promote the occurrence and development of tumors [178]. Pancreatic ductal carcinoma (PDAC) is a malignant tumor with low survival rate. The vast majority of PDAC patients have KRAS mutations [202]. Rason is a 108aa peptide encoded by LINC00673. Rason prolongs the active state of KRAS signaling by binding to KRAS in order to drive tumorigenesis and metastasis [192]. Melanoma is a type of skin cancer caused by melanocytes, the pigment-producing cells found in tissues such as the epidermis, hair follicles, and the iris. In most countries, the incidence of melanoma has been increasing over the past few decades [203, 204]. Delphine et a.l found two new polypeptides, MELOE-1 and MELOE-2, which are involved in immunosurveillance [205]. The same researchers also discovered MELOE-3, a 54aa functional small peptide encoded by lncRNA meloe, which is expressed in both melanoma cells and normal melanocytes. MELOE-3 has poor immunogenicity in melanoma cells, and the protein expressed in the physiological state is also associated with immune tolerance [189]. This provides a promising T cell target for melanoma immunotherapy.

In addition to the above cancers, peptides encoded by lncRNA have been identified to drive the progression of other cancers, including head and neck squamous cell carcinoma (HNSCC), glioma and ovarian cancer. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer globally with a high mortality rate of 40 to 50% [206]. MIAC is the first micropeptide, 51aa in length, found in head and neck squamous cell carcinoma, which is encoded by lncRNA RP11-469H8.6. MIAC directly binds to aquaporin 2 (AQP2) to suppress the expression of SEPT2 & ITGB4. It then inhibits the actin cytoskeleton, which is a key regulatory factor in the migration and invasion of cancer cells, thus suppressing tumor growth and metastasis [194]. Glioma is the most common primary tumor in the brain, accounting for up to 81% of malignant brain tumors. Although relatively rare, it has a high mortality rate [207]. Cao et al. predicted lncRNAs which may encode small transmembrane peptides in gliomas using in silico approaches. They demonstrated that lncRNA DLEU1 has two smORFs (ORF1 and ORF8). DLEU1 encodes small peptides, ORF1 and ORF8, which can aggregate to form similar ion channels and lead to an increase in the permeability of the glioma [187]. However, the expression and function of these peptides need to be verified by further experiments. Ovarian cancer is a type of tumor among females which has a high degree of malignancy. It has a low survival rate and threatens the health of most women [208]. DDUP is a polypeptide of 186aa encoded by lncRNA CTBP1. In ovarian cancer, DDUP can bind to ATR kinase activated by DNA damage and can be phosphorylated, resulting in structural changes in DDUP. Conformational changes aggravate the binding ability of DDUP to rapidly phosphorylated histone H2AX (γ-H2AX) and RAD18 (transduction of DNA damage signal), resulting in the formation of a stable γ-H2AX/DDUP/RAD18 complex and persistent retention of RAD18 foci at the injured site [193]. This leads to resistance to chemotherapy and radiotherapy based on DNA damage.

Conclusions and future perspectives

Recent studies have shown that some originally defined lncRNAs can participate in the regulation of multi-organ tumors by encoding functional small peptides, which totally changed our understanding of these supposedly non-coding RNAs. Functional peptides, such as HOXB-AS3, pep-AP, ASAP, PVT1, SRSP, etc. are encoded by lncRNAs and promote or inhibit the development of CRC through a series of regulatory processes. In other cancers, there are similar small peptides/proteins, which are encoded by lncRNAs and affect the occurrence and development of tumors. Research in this area has just begun and these new findings may lead to change in the classification of these lncRNAs in the future.

At present, it has been confirmed that lncRNAs can regulate the occurrence and development of tumors either by themselves or through encoded small peptides. Although this has great application value, such as the development of new antineoplastic drugs and/or new cancer biomarkers, deeper research is needed before the real clinical applications are possible. At the current stage, several questions remain to be answered. First, until now, studies have only identified a small number of lncRNA-encoded peptides. There are still an inestimable number of functional peptides encoded by lncRNA waiting to be discovered. Over time, we will get a clearer picture of the role of these peptides and will be able to determine whether they are evolutionarily important. Second, with the development of technology, more advanced tools are needed to accurately predict and validate the coding potential of lncRNAs. Third, the mechanism by which many small peptides affect tumor progression is not clear. It remains unclear as to whether the small peptides themselves or lncRNAs or both are functional. Scientists need to carefully discriminate between the functions of lnRNA and that of the encoded peptides or to determine whether the lncRNA is bifunctional. Finally, if these micropeptides are going to be used as potential anti-tumor targets, the upstream regulatory mechanism and downstream binding proteins need to be further clarified. Despite these problems, lncRNA-encoded peptides are a promising resource for the development of new diagnostic and prognostic biomarkers and/or therapeutic targets in cancer, which merits further and an in-depth investigation. It will be a widely-studied topic in cancer research in years to come and will undoubtedly push research into human biomedicine to a new level.

Availability of data and materials

Not applicable.

References

  1. Bray F, Laversanne M, Weiderpass E, Soerjomataram I. The ever-increasing importance of cancer as a leading cause of premature death worldwide. Cancer. 2021;127(16):3029–30.

    Article  PubMed  Google Scholar 

  2. Goodall GJ, Wickramasinghe VO. RNA in cancer. Nat Rev Cancer. 2021;21(1):22–36.

    Article  CAS  PubMed  Google Scholar 

  3. Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018.

  4. Birney E, Stamatoyannopoulos JA, Dutta A, Guigó R, Gingeras TR, Margulies EH, Weng Z, Snyder M, Dermitzakis ET, Thurman RE, et al. Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project. Nature. 2007;447(7146):799–816.

    Article  CAS  PubMed  Google Scholar 

  5. Schmitt AM, Chang HY. Long noncoding RNAs in cancer pathways. Cancer Cell. 2016;29(4):452–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai M-C, Hung T, Argani P, Rinn JL, et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464(7291):1071–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kino T, Hurt DE, Ichijo T, Nader N, Chrousos GP. Noncoding RNA gas5 is a growth arrest- and starvation-associated repressor of the glucocorticoid receptor. Sci Signal. 2010.

  8. Wu D, Yang B, Chen J, Xiong H, Li Y, Pan Z, Cao Y, Chen J, Li T, Zhou S, et al. Upregulation of long non-coding RNA RAB1A-2 induces FGF1 expression worsening lung cancer prognosis. Cancer Lett. 2018;438:116–25.

    Article  CAS  PubMed  Google Scholar 

  9. Yang F, Zhang H, Mei Y, Wu M. Reciprocal regulation of HIF-1α and lincRNA-p21 modulates the Warburg effect. Mol Cell. 2014.

  10. Salameh A, Lee AK, Cardó-Vila M, Nunes DN, Efstathiou E, Staquicini FI, Dobroff AS, Marchiò S, Navone NM, Hosoya H, et al. PRUNE2 is a human prostate cancer suppressor regulated by the intronic long noncoding RNA PCA3. Proc Natl Acad Sci U S A. 2015;112(27):8403–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gong C, Maquat LE. lncRNAs transactivate STAU1-mediated mRNA decay by duplexing with 3’ UTRs via Alu elements. Nature. 2011;470(7333):284–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Bernard D, Prasanth KV, Tripathi V, Colasse S, Nakamura T, Xuan Z, Zhang MQ, Sedel F, Jourdren L, Coulpier F, et al. A long nuclear-retained non-coding RNA regulates synaptogenesis by modulating gene expression. EMBO J. 2010;29(18):3082–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nie W, Ge H-j, Yang X-q, Sun X, Huang H, Tao X, Chen W-s, Li B. LncRNA-UCA1 exerts oncogenic functions in non-small cell lung cancer by targeting miR-193a-3p. Cancer Lett. 2016.

  14. Ingolia NT, Lareau LF, Weissman JS. Ribosome profiling of mouse embryonic stem cells reveals the complexity and dynamics of mammalian proteomes. Cell. 2011;147(4):789–802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kim M-S, Pinto SM, Getnet D, Nirujogi RS, Manda SS, Chaerkady R, Madugundu AK, Kelkar DS, Isserlin R, Jain S, et al. A draft map of the human proteome. Nature. 2014;509(7502):575–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Slavoff SA, Mitchell AJ, Schwaid AG, Cabili MN, Ma J, Levin JZ, Karger AD, Budnik BA, Rinn JL, Saghatelian A. Peptidomic discovery of short open reading frame-encoded peptides in human cells. Nat Chem Biol. 2013;9(1):59–64.

    Article  CAS  PubMed  Google Scholar 

  17. Ji Z, Song R, Regev A, Struhl K. Many lncRNAs, 5’UTRs, and pseudogenes are translated and some are likely to express functional proteins. Elife. 2015;4: e08890.

    Article  PubMed  Google Scholar 

  18. Apostolopoulos V, Bojarska J, Chai T-T, Elnagdy S, Kaczmarek K, Matsoukas J, New R, Parang K, Lopez OP, Parhiz H, et al. A global review on short peptides: frontiers and perspectives. Molecules. 2021.

  19. Bojarska J, Kaczmarek K, Zabrocki J. Wolf WJIJNS: amino acids: molecules of life. Int J Nutr Sci. 2019;4:1035–7.

    Google Scholar 

  20. Huang J-Z, Chen M, Chen D, Gao X-C, Zhu S, Huang H, Hu M, Zhu H, Yan G-R. A peptide encoded by a putative lncRNA HOXB-AS3 suppresses colon cancer growth. Mol Cell. 2017.

  21. Li CM, Haratipour P, Lingeman RG, Perry JJP, Gu L, Hickey RJ, Malkas LH. Novel peptide therapeutic approaches for cancer treatment. Cells. 2021.

  22. Soon TN, Chia AY, Yap WH, Tang Y-QJP. letters p: anticancer mechanisms of bioactive peptides. Protein Pept Lett. 2020;27(9):823–30.

    Article  CAS  PubMed  Google Scholar 

  23. Xie M, Liu D, Yang Y. Anti-cancer peptides: classification, mechanism of action, reconstruction and modification. Open Biol. 2020;10(7): 200004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Liu J, Liu Z-X, Wu Q-N, Lu Y-X, Wong C-W, Miao L, Wang Y, Wang Z, Jin Y, He M-M, et al. Long noncoding RNA AGPG regulates PFKFB3-mediated tumor glycolytic reprogramming. Nat Commun. 2020;11(1):1507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Yin D, Lu X, Su J, He X, De W, Yang J, Li W, Han L, Zhang E. Long noncoding RNA AFAP1-AS1 predicts a poor prognosis and regulates non-small cell lung cancer cell proliferation by epigenetically repressing p21 expression. Mol Cancer. 2018;17(1):92.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Wang Y, Wu S, Zhu X, Zhang L, Deng J, Li F, Guo B, Zhang S, Wu R, Zhang Z, et al. LncRNA-encoded polypeptide ASRPS inhibits triple-negative breast cancer angiogenesis. J Exp Med. 2020.

  27. Bhan A, Soleimani M, Mandal SS. Long noncoding RNA and cancer: a new paradigm. Cancer Res. 2017;77(15):3965–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Han P, Chang C-P. Long non-coding RNA and chromatin remodeling. RNA Biol. 2015;12(10):1094–8.

    Article  PubMed  PubMed Central  Google Scholar 

  29. He W, Liang B, Wang C, Li S, Zhao Y, Huang Q, Liu Z, Yao Z, Wu Q, Liao W, et al. MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene. 2019;38(23):4637–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Huang W, Sun Y-M, Pan Q, Fang K, Chen X-T, Zeng Z-C, Chen T-Q, Zhu S-X, Huang L-B, Luo X-Q, et al. The snoRNA-like lncRNA LNC-SNO49AB drives leukemia by activating the RNA-editing enzyme ADAR1. Cell Discov. 2022;8(1):117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Luo Y, Zheng S, Wu Q, Wu J, Zhou R, Wang C, Wu Z, Rong X, Huang N, Sun L, et al. Long noncoding RNA (lncRNA) EIF3J-DT induces chemoresistance of gastric cancer via autophagy activation. Autophagy. 2021;17(12):4083–101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. McCabe EM, Rasmussen TP. lncRNA involvement in cancer stem cell function and epithelial-mesenchymal transitions. Semin Cancer Biol. 2021;75:38–48.

    Article  CAS  PubMed  Google Scholar 

  33. Statello L, Guo C-J, Chen L-L, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021.

  34. Yang X, Zhang S, He C, Xue P, Zhang L, He Z, Zang L, Feng B, Sun J, Zheng M. METTL14 suppresses proliferation and metastasis of colorectal cancer by down-regulating oncogenic long non-coding RNA XIST. Mol Cancer. 2020;19(1):46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu H, Deng H, Zhao Y, Li C, Liang Y. LncRNA XIST/miR-34a axis modulates the cell proliferation and tumor growth of thyroid cancer through MET-PI3K-AKT signaling. J Exp Clin Cancer Res. 2018;37(1):279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Shen J, Hong L, Yu D, Cao T, Zhou Z, He S. LncRNA XIST promotes pancreatic cancer migration, invasion and EMT by sponging miR-429 to modulate ZEB1 expression. Int J Biochem Cell Biol. 2019;113:17–26.

    Article  CAS  PubMed  Google Scholar 

  37. Wang J, Cai H, Dai Z, Wang G. Down-regulation of lncRNA XIST inhibits cell proliferation via regulating miR-744/RING1 axis in non-small cell lung cancer. Clin Sci (Lond). 2019;133(14):1567–79.

    Article  CAS  PubMed  Google Scholar 

  38. Sun L, Zhang M, Qu H. lncRNA XIST regulates cell proliferation, migration and invasion via regulating miR-30b and RECK in nasopharyngeal carcinoma. Oncol Lett. 2021;21(4):256.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Luo C, Quan Z, Zhong B, Zhang M, Zhou B, Wang S, Luo X, Tang C. lncRNA XIST promotes glioma proliferation and metastasis through miR-133a/SOX4. Exp Ther Med. 2020;19(3):1641–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Ma SQ, Wang YC, Li Y, Li XY, Yang J, Sheng YM. LncRNA XIST promotes proliferation and cisplatin resistance of oral squamous cell carcinoma by downregulating miR-27b-3p. J Biol Regul Homeost Agents. 2020;34(6):1993–2001.

    CAS  PubMed  Google Scholar 

  41. Zeng Z-L, Lu J-H, Wang Y, Sheng H, Wang Y-N, Chen Z-H, Wu Q-N, Zheng J-B, Chen Y-X, Yang D-D, et al. The lncRNA XIST/miR-125b-2-3p axis modulates cell proliferation and chemotherapeutic sensitivity via targeting wee1 in colorectal cancer. Cancer Med. 2021;10(7):2423–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang Y, Zhu Z, Huang S, Zhao Q, Huang C, Tang Y, Sun C, Zhang Z, Wang L, Chen H, et al. lncRNA XIST regulates proliferation and migration of hepatocellular carcinoma cells by acting as miR-497-5p molecular sponge and targeting PDCD4. Cancer Cell Int. 2019;19:198.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Zhuang X, Tong H, Ding Y, Wu L, Cai J, Si Y, Zhang H, Shen M. Long noncoding RNA ABHD11-AS1 functions as a competing endogenous RNA to regulate papillary thyroid cancer progression by miR-199a-5p/SLC1A5 axis. Cell Death Dis. 2019;10(8):620.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Lei X, Li L, Duan X. Long non-coding RNA ABHD11-AS1 promotes colorectal cancer development through regulation of miR-133a/SOX4 axis. 2018. Biosci Rep. https://doi.org/10.1042/BSR20181386.

  45. Qiao X, Lv SX, Qiao Y, Li QP, Ye B, Wang CC, Miao L. Long noncoding RNA ABHD11-AS1 predicts the prognosis of pancreatic cancer patients and serves as a promoter by activating the PI3K-AKT pathway. Eur Rev Med Pharmacol Sci. 2018;22(24):8630–9.

    CAS  PubMed  Google Scholar 

  46. Wu D-D, Chen X, Sun K-X, Wang L-L, Chen S, Zhao Y. Role of the lncRNA ABHD11-AS in the tumorigenesis and progression of epithelial ovarian cancer through targeted regulation of RhoC. Mol Cancer. 2017;16(1):138.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ye Y, Chen J, Zhou Y, Fu Z, Zhou Q, Wang Y, Gao W, Zheng S, Zhao X, Chen T, et al. High expression of AFAP1-AS1 is associated with poor survival and short-term recurrence in pancreatic ductal adenocarcinoma. J Transl Med. 2015;13:137.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Liu X-H, Sun M, Nie F-Q, Ge Y-B, Zhang E-B, Yin D-D, Kong R, Xia R, Lu K-H, Li J-H, et al. Lnc RNA HOTAIR functions as a competing endogenous RNA to regulate HER2 expression by sponging miR-331-3p in gastric cancer. Mol Cancer. 2014;13:92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Li X, Hou L, Yin L, Zhao S. LncRNA XIST interacts with miR-454 to inhibit cells proliferation, epithelial mesenchymal transition and induces apoptosis in triple-negative breast cancer. J Biosci. 2020.

  50. Wang Y, Chen W, Lian J, Zhang H, Yu B, Zhang M, Wei F, Wu J, Jiang J, Jia Y, et al. The lncRNA PVT1 regulates nasopharyngeal carcinoma cell proliferation via activating the KAT2A acetyltransferase and stabilizing HIF-1α. Cell Death Differ. 2020;27(2):695–710.

    Article  CAS  PubMed  Google Scholar 

  51. Wang X, Cheng Z, Dai L, Jiang T, Li P, Jia L, Jing X, An L, Liu M, Wu S, et al. LncRNA PVT1 facilitates proliferation, migration and invasion of NSCLC cells via miR-551b/FGFR1 Axis. Onco Targets Ther. 2021;14:3555–65.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Wang F, Ji X, Wang J, Ma X, Yang Y, Zuo J, Cui J. LncRNA PVT1 enhances proliferation and cisplatin resistance via regulating mir-194-5p/HIF1A axis in oral squamous cell carcinoma. Onco Targets Ther. 2020;13:243–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Pan Y, Liu L, Cheng Y, Yu J, Feng Y. Amplified LncRNA PVT1 promotes lung cancer proliferation and metastasis by facilitating VEGFC expression. Biochem Cell Biol. 2020;98(6):676–82.

    Article  CAS  PubMed  Google Scholar 

  54. Liu Y, Wu Y, Zhu Z, Gong J, Dou W. Knockdown of lncRNA PVT1 inhibits the proliferation and accelerates the apoptosis of colorectal cancer cells via the miR-761/MAPK1 axis. Mol Med Rep. 2021.

  55. Chen J, Yu Y, Li H, Hu Q, Chen X, He Y, Xue C, Ren F, Ren Z, Li J, et al. Long non-coding RNA PVT1 promotes tumor progression by regulating the miR-143/HK2 axis in gallbladder cancer. Mol Cancer. 2019;18(1):33.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Chang Q-Q, Chen C-Y, Chen Z, Chang S. LncRNA PVT1 promotes proliferation and invasion through enhancing Smad3 expression by sponging miR-140-5p in cervical cancer. Radiol Oncol. 2019;53(4):443–52.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Zhao L, Kong H, Sun H, Chen Z, Chen B, Zhou M. LncRNA-PVT1 promotes pancreatic cancer cells proliferation and migration through acting as a molecular sponge to regulate miR-448. J Cell Physiol. 2018;233(5):4044–55.

    Article  CAS  PubMed  Google Scholar 

  58. Ding Y, Fang Q, Li Y, Wang Y. Amplification of lncRNA PVT1 promotes ovarian cancer proliferation by binding to miR-140. Mamm Genome. 2019;30(7–8):217–25.

    Article  CAS  PubMed  Google Scholar 

  59. Tian Z, Cao S, Li C, Xu M, Wei H, Yang H, Sun Q, Ren Q, Zhang L. LncRNA PVT1 regulates growth, migration, and invasion of bladder cancer by miR-31/ CDK1. J Cell Physiol. 2019;234(4):4799–811.

    Article  CAS  PubMed  Google Scholar 

  60. Zhou Y, Wang Y, Lin M, Wu D, Zhao M. LncRNA HOTAIR promotes proliferation and inhibits apoptosis by sponging miR-214-3p in HPV16 positive cervical cancer cells. Cancer Cell Int. 2021;21(1):400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Porman AM, Roberts JT, Duncan ED, Chrupcala ML, Levine AA, Kennedy MA, Williams MM, Richer JK, Johnson AM. A single N6-methyladenosine site regulates lncRNA HOTAIR function in breast cancer cells. PLoS Biol. 2022;20(11): e3001885.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Liang H, Peng J. LncRNA HOTAIR promotes proliferation, invasion and migration in NSCLC cells via the CCL22 signaling pathway. PLoS ONE. 2022;17(2): e0263997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gao C, Guo C, Liu K, Guo N. LncRNA HOTAIR promotes proliferation and migration of OSCC cells via targeting miR-126. Minerva Gastroenterol (Torino). 2022;68(2):245–7.

    PubMed  Google Scholar 

  64. Guo S, King P, Liang E, Guo AA, Liu M. LncRNA HOTAIR sponges miR-301a-3p to promote glioblastoma proliferation and invasion through upregulating FOSL1. Cell Signal. 2022;94: 110306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Wu D, Zhu J, Fu Y, Li C, Wu B. LncRNA HOTAIR promotes breast cancer progression through regulating the miR-129-5p/FZD7 axis. Cancer Biomark. 2021;30(2):203–12.

    Article  CAS  PubMed  Google Scholar 

  66. Lu M, Qin X, Zhou Y, Li G, Liu Z, Yue H, Geng X. LncRNA HOTAIR suppresses cell apoptosis, autophagy and induces cell proliferation in cholangiocarcinoma by modulating the miR-204-5p/HMGB1 axis. Biomed Pharmacother. 2020;130: 110566.

    Article  CAS  PubMed  Google Scholar 

  67. Fan L, Lei H, Lin Y, Zhou Z, Li J, Wu A, Shu G, Roger S, Yin G. Hotair promotes the migration and proliferation in ovarian cancer by miR-222-3p/CDK19 axis. Cell Mol Life Sci. 2022;79(5):254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Wang J, Wang N, Zheng Z, Che Y, Suzuki M, Kano S, Lu J, Wang P, Sun Y, Homma A. Exosomal lncRNA HOTAIR induce macrophages to M2 polarization via PI3K/ p-AKT /AKT pathway and promote EMT and metastasis in laryngeal squamous cell carcinoma. BMC Cancer. 2022;22(1):1208.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Peng CL, Zhao XJ, Wei CC, Wu JW. LncRNA HOTAIR promotes colon cancer development by down-regulating miRNA-34a. Eur Rev Med Pharmacol Sci. 2019;23(13):5752–61.

    PubMed  Google Scholar 

  70. Fu WM, Zhu X, Wang WM, Lu YF, Hu BG, Wang H, Liang WC, Wang SS, Ko CH, Waye MM, et al. Hotair mediates hepatocarcinogenesis through suppressing miRNA-218 expression and activating P14 and P16 signaling. J Hepatol. 2015;63(4):886–95.

    Article  CAS  PubMed  Google Scholar 

  71. Zhou Q, Liu ZZ, Wu H, Kuang WL. LncRNA H19 promotes cell proliferation, migration, and angiogenesis of glioma by regulating Wnt5a/β-catenin pathway via targeting miR-342. Cell Mol Neurobiol. 2022;42(4):1065–77.

    Article  CAS  PubMed  Google Scholar 

  72. Liu HC, Zhu WY, Ren LY. LncRNA H19 inhibits proliferation and enhances apoptosis of nephroblastoma cells by regulating the miR-675/TGFBI axis. Eur Rev Med Pharmacol Sci. 2022;26(11):3800–6.

    PubMed  Google Scholar 

  73. Sun Y, Zhu Q, Yang W, Shan Y, Yu Z, Zhang Q, Wu H. LncRNA H19/miR-194/PFTK1 axis modulates the cell proliferation and migration of pancreatic cancer. J Cell Biochem. 2019;120(3):3874–86.

    Article  CAS  PubMed  Google Scholar 

  74. Zhang Y, Zhu R, Wang J, Cui Z, Wang Y, Zhao Y. Upregulation of lncRNA H19 promotes nasopharyngeal carcinoma proliferation and metastasis in let-7 dependent manner. Artif Cells Nanomed Biotechnol. 2019;47(1):3854–61.

    Article  CAS  PubMed  Google Scholar 

  75. Zhao Y, Feng C, Li Y, Ma Y, Cai R. LncRNA H19 promotes lung cancer proliferation and metastasis by inhibiting miR-200a function. Mol Cell Biochem. 2019;460(1–2):1–8.

    Article  CAS  PubMed  Google Scholar 

  76. Zhou Y, Fan RG, Qin CL, Jia J, Wu XD, Zha WZ. LncRNA-H19 activates CDC42/PAK1 pathway to promote cell proliferation, migration and invasion by targeting miR-15b in hepatocellular carcinoma. Genomics. 2019;111(6):1862–72.

    Article  CAS  PubMed  Google Scholar 

  77. Ma F, Wang SH, Cai Q, Zhang MD, Yang Y, Ding J. Overexpression of LncRNA AFAP1-AS1 predicts poor prognosis and promotes cells proliferation and invasion in gallbladder cancer. Biomed Pharmacother. 2016;84:1249–55.

    Article  CAS  PubMed  Google Scholar 

  78. Zhang JY, Weng MZ, Song FB, Xu YG, Liu Q, Wu JY, Qin J, Jin T, Xu JM. Long noncoding RNA AFAP1-AS1 indicates a poor prognosis of hepatocellular carcinoma and promotes cell proliferation and invasion via upregulation of the RhoA/Rac2 signaling. Int J Oncol. 2016;48(4):1590–8.

    Article  CAS  PubMed  Google Scholar 

  79. Li Y, Hu J, Guo D, Ma W, Zhang X, Zhang Z, Lu G, He S. LncRNA SNHG5 promotes the proliferation and cancer stem cell-like properties of HCC by regulating UPF1 and Wnt-signaling pathway. Cancer Gene Ther. 2022;29(10):1373–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Xu W, Ding M, Wang B, Cai Y, Guo C, Yuan C. Molecular mechanism of the canonical oncogenic lncRNA MALAT1 in gastric cancer. Curr Med Chem. 2021;28(42):8800–9.

    Article  CAS  PubMed  Google Scholar 

  81. Wang P, Hu L, Fu G, Lu J, Zheng Y, Li Y, Jia L. LncRNA MALAT1 promotes the proliferation, migration, and invasion of melanoma cells by downregulating miR-23a. Cancer Manag Res. 2020;12:6553–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Jin Y, Feng SJ, Qiu S, Shao N, Zheng JH. LncRNA MALAT1 promotes proliferation and metastasis in epithelial ovarian cancer via the PI3K-AKT pathway. Eur Rev Med Pharmacol Sci. 2017;21(14):3176–84.

    CAS  PubMed  Google Scholar 

  83. Yue X, Wu WY, Dong M, Guo M. LncRNA MALAT1 promotes breast cancer progression and doxorubicin resistance via regulating miR-570-3p. Biomed J. 2021;44(6 Suppl 2):S296-s304.

    Article  CAS  PubMed  Google Scholar 

  84. Wu Q, Meng WY, Jie Y, Zhao H. LncRNA MALAT1 induces colon cancer development by regulating miR-129-5p/HMGB1 axis. J Cell Physiol. 2018;233(9):6750–7.

    Article  CAS  PubMed  Google Scholar 

  85. Zhou D, Wang Y, Hu H, Liu H, Deng J, Li L, Zheng C. lncRNA MALAT1 promotes HCC metastasis through the peripheral vascular infiltration via miRNA-613: a primary study using contrast ultrasound. World J Surg Oncol. 2022;20(1):203.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Wang D, Zhang S, Zhao M, Chen F. LncRNA MALAT1 accelerates non-small cell lung cancer progression via regulating miR-185-5p/MDM4 axis. Cancer Med. 2020;9(23):9138–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Zhan FL, Chen CF, Yao MZ. LncRNA TUG1 facilitates proliferation, invasion and stemness of ovarian cancer cell via miR-186-5p/ZEB1 axis. Cell Biochem Funct. 2020;38(8):1069–78.

    Article  CAS  PubMed  Google Scholar 

  88. Xie C, Chen B, Wu B, Guo J, Cao Y. LncRNA TUG1 promotes cell proliferation and suppresses apoptosis in osteosarcoma by regulating miR-212-3p/FOXA1 axis. Biomed Pharmacother. 2018;97:1645–53.

    Article  CAS  PubMed  Google Scholar 

  89. Zhao W, Jiang X, Yang S. lncRNA TUG1 promotes cell proliferation, migration, and invasion in hepatocellular carcinoma via regulating miR-29c-3p/COL1A1 axis. Cancer Manag Res. 2020;12:6837–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Tan J, Liu B, Zhou L, Gao J, Wang XK, Liu Y, Wang JR. LncRNA TUG1 promotes bladder cancer malignant behaviors by regulating the miR-320a/FOXQ1 axis. Cell Signal. 2022;91: 110216.

    Article  CAS  PubMed  Google Scholar 

  91. Zheng Y, Zheng B, Meng X, Yan Y, He J, Liu Y. LncRNA DANCR promotes the proliferation, migration, and invasion of tongue squamous cell carcinoma cells through miR-135a-5p/KLF8 axis. Cancer Cell Int. 2019;19:302.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Tang Y, Wu L, Zhao M, Zhao G, Mao S, Wang L, Liu S, Wang X. LncRNA SNHG4 promotes the proliferation, migration, invasiveness, and epithelial-mesenchymal transition of lung cancer cells by regulating miR-98-5p. Biochem Cell Biol. 2019;97(6):767–76.

    Article  CAS  PubMed  Google Scholar 

  93. Zhang L, Liu SK, Song L, Yao HR. SP1-induced up-regulation of lncRNA LUCAT1 promotes proliferation, migration and invasion of cervical cancer by sponging miR-181a. Artif Cells Nanomed Biotechnol. 2019;47(1):556–64.

    CAS  PubMed  Google Scholar 

  94. Aichen Z, Kun W, Xiaochun S, Lingling T. LncRNA FGD5-AS1 promotes the malignant phenotypes of ovarian cancer cells via targeting miR-142-5p. Apoptosis. 2021;26(5–6):348–60.

    Article  PubMed  Google Scholar 

  95. 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.

    Article  CAS  PubMed  Google Scholar 

  96. Zhang W, Li X, Zhang W, Lu Y, Lin W, Yang L, Zhang Z, Li X. The LncRNA CASC11 promotes colorectal cancer cell proliferation and migration by adsorbing miR-646 and miR-381-3p to upregulate their target RAB11FIP2. Front Oncol. 2021;11: 657650.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Li J, Xia R, Liu T, Cai X, Geng G. LncRNA-ATB promotes lung squamous carcinoma cell proliferation, migration, and invasion by targeting microRNA-590-5p/NF90 Axis. DNA Cell Biol. 2020;39(3):459–73.

    Article  CAS  PubMed  Google Scholar 

  98. Li N, Yang G, Luo L, Ling L, Wang X, Shi L, Lan J, Jia X, Zhang Q, Long Z, et al. lncRNA THAP9-AS1 promotes pancreatic ductal adenocarcinoma growth and leads to a poor clinical outcome via sponging miR-484 and interacting with YAP. Clin Cancer Res. 2020;26(7):1736–48.

    Article  PubMed  Google Scholar 

  99. Jiang W, Kai J, Li D, Wei Z, Wang Y, Wang W. lncRNA HOXB-AS3 exacerbates proliferation, migration, and invasion of lung cancer via activating the PI3K-AKT pathway. J Cell Physiol. 2020;235(10):7194–203.

    Article  CAS  PubMed  Google Scholar 

  100. Wong RSY. Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res. 2011;30(1):87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell. 2011;147(4):728–41.

    Article  CAS  PubMed  Google Scholar 

  102. Baehrecke EH. Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol. 2005;6(6):505–10.

    Article  CAS  PubMed  Google Scholar 

  103. Chen J-F, Wu P, Xia R, Yang J, Huo X-Y, Gu D-Y, Tang C-J, De W, Yang F. STAT3-induced lncRNA HAGLROS overexpression contributes to the malignant progression of gastric cancer cells via mTOR signal-mediated inhibition of autophagy. Mol Cancer. 2018;17(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Cheng Z, Liu G, Huang C, Zhao X. KLF5 activates lncRNA DANCR and inhibits cancer cell autophagy accelerating gastric cancer progression. NPJ Genom Med. 2021;6(1):75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Wu Q, Ma J, Wei J, Meng W, Wang Y, Shi M. lncRNA SNHG11 promotes gastric cancer progression by activating the Wnt/β-catenin pathway and oncogenic autophagy. Mol Ther. 2021;29(3):1258–78.

    Article  CAS  PubMed  Google Scholar 

  106. Xu N, Liu B, Lian C, Doycheva DM, Fu Z, Liu Y, Zhou J, He Z, Yang Z, Huang Q, et al. Long noncoding RNA AC003092.1 promotes temozolomide chemosensitivity through miR-195/TFPI-2 signaling modulation in glioblastoma. Cell Death Dis. 2018;9(12):1139.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Yang Y, Chen D, Liu H, Yang K. Increased expression of lncRNA CASC9 promotes tumor progression by suppressing autophagy-mediated cell apoptosis via the AKT/mTOR pathway in oral squamous cell carcinoma. Cell Death Dis. 2019;10(2):41.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Sheng J-Q, Wang M-R, Fang D, Liu L, Huang W-J, Tian D-A, He X-X, Li P-Y. LncRNA NBR2 inhibits tumorigenesis by regulating autophagy in hepatocellular carcinoma. Biomed Pharmacother. 2021;133: 111023.

    Article  CAS  PubMed  Google Scholar 

  109. Brabletz T. To differentiate or not–routes towards metastasis. Nat Rev Cancer. 2012;12(6):425–36.

    Article  CAS  PubMed  Google Scholar 

  110. De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 2013.

  111. Nieto MA, Huang RY-J, Jackson RA, Thiery JP. EMT: 2016. Cell. 2016;166(1):21–45.

    Article  CAS  PubMed  Google Scholar 

  112. Pastushenko I, Blanpain C. EMT transition states during tumor progression and metastasis. Trends Cell Biol. 2019;29(3):212–26.

    Article  CAS  PubMed  Google Scholar 

  113. Lu W, Zhang H, Niu Y, Wu Y, Sun W, Li H, Kong J, Ding K, Shen H-M, Wu H, et al. Long non-coding RNA linc00673 regulated non-small cell lung cancer proliferation, migration, invasion and epithelial mesenchymal transition by sponging miR-150-5p. Mol Cancer. 2017;16(1):118.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Song S, Qiu X. LncRNA miR503HG inhibits epithelial-mesenchymal transition and angiogenesis in hepatocellular carcinoma by enhancing PDCD4 via regulation of miR-15b. Dig Liver Dis. 2021;53(1):107–16.

    Article  CAS  PubMed  Google Scholar 

  115. Xue J, Liao L, Yin F, Kuang H, Zhou X, Wang Y. LncRNA AB073614 induces epithelial- mesenchymal transition of colorectal cancer cells via regulating the JAK/STAT3 pathway. Cancer Biomark. 2018;21(4):849–58.

    Article  CAS  PubMed  Google Scholar 

  116. Zhang C, Hao Y, Wang Y, Xu J, Teng Y, Yang X. TGF-β/SMAD4-regulated LncRNA-LINP1 inhibits epithelial-mesenchymal transition in lung cancer. Int J Biol Sci. 2018;14(12):1715–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Dikomey E, Borgmann K, Köcher S, Kriegs M, Mansour W, Parplys AC, Rieckmann T, Rothkamm K. Radiation DNA damage and use in cancer/therapeutics-translation of radiation modifiers. In: Kelley MR, Fishel ML, editors. DNA Repair in Cancer Therapy. Boston: Academic Press; 2016.

    Google Scholar 

  118. Ren J, Ding L, Zhang D, Shi G, Xu Q, Shen S, Wang Y, Wang T, Hou Y. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018;8(14):3932–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Ma F, Liu X, Zhou S, Li W, Liu C, Chadwick M, Qian C. Long non-coding RNA FGF13-AS1 inhibits glycolysis and stemness properties of breast cancer cells through FGF13-AS1/IGF2BPs/Myc feedback loop. Cancer Lett. 2019;450:63–75.

    Article  CAS  PubMed  Google Scholar 

  120. Rohrig H, Schmidt J, Miklashevichs E, Schell J, John M. Soybean ENOD40 encodes two peptides that bind to sucrose synthase. Proc Natl Acad Sci U S A. 2002;99(4):1915–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Kondo T, Hashimoto Y, Kato K, Inagaki S, Hayashi S, Kageyama Y. Small peptide regulators of actin-based cell morphogenesis encoded by a polycistronic mRNA. Nat Cell Biol. 2007;9(6):660–5.

    Article  CAS  PubMed  Google Scholar 

  122. Zhang C, Zhou B, Gu F, Liu H, Wu H, Yao F, Zheng H, Fu H, Chong W, Cai S, et al. Micropeptide PACMP inhibition elicits synthetic lethal effects by decreasing CtIP and poly(ADP-ribosyl)ation. Mol Cell. 2022.

  123. Papaioannou D, Petri A, Dovey OM, Terreri S, Wang E, Collins FA, Woodward LA, Walker AE, Nicolet D, Pepe F, et al. The long non-coding RNA HOXB-AS3 regulates ribosomal RNA transcription in NPM1-mutated acute myeloid leukemia. Nat Commun. 2019;10(1):5351.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Shin SY. Shin CJPBR: Regulatory non-coding RNAs in plants: potential gene resources for the improvement of agricultural traits. Plant Biotechnol Rep. 2016;10(2):35–47.

    Article  Google Scholar 

  125. Lv D, Chang Z, Cai Y, Li J, Wang L, Jiang Q, Xu K, Ding N, Li X, Xu J, et al. TransLnc: a comprehensive resource for translatable lncRNAs extends immunopeptidome. Nucleic Acids Res. 2022;50(D1):D413–20.

    Article  CAS  PubMed  Google Scholar 

  126. Lin MF, Jungreis I, Kellis M. PhyloCSF: a comparative genomics method to distinguish protein coding and non-coding regions. Bioinformatics. 2011;27(13):i275–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Hu L, Xu Z, Hu B, Lu ZJ. COME: a robust coding potential calculation tool for lncRNA identification and characterization based on multiple features. Nucleic Acids Res. 2017;45(1): e2.

    Article  PubMed  Google Scholar 

  128. Wang L, Park HJ, Dasari S, Wang S, Kocher J-P, Li W. CPAT: coding-potential assessment tool using an alignment-free logistic regression model. Nucleic Acids Res. 2013;41(6): e74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Arrial RT, Togawa RC. Brigido MdM: screening non-coding RNAs in transcriptomes from neglected species using PORTRAIT: case study of the pathogenic fungus paracoccidioides brasiliensis. BMC Bioinfor. 2009;10:239.

    Article  Google Scholar 

  130. Liu J, Gough J, Rost B. Distinguishing protein-coding from non-coding RNAs through support vector machines. PLoS Genet. 2006;2(4): e29.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Kong L, Zhang Y, Ye Z-Q, Liu X-Q, Zhao S-Q, Wei L, Gao G. CPC: assess the protein-coding potential of transcripts using sequence features and support vector machine. Nucleic Acids Res. 2007;35:W345–9.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Kang Y-J, Yang D-C, Kong L, Hou M, Meng Y-Q, Wei L, Gao G. CPC2: a fast and accurate coding potential calculator based on sequence intrinsic features. Nucleic Acids Res. 2017;45(W1):W12–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Washietl S, Findeiss S, Müller SA, Kalkhof S, von Bergen M, Hofacker IL, Stadler PF, Goldman N. RNAcode: robust discrimination of coding and noncoding regions in comparative sequence data. RNA. 2011;17(4):578–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Hanada K, Akiyama K, Sakurai T, Toyoda T, Shinozaki K, Shiu S-H. sORF finder: a program package to identify small open reading frames with high coding potential. Bioinformatics. 2010;26(3):399–400.

    Article  CAS  PubMed  Google Scholar 

  135. Michel AM, Kiniry SJ, O’Connor PBF, Mullan JP, Baranov PV. GWIPS-viz: 2018 update. Nucleic Acids Res. 2018;46(D1):D823–30.

    Article  CAS  PubMed  Google Scholar 

  136. Michel AM, Fox G, Kiran MA, De Bo C, O’connor PBF, Heaphy SM, Mullan JPA, Donohue CA, Higgins DG, Baranov PV. GWIPS-viz: development of a ribo-seq genome browser. Nucleic Acids Res. 2014;42:D859–64.

    Article  CAS  PubMed  Google Scholar 

  137. Zhu M, Gribskov M. MiPepid: micropeptide identification tool using machine learning. BMC Bioinformatics. 2019;20(1):559.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Sun K, Wang H, Sun H: NAMS webserver: coding potential assessment and functional annotation of plant transcripts. Brief Bioinform 2021, 22(3).

  139. Zhang Y, Jia C, Fullwood MJ, Kwoh CK. DeepCPP: a deep neural network based on nucleotide bias information and minimum distribution similarity feature selection for RNA coding potential prediction. Brief Bioinform. 2021;22(2):2073–84.

    Article  CAS  PubMed  Google Scholar 

  140. Bazzini AA, Johnstone TG, Christiano R, Mackowiak SD, Obermayer B, Fleming ES, Vejnar CE, Lee MT, Rajewsky N, Walther TC, et al. Identification of small ORFs in vertebrates using ribosome footprinting and evolutionary conservation. EMBO J. 2014;33(9):981–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Suenaga Y, Kato M, Nagai M, Nakatani K, Kogashi H, Kobatake M, Makino T. Open reading frame dominance indicates protein-coding potential of RNAs. EMBO Rep. 2022;23(6): e54321.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Choi S-W, Kim H-W, Nam J-W. The small peptide world in long noncoding RNAs. Brief Bioinform. 2019;20(5):1853–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Patraquim P, Magny EG, Pueyo JI, Platero AI, Couso JP. Translation and natural selection of micropeptides from long non-canonical RNAs. Nat Commun. 2022;13(1):6515.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Sun L, Luo H, Bu D, Zhao G, Yu K, Zhang C, Liu Y, Chen R, Zhao Y. Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res. 2013;41(17): e166.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Zhang M, Zhao J, Li C, Ge F, Wu J, Jiang B, Song J, Song X. csORF-finder: an effective ensemble learning framework for accurate identification of multi-species coding short open reading frames. Brief Bioinform. 2022.

  146. Leung MKK, Delong A, Alipanahi B, Frey BJ. Machine learning in genomic medicine: a review of computational problems and data sets. Proc IEEE. 2015;104(1):176–97.

    Article  Google Scholar 

  147. Yip KY, Cheng C, Gerstein M. Machine learning and genome annotation: a match meant to be? Genome Biol. 2013;14(5):205.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Alipanahi B, Delong A, Weirauch MT, Frey BJ. Predicting the sequence specificities of DNA- and RNA-binding proteins by deep learning. Nat Biotechnol. 2015;33(8):831–8.

    Article  CAS  PubMed  Google Scholar 

  149. Ballester PJ, Mitchell JBO. A machine learning approach to predicting protein-ligand binding affinity with applications to molecular docking. Bioinformatics. 2010;26(9):1169–75.

    Article  CAS  PubMed  Google Scholar 

  150. Califano A, Alvarez MJ. The recurrent architecture of tumour initiation, progression and drug sensitivity. Nat Rev Cancer. 2017;17(2):116–30.

    Article  CAS  PubMed  Google Scholar 

  151. Carro MS, Lim WK, Alvarez MJ, Bollo RJ, Zhao X, Snyder EY, Sulman EP, Anne SL, Doetsch F, Colman H, et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature. 2010;463(7279):318–25.

    Article  CAS  PubMed  Google Scholar 

  152. Camacho DM, Collins KM, Powers RK, Costello JC, Collins JJ. Next-generation machine learning for biological networks. Cell. 2018;173(7):1581–92.

    Article  CAS  PubMed  Google Scholar 

  153. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Schlesinger F, et al. Landscape of transcription in human cells. Nature. 2012;489(7414):101–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol. 2013;31(9):814–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Marbach D, Costello JC, Küffner R, Vega NM, Prill RJ, Camacho DM, Allison KR, Kellis M, Collins JJ, Stolovitzky G. Wisdom of crowds for robust gene network inference. Nat Methods. 2012;9(8):796–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Xie R, Wen J, Quitadamo A, Cheng J, Shi X. A deep auto-encoder model for gene expression prediction. BMC Genomics. 2017;18(Suppl 9):845.

    Article  PubMed  PubMed Central  Google Scholar 

  157. Jha A, Gazzara MR, Barash Y. Integrative deep models for alternative splicing. Bioinformatics. 2017;33(14):i274–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Hill ST, Kuintzle R, Teegarden A, Merrill E, Danaee P, Hendrix DA. A deep recurrent neural network discovers complex biological rules to decipher RNA protein-coding potential. Nucleic Acids Res. 2018;46(16):8105–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Liu X-Q, Li B-X, Zeng G-R, Liu Q-Y, Ai D-M. Prediction of long non-coding RNAs based on deep learning. Genes. 2019.

  160. Yu N, Yu Z, Pan Y. A deep learning method for lincRNA detection using auto-encoder algorithm. BMC Bioinformatics. 2017;18(Suppl 15):511.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Zou J, Huss M, Abid A, Mohammadi P, Torkamani A, Telenti A. A primer on deep learning in genomics. Nat Genet. 2019;51(1):12–8.

    Article  CAS  PubMed  Google Scholar 

  162. Zhu S, Wang J-Z, Chen D, He Y-T, Meng N, Chen M, Lu R-X, Chen X-H, Zhang X-L, Yan G-R. An oncopeptide regulates m6A recognition by the m6A reader IGF2BP1 and tumorigenesis. Nat Commun. 2020;11(1):1685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Hirano S. Western blot analysis. Methods Mol Biol. 2012;926:87–97.

    Article  CAS  PubMed  Google Scholar 

  164. Hnasko TS, Hnasko RM. The Western Blot. Methods Mol Biol. 2015;1318:87–96.

    Article  PubMed  Google Scholar 

  165. Odell ID, Cook D. Immunofluorescence techniques. J Invest Dermatol. 2013;133(1): e4.

    Article  CAS  PubMed  Google Scholar 

  166. Terpe K. Overview of tag protein fusions: from molecular and biochemical fundamentals to commercial systems. Appl Microbiol Biotechnol. 2003;60(5):523–33.

    Article  CAS  PubMed  Google Scholar 

  167. Wu P, Mo Y, Peng M, Tang T, Zhong Y, Deng X, Xiong F, Guo C, Wu X, Li Y, et al. Emerging role of tumor-related functional peptides encoded by lncRNA and circRNA. Mol Cancer. 2020;19(1):22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Lin J-S, Lai E-M. Protein-protein interactions: co-immunoprecipitation. Methods Mol Biol. 2017;1615:211–9.

    Article  PubMed  Google Scholar 

  169. Chen Y, Long W, Yang L, Zhao Y, Wu X, Li M, Du F, Chen Y, Yang Z, Wen Q, et al. Functional peptides encoded by long non-coding RNAs in gastrointestinal cancer. Front Oncol. 2021;11: 777374.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Zhu S, Wang J-Z, Chen D, He Y-T, Meng N, Chen M, Lu R-X, Chen X-H, Zhang X-L, Yan G-R. An oncopeptide regulates mA recognition by the mA reader IGF2BP1 and tumorigenesis. Nat Commun. 2020;11(1):1685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Nita A, Matsumoto A, Tang R, Shiraishi C, Ichihara K, Saito D, Suyama M, Yasuda T, Tsuji G, Furue M, et al. A ubiquitin-like protein encoded by the “noncoding” RNA TINCR promotes keratinocyte proliferation and wound healing. PLoS Genet. 2021;17(8): e1009686.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Szafron LM, Balcerak A, Grzybowska EA, Pienkowska-Grela B, Felisiak-Golabek A, Podgorska A, Kulesza M, Nowak N, Pomorski P, Wysocki J, et al. The novel gene crnde encodes a nuclear peptide (CRNDEP) which is overexpressed in highly proliferating tissues. PLoS ONE. 2015;10(5): e0127475.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Wang X, Zhang H, Yin S, Yang Y, Yang H, Yang J, Zhou Z, Li S, Ying G, Ba Y. lncRNA-encoded pep-AP attenuates the pentose phosphate pathway and sensitizes colorectal cancer cells to Oxaliplatin. EMBO Rep. 2022;23(1): e53140.

    Article  CAS  PubMed  Google Scholar 

  174. Ge Q, Jia D, Cen D, Qi Y, Shi C, Li J, Sang L, Yang L-J, He J, Lin A, et al. Micropeptide ASAP encoded by LINC00467 promotes colorectal cancer progression by directly modulating ATP synthase activity. J Clin Invest. 2021.

  175. Kikuchi Y, Tokita S, Hirama T, Kochin V, Nakatsugawa M, Shinkawa T, Hirohashi Y, Tsukahara T, Hata F, Takemasa I, et al. CD8 T-cell Immune Surveillance against a Tumor antigen encoded by the oncogenic long noncoding RNA. Cancer Immunol Res. 2021;9(11):1342–53.

    Article  CAS  PubMed  Google Scholar 

  176. Li XL, Pongor L, Tang W, Das S, Muys BR, Jones MF, Lazar SB, Dangelmaier EA, Hartford CC, Grammatikakis I, et al. A small protein encoded by a putative lncRNA regulates apoptosis and tumorigenicity in human colorectal cancer cells. Elife. 2020.

  177. Meng N, Chen M, Chen D, Chen X-H, Wang J-Z, Zhu S, He Y-T, Zhang X-L, Lu R-X, Yan G-R. Small protein hidden in lncRNA promotes “cancerous” RNA splicing and tumorigenesis. Adv Sci. 2020;7(10):1903233.

    Article  CAS  Google Scholar 

  178. Lu S, Zhang J, Lian X, Sun L, Meng K, Chen Y, Sun Z, Yin X, Li Y, Zhao J, et al. A hidden human proteome encoded by “non-coding” genes. Nucleic Acids Res. 2019;47(15):8111–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Guo B, Wu S, Zhu X, Zhang L, Deng J, Li F, Wang Y, Zhang S, Wu R, Lu J, et al. Micropeptide CIP2A-BP encoded by LINC00665 inhibits triple-negative breast cancer progression. EMBO J. 2020;39(1): e102190.

    Article  CAS  PubMed  Google Scholar 

  180. Wu S, Guo B, Zhang L, Zhu X, Zhao P, Deng J, Zheng J, Li F, Wang Y, Zhang S, et al. A micropeptide XBP1SBM encoded by lncRNA promotes angiogenesis and metastasis of TNBC via XBP1s pathway. Oncogene. 2022;41(15):2163–72.

    Article  CAS  PubMed  Google Scholar 

  181. Pang Y, Liu Z, Han H, Wang B, Li W, Mao C, Liu S. Peptide SMIM30 promotes HCC development by inducing SRC/YES1 membrane anchoring and MAPK pathway activation. J Hepatol. 2020;73(5):1155–69.

    Article  CAS  PubMed  Google Scholar 

  182. Xu W, Deng B, Lin P, Liu C, Li B, Huang Q, Zhou H, Yang J, Qu L. Ribosome profiling analysis identified a KRAS-interacting microprotein that represses oncogenic signaling in hepatocellular carcinoma cells. Sci China Life Sci. 2020;63(4):529–42.

    Article  CAS  PubMed  Google Scholar 

  183. Lun Y-Z, Pan Z-P, Liu S-A, Sun J, Han M, Liu B, Dong W, Pan L-H, Cheng J. The peptide encoded by a novel putative lncRNA HBVPTPAP inducing the apoptosis of hepatocellular carcinoma cells by modulating JAK/STAT signaling pathways. Virus Res. 2020;287: 198104.

    Article  CAS  PubMed  Google Scholar 

  184. Xiang X, Fu Y, Zhao K, Miao R, Zhang X, Ma X, Liu C, Zhang N, Qu K. Cellular senescence in hepatocellular carcinoma induced by a long non-coding RNA-encoded peptide PINT87aa by blocking FOXM1-mediated. Theranostics. 2021;11(10):4929–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Guo Z-W, Meng Y, Zhai X-M, Xie C, Zhao N, Li M, Zhou C-L, Li K, Liu T-C, Yang X-X, et al. Translated long non-coding ribonucleic acid ZFAS1 promotes cancer cell migration by elevating reactive oxygen species production in hepatocellular carcinoma. Front Genet. 2019;10:1111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Xu W, Liu C, Deng B, Lin P, Sun Z, Liu A, Xuan J, Li Y, Zhou K, Zhang X, et al. TP53-inducible putative long noncoding RNAs encode functional polypeptides that suppress cell proliferation. Genome Res. 2022;32(6):1026–41.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Cao Y, Yang R, Lee I, Zhang W, Sun J, Meng X, Wang W. Prediction of LncRNA-encoded small peptides in glioma and oligomer channel functional analysis using in silico approaches. PLoS ONE. 2021;16(3): e0248634.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Leng F, Miu Y-Y, Zhang Y, Luo H, Lu X-L, Cheng H, Zheng Z-G. A micro-peptide encoded by HOXB-AS3 promotes the proliferation and viability of oral squamous cell carcinoma cell lines by directly binding with IGF2BP2 to stabilize c-Myc. Oncol Lett. 2021;22(4):697.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Charpentier M, Croyal M, Carbonnelle D, Fortun A, Florenceau L, Rabu C, Krempf M, Labarrière N, Lang F. IRES-dependent translation of the long non coding RNA meloe in melanoma cells produces the most immunogenic MELOE antigens. Oncotarget. 2016;7(37):59704–13.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Polycarpou-Schwarz M, Groß M, Mestdagh P, Schott J, Grund SE, Hildenbrand C, Rom J, Aulmann S, Sinn H-P, Vandesompele J, et al. The cancer-associated microprotein CASIMO1 controls cell proliferation and interacts with squalene epoxidase modulating lipid droplet formation. Oncogene. 2018;37(34):4750–68.

    Article  CAS  PubMed  Google Scholar 

  191. Wu S, Zhang L, Deng J, Guo B, Li F, Wang Y, Wu R, Zhang S, Lu J, Zhou Y. A novel micropeptide encoded by Y-Linked LINC00278 links cigarette smoking and AR signaling in male esophageal squamous cell carcinoma. Cancer Res. 2020;80(13):2790–803.

    Article  CAS  PubMed  Google Scholar 

  192. Cheng R, Li F, Zhang M, Xia X, Wu J, Gao X, Zhou H, Zhang Z, Huang N, Yang X, et al. A novel protein RASON encoded by a lncRNA controls oncogenic RAS signaling in KRAS mutant cancers. Cell Res. 2023;33(1):30–45.

    Article  CAS  PubMed  Google Scholar 

  193. Yu R, Hu Y, Zhang S, Li X, Tang M, Yang M, Wu X, Li Z, Liao X, Xu Y, et al. LncRNA CTBP1-DT-encoded microprotein DDUP sustains DNA damage response signalling to trigger dual DNA repair mechanisms. Nucleic Acids Res. 2022;50(14):8060–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Li M, Li X, Zhang Y, Wu H, Zhou H, Ding X, Zhang X, Jin X, Wang Y, Yin X, et al. Micropeptide MIAC inhibits HNSCC progression by interacting with aquaporin 2. J Am Chem Soc. 2020;142(14):6708–16.

    Article  CAS  PubMed  Google Scholar 

  195. Villanueva A. Hepatocellular carcinoma. N Engl J Med. 2019;380(15):1450–62.

    Article  CAS  PubMed  Google Scholar 

  196. Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet. 2014;383(9927):1490–502.

    Article  PubMed  Google Scholar 

  197. Sung H, Ferlay J, Siegel RL, 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(3):209–49.

    Article  PubMed  Google Scholar 

  198. Denkert C, Liedtke C, Tutt A, von Minckwitz G. Molecular alterations in triple-negative breast cancer-the road to new treatment strategies. Lancet. 2017;389(10087):2430–42.

    Article  CAS  PubMed  Google Scholar 

  199. Xu S, Jia G, Zhang H, Wang L, Cong Y, Lv M, Xu J, Ruan H, Jia X, Xu P, et al. LncRNA HOXB-AS3 promotes growth, invasion and migration of epithelial ovarian cancer by altering glycolysis. Life Sci. 2021;264: 118636.

    Article  CAS  PubMed  Google Scholar 

  200. Zhang XM, Chen H, Zhou B, Zhang QY, Liao Y, Wang JS, Wang ZH. lncRNA HOXB-AS3 promotes hepatoma by inhibiting p53 expression. Eur Rev Med Pharmacol Sci. 2018;22(20):6784–92.

    PubMed  Google Scholar 

  201. Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet. 2021;398(10299):535–54.

    Article  PubMed  Google Scholar 

  202. Park W, Chawla A, O’Reilly EM. Pancreatic cancer: a review. JAMA. 2021;326(9):851–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Erdei E, Torres SM. A new understanding in the epidemiology of melanoma. Expert Rev Anticancer Ther. 2010;10(11):1811–23.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Mao L, Qi Z, Zhang L, Guo J, Si L. Immunotherapy in acral and mucosal melanoma: current status and future directions. Front Immunol. 2021;12: 680407.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Carbonnelle D, Vignard V, Sehedic D, Moreau-Aubry A, Florenceau L, Charpentier M, Mikulits W, Labarriere N, Lang F. The melanoma antigens MELOE-1 and MELOE-2 are translated from a bona fide polycistronic mRNA containing functional IRES sequences. PLoS ONE. 2013;8(9): e75233.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Moskovitz J, Moy J, Ferris RL. Immunotherapy for head and neck squamous cell carcinoma. Curr Oncol Rep. 2018;20(2):22.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Ostrom QT, Bauchet L, Davis FG, Deltour I, Fisher JL, Langer CE, Pekmezci M, Schwartzbaum JA, Turner MC, Walsh KM, et al. The epidemiology of glioma in adults: a “state of the science” review. Neuro Oncol. 2014;16(7):896–913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Kossaï M, Leary A, Scoazec J-Y, Genestie C. Ovarian cancer: a heterogeneous disease. Pathobiology. 2018;85(1–2):41–9.

    Article  PubMed  Google Scholar 

  209. Peltier DC, Roberts A, Reddy P. LNCing RNA to immunity. Trends Immunol. 2022;43(6):478–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Xu D, Cao M, Wang B, et al. Epigenetically regulated lncRNAs dissect the intratumoural heterogeneity and facilitate immune evasion of glioblastomas. Theranostics. 2023;13(5):1490–505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Li Y, Jiang T, Zhou W, et al. Pan-cancer characterization of immune-related lncRNAs identifies potential oncogenic biomarkers. Nat Commun. 2020;11(1):1000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Barczak W, Carr SM, Liu G, et al. Long non-coding RNA-derived peptides are immunogenic and drive a potent anti-tumour response. Nat Commun. 2023;14(1):1078.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Zhang Y, Wang X, Hu C, Yi H. Shiny transcriptional junk: lncRNA-derived peptides in cancers and immune responses. Life Sci. 2023;316: 121434.

    Article  CAS  PubMed  Google Scholar 

  214. Luo X, Huang Y, Li H, et al. SPENCER: a comprehensive database for small peptides encoded by noncoding RNAs in cancer patients. Nucleic Acids Res. 2022;50(D1):D1373–81.

    Article  CAS  PubMed  Google Scholar 

  215. Zhang Q, Wu E, Tang Y, et al. Deeply mining a universe of peptides encoded by long noncoding RNAs. Mol Cell Proteomics. 2021;20: 100109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Dragomir MP, Manyam GC, Ott LF, et al. FuncPEP: a database of functional peptides encoded by non-coding RNAs. Noncoding RNA. 2020;6(4):41.

    CAS  PubMed  PubMed Central  Google Scholar 

  217. Liu H, Zhou X, Yuan M, et al. ncEP: a manually curated database for experimentally validated ncrna-encoded proteins or peptides. J Mol Biol. 2020;432(11):3364–8.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This work was supported by National Natural Science Foundation of China (No. 81972643 and 82172962) and Sichuan Science and Technology Project (2021YJ0201).

Author information

Authors and Affiliations

Authors

Contributions

HT searched and reviewed published articles and wrote the manuscript. LT searched literature and drawings. YX, QM, MY and HY conducted and critically reviewed the article. ZX and JS made substantial contributions to the conception, design of the study and revised the manuscript. All authors contributed to the article and approved the submitted version.

Corresponding authors

Correspondence to Zhangang Xiao or Jing Shen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All the authors have approved the final manuscript for publication.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tian, H., Tang, L., Yang, Z. et al. Current understanding of functional peptides encoded by lncRNA in cancer. Cancer Cell Int 24, 252 (2024). https://doi.org/10.1186/s12935-024-03446-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-024-03446-7

Keywords