Skip to main content

Notch-associated lncRNAs profiling circuiting epigenetic modification in colorectal cancer

Abstract

Background

Colorectal cancer (CRC) is one of the most prevalent digestive cancers, ranking the 2nd cause of cancer-related fatality worldwide. The worldwide burden of CRC is predicted to rise by 60% by 2030. Environmental factors drive, first, inflammation and hence, cancer incidence increase.

Main

The Notch-signaling system is an evolutionarily conserved cascade, has role in the biological normal developmental processes as well as malignancies. Long non-coding RNAs (LncRNAs) have become major contributors in the advancement of cancer by serving as signal pathways regulators. They can control gene expression through post-translational changes, interactions with micro-RNAs or down-stream effector proteins. Recent emerging evidence has emphasized the role of lncRNAs in controlling Notch-signaling activity, regulating development of several cancers including CRC.

Conclusion

Notch-associated lncRNAs might be useful prognostic biomarkers or promising potential therapeutic targets for CRC treatment.

Therefore, here-in we will focus on the role of “Notch-associated lncRNAs in CRC” highlighting “the impact of Notch-associated lncRNAs as player for cancer induction and/or progression.”

Graphical Abstract

Introduction

Background

CRC is the third most ubiquitous malignancy as well as the second major cause of cancer-correlated death worldwide [1]. Surprisingly, CRC is now becoming more common among adolescents and young adults under the age of 45, who frequently come with advanced disease [2]. the prevalence of CRC is globally estimated to grow, environmental factors, such as increased sedentary behavior and obesity or processed food are thought to be one of the driving risk factors to this increase [3]. Even after surgical resection, chemotherapy/radiotherapy, using immunotherapy or the state-of-the-art targeted therapy, but, unfortunately, the 5-year survival rate remains low [2]. Cancer recurrence and distant metastases are the cause for these poor outcomes, especially for more advanced stage CRC [4].

CRC develops, throughout time, like other cancer types, as a result of a cascade of epigenetic alterations, mostly, affecting the genetic component, driving the normal colonic mucosa conversion into malignant tumor [5]. This interconversion is generated from alterations occurring within polyps, mostly adenomas [6]. Recent evidence shows that aberrant Notch cascade is crucial for CRC evolution.

Notch gene was discovered in 1917 by Morgan et al. in mutant Drosophila. The gene is known as 'Notch' because it causes a “breach” in the wings of Drosophila [7]. Notch cascade is conserved, among species, to control variety of biological activities as cell proliferation, differentiation as well as regulating cell fate decisions [8, 9].

Deregulation of Notch pathway is related to the development of hematological and solid malignancies, via pro-tumorigenic effect [10,11,12,13]. In the intestine, Notch pathway controls the homeostatic self-renewal processes and can cause ulcerative colitis, if the Notch pathway was dysregulated [14], which would cause the tumorigenic transformation of epithelia [15].

Notch pathway is a key player in CRC from initiation to resistance and metastasis, driving CRC progression and/or poor overall survival (OS) [16,17,18,19]. Positive association has been shown between the Notch receptor, Notch1, expression and deeper invasion of tumor-lymph node-metastasis (TNM) in CRC [20]. Patients with Notch1-positive malignancies had a worse OS rate than those with Notch1-negative ones [20]. Moreover, Notch-signaling is an ultimate regulator of epithelial-mesenchymal transition (EMT) process [21]. Notch-induced-EMT is a fundamental factor in CRC stemness and aggressiveness [22]. Also, increased expression of Notch as well as its target genes was shown to contribute to CRC chemoresistance [23,24,25,26].

Epigenetics-influenced activation of the Notch pathway would be led by non-protein coding RNAs (ncRNAs) expression dysregulation [27, 28]. LncRNAs are non-protein producing transcripts, performing a crucial role in the epigenetic regulation(s) affecting gene expression [29]. LncRNAs can control Notch-activation through regulation of Notch receptors or Notch ligands expression, either on transcriptional or post-transcriptional levels [30]. On the other hand, some lncRNAs are Notch-signaling downstream targets [31]. Several studies have showed that dysregulated lncRNAs have implications in CRC development, progression, metastasis as well as developing chemoresistance affecting the disease clinical outcomes [32,33,34].

Therefore, the interest in this review is to focus on the “Impact of the Notch-associated lncRNAs in CRC”. The review first aims to briefly discuss lncRNAs', Notch-signaling pathway and Notch-associated-lncRNAs mechanism(s) profiling in cancer. LncRNAs interacting with the Notch-cascade contributing to the development of various tumors are presented in the review. Second, we will highlight the role of Notch-associated lncRNAs as a player in cancer induction and progression, after defining specifically CRC types. Moreover, describing “Notch-associated lncRNAs impact on CRC clinical outcomes” and the “Notch-associated lncRNAs relationship to multidrug resistance (MDR), metastasis or recurrence.”

Non-protein coding RNA

Non-protein coding (non-coding) regions of the genome, generates numerous families of ncRNAs [35, 36], that controls gene expression and function. ncRNAs are classified based on their length, location and function into micro-RNAs (miRNAs), lncRNAs, small nucleolar RNAs (snoRNAs), small nuclear RNAs (snRNAs), small interfering RNAs (siRNAs) and PIWI-interacting RNAs (piRNAs) [37,38,39].

Long non-coding RNA

LncRNA are molecules with a length of more than 200 nucleotides [40]. LncRBase; The lncRNA sequence database; LncRBase is freely available at http://www.lncRbase.org

LncRNAs were originally described in mice through large-scale sequencing of entire cDNA libraries [41].

LncRNAs are named after their biogenesis locations in relation to the coding genes [42], which is illustrated in Fig. 1. LncRNAs can be intergenic (lincRNAs) which are derived from gaps between genes, usually placed between protein-coding genes, intronic-lncRNAs which originate from a protein-coding genes' intronic regions, sense-lncRNAs which are produced from same strand and direction of neighboring protein-coding genes. On the other hand, the antisense-lncRNAs (aslncRNA) called natural antisense transcripts (NATs) are generated from transcription of complementary strands of protein-coding genes. Likewise, the bidirectional-lncRNAs which are derived from sequences that are close to protein-coding genes' transcription start sites, but from reverse strand also. Enhancer RNAs (eRNAs) which are generated from protein-coding genes' upstream enhancer and promoter regions [38, 43, 44].

LncRNA structure

The biogenesis of lncRNAs is mediated by RNA polymerase II, similar to that of messenger RNA (mRNA) [45]. As a result, many lncRNAs have caps on the 5′ end and poly(A) tails on the 3′ end [46]. The majority of lncRNAs are thought to have more than two exons, as well as secondary and tertiary structures [47]. For each transcriptional start of a given lncRNA, nearly two distinct 3′ ends can be detected. Alternate cleavage and polyadenylation are the two processes that contribute to alternative 3′ ends, resulting in generation of different isoforms of lncRNAs from the same site, which can be increased even more by alternative splicing events [48, 49]. On the other hand, there is an exception in some lncRNAs which can be un-polyadenylated [38].

LncRNA-encoding genes generally have their own promoters, transcription factors (TFs) and distinctive DNA motifs, suggesting that transcription of lncRNAs may be an independent epigenetic modification [47]. Moreover, other epigenetic factors as DNA methylation can regulate lncRNAs transcription [47]. LncRNAs can be found in the nucleus, cytoplasm, as well as body vesicles such as exosomes and mitochondria [50]. More than half of the expressed lncRNAs are cytoplasmic, where they relate to polysome fractions, regulating mRNAs stability and translation [51].

LncRNAs as epigenetic regulators

LncRNAs have the capacity to regulate several biological processes in both the normal and the disease states [52, 53]. LncRNAs play a key role in regulation of gene expression (54), which is clarified in Fig. 2.

LncRNAs can act as chromatin modifiers as guide lncRNA, interacting with chromatin-modifying-enzymes, mediating epigenetic modification by recruiting the developed chromatin remodeler complex to a specific gene locus [45]. On the other hand, scaffold lncRNA can assist in ribonucleoprotein (RNP) complexes assembly by interacting and placing proteins close to each other [29], Fig. 2A. And, depending on the proteins and RNAs present, transcriptional activation or repression is the result, once the complexes have been wholly developed [55].

Fig. 1
figure 1

LncRNAs' classification based on their biogenesis site in relation to the coding genes. Depending on the biogenesis location, lncRNAs are classified into, intergenic which is transcribed from gaps between genes. Intronic which is transcribed from intronic regions of protein coding genes, sense which is transcribed in same direction and on same strand of neighboring coding genes; Both exonic and intronic sense lncRNAs are possible. Antisense which can be multiple exonic and intronic also but is transcribed from the reverse strand of neighboring coding gene. Bidirectional which is transcribed from region near to promoter of neighboring coding gene but from opposite strand. Enhancer which is transcribed from coding gene's enhancer region

Fig. 2
figure 2

LncRNAs as epigenetic regulators. In A lncRNA can control gene expression via modfying chromatin architecture; guide lncRNA interacts with chromatin modfying enzyme firstly, then guiding it to specfic gene locus. While scaffold serves as platfrom for developing of RNP complexes through interaction with ribonucleoproteins. In B lncRNA can control gene transcription, Decoy lncRNA inhibits transcription by trapping TFs; while enhancer or signal lncRNAs activate transcription by acting as TF or TF activator respectively. In C lncRNAs can post transcriptionally control gene expression by acting as ceRNA which sponges other regulatory ncRNAs, miRNAs, preventing their interaction with target mRNA or via lncRNA direct interaction with mRNA

LncRNAs can act as transcriptional regulators, including decoy lncRNA which repress transcription of its neighboring coding gene by trapping regulatory factors including TFs [56]. Enhancer lncRNA which can function as a transcription factor-like molecule or enhancer, to boost gene expression [57]. Moreover, signal lncRNAs act as a molecular signal to control transcription in response to diverse stimuli [55]. As a result, its presence and synthesis can be used as a measure of transcriptional activity [55], Fig. 2B.

LncRNAs can act as post-transcriptional regulators, including competitive endogenous RNA (ceRNA) acting as sponge for miRNAs and hence, silencing its target mRNA [58] or lncRNAs-mRNA direct interaction via recognizing complimentary sequences, with an overall regulation of capping, splicing and mRNA stability [54], Fig. 2C.

Notch-signaling mechanism (Fig. 3)

Fig. 3
figure 3

Notch-signaling and Notch-activation pathways in cancer, in relation to Notch associated-lncRNAs target genes. Notch receptors; Notch1, Notch2, Notch3, Notch4, Notch ligands; DLL; Delta-like ligand DLL1, DLL3, DLL4, and JAG, Jagged; JAG1, JAG2, ADAM; A Disintigrin and metalloproteases,, Co-R; co-repressors, co-A; co-activators, CSL transcription factor; (CBF1 Suppressor of Hairless, Lag-1), Hes1 hairy and enhancer of split-1, MAM mastermind proteins, NICD Notch intracellular domain, NECD Notch extracellular domain, Ƴ secretase gamma secretase. Notch signaling mechanism can be reinforced in cancer cells by lncRNAs, which can directly engage the Notch signal genes, boosting their expression, or disrupt miRNAs implicated in Notch signal suppression at specific stage

The binding of Notch ligand on one cell's membrane to a Notch receptor (Notch1, Notch2, Notch3) on the contacting cell's membrane initiates Notch signaling [59]. A two-step proteolysis cleavage process of Notch receptors starts once the ligand binds to them on the cell surface [60]. The ADAM enzymes (a disintegrin and metalloproteinase) catalyze the initial cleavage, resulting in the loss of the Notch's extracellular domain (NECD), which is then released by endocytosis [61]. While, the second cleavage is triggered by gamma-secretase, leading to release of the active Notch intracellular domain (NICD) [62, 63]. NICD enters the cell nucleus and interacts with the transcription factor CSL (CBF1, Suppressor of Hairless, Lag-1) and co-A activators mastermind (co-A MAM), all forming CSL-Notch-Mastermind transcription factor complex [62, 64], which is responsible for activating the Notch-target molecules transcription, like hairy and enhancer of split-1 (Hes1) and p300 [65]. Hes and Hey families' members are the most well-known Notch targets, which contribute to control many gene expression features related to cell fate regulation such as proliferation, differentiation and apoptosis [61]. The Hes family of transcription factors, specifically Hes1 in the gut, are the best identified Notch targets [66].

Notch-associated-lncRNAs mechanism(s) profiling in cancer

Physiologically, several lncRNAs have been found to have a positive or negative association with the Notch-signaling pathway as well as micro-RNAs or mRNA as the Notch-related molecules [30]. In cancer generally, and colorectal specifically, activation of Notch-signaling can be influenced by several dysregulated lncRNAs, on the other hand, Notch-activation controls the expression of same or other lncRNAs, as depicted in Fig. 3. A list of lncRNAs interacting with the Notch-cascade contributing to various tumors development are presented in Tables 1 and 2.

Table 1 List of down and upregulated lncRNAs expressions in different cancers and their Notch-target gene(s)
Table 2 List of cancer types with lncRNAs expressions down and upregulated and their Notch-target gene(s)

LncRNA-low expression in tumor (lncRNA-LET) a newly discovered lncRNA, was detected on chromosome 15q24.1 [136]. In NSCLC, LET demonstrated a tumor-suppressive effect; its overexpression in cells decreased NICD1 level [67]. As well, Neighbor of BRCA1 gene 2 (NBR2) is lncRNA that is encoded from the gene which locates near to the tumor suppressor gene BRCA1 [137]. NBR2 acts as tumor suppressor by inhibiting Notch1 expression in NSCLC and osteosarcoma [68, 69]. Additionally, Maternally expressed gene 3 (MEG3) is an imprinted gene in humans locating on chromosome 14q32.3, encodes lncRNA MEG3 [138]. lncRNA MEG3 inhibits endometrial tumor growth by negatively regulating Notch1 and Hes1 levels [70].

Human miR-22 host gene (MIR22HG) is a tumor suppressor lncRNA stimulating the expression of miR-22 [139]. MIR22HG suppresses Notch2 signaling, inhibiting progression of gastric cancer [71]. Likewise, LincRNA-p21 is 15 kb upstream from p21 gene, that can control both mRNA translation as well as protein stability [140]. LincRNA-p21 enhanced level reduces expression of Notch proteins, Hes1 and NICD, inhibiting hepatocellular carcinoma invasion and metastasis [72]. Moreover, CCAAT Enhancer Binding Protein Alpha (CEBPA) is a transcription factor that can regulate cell cycle with oncogenic functions [141]. CEBPA-AS1 is the CEBPA antisense RNA1 [142]. CEBPA-AS1 attenuates osteosarcoma progression via inhibiting Notch pathway members, Hes1 and RBPJ [74].

Receptor activity modifying protein 2 (RAMP2) is a single-transmembrane domain protein that plays key role in endothelial homeostasis. lncRNA RAMP2-AS1 is transcribed from RAMP2 antisense [143]. RAMP2-AS1 overexpression in cells represses Notch3 expression, impeding glioblastoma progression [75]. Besides, PAX6 upstream antisense RNA (PAUPAR) is lncRNA that could control expression of its adjacent gene Pax6, a transcription factor which controls neuronal differentiation [144]. PAUPAR serves as tumor suppressor in uveal melanoma via negatively regulation of Hes1 expression [77].

Xist (X inactive specific transcript) is the key regulator of X chromosome inactivation, which results in the stable and reliable one X chromosome silencing in somatic cells of female mammals in early development stages [145]. Xist acts as oncogenic lncRNA in NSCLC & pancreatic cancer via sponging miR-137, promoting Notch1 expression [78, 79]. Additionally, Ladybird-like homeobox gene 2 (LBX2) is a transcription factor encoding gene located on chromosome 2p13.1, involved in regulation of heart development as well as tumorigenesis of CRC [146]. LBX2-AS1, LBX2 antisense1 is lncRNA transcribed from an intron of the same chromosome [147]. In NSCLC, LBX2-AS1 functions as tumor promoter that positively regulates Notch signal markers, Notch1, p21 and Hes1, expressions [80].

Plasmacytoma variant translocation 1 (PVT1) lncRNA was firstly discovered in murine leukemia virus-induced T lymphomas as a ubiquitous retroviral integration site; PVT1 acts as oncogenic lncRNA in many cancers [148]. Upregulation of PVT1 promotes NSCLC progression through Yes-associated protein 1 (YAP1) mediated Notch pathway activation, boosting Notch1, NICD and Hes1 levels [81]. Likewise, Notch 1 associated lncRNA in T cell acute lymphoblastic leukemia1 (NALT1) is identified to cis-regulate its neighboring gene, Notch1, supposing that NALT1 actions is relayed on Notch signaling [83]. NALT1 overexpression activates Notch 1 expression in both gastric cancer and pediatric T cell acute lymphoblastic leukemia [83, 84].

Small nucleolar host gene 1 (SNHG1) is host for 8 small nucleolar RNAs, which contributes to ribosomal RNA modifications [149]. Overexpressed SNHG1 positively regulates Notch 1 and Doublecortin-like kinase 1 (DCLK1) expressions via modulation of miR-15b, inducing EMT in gastric cancer [85]. Besides, Deleted in Lymphocytic Leukemia 2 (DLEU2) is an RNA gene which is firstly discovered in chromosome 13q14 genomic region, a region that is usually eliminated in B-cell chronic lymphocytic leukemia [150]. DLEU2 promotes gastric cancer progression via serving as ceRNA for miR-23b-3p enhancing Notch2 expression [87]. While, upregulated DLEU2 induces cervical cancer proliferation by inhibition Notch pathway activity, Notch1 and RBPJ, through impeding p53 expression [88].

Steroid receptor RNA activator (SRA) is lncRNA that can activate transcriptional activity of steroid receptor [151]. SRA upregulation contributes to cervical tumorigenesis through enhancing Notch signal, promoting Notch1, Hes1 and p300 levels [89]. Additionally, lncRNA DARS antisense RNA 1 (DARS-AS1) that can also regulate its neighboring gene DARS (aspartyl-tRNA synthetase) is identified as tumor enhancer in various cancers [152]. DARS-AS1 enhanced expression positively regulates JAG1 through sponging miR-628-5p, inducing cervical tumorigenesis via Notch activation [90].

HOX transcript antisense RNA (HOTAIR) is lncRNA, transcribed from antisense strand of Homeobox C (HOXC) cluster gene; HOX genes encode essential embryonic development regulators. HOTAIR is a crucial regulator of chromatin structure and organization that controls expression of HOXD cluster genes [153, 154]. HOTAIR serves as tumor promoter, increasing Notch1 and JAG1 expressions in retinoblastoma [91]. Also, HOTAIR overexpression increases Notch markers levels, Notch1, Hes1 and P300, enhancing cervical carcinogenesis [92]. Moreover, HOTAIR positively regulates Notch 3 through serving as ceRNA for miR-613 in pancreatic cancer [93].

Regulator of reprogramming (ROR) is promoter lncRNA for reprogramming of pluripotent stem cells. ROR is a key player in human embryonic stem cells self-renewal and differentiation [155]. ROR higher levels activates Notch1 expression via negative regulation of miR-32, stimulating EMT in retinoblastoma [94]. While ROR enhances Notch1 expression in endometrial cancer via regulating of miR34a [95]. As well, gastric carcinoma highly expressed transcript 1 (GHET1) is a confirmed oncogene lncRNA in multiple tumors [156]. Upregulated GHET1 increases prostate cancer proliferation through inducing HIF-1α and Notch 1 signal via negative regulation of Kruppel-like factor 2 (KLF2) [96].

FEZF1-AS1 is FEZ family Zinc Finger 1-Antisense RNA 1, a novel oncogenic lncRNA in various tumors [157]. FEZF1-AS1 is a tumor promoter in prostate cancer via Notch signal activation, overexpressed FEZF1-AS1 contributes to higher levels of Notch1, p21 and Hes1 [97]. On the other hand, FEZF1-AS1 upregulates Notch1 in NSCLC and glioblastoma via negative regulation of miR-34a [27, 98]. Besides, Differentiation antagonizing non-protein coding RNA (DANCR) is a lncRNA prevents differentiation of epidermal progenitor cells into osteoblasts [158]. DANCR overexpression positively regulates JAG1 via targeting miR-34a-5p, aggravating prostatic cancer resistance to docetaxel [99].

Linc-OIP5 (opa interacting protein5) is identified to regulate neurogenesis [159]. Linc-OIP5 overexpression enhances glioma tumorigenesis through Notch activation, upregulating JAG1, Notch-1 and Hes1 expressions [100]. Also, Linc-OIP5 knockdown in breast cancer cells was highly associated with JAG1 expression downregulation [101]. On the other hand, Zinc Finger NFX1-Type Containing 1 (ZNFX1) antisense RNA (ZFAS1) is an lncRNA transcribed from antisense strand next to ZNFX1 protein coding gene; ZFAS1 is identified as a regulator of alveolar and epithelial cell development in the mammary gland [160]. ZFAS1 serves as tumor promoter in glioma cells through upregulating of Notch pathway, enhancing Hes-1 and NICD levels [102]. Likewise, Prostate cancer-up-regulated RNA 1 (PlncRNA-1) is lncRNA transcript that is firstly recognized to be overexpressed in prostate cancer [161]. PlncRNA-1 overexpression induces glioma progression through boosting expressions of Notch1, JAG1 and Hes1, stimulating Notch signal [104].

Colorectal Neoplasia Differentially Expressed (CRNDE) lncRNA exhibits tissue-specific and time-specific expression patterns, is firstly discovered with its upregulated expression in colorectal adenomas and carcinomas [162]. overexpressed CRNDE lncRNA functions as oncogene in osteosarcoma cells via upregulation of Notch1, JAG1 and EMT related proteins [109]. Additionally, Urothelial carcinoma associated 1 (UCA1) is a lncRNA that is primarily discovered from cell lines of bladder cancer [163]. UCA1 elevated level positively regulates JAG1 and Notch1 through targeting miR-124, promoting tongue carcinoma [114].

Metastasis associated in lung adenocarcinoma transcript 1 (MALAT1) lnRNA is originally recognized in non-small-cell lung cancer (NSCLC) primary stages study, so was given that name. MALAT1 clinical relevance is related to metastasis prediction and survival in NSCLC early stages [164]. By blocking the Notch1 signalling pathway, MALAT-1 knockdown increased the chemosensitivity of ovarian cancer cells to cisplatin [115]. Besides, Distal-less homeobox 6 antisense 1 (DLX6-AS1) is a lncRNA expressed in normal brain tissue [165]. Upregulated DLX6-AS1 is epithelial ovarian tumor enhancer through modulating of Notch signal, DLX6-AS1 silencing was highly associated with decreasing levels of Notch1, p21, and Hes1 [116].

LncRNA for neurodevelopment (LncND) is linked to a neuro-developmental diseases in humans [117]. LncND overexpression upregulates Notch1 & Notch2 in neuroblastoma cells through inhibiting of miR-143-3p [117]. On the other hand, Hepatocyte nuclear factor-1 homeobox A (HNF1A) antisense RNA 1 (HNF1A-AS1) lncRNA is transcribed in the reverse direction of the HNF1A gene [166]. HNF1A-AS1 increased expression stimulates Notch pathway in oral squamous cell carcinoma; suppression of HNF1A-AS1 inhibits the expression of Notch1 and Hes1 [118].

Colorectal cancer-associated lncRNA (CCAL) has a verified oncogenic functions in CRC cells [167]. Upregulated CCAL activates Notch1 pathway, which aided in the progression of papillary thyroid cancer [119]. Additionally, BRAF-activated non-coding RNA (BANCR) is lncRNA discovered in melanoma cells at first, then its aberrant expression was verified in several cancers including CRC [168]. In melanoma, BANCR is a tumor promoter, negatively targets miR‑204, enhancing Notch 2 expression [121].

HOXA cluster antisense RNA2 (HOXA-AS2) lncRNA is transcribed from region between HOXA3 and HOXA4 regions [169]. In cervical cancer cells, HOXA-AS2 triggers Notch signal, downregulation of HOXA-AS2 contributes to decreased NICD protein level as well as significantly reducing interaction between NICD and the transcription factor RBP-JK [124].

Molecular mechanisms driving CRC in relation to Notch-Signaling

Mechanisms accompanied by oncogene(s) activation and inhibition of tumor suppressors expression [5], though driving CRC formation as in Fig. 4 are chromosomal instability (CIN), CpG island methylation (CIM), and microsatellite instability (MSI) [170].

The conventional chromosomal instability mechanism is characterized by the accumulation of mutations that are initiated after mutational inactivation in the adenomatous polyposis coli (APC), accompanied by oncogenes activations including ki-ras2 Kirsten rat sarcoma viral oncogene homolog (Kras), cyclooxygenase-2 (COX2) and v-raf murine sarcoma viral oncogene homolog B1 (BRAF), tumor suppressor genes silencing including TP53, Deleted in colon cancer/Deleted in pancreatic cancer locus4 (DCC/DPC4) and loss of heterozygosity of chromosome 18 [171, 172].

Microsatellite instability is caused by faulty or inactive DNA mismatch repair genes (MMR). Mutations in MMR genes create genetic abnormalities in other tumor suppressor genes that are associated with growth control [171]. Hereditary non-polyposis CRC syndrome and Lynch syndrome are characterized by microsatellite instability, resulting from one of MMR genes hereditary mutations such as MLH1, MSH2, and PMS2 [170, 173].

CpG island methylation pathway is identified by hypermethylation of CpG islands in gene promotors, contributing to transcriptional inhibition of varies tumor suppressor genes including MMR genes [173].

It is noteworthy to mention that inflammation-driven CRC promoted by a mutant version of the tumor protein 53 (p53(, is mediated via the nuclear factor kappa B cell (NF-κB) prolonged activation [174]. NF-κB activation is enhanced by Notch1 overexpression, which upregulates the transcriptional activity of NF-kappa-B p65 subunit [175]. Furthermore, inactivation of TP53 enhances the progression of Notch-induced invasive adenocarcinoma (in the glandular tissue) with EMT characteristics [176]. In other words, we can infer that combination of Notch1 hyperactivity with oncogenic Kras activation and TP53 inactivation promotes high rates of metastasis of intestinal adenocarcinoma [17] (Fig. 4).

Fig. 4
figure 4

Molecular mechanisms driving CRC in relation to Notch-signaling. APC adenomatous polyposis coli, BRAF v-raf murine sarcoma viral oncogene homolog B1, CIM CpG island methylation, CIN Chromosomal instability, Kras Ki-ras2 Kirsten rat sarcoma viral oncogene homolog, MSI microsatellite instability, NF-κB nuclear factor kappa B cell, TP53 tumor protein p53

Notch signaling as a regulator of CRC immune response

Substantial evidence supports the critical function of the Notch pathway in the immune system [177]. Notch signal controls the activation of, CD8+ cytotoxic T cell, which is the key player in the anti-tumor immunological function [178]. Additionally, the crosstalk between tumor cells and dendritic cells, which is necessary for the generation and proliferation of T regulatory (Treg) cells in the TME, is significantly influenced by JAG1-induced Notch activation [179]. JAG1-Notch3 signal activation has been revealed to be crucial for Treg generation and expansion driven by OX40 ligand [180]. Besides, JAG1-mediated maturation of dendritic cells encourages Treg survival and proliferation [181].

In CRC, both peripheral blood samples and tissues showed increased Notch1, Hes1 and Hes5 mRNA expressions in CD8+ T cells, while Notch2 mainly displayed enhanced level in tissue specimens [182]. Notch signal has potential immunosuppressive effect, which inhibits CD8+ T lymphocytes' cytolytic and noncytolytic activities by inducing programmed cell death protein-1 (PD-1). Silencing the Notch pathway enhances the cytotoxicity of tumor-infiltrating CD8+ T cells via increasing their production of pro-inflammatory cytokines such as interferon gamma (IFN-γ), interleukin-1β (IL-1β), IL-6, IL-8, tumor necrosis factor alpha (TNF-α), and vascular endothelial growth factor (VEGF) as well as reducing their PD-1 expression [182]. Likewise, it was showed that mutations in the Notch system were related with a rise in amount of tumor CD8+ T cells and a decline in Treg cells, with increasing expressions of immune checkpoints, chemokines and some effector molecules [183]. Notch mutation- induced immune checkpoints upregulation can stimulate better anti-tumor immunological response, suggesting that patients with these mutations may be more responsive to immune checkpoint blockades, which is a promising therapeutic approach intended to restore anticancer immune responses [183, 184].

Moreover, over activation of Notch1 signal drives metastasis in CRC in neutrophil-dependent manner via promoting chemokine CXCL5 (C-X-C Motif Chemokine Ligand 5) and transforming Growth Factor β (TGF-β) productions, triggering an inhibitory strategy to suppress T cell responses in the TME and create an immune-suppressed environment [17]. Additionally, accumulation of myeloid derived CD11b expressing cells in regions, where cells experienced EMT, requires Jag2 expression stimulating Notch signal and EMT [185]. Furthermore, it has been shown that measuring the proportion of circulating CD11b-Jag2 cells in patients may provide a sign of CRC development into a metastatic state [185].

Mechanism(s) by which upregulated-Notch-associated lncRNAs cause CRC (Table 3)

Table 3 Mechanisms by which upregulated-Notch-associated lncRNAs cause CRC

LINC00152 named cytoskeleton regulator (CYTOR) was an overexpressed lncRNA in CRC tissues, acting via sponging miR-139-5p, leading to positive regulation of Notch1 expression. knockdown of Notch1 expression was shown to effectively inhibit CRC cell growth caused by LINC00152 upregulation, proving evidence that activities of LINC00152 are relayed on Notch1 activation [127].

Likewise, LINC00707 expression was highly elevated in CRC tissues, to sponge miR-206 and regulate expression of its target molecules, Notch3 and transmembrane 4 L six family member 1 (TM4SF1) [130].

Additionally, lncRNA Down syndrome cell adhesion molecule Antisense RNA 1 (DSCAM-AS1) is a tumor promoter that is upregulated in CRC tissues. DSCAM-AS1 positively regulates Notch1 through targeting miR-137. Where, the inhibitory effects caused by silencing of DSCAM-AS1 in CRC cells, could be reversed by Notch1 overexpression or miR-137 suppression [128].

Besides, LncRNA FOXD2 adjacent opposite strand RNA 1 (FOXD2-AS1) was significantly overexpressed in CRC tissues. Inhibition of FOXD2-AS1 expression in CRC cells resulted in an enhancement of E-cadherin protein expression, decreasing the expression of N-cadherin and the Snail protein as well as significant decrement in the Notch-related proteins (NICD and Hes-1) expression, suggesting that FOXD2-AS1 promoted CRC progression through regulation of EMT and Notch pathway [125].

In addition, ENST00000455974 is an upregulated lncRNA in DNA mismatch repair-proficient colon cancer tissues. ENST00000455974 can regulate both mRNA and the Notch-signaling ligand JAG2 protein expression [131].

Moreover, Yuqin Zhang et al., verified that lncRNAs GNAS Antisense RNA 1 (GNAS-AS1) and RP11-465L10.10 expressions were significantly elevated in CRC tissues, being involved in CRC development through direct binding to the Notch downstream target Hes1132. Hes1 is involved in CRC stem cells self-renewal and tumorgenicity, promoting cell proliferation and migration [186, 187].

Furthermore, the expression of up-regulated in CRC liver metastasis (UICLM) lncRNA was elevated in CRC tissues, when being silenced, contributing to the downregulation of essential stemness-related-genes, including Notch1 [135].

In addition, the lncRNA-34a (Lnc34a) is increased in CRC and epigenetically suppresses miR-34a expression [133].

Besides, the expression of lncRNA FAM83H antisense RNA1 (FAM83H‑AS1) was enhanced in CRC tissues, and when was knocked down in CRC cell lines, resulted in suppression in both mRNA and Notch1 and Hes1 protein levels, countered through the Notch-signal activator JAG1 [126].

LINC01198 was an another upregulated lncRNA in CRC tissues that regulates Notch-pathway markers, namely, Notch1, p300 and Hes1 [129].

Moreover, Leukemia-Associated Non-Coding IGF1R Activator RNA 1 (LUNAR1), a novel Notch-regulated lncRNA, was recently reported to be significantly upregulated in CRC tissues [188, 189], induced by Notch1 activation, enhancing CRC progression through sustaining insulin-like growth factor 1 receptor (IGF-1R) expression [188].

Notch-associated lncRNAs impact on CRC clinical outcome (Table 4).

Table 4 Notch-associated lncRNAs impact on CRC clinical outcome

Upregulation of FOXD2-AS1 is a predictor for poor survival in CRC, where FOXD2-AS1 higher expression was associated with clinical lower survival rate [190]. Furthermore, 12-year follow-up study after surgery, the survival rate analysis showed patients with enhanced FOXD2-AS1 expression, significantly exhibited 6-year survival rate, not 12-year, relative to those with lower expression [191].

FAM83H‑AS1 is an independent prognostic indicator in colon cancer. Where, patients with greater level of FAM83H‑AS1 had shorter OS time compared to patients with lower level [192]. Furthermore, FAM83H‑AS1 higher levels were significantly associated with larger tumor size and advanced tumor stage [126].

Additionally, overexpression of LINC00152 was associated with poor CRC prognosis, advanced tumor stage and worse OS as well as disease-free survival [127, 193]. Moreover, Linc00152 can be used as predictor for response of Oxaliplatin-receiving-patients, after radical colectomy, where high Linc00152 expression in Oxaliplatin-receiving-patients was associated with an increased N stage, recurrence, shorter OS and recurrence-free survival in comparison to patients with lower expression [194].

Likewise, LINC00707 and DSCAM-AS1 elevated levels are associated with poor patients' prognosis, shorter OS relative to those with lower expression [128, 195, 196]. LINC00707 enhanced expression was positively correlated with larger tumor size, advanced TNM stage, lymphatic metastasis and distant metastasis [130, 196].

DSCAM-AS1 upregulation was correlated to advanced clinical stage and metastasis status [128].

Besides, patients with higher levels of ENST00000455974 or UICLM had worse progression-free survival [131, 135]. Higher levels of UICLM were significantly correlated with CRC larger tumor size, advanced tumor stage as well as liver metastasis [135].

Again, LUNAR1 upregulation is associated with aggressive CRC, advanced tumor stage, poor differentiation status (high grade and stage), deeper tumor invasion and TNM, being attributed to unfavorable disease-free survival as well as OS [188].

Notch-associated lncRNAs in relation to multi-drug resistance (MDR) in CRC (Table 5)

Table 5 Notch-associated lncRNAs in relation to multi-drug resistance in CRC

Cancer cells' adaptation reaction, to a diversity of cytotoxic drugs, is MDR, an obstacle to achieve effective chemotherapy [197].

Overexpression of linc00152 promoted colon cancer resistance to oxaliplatin-induced apoptosis. Linc00152 mediates drug resistance, through modulating erb-b2 receptor tyrosine kinase 4 (ERBB4) expression, by functioning as ceRNA negatively regulating miR-193a-3p expression with AKT (Protein kinase B) signaling activation [194]. Additionally, linc00152 overexpression inhibits 5-fluorouracil (5-FU) induced cell death in CRC, through activation of Notch1 and sponging miR-139-5p [127].

LINC00707 acts as ceRNA targeting miR-206 and inhibiting its expression, promoting its target Notch3 expression [130, 198]. MiR-206 downregulation, enhances resistance towards 5-FU by positively regulating B-cell lymphoma-2 (Bcl-2) protein level in CRC [199]. Additionally, downregulation of Notch3 in CRC cells was shown to improve the cells' chemosensitivity to topotecan as well as cisplatin with astragaloside IV coadministration [200, 201]. Therefore, LINC00707 may contribute to MDR through regulation of miR-206/Notch3 axis.

Likewise, DSCAM-AS1 targets miR-137 and negatively regulates its expression in CRC and breast cancer [128, 202]. Hence, downregulation of miR-137, promotes oxaliplatin-resistance via targeting Y-Box Binding Protein 1 (YBX1) [203].

Also, Lnc34a acts as ceRNA negatively regulating miR-34a expression [133]. miR-34a downregulation was shown to be associated with 5-FU resistance in colon cancer cells, via upregulation of its target protein levels lactate dehydrogenase [204].

Besides, ENST00000455974 positively regulates JAG2 expression. In CRC, JAG2 increased expression was shown to enhance chemoresistance to doxorubicin-induced cytotoxicity. Silencing JAG2, induced CRC cells apoptosis via suppression of p21 expression [205]. Additionally, knockdown of JAG2 expression was found to increase CRC cells chemosensitivity to 5-FU and oxaliplatin [205].

Moreover, studies identified that Notch1 could regulate MDR related genes multidrug resistance protein 1 (MRP1)/ ATP binding cassette subfamily C member 1 (ABCC1) and BcL-2 in cancer cells [206, 207]. In CRC, Notch1 overexpression is contributed to 5-FU resistance, Notch1 suppression via miR-139-5p overexpression increases 5-FU sensitization in CRC cells, depending on Notch1 downstream targets MRP-1 and BcL-2 downregulation [25]. Likewise, downregulation of Notch1 by miR-139-5p overexpression was associated with increasing drug sensitivity in MDR related to non-kinase transmembrane glycoprotein (CD44+) and CD133+ (prominin-1) CRC cells [208].

Furthermore, Hes1 elevated expression induces 5-FU resistance in CRC via enhancing EMT and ATP-binding cassette (ABC) transporters. Hes1 increased levels in CRC cells were highly associated with N-cadherin increasing expression, and E-cadherin suppressing expression which promote EMT [23]. Additionally, overexpression of Hes1 was contributed to upregulation of several ABC transporters, ABCC1, ABCC2 and p-glycoprotein 1 (P-gp1) which are crucial components in the metabolism of drugs [23]. ABC transporters are regarded as primary cause of treatment failure via reducing drug uptake and accumulation in cells. Resistance to a wide spectrum of anticancer medicines is conferred by overexpression of the ABCC1 transporter [23, 209].

In addition, LUNAR1 may be involved in conferring 5-FU resistance, being positively regulator of IGF-1R expression [188]. In human CRC cells, IGF-1R suppression could improve 5-FU-induced cell apoptosis and viability inhibition [210].

Notch-associated lncRNAs in relation to metastasis or recurrence in CRC (Table 6)

Table 6 Notch-associated lncRNAs in relation to metastasis or recurrence in CRC

UICLM increased level promotes liver metastasis in CRC through positively regulation of zinc finger E-box binding homeobox 2 (ZEB2) expression by sponging miR-215 [135]. ZEB2 is an E-cadherin transcriptional repressor regulates EMT [211, 212]. Additionally, expression of DSCAM-AS1 was significantly increased in patients with tumor metastasis compared to non-metastasis patients [128, 213]. DSCAM-AS1 induces invasion and migration of CRC cells through negative regulation of miR-216b [213].

Moreover, LINC00152 upregulation promotes CRC metastasis through positively regulation of Fascin actin-bundling protein 1 (FSCN1) expression via sponging miR-185-3p and miR-632. Fascin plays an important role in creating actin-based cellular protrusions, promoting motility and migration of CRC cells [214,215,216]. LINC00152 silencing in colon cancer cells was associated with increased E-cadherin level and decreased levels of mesenchymal markers vimentin and N-cadherin [217]. LINC00152 overexpression consistently contributed to epithelial properties loss and the development of mesenchymal traits in colon cells, promoting colon cancer cellular invasion and metastasis through interacting with β-catenin [217]. Beside, LINC00707 positively regulates Notch3 expression [130]. Notch3 increased nuclear expression has been attributed to tumor recurrence and could be used as a potential predictor in recurrent stages II and III CRC [218].

Notch activation to facilitate CRC metastasis, mediated via EMT process

EMT has contributed to a crosstalk between Notch receptors and their ligands in CRC [219] (Fig. 5A and 5B).

Fig. 5
figure 5

A Notch signaling induced EMT in CRC. MMP matrix metalloproteinases, SLUG snail family transcriptional repressor 2, Smad-3; SMAD family member 3, TGF β Transforming Growth Factor β. B Notch signaling induced EMT in CRC

Prolonged Notch1 activation in the epithelial cells cause a senescence-like state, allowing tumor cells to trans-migrate from the main tumor and recruitment to distant locations [21]. Notch1 overexpression enhances Snail expression and inhibits E-cadherin expression in the immortalized endothelium cells, with induction of EMT and malignant transformation [220]. Activation of Notch-signaling contributes to hypoxia induced tumor cells invasion and migration [221].

In CRC, Notch1-signal enhances EMT, due to its interaction with transcription factor controlling EMT and TGF-β, promoting more TME driving metastasis [16]. Likewise, Fender et al., showed that higher level of the EMT-related proteins CD44, snail family transcriptional repressor 2 (SLUG), and SMAD family member 3(Smad-3), as well as phenotypic alterations in CRC cells, emerged from constitutive activation of NICD1 in CRC cells [22].

Activity of Jagged-1 is regulated by Notch1, which then activates Notch3, leading to an increased production of SLUG and CD44 [22].

Sonoshita et al., showed that inhibition of Notch-signal causes suppression in CRC tumor invasion and intravasation activated by knockdown of Amino-terminal enhancer of split (Aes) gene in ApcΔ716mice intestinal polyposis, pointing out to Notch-signal inhibition as a potential player during CRC metastasis prevention and treatment [222].

The Notch-related protein, Hes1, enhances CRC metastasis through induction of EMT; Upregulation of Hes1 contributed to loss of cell adhesion via repressing E-cadherin expression and enhancing N-cadherin, vimentin as well as the EMT inducer Twist-1 expressions [223]. Moreover, Hes1 overexpression was associated with an increased matrix metalloproteinases members (MMP2 and MMP9) mRNA levels in CRC cells, promoting tumor invasion [223]. Furthermore, Hes1 increases invasion via positively regulating MMP14 expression, mediated through STAT3 activity upregulation [224]. Therefore, patients with an increased Hes1 in stage II CRC, would have a higher recurrence rate chances after treatment [23].

Conclusion and prospective

CRC has recognized as a dominant public health issue due to its high frequency and fatality rates [1]. Patients' prognosis remains poor despite substantial advancements in its treatments. Additionally, post-surgical relapse and metastases occur frequently [2, 4, 225]. Therefore, it is crucial to consider attentively the developments of novel biomarkers for CRC prognosis and treatment. Since the disruption of molecular pathways is a distinct characteristic of CRC, a variety of evaluations have suggested that pathways could be used as CRC treatment targets [226].

Recent studies have confirmed the major role of Notch signal in CRC progress. Notch signaling is capable of controlling both the homeostatic self-renewal and tumorigenic transformation of intestinal epithelial cells [15]. Additionally, epigenetic modifications have been shown to greatly contribute to occurrence and progression of inflammation enhanced CRC, and understanding of these alterations will aid to novel therapeutic alternatives detection [227]. The interplay of lncRNAs and Notch signal introduces innovative suggestion for CRC medication development.

In this review, we illustrated that Notch-associated lncRNAs displayed pivotal epigenetic regulatory role among cancer different aspects (growth, resistance, recurrence, and metastasis). The review summarized these regulatory control/involvement to come to a clearer understanding of Notch-related lncRNAs and their mechanisms upon cancer cells and the reverse, in CRC or other various cancer types. We enumerated a list of lncRNAs, described to influence, or are influenced by Notch-signaling activation, leading to colorectal tumorigenesis. Dysregulation of Notch-associated lncRNAs revealed to be highly associated with CRC progression/recurrence or conferring MDR as well as being involved in CRC metastasis. Thence, Notch-associated lncRNAs might be useful prognostic biomarkers or promising potential therapeutic targets for CRC treatment.

However, the impact of Notch associated lncRNAs in CRC, Few studies are available about lncRNAs GNAS-AS1 and RP11-89K10.1 and their impact on CRC clinical outcomes is still unknown, and their function(s) in CRC require further identification. The direct interaction between lnc34a and Notch-signaling is not fully elucidated. Additionally, other transcriptional regulators as histone modification, chromatin remodeling, and X chromosome inactivation to be addressed in another review.

Availability of data and materials

Not applicable.

Abbreviations

ABC:

ATP-binding cassette

ADAM:

A disintegrin and metalloproteinase

Aes:

Amino-terminal enhancer of split

APC:

Adenomatous polyposis coli

AKT:

Protein kinase B

Bcl-2:

B-cell lymphoma-2

BRAF:

V-raf murine sarcoma viral oncogene homolog B1

CD133:

Prominin-1

ceRNA:

Competitive endogenous RNA

CRC:

Colorectal cancer

CSL:

CBF1, Suppressor of Hairless, Lag-1

Co-A MAM:

Co-A activators mastermind

CIN:

Chromosomal instability

CIM:

CpG island methylation

COX2:

Cyclooxygenase-2

CXCL5:

C-X-C Motif Chemokine Ligand 5

DCLK1:

Doublecortin-like kinase 1

EMT:

Epithelial-mesenchymal transition

eRNAs:

Enhancer RNAs

ERBB4:

Erb-b2 receptor tyrosine kinase 4

FSCN1:

Fascin actin-bundling protein 1

5-FU:

5-Fluorouracil

Hes1:

Hairy and enhancer of split-1

HCG11:

18; Human leucocyte antigen complex group 11, 18

HEY1:

Hairy/enhancer-of-split related with YRPW motif protein1

HIF1α:

Hypoxia-inducible factor 1α

IGF-1R:

Insulin-like growth factor 1 receptor

IFN-γ:

Interferon gamma

IL-1β:

Interleukin-1β

Kras:

Ki-ras2 Kirsten rat sarcoma viral oncogene homolog

KLF2:

Kruppel-like factor 2

LncRNAs:

Long non-coding RNAs

LINC00261:

Long Intergenic Non-Protein Coding RNA 261

MAML:

Mastermind like transcriptional coactivator

MATH1:

A mouse homolog of the Drosophila proneural gene atonal

MDR:

Multidrug resistance

MiRNAs:

Micro-RNAs

MSI:

Microsatellite instability

MMR:

Mismatch repair genes

MMP:

Matrix metalloproteinases

MLH1 :

mutL homolog 1

MSH2 :

mutS homolog 2

NATs:

Natural antisense transcripts

NcRNAs:

Non-protein coding RNAs

NECD:

Notch extracellular domain

NICD:

Notch intracellular domain

NSCLC:

Non-small cell lung cancer

OS:

Overall survival

PD-1:

Programmed cell death protein-1

P-gp1:

P-glycoprotein 1

PMS2:

PMS1 homolog 2

RBPJ transcription factor:

Recombination signal binding protein for immunoglobulin kappa J region

RNP:

Ribonucleoprotein

SLUG:

Snail family transcriptional repressor 2

Smad3:

SMAD family member 3

SNHG3:

7, 12, Small nucleolar RNA host gene 3, 7, 12

Stat3:

Signal transducer and activator of transcription 3

TFs:

Transcription factors

TGF β:

Transforming Growth Factor β

TNF-α:

Tumor necrosis factor alpha

TNM:

Tumor-lymph node-metastasis

TM4SF1:

Transmembrane 4 L six family member 1

TP53 or p53:

Tumor protein p53

VEGF:

Vascular endothelial growth factor

YAP1:

Yes-associated protein 1

YBX1:

Y-Box Binding Protein 1

ZEB2:

Zinc finger E-box binding homeobox 2

References

  1. Bray F, Ferlay J, Soerjomataram I. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J Clin. 2018;68(6):394–424.

    Article  Google Scholar 

  2. Tang XJ, Wang W, Hann SS. Interactions among lncRNAs, miRNAs and mRNA in colorectal cancer. Biochimie. 2019;163:58–72.

    Article  CAS  PubMed  Google Scholar 

  3. Rawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gastroenterol. 2019;14(2):89–103.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Sheng H, Wei X, Mao M, He J, Luo T, Lu S, et al. Adenocarcinoma with mixed subtypes is a rare but aggressive histologic subtype in colorectal cancer. BMC Cancer. 2019;19(1):1–11.

    Article  Google Scholar 

  5. He J, Wu F, Han Z, Hu M, Lin W, Li Y, et al. Biomarkers (mRNAs and non-coding RNAs) for the diagnosis and prognosis of colorectal cancer—from the body fluid to tissue level. Front Oncol. 2021;11(April):1–14.

    Google Scholar 

  6. Simon K. Colorectal cancer development and advances in screening. Clin Interv Aging. 2016;11:967–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Morgan TH. The theory of the gene. Am Nat. 1917;51(609):513–44. https://doi.org/10.1086/279629.

    Article  Google Scholar 

  8. Luo Z, Shang X, Zhang H, Wang G, Massey PA, Barton SR, et al. Notch signaling in osteogenesis, osteoclastogenesis, and angiogenesis. Am J Pathol. 2019;189(8):1495–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lai EC. Notch signaling: control of cell communication and cell fate. Development. 2004;131(5):965–73. https://doi.org/10.1242/dev.01074.

    Article  CAS  PubMed  Google Scholar 

  10. Weng AP, Ferrando AA, Lee W, Morris JP, Silverman LB, Sanchez-Irizarry C, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science (80-). 2004;306(5694):269–71.

    Article  CAS  Google Scholar 

  11. McCarter AC, Wang Q, Chiang M. Notch in leukemia. In: Borggrefe T, Giaimo BD, editors. Molecular mechanisms of notch signaling. Cham: Springer International Publishing; 2018. p. 355–94. https://doi.org/10.1007/978-3-319-89512-3_18.

    Chapter  Google Scholar 

  12. Zou B, Zhou X, Lai S, Liu J. Notch signaling and non-small cell lung cancer (Review). Oncol Lett. 2018;15(3):3415–21. https://doi.org/10.3892/ol.2018.7738.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Brzozowa-Zasada M, Piecuch A, Dittfeld A, Mielańczyk Ł, Michalski M, Wyrobiec G, et al. Notch signalling pathway as an oncogenic factor involved in cancer development. Contemp Oncol (Poznan, Poland). 2016;20(4):267–72.

    CAS  Google Scholar 

  14. Ghorbaninejad M, Heydari R, Mohammadi P, Shahrokh S, Haghazali M, Khanabadi B, et al. Contribution of NOTCH signaling pathway along with TNF-α in the intestinal inflammation of ulcerative colitis. Gastroenterol Hepatol from Bed to Bench. 2019;12(Suppl1):S80–6.

    Google Scholar 

  15. Radtke F, Clevers H. Self-renewal and cancer of the gut: two sides of a coin. Science (80-). 2005;307(5717):1904–9.

    Article  CAS  Google Scholar 

  16. Jackstadt R, van Hooff SR, Leach JD, Cortes-Lavaud X, Lohuis JO, Ridgway RA, et al. Epithelial NOTCH signaling rewires the tumor microenvironment of colorectal cancer to drive poor-prognosis subtypes and metastasis. Cancer Cell. 2019;36(3):319-336.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ruland J. Colon cancer: epithelial notch signaling recruits neutrophils to drive metastasis. Cancer Cell. 2019;36(3):213–4.

    Article  CAS  PubMed  Google Scholar 

  18. Zhang Y, Li B, Ji ZZ, Zheng PS. Notch1 regulates the growth of human colon cancers. Cancer. 2010;116(22):5207–18.

    Article  CAS  PubMed  Google Scholar 

  19. Shaik JP, Alanazi IO, Pathan AAK, Parine NR, Almadi MA, Azzam NA, et al. Frequent activation of notch signaling pathway in colorectal cancers and its implication in patient survival outcome. Buyukhatipoglu H, editor. J Oncol 2020;2020:6768942. https://doi.org/10.1155/2020/6768942

  20. Chu D, Li Y, Wang W, Zhao Q, Li J, Lu Y, et al. High level of notch1 protein is associated with poor overall survival in colorectal cancer. Ann Surg Oncol. 2010;17(5):1337–42. https://doi.org/10.1245/s10434-009-0893-7.

    Article  PubMed  Google Scholar 

  21. Wieland E, Rodriguez-Vita J, Liebler SS, Mogler C, Moll I, Herberich SE, et al. Endothelial notch1 activity facilitates metastasis. Cancer Cell. 2017;31(3):355–67.

    Article  CAS  PubMed  Google Scholar 

  22. Fender AW, Nutter JM, Fitzgerald TL, Bertrand FE, Sigounas G. Notch-1 promotes stemness and epithelial to mesenchymal transition in colorectal cancer. J Cell Biochem. 2015;116(11):2517–27. https://doi.org/10.1002/jcb.25196.

    Article  CAS  PubMed  Google Scholar 

  23. Sun L, Ke J, He Z, Chen Z, Huang Q, Ai W, et al. HES1 promotes colorectal cancer cell resistance to 5-Fu by inducing Of EMT and ABC transporter proteins. J Cancer. 2017;8(14):2802–8.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Jin Y, Wang M, Hu H, Huang Q, Chen Y, Wang G. Overcoming stemness and chemoresistance in colorectal cancer through miR-195–5p-modulated inhibition of notch signaling. Int J Biol Macromol. 2018;117:445–53.

    Article  CAS  PubMed  Google Scholar 

  25. Liu H, Yin Y, Hu Y, Feng Y, Bian Z, Yao S, et al. miR-139–5p sensitizes colorectal cancer cells to 5-fluorouracil by targeting NOTCH-1. Pathol Res Pract. 2016;212(7):643–9.

    Article  CAS  PubMed  Google Scholar 

  26. Zhao Q, Zhuang K, Han K, Tang H, Wang Y, Si W, et al. Silencing DVL3 defeats MTX resistance and attenuates stemness via Notch Signaling Pathway in colorectal cancer. Pathol Res Pract. 2020;216(8):152964.

    Article  CAS  PubMed  Google Scholar 

  27. Huang S, Li C, Huang J, Luo P, Mo D, Wang H. LncRNA FEZF1-AS1 promotes non-small lung cancer cell migration and invasion through the up-regulation of NOTCH1 by serving as a sponge of miR-34a. BMC Pulm Med. 2020;20(1):110. https://doi.org/10.1186/s12890-020-1154-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jiang H, Li X, Wang W, Dong H. Long non-coding RNA SNHG3 promotes breast cancer cell proliferation and metastasis by binding to microRNA-154–3p and activating the notch signaling pathway. BMC Cancer. 2020;20(1):838. https://doi.org/10.1186/s12885-020-07275-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wang C, Wang L, Ding Y, Lu X, Zhang G, Yang J, et al. LncRNA structural characteristics in epigenetic regulation. Int J Mol Sci. 2017;18(12):2659.

    Article  PubMed Central  Google Scholar 

  30. Reicher A, Foßelteder J, Kwong LN, Pichler M. Crosstalk between the Notch signaling pathway and long non-coding RNAs. Cancer Lett. 2018;420:91–6. https://doi.org/10.1016/j.canlet.2018.01.070.

    Article  CAS  PubMed  Google Scholar 

  31. Trimarchi T, Bilal E, Ntziachristos P, Fabbri G, Dalla-Favera R, Tsirigos A, et al. Genome-wide mapping and characterization of notch-regulated long noncoding RNAs in acute leukemia. Cell. 2014;158(3):593–606. https://doi.org/10.1016/j.cell.2014.05.049.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Bian Z, Jin L, Zhang J, Yin Y, Quan C, Hu Y, et al. LncRNA - UCA1 enhances cell proliferation and 5-fluorouracil resistance in colorectal cancer by inhibiting MIR-204-5p. Sci Rep. 2016;6(March):1–12.

    Google Scholar 

  33. Luo J, Qu J, Wu D-K, Lu Z-L, Sun Y-S, Qu Q. Long non-coding RNAs: a rising biotarget in colorectal cancer. Oncotarget. 2017;8(13):22187–202.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Zhu Y, Li B, Liu Z, Jiang L, Wang G, Lv M, et al. Up-regulation of lncRNA SNHG1 indicates poor prognosis and promotes cell proliferation and metastasis of colorectal cancer by activation of the Wnt/β-catenin signaling pathway. Oncotarget. 2017;8(67):111715–27.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Yao RW, Wang Y, Chen LL. Cellular functions of long noncoding RNAs. Nat Cell Biol. 2019;21(5):542–51. https://doi.org/10.1038/s41556-019-0311-8.

    Article  CAS  PubMed  Google Scholar 

  36. Mahmoud MM, Sanad EF, Hamdy NM. MicroRNAs’ role in the environment-related non-communicable diseases and link to multidrug resistance, regulation, or alteration. Environ Sci Pollut Res. 2021;28(28):36984–7000. https://doi.org/10.1007/s11356-021-14550-w.

    Article  CAS  Google Scholar 

  37. Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem. 2012;81(1):145–66. https://doi.org/10.1146/annurev-biochem-051410-092902.

    Article  CAS  PubMed  Google Scholar 

  38. Fernandes JCR, Acuña SM, Aoki JI, Floeter-Winter LM, Muxel SM. Long non-coding RNAs in the regulation of gene expression: Physiology and disease. Vol. 5, Non-coding RNA. 2019. p. 17.

  39. Ulitsky I, Bartel DP. LincRNAs: genomics, evolution, and mechanisms. Cell. 2013;154(1):26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Karlsson O, Baccarelli AA. Environmental health and long non-coding RNAs. Curr Environ Heal Rep. 2016;3(3):178–87. https://doi.org/10.1007/s40572-016-0092-1.

    Article  CAS  Google Scholar 

  41. Okazaki Y, Furuno M, Kasukawa T, Adachi J, Bono H, Kondo S, et al. Analysis of the mouse transcriptome based on functional annotation of 60,770 full-length cDNAs. Nature. 2002;420(6915):563–73.

    Article  PubMed  Google Scholar 

  42. Salviano-Silva A, Lobo-Alves SC, de Almeida RC, Malheiros D, Petzl-Erler ML. Besides pathology: long non-coding RNA in cell and tissue homeostasis. Non-coding RNA. 2018;4(1):3.

    Article  PubMed Central  Google Scholar 

  43. Khandelwal A, Bacolla A, Vasquez KM, Jain A. Long non-coding RNA: a new paradigm for lung cancer. Mol Carcinog. 2015;54(11):1235–51.

    Article  CAS  PubMed  Google Scholar 

  44. Hermans-Beijnsberger S, van Bilsen M, Schroen B. Long non-coding RNAs in the failing heart and vasculature. Non-coding RNA Res. 2018;3(3):118–30.

    Article  CAS  Google Scholar 

  45. Morlando M, Fatica A. Alteration of epigenetic regulation by long noncoding RNAs in cancer. Int J Mol Sci. 2018;19(2):570.

    Article  PubMed Central  Google Scholar 

  46. Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 2016;17(1):47–62.

    Article  CAS  PubMed  Google Scholar 

  47. Dempsey JL, Cui JY. Long non-coding RNAs: A novel paradigm for toxicology. Toxicol Sci. 2017;155(1):3–21.

    Article  CAS  PubMed  Google Scholar 

  48. Kawaji H, Severin J, Lizio M, Waterhouse A, Katayama S, Irvine KM, et al. The FANTOM web resource: from mammalian transcriptional landscape to its dynamic regulation. Genome Biol. 2009;10(4):R40. https://doi.org/10.1186/gb-2009-10-4-r40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ziegler C, Kretz M. The more the Merrier-Complexity in long non-coding RNA loci. Vol. 8, Frontiers in endocrinology. 2017. p. 1–6.

  50. Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22(2):96–118. https://doi.org/10.1038/s41580-020-00315-9.

    Article  CAS  PubMed  Google Scholar 

  51. Carlevaro-Fita J, Rahim A, Guigó R, Vardy LA, Johnson R. Cytoplasmic long noncoding RNAs are frequently bound to and degraded at ribosomes in human cells. RNA. 2016;22(6):867–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Dahariya S, Paddibhatla I, Kumar S, Raghuwanshi S, Pallepati A, Gutti RK. Long non-coding RNA: classification, biogenesis and functions in blood cells. Mol Immunol. 2019;112(May):82–92.

    Article  CAS  PubMed  Google Scholar 

  53. AbdelHamid SG, Refaat AA, Benjamin AM, Elmawardy LA, Elgendy LA, Manolly MM, et al. Deciphering epigenetic(s) role in modulating susceptibility to and severity of COVID-19 infection and/or outcome: a systematic rapid review. Environ Sci Pollut Res. 2021;28(39):54209–21. https://doi.org/10.1007/s11356-021-15588-6.

    Article  CAS  Google Scholar 

  54. Bhat SA, Ahmad SM, Mumtaz PT, Malik AA, Dar MA, Urwat U, et al. Long non-coding RNAs: mechanism of action and functional utility. Non-coding RNA Res. 2016;1(1):43–50. https://doi.org/10.1016/j.ncrna.2016.11.002.

    Article  Google Scholar 

  55. Fang Y, Fullwood MJ. Roles, functions, and mechanisms of long non-coding RNAs in cancer. Genom Proteom Bioinform. 2016;14(1):42–54. https://doi.org/10.1016/j.gpb.2015.09.006.

    Article  Google Scholar 

  56. Li X, Wu Z, Fu X, Han W. LncRNAs: Insights into their function and mechanics in underlying disorders. Mutat Res Rev Mutat Res. 2014;762:1–21.

    Article  CAS  PubMed  Google Scholar 

  57. O’Brien SJ, Bishop C, Hallion J, Fiechter C, Scheurlen K, Paas M, et al. Long non-coding RNA (lncRNA) and epithelial-mesenchymal transition (EMT) in colorectal cancer: a systematic review. Cancer Biol Ther. 2020;21(9):769–81. https://doi.org/10.1080/15384047.2020.1794239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Mahmoud MM, Sanad EF, Elshimy RAA, Hamdy NM. Competitive endogenous role of the LINC00511/miR-185–3p axis and miR-301a-3p from liquid biopsy as molecular markers for breast cancer diagnosis. Front Oncol. 2021. https://doi.org/10.3389/fonc.2021.749753.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Andersson ER, Sandberg R, Lendahl U. Notch signaling: simplicity in design, versatility in function. Development. 2011;138(17):3593–612. https://doi.org/10.1242/dev.063610.

    Article  CAS  PubMed  Google Scholar 

  60. Brou C, Logeat F, Gupta N, Bessia C, LeBail O, Doedens JR, et al. A novel proteolytic cleavage involved in notch signaling: the role of the disintegrin-metalloprotease TACE. Mol Cell. 2000;5(2):207–16.

    Article  CAS  PubMed  Google Scholar 

  61. Guruharsha KG, Kankel MW, Artavanis-Tsakonas S. The Notch signalling system: recent insights into the complexity of a conserved pathway. Nat Rev Genet. 2012;13(9):654–66. https://doi.org/10.1038/nrg3272.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Kopan R, Ilagan MXG. The canonical notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137(2):216–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Schroeter EH, Kisslinger JA, Kopan R. Notch-1 signalling requires ligand-induced proteolytic release of intracellular domain. Nature. 1998;393(6683):382–6.

    Article  CAS  PubMed  Google Scholar 

  64. Miele L. Notch signaling. Clin Cancer Res. 2006;12(4):1074–9.

    Article  CAS  PubMed  Google Scholar 

  65. Bray SJ. Notch signalling: a simple pathway becomes complex. Nat Rev Mol Cell Biol. 2006;7(9):678–89.

    Article  CAS  PubMed  Google Scholar 

  66. Sikandar SS, Pate KT, Anderson S, Dizon D, Edwards RA, Waterman ML, et al. NOTCH signaling is required for formation and self-renewal of tumor-initiating cells and for repression of secretory cell differentiation in colon cancer. Cancer Res. 2010;70(4):1469–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Li S, Zhao H, Li J, Zhang A, Wang H. Downregulation of long non-coding RNA LET predicts poor prognosis and increases Notch signaling in non-small cell lung cancer. Oncotarget. 2018;9(1):1156–68.

    Article  PubMed  Google Scholar 

  68. Cai W, Wu B, Li Z, He P, Wang B, Cai A, et al. LncRNA NBR2 inhibits epithelial-mesenchymal transition by regulating Notch1 signaling in osteosarcoma cells. J Cell Biochem. 2019;120(2):2015–27.

    Article  CAS  Google Scholar 

  69. Gao YP, Li Y, Li HJ, Zhao B. LncRNA NBR2 inhibits EMT progression by regulating Notch 1 pathway in NSCLC. Eur Rev Med Pharmacol Sci. 2019;23(18):7950–8.

    PubMed  Google Scholar 

  70. Guo Q, Qian Z, Yan D, Li L, Huang L. LncRNA-MEG3 inhibits cell proliferation of endometrial carcinoma by repressing Notch signaling. Biomed Pharmacother. 2016;82:589–94.

    Article  CAS  PubMed  Google Scholar 

  71. Li H, Wang Y. Long noncoding RNA (IncRNA) MIR22HG suppresses gastric cancer progression through attenuating NOTCH2 signaling. Med Sci Monit. 2019;25:656–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Jia M, Jiang L, Wang YD, Huang JZ, Yu M, Xue HZ. lincRNA-p21 inhibits invasion and metastasis of hepatocellular carcinoma through Notch signaling-induced epithelial–mesenchymal transition. Hepatol Res. 2016;46(11):1137–44. https://doi.org/10.1111/hepr.12659.

    Article  CAS  PubMed  Google Scholar 

  73. Zhang H-F, Li W, Han Y-D. LINC00261 suppresses cell proliferation, invasion and Notch signaling pathway in hepatocellular carcinoma. Cancer Biomark. 2018;21:575–82.

    Article  CAS  PubMed  Google Scholar 

  74. Xia P, Gu R, Zhang W, Sun YF. lncRNA CEBPA-AS1 overexpression inhibits proliferation and migration and stimulates apoptosis of OS cells via notch signaling. Mol Ther Nucleic Acids. 2020;6(19):1470–81.

    Article  Google Scholar 

  75. Liu S, Mitra R, Zhao MM, Fan W, Eischen CM, Yin F, et al. The potential roles of long noncoding RNAs (lncRNA) in glioblastoma development. Mol Cancer Ther. 2016;15(12):2977–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Xu Z, Huang B, Zhang Q, He X, Wei H, Zhang D. NOTCH1 regulates the proliferation and migration of bladder cancer cells by cooperating with long non-coding RNA HCG18 and microRNA-34c-5p. J Cell Biochem. 2019;120(4):6596–604.

    Article  CAS  PubMed  Google Scholar 

  77. Ding X, Wang X, Lin M, Xing Y, Ge S, Jia R, et al. PAUPAR lncRNA suppresses tumourigenesis by H3K4 demethylation in uveal melanoma. FEBS Lett. 2016;590:1729–38.

    Article  CAS  PubMed  Google Scholar 

  78. Liu PJ, Pan YH, Wang DW, You D. Long non-coding RNA XIST promotes cell proliferation of pancreatic cancer through miR-137 and Notch1 pathway. Eur Rev Med Pharmacol Sci. 2020;24(23):12161–70.

    PubMed  Google Scholar 

  79. Wang X, Zhang G, Cheng Z, Dai L, Jia L, Jing X, et al. Knockdown of LncRNA-XIST suppresses proliferation and TGF-β1-induced EMT in NSCLC through the Notch-1 pathway by regulation of miR-137. Genet Test Mol Biomarkers. 2018;22(6):333–42.

    Article  CAS  PubMed  Google Scholar 

  80. Tang LX, Su SF, Wan Q, He P, Xhang Y, Cheng XM. Novel long non-coding RNA LBX2-AS1 indicates poor prognosis and promotes cell proliferation and metastasis through Notch signaling in non-small cell lung cancer. Eur Rev Med Pharmacol Sci. 2019;23(17):7419–29.

    PubMed  Google Scholar 

  81. Zeng SG, Xie JH, Zeng QY, Dai SH, Wang Y, Wan XM, et al. lncRNA PVT1 promotes metastasis of non-small cell lung cancer through EZH2-mediated activation of Hippo/NOTCH1 signaling pathways. Cell J. 2021;23(1):21–31.

    PubMed  PubMed Central  Google Scholar 

  82. Deng Y, Zhang L, Luo R. LINC01783 facilitates cell proliferation, migration and invasion in non-small cell lung cancer by targeting miR-432-5p to activate the notch pathway. Cancer Cell Int. 2021;21(1):234. https://doi.org/10.1186/s12935-021-01912-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Piao HY, Guo S, Wang Y, Zhang J. Long noncoding RNA NALT1-induced gastric cancer invasion and metastasis via NOTCH signaling pathway. World J Gastroenterol. 2019;25(44):6508–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Wang Y, Wu P, Lin R, Rong L, Xue Y, Fang Y. LncRNA NALT interaction with NOTCH1 promoted cell proliferation in pediatric T cell acute lymphoblastic leukemia. Sci Rep. 2015;5(1):13749. https://doi.org/10.1038/srep13749.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Liu Z-Q, He W-F, Wu Y-J, Zhao S-L, Wang L, Ouyang Y-Y, et al. LncRNA SNHG1 promotes EMT process in gastric cancer cells through regulation of the miR-15b/DCLK1/Notch1 axis. BMC Gastroenterol. 2020;20(1):156. https://doi.org/10.1186/s12876-020-01272-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Zhao X, Zhao Z. Linc01555 promotes proliferation, migration and invasion of gastric carcinoma cells by interacting with Notch signaling pathway. J BUON. 2020;25(2):1007–12.

    CAS  PubMed  Google Scholar 

  87. Li G, Zhang Z, Chen Z, Liu B, Wu H. LncRNA DLEU2 is activated by STAT1 and induces gastric cancer development via targeting miR-23b-3p/NOTCH2 axis and Notch signaling pathway. Life Sci. 2021;277(247):119419. https://doi.org/10.1016/j.lfs.2021.119419.

    Article  CAS  PubMed  Google Scholar 

  88. He M, Wang Y, Cai J, Xie Y, Tao C, Jiang Y, et al. LncRNA DLEU2 promotes cervical cancer cell proliferation by regulating cell cycle and NOTCH pathway. Exp Cell Res. 2021;402(1):112551. https://doi.org/10.1016/j.yexcr.2021.112551.

    Article  CAS  PubMed  Google Scholar 

  89. Eoh Jin K, Paek J, Kim Wun S, Kim Jung H, Lee Yeon H, Lee Kil S, et al. Long non-coding RNA, steroid receptor RNA activator (SRA), induces tumor proliferation and invasion through the NOTCH pathway in cervical cancer cell lines. Oncol Rep. 2017;38(6):3481–8. https://doi.org/10.3892/or.2017.6023.

    Article  CAS  Google Scholar 

  90. Chen Y, Wu Q, Lin J, Wei J. DARS-AS1 accelerates the proliferation of cervical cancer cells via miR-628–5p/JAG1 axis to activate Notch pathway. Cancer Cell Int. 2020;20(1):535. https://doi.org/10.1186/s12935-020-01592-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Dong C, Liu S, Lv Y, Zhang C, Gao H, Tan L, et al. Long non-coding RNA HOTAIR regulates proliferation and invasion via activating Notch signalling pathway in retinoblastoma. J Biosci. 2016;41(4):677–87.

    Article  CAS  PubMed  Google Scholar 

  92. Lee M, Kim HJ, Kim SW, Park SA, Chun KH, Cho NH, et al. The long non-coding RNA HOTAIR increases tumour growth and invasion in cervical cancer by targeting the Notch pathway. Oncotarget. 2016;7(28):44558–71.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Cai H, Yao J, An Y, Chen X, Chen W, Wu D, et al. LncRNA HOTAIR acts a competing endogenous RNA to control the expression of notch3 via sponging miR-613 in pancreatic cancer. Oncotarget. 2017;8(20):32905–17.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Gao Y, Luo X, Zhang J. LincRNA-ROR is activated by H3K27 acetylation and induces EMT in retinoblastoma by acting as a sponge of miR-32 to activate the Notch signaling pathway. Cancer Gene Ther. 2021;28(12):42–54. https://doi.org/10.1038/s41417-020-0181-z.

    Article  CAS  PubMed  Google Scholar 

  95. Zeng SY, Liu CQ, Zhuang Y, Chen Y, Gu LL, Shi SQ. LncRNA ROR promotes proliferation of endometrial cancer cells via regulating Notch1 pathway. Eur Rev Med Pharmacol Sci. 2020;24(11):5970–8.

    PubMed  Google Scholar 

  96. Zhu Y, Tong Y, Wu J, Liu Y, Zhao M. Knockdown of LncRNA GHET1 suppresses prostate cancer cell proliferation by inhibiting HIF-1α/Notch-1 signaling pathway via KLF2. BioFactors. 2019;45(3):364–73.

    Article  CAS  PubMed  Google Scholar 

  97. Zhu LF, Song LD, Xu Q, Zhan JF. Highly expressed long non-coding RNA FEZF1-AS1 promotes cells proliferation and metastasis through Notch signaling in prostate cancer. Vol. 23, European review for medical and pharmacological sciences. 2019. p. 5122–32.

  98. Luo L, Zhang Y, He H, Chen C, Zhang B, Cai M. LncRNA FEZF1-AS1 sponges miR-34a to upregulate Notch-1 in glioblastoma. Cancer Manag Res. 2020;12:1827–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Ma Y, Fan B, Ren Z, Liu B, Wang Y. Long noncoding RNA DANCR contributes to docetaxel resistance in prostate cancer through targeting the miR-34a-5p/JAG1 pathway. Onco Targets Ther. 2019;12:5485–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Hu G, Wu L, Kuang W, Chen Y, Zhu X, Guo H, et al. Knockdown of linc-OIP5 inhibits proliferation and migration of glioma cells through down-regulation of YAP-NOTCH signaling pathway. Gene. 2017;610:24–31. https://doi.org/10.1016/j.gene.2017.02.006.

    Article  CAS  PubMed  Google Scholar 

  101. Zhu Q, Li J, Wu Q, Cheng Y, Zheng H, Zhan T, et al. Linc—OIP5 in the breast cancer cells regulates angiogenesis of human umbilical vein endothelial cells through YAP1 / Notch / NRP1 signaling circuit at a tumor microenvironment. Biol Res. 2020;53(1):1–12.

    Article  CAS  Google Scholar 

  102. Gao K, Ji Z, She K, Yang Q, Shao L. Long non-coding RNA ZFAS1 is an unfavourable prognostic factor and promotes glioma cell progression by activation of the Notch signaling pathway. Biomed Pharmacother. 2017;87:555–60. https://doi.org/10.1016/j.biopha.2017.01.014.

    Article  CAS  PubMed  Google Scholar 

  103. Wu J, Wang N, Yang Y, Jiang G, Zhan H, Li F. LINC01152 upregulates MAML2 expression to modulate the progression of glioblastoma multiforme via Notch signaling pathway. Cell Death Dis. 2021;12(1):115. https://doi.org/10.1038/s41419-020-03163-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Wang X, Yan Y, Zhang C, Wei W, Ai X, Pang Y, et al. Upregulation of lncRNA PlncRNA-1 indicates the poor prognosis and promotes glioma progression by activation of Notch signal pathway. Biomed Pharmacother. 2018;103(5):216–21. https://doi.org/10.1016/j.biopha.2018.03.150.

    Article  CAS  PubMed  Google Scholar 

  105. Zhao X, Shen F, Yang B. LncRNA LINC01410 induced by MYC accelerates glioma progression via sponging miR-506–3p and modulating NOTCH2 expression to motivate Notch signaling pathway. Cell Mol Neurobiol. 2021. https://doi.org/10.1007/s10571-021-01042-1.

    Article  PubMed  Google Scholar 

  106. Sun X, Huang T, Liu Z, Sun M, Luo S. LncRNA SNHG7 contributes to tumorigenesis and progression in breast cancer by interacting with miR-34a through EMT initiation and the Notch-1 pathway. Eur J Pharmacol. 2019;856(January):172407. https://doi.org/10.1016/j.ejphar.2019.172407.

    Article  CAS  PubMed  Google Scholar 

  107. Zhou S, Yu L, Xiong M, Dai G. Biochemical and biophysical research communications LncRNA SNHG12 promotes tumorigenesis and metastasis in osteosarcoma by upregulating Notch2 by sponging miR-195–5p. Biochem Biophys Res Commun. 2018;495(2):1822–32. https://doi.org/10.1016/j.bbrc.2017.12.047.

    Article  CAS  PubMed  Google Scholar 

  108. Liu Z-B, Tang C, Jin X, Liu S-H, Pi W. Increased expression of lncRNA SNHG12 predicts a poor prognosis of nasopharyngeal carcinoma and regulates cell proliferation and metastasis by modulating Notch signal pathway. Cancer Biomark. 2018;23:603–13.

    Article  CAS  PubMed  Google Scholar 

  109. Li Z, Tang Y, Xing W, Dong W, Wang Z. LncRNA, CRNDE promotes osteosarcoma cell proliferation, invasion and migration by regulating Notch1 signaling and epithelial-mesenchymal transition. Exp Mol Pathol. 2018;104(1):19–25.

    Article  CAS  PubMed  Google Scholar 

  110. Huang R, Nie W, Yao K, Chou J. Depletion of the lncRNA RP11-567G11.1 inhibits pancreatic cancer progression. Biomed Pharmacother. 2019;112(138):108685. https://doi.org/10.1016/j.biopha.2019.108685.

    Article  CAS  PubMed  Google Scholar 

  111. Liu S, Liu WH, Diao ZL, Zhang AH, Guo W, Han X, et al. LncRNA RP11-567G11.1 accelerates the proliferation and invasion of renal cell carcinoma through activating the Notch pathway. Eur Rev Med Pharmacol Sci. 2020;24(9):4738–44.

    CAS  PubMed  Google Scholar 

  112. Xu J, Xu W, Yang X, Liu Z, Sun Q. LncRNA HCG11/miR-579–3p/MDM2 axis modulates malignant biological properties in pancreatic carcinoma via Notch/Hes1 signaling pathway. Aging (Albany NY). 2021;13(12):16471–84.

    Article  CAS  Google Scholar 

  113. Chen H, Liu JZ, Hu GJ, Shi LL, Lan T. Promotion of proliferation and metastasis of hepatocellular carcinoma by LncRNA00673 based on the targeted-regulation of notch signaling pathway. Eur Rev Med Pharmacol Sci. 2017;21(15):3412–20.

    CAS  PubMed  Google Scholar 

  114. Zhang TH, Liang LZ, Liu XL, Wu JN, Su K, Chen JY, et al. LncRNA UCA1/miR-124 axis modulates TGFβ1-induced epithelial-mesenchymal transition and invasion of tongue cancer cells through JAG1/Notch signaling. J Cell Biochem. 2019;120(6):10495–504. https://doi.org/10.1002/jcb.28334.

    Article  CAS  PubMed  Google Scholar 

  115. Bai L, Wang A, Zhang Y, Xu X, Zhang X. Knockdown of MALAT1 enhances chemosensitivity of ovarian cancer cells to cisplatin through inhibiting the Notch1 signaling pathway. Exp Cell Res. 2018;366(2):161–71. https://doi.org/10.1016/j.yexcr.2018.03.014.

    Article  CAS  PubMed  Google Scholar 

  116. Zhao J, Liu HR. Down-regulation of long noncoding RNA DLX6-AS1 defines good prognosis and inhibits proliferation and metastasis in human epithelial ovarian cancer cells via Notch signaling pathway. Eur Rev Med Pharmacol Sci. 2019;23(8):3243–52.

    CAS  PubMed  Google Scholar 

  117. Rani N, Nowakowski TJ, Zhou H, Godshalk SE, Lisi V, Kriegstein AR, et al. A primate lncRNA mediates Notch signaling during neuronal development by sequestering miRNA. Neuron. 2016;90(6):1174–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Liu Z, Li H, Fan S, Lin H, Lian W. STAT3-induced upregulation of long noncoding RNA HNF1A-AS1 promotes the progression of oral squamous cell carcinoma via activating Notch signaling pathway. Cancer Biol Ther. 2019;20(4):444–53. https://doi.org/10.1080/15384047.2018.1529119.

    Article  CAS  PubMed  Google Scholar 

  119. Ye Y, Song Y, Zhuang J, He S, Ni J, Xia W. Long noncoding RNA CCAL promotes papillary thyroid cancer progression by activation of NOTCH1 pathway. Oncol Res. 2018;26(9):1383–90.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Zhang M, Han Y, Zheng Y, Zhang Y, Zhao X, Gao Z, et al. ZEB1-activated LINC01123 accelerates the malignancy in lung adenocarcinoma through NOTCH signaling pathway. Cell Death Dis. 2020;11(11):981. https://doi.org/10.1038/s41419-020-03166-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Cai B, Zheng Y, Ma S, Xing Q, Wang X, Yang B, et al. BANCR contributes to the growth and invasion of melanoma by functioning as a competing endogenous RNA to upregulate Notch2 expression by sponging miR-204. Int J Oncol. 2017;51(6):1941–51.

    Article  CAS  PubMed  Google Scholar 

  122. Wang Y, Li M, Dong C, Ma Y, Xiao L, Zuo S, et al. Linc00152 knockdown inactivates the Akt / mTOR and Notch1 pathways to exert its anti-hemangioma effect. Life Sci. 2019;223(February):22–8. https://doi.org/10.1016/j.lfs.2019.03.006.

    Article  CAS  PubMed  Google Scholar 

  123. Ma Q, Dai X, Lu W, Qu X, Liu N, Zhu C. Biochemical and Biophysical Research Communications Silencing long non-coding RNA MEG8 inhibits the proliferation and induces the ferroptosis of hemangioma endothelial cells by regulating miR-497–5p / NOTCH2 axis. Biochem Biophys Res Commun. 2021;556:72–8. https://doi.org/10.1016/j.bbrc.2021.03.132.

    Article  CAS  PubMed  Google Scholar 

  124. Wu Q, Lu S, Zhang L, Zhao L. LncRNA HOXA-AS2 activates the notch pathway to promote cervical cancer cell proliferation and migration. Reprod Sci. 2021;28(10):3000–9. https://doi.org/10.1007/s43032-021-00626-y.

    Article  CAS  PubMed  Google Scholar 

  125. Yang X, Duan B, Zhou X. Long non-coding RNA FOXD2-AS1 functions as a tumor promoter in colorectal cancer by regulating EMT and Notch signaling pathway. Eur Rev Med Pharmacol Sci. 2017;21(16):3586–91.

    CAS  PubMed  Google Scholar 

  126. Lu S, Dong W, Zhao P, Liu Z. LncRNA FAM83H-AS1 is associated with the prognosis of colorectal carcinoma and promotes cell proliferation by targeting the notch signaling pathway. Oncol Lett. 2018;15(2):1861–8.

    PubMed  Google Scholar 

  127. Bian Z, Zhang J, Li M, Feng Y, Yao S, Song M, et al. Long non-coding RNA LINC00152 promotes cell proliferation, metastasis, and confers 5-FU resistance in colorectal cancer by inhibiting miR-139-5p. Oncogenesis. 2017;6(11):395. https://doi.org/10.1038/s41389-017-0008-4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Xu J, Wu G, Zhao Y, Han Y, Zhang S, Li C, et al. Long noncoding RNA DSCAM-AS1 facilitates colorectal cancer cell proliferation and migration via miR-137/notch1 axis. J Cancer. 2020;11(22):6623–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. D’Abbronzo G, Franco R. LINC01198 promotes colorectal cancer cell proliferation and inhibits apoptosis via notch signaling pathway. Eur Rev Med Pharmacol Sci. 2020;24(19):9776–7.

    PubMed  Google Scholar 

  130. Zhu H, He G, Wang Y, Hu Y, Zhang Z, Qian X, et al. Long intergenic noncoding RNA 00707 promotes colorectal cancer cell proliferation and metastasis by sponging miR-206. Onco Targets Ther. 2019;12:4331–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Lao Y, Li Q, Li N, Liu H, Liu K, Jiang G, et al. Long noncoding RNA ENST00000455974 plays an oncogenic role through up-regulating JAG2 in human DNA mismatch repair-proficient colon cancer. Biochem Biophys Res Commun. 2019;508(2):339–47. https://doi.org/10.1016/j.bbrc.2018.11.088.

    Article  CAS  PubMed  Google Scholar 

  132. Zhang Y, Zheng L, Lao X, Wen M, Qian Z, Liu X, et al. Hes1 is associated with long non-coding RNAs in colorectal cancer. Ann Transl Med. 2019;7(18):459.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Wang L, Bu P, Ai Y, Srinivasan T, Chen HJ, Xiang K, et al. A long non-coding RNA targets microRNA miR-34a to regulate colon cancer stem cell asymmetric division. Green MR, editor. Elife 2016;5:e14620. https://doi.org/10.7554/eLife.14620

  134. Bu P, Chen KY, Chen JH, Wang L, Walters J, Shin YJ, et al. A microRNA miR-34a-regulated bimodal switch targets notch in colon cancer stem cells. Cell Stem Cell. 2013;12(5):602–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Chen DL, Lu YX, Zhang JX, Wei XL, Wang F, Zeng ZL, et al. Long non-coding RNA UICLM promotes colorectal cancer liver metastasis by acting as a ceRNA for microRNA-215 to regulate ZEB2 expression. Theranostics. 2017;7(19):4836–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Ye Z, Duan J, Wang L, Ji Y, Qiao B. LncRNA-LET inhibits cell growth of clear cell renal cell carcinoma by regulating miR-373-3p. Cancer Cell Int. 2019;19(1):311. https://doi.org/10.1186/s12935-019-1008-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Auriol E, Billard L-M, Magdinier F, Dante R. Specific binding of the methyl binding domain protein 2 at the BRCA1-NBR2 locus. Nucleic Acids Res. 2005;33(13):4243–54. https://doi.org/10.1093/nar/gki729.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Jia H-Y, Zhang K, Lu W-J, Xu G-W, Zhang J-F, Tang Z-L. LncRNA MEG3 influences the proliferation and apoptosis of psoriasis epidermal cells by targeting miR-21/caspase-8. BMC Mol cell Biol. 2019;20(1):46.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Zhang D-Y, Zou X-J, Cao C-H, Zhang T, Lei L, Qi X-L, et al. Identification and functional characterization of long non-coding RNA MIR22HG as a tumor suppressor for hepatocellular carcinoma. Theranostics. 2018;8(14):3751–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Hall JR, Messenger ZJ, Tam HW, Phillips SL, Recio L, Smart RC. Long noncoding RNA lincRNA-p21 is the major mediator of UVB-induced and p53-dependent apoptosis in keratinocytes. Cell Death Dis. 2015;6(3):e1700. https://doi.org/10.1038/cddis.2015.67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Szmajda D, Krygier A, Jamroziak K, Żebrowska-Nawrocka M, Balcerczak E. Expression level of CEBPA gene in acute lymphoblastic leukemia individuals. Sci Rep. 2019;9(1):15640. https://doi.org/10.1038/s41598-019-52104-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Ke D, Li H, Zhang Y, An Y, Fu H, Fang X, et al. The combination of circulating long noncoding RNAs AK001058, INHBA-AS1, MIR4435-2HG, and CEBPA-AS1 fragments in plasma serve as diagnostic markers for gastric cancer. Oncotarget 2017;8(13):21516–25. https://www.oncotarget.com/article/15628/text/

  143. Lai CH, Chen AT, Burns AB, Sriram K, Luo Y, Tang X, et al. RAMP2-AS1 regulates endothelial homeostasis and aging. Front Cell Dev. 2021. https://doi.org/10.3389/fcell.2021.635307.

    Article  Google Scholar 

  144. Vance KW, Sansom SN, Lee S, Chalei V, Kong L, Cooper SE, et al. The long non-coding RNA paupar regulates the expression of both local and distal genes. EMBO J. 2014;33(4):296–311. https://doi.org/10.1002/embj.201386225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Cerase A, Pintacuda G, Tattermusch A, Avner P. Xist localization and function: new insights from multiple levels. Genome Biol. 2015;16(1):166. https://doi.org/10.1186/s13059-015-0733-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Huang X, Yang Y, Yang C, Li H, Cheng H, Zheng Y. Overexpression of LBX2 associated with tumor progression and poor prognosis in colorectal cancer. Oncol Lett. 2020;19(6):3751–60. https://doi.org/10.3892/ol.2020.11489.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Ma Y-N, Hong Y-G, Yu G-Y, Jiang S, Zhao B, Guo A, et al. LncRNA LBX2-AS1 promotes colorectal cancer progression and 5-fluorouracil resistance. Cancer Cell Int. 2021;21(1):501. https://doi.org/10.1186/s12935-021-02209-y.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Lu D, Luo P, Wang Q, Ye Y, Wang B. lncRNA PVT1 in cancer: a review and meta-analysis. Clin Chim Acta. 2017;474:1–7.

    Article  CAS  PubMed  Google Scholar 

  149. Thin KZ, Tu JC, Raveendran S. Long non-coding SNHG1 in cancer. Clin Chim Acta. 2019;494:38–47.

    Article  CAS  PubMed  Google Scholar 

  150. Ghafouri-Fard S, Dashti S, Farsi M, Taheri M. Deleted in lymphocytic leukemia 2 (DLEU2): an lncRNA with dissimilar roles in different cancers. Biomed Pharmacother. 2021;133:111093.

    Article  CAS  PubMed  Google Scholar 

  151. Yan R, Wang K, Peng R, Wang S, Cao J, Wang P, et al. Genetic variants in lncRNA SRA and risk of breast cancer. Oncotarget. 2016;7(16):22486–96.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Jiao M, Guo H, Chen Y, Li L, Zhang L. DARS-AS1 promotes clear cell renal cell carcinoma by sequestering miR-194–5p to up-regulate DARS. Biomed Pharmacother. 2020;128:110323.

    Article  CAS  PubMed  Google Scholar 

  153. Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129(7):1311–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Woo CJ, Kingston RE. HOTAIR lifts noncoding RNAs to new levels. Cell. 2007;129(7):1257–9.

    Article  CAS  PubMed  Google Scholar 

  155. Pan Y, Li C, Chen J, Zhang K, Chu X, Wang R, et al. The emerging roles of long noncoding RNA ROR (lincRNA-ROR) and its possible mechanisms in human cancers. Cell Physiol Biochem. 2016;40(1–2):219–29. https://doi.org/10.1159/000452539.

    Article  CAS  PubMed  Google Scholar 

  156. Jiang YF, Zhang HY, Ke J, Shen H, Ou HB, Liu Y. Overexpression of LncRNA GHET1 predicts an unfavourable survival and clinical parameters of patients in various cancers. J Cell Mol Med. 2019;23(8):4891–9. https://doi.org/10.1111/jcmm.14486.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Shi C, Sun L, Song Y. FEZF1-AS1: a novel vital oncogenic lncRNA in multiple human malignancies. Biosci Rep. 2019;39(6):BSR20191202. https://doi.org/10.1042/BSR20191202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Jin SJ, Jin MZ, Xia BR, Jin WL. Long non-coding RNA DANCR as an emerging therapeutic target in human cancers. Front Oncol. 2019;9:1225. https://doi.org/10.3389/fonc.2019.01225.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Ulitsky I, Shkumatava A, Jan CH, Sive H, Bartel DP. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution. Cell. 2011;147(7):1537–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Askarian-Amiri ME, Crawford J, French JD, Smart CE, Smith MA, Clark MB, et al. SNORD-host RNA Zfas1 is a regulator of mammary development and a potential marker for breast cancer. RNA. 2011;17(5):878–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Cui Z, Ren S, Lu J, Wang F, Xu W, Sun Y, et al. The prostate cancer-up-regulated long noncoding RNA PlncRNA-1 modulates apoptosis and proliferation through reciprocal regulation of androgen receptor. Urol Oncol Semin Orig Investig. 2013;31(7):1117–23.

    CAS  Google Scholar 

  162. Ellis BC, Molloy PL, Graham LD. CRNDE: A long non-coding RNA involved in cancer neurobiology, and development. Front Genet. 2012;3:1–15. https://doi.org/10.3389/fgene.2012.00270.

    Article  CAS  Google Scholar 

  163. Wang X-S, Zhang Z, Wang H-C, Cai J-L, Xu Q-W, Li M-Q, et al. Rapid Identification of UCA1 as a very sensitive and specific unique marker for human bladder carcinoma. Clin Cancer Res. 2006;12(16):4851–8.

    Article  CAS  PubMed  Google Scholar 

  164. Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, et al. MALAT-1, a novel noncoding RNA, and thymosin β4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003;22(39):8031–41. https://doi.org/10.1038/sj.onc.1206928.

    Article  CAS  PubMed  Google Scholar 

  165. Alizadeh A, Jebelli A, Baradaran B, Amini M, Oroojalian F, Hashemzaei M, et al. Crosstalk between long non-coding RNA DLX6-AS1, microRNAs and signaling pathways: a pivotal molecular mechanism in human cancers. Gene. 2021;769:145224.

    Article  CAS  PubMed  Google Scholar 

  166. Wu Y, Liu H, Shi X, Yao Y, Yang W, Song Y. The long non-coding RNA HNF1A-AS1 regulates proliferation and metastasis in lung adenocarcinoma. Oncotarget. 2015;6(11):9160–72.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Deng X, Ruan H, Zhang X, Xu X, Zhu Y, Peng H, et al. Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells. Int J Cancer. 2020;146(6):1700–16. https://doi.org/10.1002/ijc.32608.

    Article  CAS  PubMed  Google Scholar 

  168. Zhou T, Gao Y. Increased expression of LncRNA BANCR and its prognostic significance in human hepatocellular carcinoma. World J Surg Oncol. 2016;14(1):8. https://doi.org/10.1186/s12957-015-0757-5.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Wang J, Su Z, Lu S, Fu W, Liu Z, Jiang X, et al. LncRNA HOXA-AS2 and its molecular mechanisms in human cancer. Clin Chim Acta. 2018;485:229–33.

    Article  CAS  PubMed  Google Scholar 

  170. Tariq K, Ghias K. Colorectal cancer carcinogenesis: a review of mechanisms. Cancer Biol Med. 2016;13(1):120–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Lizarbe MA, Calle-Espinosa J, Fernández-Lizarbe E, Fernández-Lizarbe S, Robles MÁ, Olmo N, et al. Colorectal cancer: from the genetic model to posttranscriptional regulation by noncoding RNAs. Linnebacher M, editor. Biomed Res Int 2017;2017:7354260. https://doi.org/10.1155/2017/7354260

  172. Malki A, Elruz RA, Gupta I, Allouch A, Vranic S, Al Moustafa AE. Molecular mechanisms of colon cancer progression and metastasis: recent insights and advancements. Int J Mol Sci. 2021;22:1–24.

    Google Scholar 

  173. Arends MJ. Pathways of colorectal carcinogenesis. Appl Immunohistochem Mol Morphol 2013;21(2):97–102. https://journals.lww.com/appliedimmunohist/Fulltext/2013/03000/Pathways_of_Colorectal_Carcinogenesis.1.aspx

  174. Cooks T, Pateras IS, Tarcic O, Solomon H, Schetter AJ, Wilder S, et al. Mutant p53 prolongs NF-κB activation and promotes chronic inflammation and inflammation-associated colorectal cancer. Cancer Cell. 2013;23(5):634–46. https://doi.org/10.1016/j.ccr.2013.03.022.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Chu D, Zhou Y, Zhang Z, Li Y, Li J, Zheng J, et al. Notch1 expression, which is related to p65 status, is an independent predictor of prognosis in colorectal cancer. Clin Cancer Res. 2011;17(17):5686–94.

    Article  CAS  PubMed  Google Scholar 

  176. Nakayama M, Oshima M. Mutant p53 in colon cancer. J Mol Cell Biol. 2019;11(4):267–76. https://doi.org/10.1093/jmcb/mjy075.

    Article  CAS  PubMed  Google Scholar 

  177. Mathern DR, Laitman LE, Hovhannisyan Z, Dunkin D, Farsio S, Malik TJ, et al. Mouse and human Notch-1 regulate mucosal immune responses. Mucosal Immunol. 2014;7(4):995–1005. https://doi.org/10.1038/mi.2013.118.

    Article  CAS  PubMed  Google Scholar 

  178. Meurette O, Mehlen P. Notch signaling in the tumor microenvironment. Cancer Cell. 2018;34(4):536–48.

    Article  CAS  PubMed  Google Scholar 

  179. Li D, Masiero M, Banham AH, Harris AL. The notch ligand jagged1 as a target for anti-tumor therapy. Front Oncol. 2014. https://doi.org/10.3389/fonc.2014.00254.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Gopisetty A, Bhattacharya P, Haddad C, Bruno JC, Vasu C, Miele L, et al. OX40L/Jagged1 cosignaling by GM-CSF–induced bone marrow-derived dendritic cells is required for the expansion of functional regulatory T cells. J Immunol. 2013;190(11):5516–25.

    Article  CAS  PubMed  Google Scholar 

  181. Bugeon L, Gardner LM, Rose A, Gentle M, Dallman MJ. Cutting edge: Notch signaling induces a distinct cytokine profile in dendritic cells that supports T cell-mediated regulation and IL-2-dependent IL-17 production. J Immunol. 2008;181(12):8189–93.

    Article  CAS  PubMed  Google Scholar 

  182. Yu W, Wang Y, Guo P. Notch signaling pathway dampens tumor-infiltrating CD8+ T cells activity in patients with colorectal carcinoma. Biomed Pharmacother. 2018;97:535–42.

    Article  CAS  PubMed  Google Scholar 

  183. Wang F, Long J, Li L, Zhao Z, Wei F, Yao Y, et al. Mutations in the notch signalling pathway are associated with enhanced anti-tumour immunity in colorectal cancer. J Cell Mol Med. 2020;24(20):12176–87. https://doi.org/10.1111/jcmm.15867.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Kamal AM, Wasfey EF, Elghamry WR, Sabry OM, Elghobary HA, Radwan SM. Genetic signature of CTLA-4, BTLA, TIM-3 and LAG-3 molecular expression in colorectal cancer patients: Implications in diagnosis and survival outcomes. Clin Biochem. 2021;96:13–8.

    Article  CAS  PubMed  Google Scholar 

  185. Caiado F, Carvalho T, Rosa I, Remédio L, Costa A, Matos J, et al. Bone marrow-derived CD11b+ Jagged2+ cells promote epithelial-to-mesenchymal transition and metastasization in colorectal cancer. Cancer Res. 2013;73(14):4233–46.

    Article  CAS  PubMed  Google Scholar 

  186. Gao F, Huang W, Zhang YQ, Tang SH, Zheng L, Ma F, et al. Hes1 promotes cell proliferation and migration by activating Bmi-1 and PTEN/Akt/GSK3ß pathway in human colon cancer. Oncotarget. 2015;6(36):38667–80.

    Article  PubMed  PubMed Central  Google Scholar 

  187. Gao F, Zhang Y, Wang S, Liu Y, Zheng L, Yang J, et al. Hes1 is involved in the self-renewal and tumourigenicity of stem-like cancer cells in colon cancer. Sci Rep. 2014;4(1):3963. https://doi.org/10.1038/srep03963.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Zhang Z, Li G, Qiu H, Yang J, Bu X, Zhu S, et al. The novel notch-induced long noncoding RNA LUNAR1 determines the proliferation and prognosis of colorectal cancer. Sci Rep. 2019;9(1):1–9. https://doi.org/10.1038/s41598-019-56536-2.

    Article  CAS  Google Scholar 

  189. Qian J, Garg A, Li F, Shen Q, Xiao K. LncRNA LUNAR1 accelerates colorectal cancer progression by targeting the miR-495-3p/MYCBP axis. Int J Oncol. 2020;57(5):1157–68.

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Zhang M, Jiang X, Jiang S, Guo Z, Zhou Q, He J. LncRNA FOXD2-AS1 regulates miR-25-3p/sema4c axis to promote the invasion and migration of colorectal cancer cells. Cancer Manag Res. 2019;11:10633–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Zhu Y, Qiao L, Zhou Y, Ma N, Wang C, Zhou J. Long non-coding RNA FOXD2-AS1 contributes to colorectal cancer proliferation through its interaction with microRNA-185-5p. Cancer Sci. 2018;109(7):2235–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Yang L, Cui J, Wang Y, Tan J. FAM83H-AS1 is upregulated and predicts poor prognosis in colon cancer. Biomed Pharmacother. 2019;118(August):109342. https://doi.org/10.1016/j.biopha.2019.109342.

    Article  CAS  PubMed  Google Scholar 

  193. Zhang J, Yin M, Huang J, Lv Z, Liang S, Miao X, et al. Long noncoding RNA LINC00152 as a novel predictor of lymph node metastasis and survival in human cancer: a systematic review and meta-analysis. Clin Chim Acta. 2018;483(March):25–32. https://doi.org/10.1016/j.cca.2018.03.034.

    Article  CAS  PubMed  Google Scholar 

  194. Yue B, Cai D, Liu C, Fang C, Yan D. Linc00152 functions as a competing endogenous RNA to confer oxaliplatin resistance and holds prognostic values in colon cancer. Mol Ther. 2016;24(12):2064–77. https://doi.org/10.1038/mt.2016.180.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Li B, Sun H, Zhang J. LncRNA DSCAM-AS1 promotes colorectal cancer progression by acting as a molecular sponge of miR-384 to modulate AKT3 expression. Aging (Albany NY). 2020;12(10):9781–92.

    Article  CAS  Google Scholar 

  196. Wang H, Luan H, Zhan T, Liu X, Song J, Dai H. Long non-coding RNA LINC00707 acts as a competing endogenous RNA to enhance cell proliferation in colorectal cancer. Exp Ther Med. 2020;19(2):1439–47. https://doi.org/10.3892/etm.2019.8350.

    Article  CAS  PubMed  Google Scholar 

  197. Palko-Łabuz A, Środa-Pomianek K, Wesołowska O, Kostrzewa-Susłow E, Uryga A, Michalak K. MDR reversal and pro-apoptotic effects of statins and statins combined with flavonoids in colon cancer cells. Biomed Pharmacother. 2018;2019(109):1511–22.

    Google Scholar 

  198. Shao HJ, Li Q, Shi T, Zhang GZ, Shao F. LINC00707 promotes cell proliferation and invasion of colorectal cancer via miR-206/FMNL2 axis. Eur Rev Med Pharmacol Sci. 2019;23(9):3749–59.

    PubMed  Google Scholar 

  199. Meng X, Fu R. MiR-206 regulates 5-FU resistance by targeting Bcl-2 in colon cancer cells. Onco Targets Ther. 2018;11:1757–65.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Xie T, Li Y, Li S-L, Luo H-F. Astragaloside IV enhances cisplatin chemosensitivity in human colorectal cancer via regulating NOTCH3. Oncol Res. 2016;24(6):447–53.

    Article  PubMed  PubMed Central  Google Scholar 

  201. Yao J, Qian CJ. Notch3 siRNA enhances chemosensitivity of colon cancer cells to topotecan. Chin J Cancer Biother. 2009;16(6):604–8.

    CAS  Google Scholar 

  202. Ma Y, Bu D, Long J, Chai W, Dong J. LncRNA DSCAM-AS1 acts as a sponge of miR-137 to enhance Tamoxifen resistance in breast cancer. J Cell Physiol. 2019;234(3):2880–94.

    Article  CAS  PubMed  Google Scholar 

  203. Guo Y, Pang Y, Gao X, Zhao M, Zhang X, Zhang H, et al. MicroRNA-137 chemosensitizes colon cancer cells to the chemotherapeutic drug oxaliplatin (OXA) by targeting YBX1. Cancer Biomark. 2017;18:1–9.

    Article  PubMed  Google Scholar 

  204. Li X, Zhao H, Zhou X, Song L. Inhibition of lactate dehydrogenase A by microRNA.34a resensitizes colon cancer cells to 5.fluorouracil. Mol Med Rep. 2015;11(1):577–82.

    Article  CAS  PubMed  Google Scholar 

  205. Vaish V, Kim J, Shim M. Jagged-2 (JAG2) enhances tumorigenicity and chemoresistance of colorectal cancer cells. Oncotarget. 2017;8(32):53262–75.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Ferreira AC, Suriano G, Mendes N, Gomes B, Wen X, Carneiro F, et al. E-cadherin impairment increases cell survival through Notch-dependent upregulation of Bcl-2. Hum Mol Genet. 2012;21(2):334–43. https://doi.org/10.1093/hmg/ddr469.

    Article  CAS  PubMed  Google Scholar 

  207. Cho S, Lu M, He X, Ee PL-R, Bhat U, Schneider E, et al. Notch1 regulates the expression of the multidrug resistance gene ABCC1/MRP1 in cultured cancer cells. Proc Natl Acad Sci. 2011;108(51):20778–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Xu K, Shen K, Liang X, Li Y, Nagao N, Li J, et al. MiR-139-5p reverses CD44+/CD133+-associated multidrug resistance by downregulating NOTCH1 in colorectal carcinoma cells. Oncotarget. 2016;7(46):75118–29.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Sodani K, Patel A, Kathawala RJ, Chen Z-S. Multidrug resistance associated proteins in multidrug resistance. Chin J Cancer. 2012;31(2):58–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Liu N, Li J, Zhao Z, Han J, Jiang T, Chen Y, et al. MicroRNA-302a enhances 5-fluorouracil-induced cell death in human colon cancer cells. Oncol Rep. 2017;37(1):631–9.

    Article  PubMed  Google Scholar 

  211. Zheng Y-B, Luo H-P, Shi Q, Hao Z-N, Ding Y, Wang Q-S, et al. miR-132 inhibits colorectal cancer invasion and metastasis via directly targeting ZEB2. World J Gastroenterol. 2014;20(21):6515–22.

    Article  PubMed  PubMed Central  Google Scholar 

  212. Sun ZF, Zhang Z, Liu Z, Qiu B, Liu K, Dong G. MicroRNA-335 inhibits invasion and metastasis of colorectal cancer by targeting ZEB2. Med Oncol. 2014;31(6):982.

    Article  PubMed  Google Scholar 

  213. Liu F, Jia J, Sun L, Yu Q, Duan H, Jiao D, et al. lncRNA DSCAM-AS1 downregulates miR-216b to promote the migration and invasion of colorectal adenocarcinoma cells. Onco Targets Ther. 2019;12:6789–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Kureishy N, Sapountzi V, Prag S, Anilkumar N, Adams JC. Fascins, and their roles in cell structure and function. BioEssays. 2002;24(4):350–61.

    Article  CAS  PubMed  Google Scholar 

  215. Ou C, Sun Z, He X, Li X, Fan S, Zheng X, et al. Targeting YAP1/LINC00152/FSCN1 signaling axis prevents the progression of colorectal cancer. Adv Sci. 2020;7(3):1901380.

    Article  CAS  Google Scholar 

  216. Adams JC. Roles of fascin in cell adhesion and motility. Curr Opin Cell Biol. 2004;16(5):590–6.

    Article  CAS  PubMed  Google Scholar 

  217. Yue B, Liu C, Sun H, Liu M, Song C, Cui R, et al. A positive feed-forward loop between LncRNA-CYTOR and Wnt/β-catenin signaling promotes metastasis of colon cancer. Mol Ther. 2018;26(5):1287–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Ozawa T, Kazama S, Akiyoshi T, Murono K, Yoneyama S, Tanaka T, et al. Nuclear Notch3 expression is associated with tumor recurrence in patients with stage II and III colorectal cancer. Ann Surg Oncol. 2014;21(8):2650–8. https://doi.org/10.1245/s10434-014-3659-9.

    Article  PubMed  Google Scholar 

  219. Tyagi A, Sharma AK, Damodaran C. A review on Notch signaling and colorectal cancer. Cells. 2020;9(6):1549.

    Article  CAS  PubMed Central  Google Scholar 

  220. Timmerman LA, Grego-Bessa J, Raya A, Bertrán E, Pérez-Pomares JM, Díez J, et al. Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev. 2004;18(1):99–115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Sahlgren C, Gustafsson MV, Jin S, Poellinger L, Lendahl U. Notch signaling mediates hypoxia-induced tumor cell migration and invasion. Proc Natl Acad Sci U S A. 2008;105(17):6392–7. https://doi.org/10.1073/pnas.0802047105.

    Article  PubMed  PubMed Central  Google Scholar 

  222. Sonoshita M, Aoki M, Fuwa H, Aoki K, Hosogi H, Sakai Y, et al. Suppression of colon cancer metastasis by aes through inhibition of notch signaling. Cancer Cell. 2011;19(1):125–37.

    Article  CAS  PubMed  Google Scholar 

  223. Yuan R, Ke J, Sun L, He Z, Zou Y, He X, et al. HES1 promotes metastasis and predicts poor survival in patients with colorectal cancer. Clin Exp Metastasis. 2015;32(2):169–79. https://doi.org/10.1007/s10585-015-9700-y.

    Article  CAS  PubMed  Google Scholar 

  224. Weng MT, Tsao PN, Lin HL, Tung CC, Change MC, Chang YT, et al. Hes1 increases the invasion ability of colorectal cancer cells via the STAT3-MMP14 pathway. PLoS ONE. 2015;10(12): e0144322.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Rizk NI, Abulsoud AI, Kamal MM, Kassem DH, Hamdy NM. Exosomal-long non-coding RNAs journey in colorectal cancer: Evil and goodness faces of key players. Life Sci. 2022;292:120325.

    Article  CAS  PubMed  Google Scholar 

  226. Wan M, Wang Y, Zeng Z, Deng B, Zhu B, Cao T, et al. Colorectal cancer (CRC) as a multifactorial disease and its causal correlations with multiple signaling pathways. Biosci Rep. 2020;40(3):00265. https://doi.org/10.1042/BSR20200265.

    Article  Google Scholar 

  227. Yang Z-H, Dang Y-Q, Ji G. Role of epigenetics in transformation of inflammation into colorectal cancer. World J Gastroenterol. 2019;25(23):2863.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB). This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

EO data curation, original draft preparation and rewriting, figures and tables construction, FE editing, rewriting, figures and tables revision, HNM conceptualization, supervision, tables and figures idea creation and revision, editing, rewriting, reviewing from submission till acceptance.

Corresponding author

Correspondence to Nadia M. Hamdy.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publications

All authors have read the manuscript and agreed to publish.

Competing interests

Authors declare there is no conflict of interest.

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

Emam, O., Wasfey, E.F. & Hamdy, N.M. Notch-associated lncRNAs profiling circuiting epigenetic modification in colorectal cancer. Cancer Cell Int 22, 316 (2022). https://doi.org/10.1186/s12935-022-02736-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-022-02736-2

Keywords