Next Article in Journal
Contemporary Biomarkers in Pulmonary Embolism Diagnosis: Moving beyond D-Dimers
Next Article in Special Issue
C10orf55, CASC2, and SFTA1P lncRNAs Are Potential Biomarkers to Assess Radiation Therapy Response in Head and Neck Cancers
Previous Article in Journal
Gender Differences in Oral Health: Self-Reported Attitudes, Values, Behaviours and Literacy among Romanian Adults
Previous Article in Special Issue
Impact of the COVID-19 Pandemic on Breast Cancer Stage at Diagnosis in a Regional Cancer Center in Poland between 2019 and 2021
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radio-lncRNAs: Biological Function and Potential Use as Biomarkers for Personalized Oncology

by
Joanna Kozłowska-Masłoń
1,2,3,*,†,
Kacper Guglas
1,2,4,†,
Anna Paszkowska
1,2,5,‡,
Tomasz Kolenda
1,2,‡,
Marta Podralska
6,
Anna Teresiak
1,2,
Renata Bliźniak
1 and
Katarzyna Lamperska
1,2,*
1
Laboratory of Cancer Genetics, Greater Poland Cancer Center, Garbary Street 15, 61-866 Poznan, Poland
2
Research and Implementation Unit, Greater Poland Cancer Center, Garbary Street 15, 61-866 Poznan, Poland
3
Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, 61-614 Poznan, Poland
4
Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki and Wigury Street 61, 02-091 Warsaw, Poland
5
Faculty of Biology, Adam Mickiewicz University, Umultowska 89, 61-614 Poznan, Poland
6
Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479 Poznan, Poland
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors contributed equally to this work.
J. Pers. Med. 2022, 12(10), 1605; https://doi.org/10.3390/jpm12101605
Submission received: 31 August 2022 / Revised: 20 September 2022 / Accepted: 26 September 2022 / Published: 29 September 2022
(This article belongs to the Special Issue Personalized Radiotherapy)

Abstract

:
Long non-coding RNAs (lncRNAs) consist of at least 200 nucleotides. Although these molecules do not code proteins, they carry many regulatory functions in normal cells, as well as in cancer cells. For instance, many of these molecules have been previously correlated with tumorigenesis of different cancers and their reaction to various stress factors, such as radiotherapy, chemotherapy, or reactive oxygen species (ROS). The lncRNAs are associated not only with dysregulation in cancers after applied treatment but also with beneficial effects that may be achieved by modulating their expression, often significantly enhancing the patients’ outcomes. A multitude of these molecules was previously considered as potential biomarkers of tumor development, progression, or cells’ response to radio- or chemotherapy. Irradiation, which is often used in treating numerous cancer types, is not always sufficient due to cells gaining resistance in multiple ways. In this review, studies considering lncRNAs and their reaction to radiotherapy were examined. These molecules were divided regarding their role in specific processes strictly related to irradiation, and their influence on this type of treatment was explained, showing how vast an impact they have on IR-supported combat with the disease. This review aims to shed some light on potential future lncRNA-based biomarkers and therapeutic targets.

1. lncRNAs Are New Players in Radiogenomics

It is well known that genetic background has a pivotal influence on the radioresistance of normal and cancer cells. Current research on radiosensitivity focuses on comprehensive genome analyses that take into account mutations in genes, as well as changes at the transcriptome and epigenome levels [1,2]. Unfortunately, to this day, it has not been possible to create specific mutation panels enabling the determination of clinical response to the applied radiotherapy. Radiogenomics is “the study of genomic changes that underlie the radioresponse of normal and tumor tissues” and is likely to enable a breakthrough in personalized radiotherapy [3]. It should be emphasized that the radiogenomic research conducted so far has focused on mRNA transcripts, protein-coding genes, and epigenetic elements—miRNAs [1,4,5]. However, the epigenome is constructed by many different types of RNA molecules, which interact and together create complex networks consisting of mRNAs, miRNAs, and other non-coding RNAs (ncRNAs) [6,7].
ncRNAs can be divided into two groups: constitutive RNAs such as transfer RNA (tRNA), ribosomal RNA (rRNA), small-nuclear RNA (snRNA), and small-nucleolar RNA (snoRNA); and regulatory RNAs including small interfering RNA (siRNA), piwi-interacting RNA (piRNA), microRNA (miRNA), natural antisense transcripts (NAT), circular RNA (circRNA), and long non-coding RNA (lncRNA) [1,2,3]. It should be mentioned that this list is incomplete, and periodically new dependencies or even elements of the epigenome are discovered. This is possible due to the development of databases, analytical techniques, bioinformatics, and the use of artificial intelligence (AI) [3,8,9,10]. As a result, the so-called genetic noise, the unknown or omitted functional RNA molecules, have been discovered and now are known as lncRNAs. In addition, they are also partly classified as pseudogenes [11,12]. Although these molecules do not encode proteins, over time they have become valuable and significant elements of cell biology. Their sequence consists of more than 200 nucleotides and very often contains components typical for mRNA, namely poly-A tails and regulatory sequences, such as those for miRNAs. The activity of lncRNAs is characterized not only by interaction with proteins and other RNAs but also by regulation of transcription and expression of genes through changes to the chromatin structure. Genes encoding the mentioned molecules can occupy intergenic or intragenic positions, of which a specific intragenic gene may be located in the intron, transcriptional enhancer elements, promoter, or 3′UTR flanking regions [11,13].
The process of lncRNA biogenesis is regulated by RNA polymerase II and is similar to mRNA formation. After transcription, the cap structure is attached to the molecule; then it undergoes polyadenylation and splicing [14]. lncRNAs appear to be pivotal in functions related to the regulation of gene transcription in the nucleus or subsequent post-transcriptional modifications in the cytoplasm. However, due to their location in the genome, it is difficult to fully understand the function of lncRNAs. It should be noted that more than 50% of lncRNAs are long intergenic non-coding RNAs (lincRNAs) [15]. The direction of transcription and the distance at which the transcript responsible for encoding the protein is located in relation to the lincRNA has made it possible to divide this group of RNAs into four classes: (i) located on the same strand, (ii) convergent, (iii) divergent, and (iv) isolated, which are located at least 50 kb from the nearest gene encoding the protein [15]. The main challenges in understanding the mechanisms of action of lncRNAs stem from the varying levels of expression depending on tissue localization [16], the frequent heterogeneity of isoforms, and the numerous repeats in regions of transcription initiation [17]. Moreover, since it has been shown that lncRNA activity changes with different levels of their expression, its specificity in various cell types and tissues should be assessed [18]. As it turned out, abnormalities in activity or biogenesis mechanisms of these molecules can appear in states of pathological conditions and indicate cancer progression by affecting a multitude of transcription factors along with the structure of the chromatin [19]. It has been demonstrated that the aberration of not only coding RNAs but also non-coding RNAs plays a crucial role in cancer biology. Although the function and activity of lncRNAs are still under investigation, in the future they may become crucial tools for predicting the development and possible treatment of cancer [19].
It should be noted that changes in the epigenetic system can be caused by even small doses of irradiation (IR). Li et al. indicated that hepatocarcinogenesis can be caused by changes in the expression of HULC lncRNA, which is an IR-induced molecule. The abovementioned RNA regulates the neighboring gene, CDKN1, by complementary base pairing, and in turn promotes cell cycle progression [20]. Additionally, apart from local, tissue-based changes in lncRNA expression, IR also causes a systematic, whole organism response. Aryankalayil et al. observed that after mice whole-body IR, using different doses of X-rays, expression levels of different lncRNAs were dramatically changed in the whole blood. The radiation-induced RNAs were connected with DNA damage response and the immune system. Moreover, it has been proven that two lncRNAs, Gm14005 (Morrbid) and Tmevpg1, could be used as potential biomarkers after radiation exposition [21].
Studies indicate that epigenetic mechanisms are crucial for the IR-induced changes in reactive oxygen species (ROS) generation, as well as that oxidative stress itself can modulate epigenetics in the cells exposed to radiation [22]. lncRNAs, due to their vast influence on cellular processes, including DNA damage repair, cell cycle arrest, apoptosis, senescence, maintaining cancer stem cell populations, the epithelial-to-mesenchymal transition (EMT) process, and autophagy, seem to play an important role in the response to IR and are new players in radiobiological research [23].
In this review, the current knowledge about lncRNAs involved in radiobiological processes, as well as their use as potential biomarkers in oncology, especially in personalized radiotherapy, is discussed.

2. Involvement of lncRNAs in Radiobiological Processes

lncRNAs are pivotal regulators of cell biology that have a significant impact on cancer development. However, these molecules also play a crucial role in adverse effects associated with radiotherapy. One such complication, radiation-induced intestinal fibrosis (RIF), is frequent in abdominal and pelvic tumor treatment. Zhou et al. determined that the lncRNA WWC2-AS1 is overexpressed in RIF compared to adjacent tissue, and its knockdown significantly inhibited proliferation, invasion, and migration of changed tissue, presumably through modulating the miR-16/FGF2 axis. A therapeutic strategy targeting this lncRNA could reduce RIF progression and lead to improvement in patients’ life quality [24]. Such discoveries emphasize the importance of RNA-based cancer research in the context of tumor biology and in the accompanying changes throughout the organism before, during, and after treatment; see Figure 1.

2.1. Cell Cycle and Proliferation

One of the lncRNAs that have a notable impact on the cancer cell cycle and proliferation is the small nucleolar RNA host gene 7 (SNHG7) lncRNA. Chen et al. observed that the SNHG7/miR-9-5p/DPP4 axis modulates tumor growth based on analyses of 131I-resistant thyroid carcinoma cell lines. The examined lncRNA acted as a molecular sponge sequestering miR-9-5p, which led to an increase in dipeptidyl-peptidase 4 (DPP4) expression levels resulting in activation of the PI3K/Akt signaling pathway. The authors emphasized the importance of this regulatory axis as a future therapeutic target [25]. Experiments by Guo et al. discovered that in esophageal cancer, lncRNA HCP5 modulates the Akt signaling pathway via regulating the miR-216a-3p/PDK1 axis. Its knockdown combined with irradiation led to suppression of proliferation, a decrease in AKT activation, and an increase in apoptosis rate, consequently improving radiosensitivity [26].
Small nucleolar RNA host gene 6 (SNHG6) is significantly overexpressed in breast cancer (BC) cells and tissues compared to normal cells. It positively correlates with laminin subunit gamma 1 (LAMC1) and negatively with miR-543. The downregulation of SNHG6 resulted in inhibition of cell migration and proliferation, impaired colony formation, enhanced apoptosis, and radiosensitivity of BC cells. Additionally, downregulation of this lncRNA also decreased protein levels of Snail and vimentin but increased E-cadherin, suggesting that its depletion suppressed EMT processes in BC cells. Interestingly, miR-543, which is negatively correlated with SNHG6, can change to the opposite of all of the above when overexpressed in BC cells. The knockdown of the abovementioned lncRNA also decreased levels of LAMC1, p-PI3K, and p-AKT in the PI3K/AKT pathway. All of the results above show how vast an impact SNHG6 in BC has on radiotherapy response [27].
Colorectal neoplasia differentially expressed (CRNDE) lncRNA is significantly upregulated in ovarian cancer cells and correlated with their radioresistance. CRNDE silencing resulted in enhanced radiosensitivity and inhibited clone formation and tumor growth in mice [28].
Another molecule influencing the Akt pathway activation is urothelial carcinoma-associated 1 (UCA1). A study by Fotouhi Ghiam et al. indicated that this molecule is significantly upregulated in prostate cancer (PCa), and its high level is associated with an unfavorable prognosis. Experimentally induced depletion of UCA1 in cell lines abrogated the aggressive phenotype and potentiated radiosensitivity via reducing proliferative capacity, leading to cell cycle arrest at the G2/M transition and inhibiting activation of the pro-survival Akt pathway. It was determined that this lncRNA has significant prognostic and therapeutic potential in PCa [29]. A different lncRNA linked with PCa irradiation (IR)-based treatment is GAS5. Yung et al. discovered that radiotherapy supported by α-Solanine administration caused upregulation of the said lncRNA and downregulation of miR-18a, which led to inhibition of cells’ proliferation ability and promotion of apoptosis via an increasing level of γ-H2AX [30]. The above interaction seems to be a fascinating new therapeutic approach, which could provide sensitization to radioresistant tumors.
The aforementioned GAS5 can also modulate G2/M arrest in malignant cells. Ma et al. discovered that it was downregulated in the BC cell line, and its level further decreased after IR. Moreover, they have proven that induced GAS5 overexpression assisted by radiotherapy suppressed cell proliferation, increased the DNA damage rate, and promoted activation of apoptosis, elevating Bax and caspase-3 expression levels. This pro-apoptotic response could be diminished by miR-21; however, it is negatively regulated by the studied lncRNA. Although the exact mechanism has not been fully described yet, the presented results imply that GAS5 plays a pivotal role in radiosensitization [31]. A different molecule regulating G2/M arrest is lncRNA MALAT1. Analysis carried out on high-risk human papillomavirus (HR-HPV)-positive cervical cancer (CC) cell lines showed that this molecule modulated radiotherapy response via sequestering miR-145. Depletion of MALAT1 resulted in an elevated ratio of G2/M checkpoint arrest and cell apoptosis, which induced radiosensitivity [32].
Colon cancer-associated transcript 1 (CCAT1) in NSCLC cells is upregulated, causing high radioresistance and a low apoptosis rate of lung cancer cells. On the other hand, downregulation of this lncRNA resulted in radiosensitivity improvement of NSCLC cells by mediating cell cycle arrest, increased apoptosis rate, and DNA damage. Downregulation of CCAT1 arrests cells at G2/M, which is believed to be the most radiosensitive moment for cells, and further promotes radiation-induced γH2AX expression, a DNA damage marker. Silencing CCAT1 may also block MAPK signaling pathways and decrease p-p38/p38, p-ERK/ERK, and p-JNK/JNK [33].
Li et al. carried out analyses indicating that lncRNA HMMR-AS1 is highly expressed in glioblastoma (GM) cell lines and stabilizes mRNA of HMMR oncogene via sense-antisense interference. Depletion of lncRNA leads to a reduction in HMMR level resulting in cell arrest at the G1/S checkpoint and consequently inhibits tumor growth, cell migration, and the mesenchymal phenotype. The described interaction might become a very interesting possible therapeutic strategy [34].
A study by Liu et al. identified the LINC00473/miR-497-5p/CDC25A axis, which modulates esophageal squamous cell carcinoma (ESCC) cell lines’ proliferative ability and response to IR. Overexpression of said lncRNA was linked with a more advanced T stage, lymph node metastasis, and less differentiated tumor tissue. LINC00473′s oncogenic function could be abrogated by induced overexpression of miR-497-5p, resulting in sensitization to radiotherapy [35]. Interestingly, Chen et al. implied that LINC00473 elevated SPIN1 expression by negatively regulating miR-374a-5p, resulting in the promotion of cell proliferation and radioresistance. Further studies showed that both LINC00473 and SPIN1 competed for miRNA, and their high expression levels were correlated with poor prognosis and weakened response to IR [36].
Tang et al. reported that DiGeorge syndrome critical region gene 5 (DGCR5) is a lncRNA associated with larynx squamous cell carcinoma (LSCC) progression and resistance to treatment. This molecule acts as a molecular sponge and negatively regulates miR-195. The authors proposed that both silencing of DGCR5 and inducing expression of miRNA have a beneficial impact on supporting effects of IR treatment [37].

2.2. Cell Death and Autophagy

Radiotherapy can induce the death of cancer cells in different pathways, such as apoptosis, pyroptosis, mitotic catastrophe, necrosis, or autophagy.
Liu et al. determined that lncRNA LINC00630 binds to EZH2 and subsequently negatively regulates the BEX1 gene via enhancing its promoter methylation in colorectal cancer (CRC) cell lines. Epigenetic silencing of BEX1 results in suppression of apoptosis, enhancement of cell viability, and strengthening of the radioresistance mechanisms. On the other hand, lncRNA silencing significantly improved the sensitivity of cancer cells after IR. Although the exact nature of this interaction has not been fully described yet, the LINC00630/EZH2/BEX1 axis seems to be a new fascinating direction of study [38]. A different lncRNA, whose knockdown might notably improve the efficacy of radiotherapy in CRC patients, is HOX transcript antisense RNA (HOTAIR). It has been proven that lncRNA acts as a molecular sponge for miR-93, reducing its level and leading to an increase of the ATG12 expression level. HOTAIR silencing resulted in a decrease in cell viability and survival rates, as well as a reduction of pro-apoptotic protein expression, which caused activation of apoptosis and an increase in levels of p62, cleaved caspase-3, and Bax in studied cell lines [39]. Contrary to the LINC000630 and HOTAIR, the effect of lincRNA-p21 overexpression in CRC enhances the beneficial effects of radiotherapy. Its level tends to rise after applied IR and reinforces radiosensitivity by potentiating cell apoptosis. Induced upregulation inhibited Wnt/β-catenin signaling and elevated the expression level of the pro-apoptotic gene—Noxa [40].
The ESCC-based study by Lin et al. indicated that simultaneous overexpression of lncRNA GAS5 and downregulation of miR-21 cause elevation of RECK expression levels, strengthening radiosensitivity after IR. The obtained effect seems to result from enhanced apoptosis. Moreover, levels of all three molecules were correlated with clinical features such as TNM staging, degree of differentiation, lymph node metastasis, and distant metastasis [41].
In nasopharyngeal carcinoma (NPC), lncRNA PVT1 has been associated with unfavorable prognosis and radioresistance promotion. He et al. proved that its silencing sensitized cell lines to the applied treatment leading to inhibition of proliferation and induction of apoptosis. The depletion of PVT1 resulted in a reduction of ATM, Chk2, and p53 phosphorylation levels, as well as the activation of pro-apoptotic proteins. The above underlines the prognostic and therapeutic potential of the said molecule [42].
Membrane-associated guanylate kinase, WW, and PDZ domain containing 2 antisense RNA 3 (MAGI2-AS3) lncRNA is poorly expressed in ESCC. It is known to negatively correlate with homeobox protein Hox-B7 (HOXB7), which is overexpressed in ESCC and contributes to cancers’ radioresistance. MAGI2-AS3 downregulates HOXB7 via histone methyletransferase EZH2 to initiate H3K27me3, which is required for the EZH2-mediated repression of various genes that are vital for carcinogenesis and tumor development. H3K27me3 has the ability to suppress HOXB7 and its functions, such as developing radioresistance in ESCC cells. MAGI2-AS3 overexpression inhibits ESCC cells’ proliferation and resistance to radiotherapy as well as promotes cell apoptosis by downregulating HOXB7. In conclusion, MAGI2-AS3 overexpression enhances the radiosensitivity of ESCC cells to radiotherapy through the downregulation of HOXB7 via EZH2 [43].
The lncRNA antisense non-coding RNA in the INK4 locus (ANRIL), the antisense RNA1 of CDKN2B, is highly expressed in colon cancer tissues and cell lines, inhibiting apoptosis and causing resistance to radiotherapy. The upregulation of ANRIL inhibited chitooligosaccharide (COS)-induced radiosensitivity in colon cancer cells by targeting miR-181a-5p. Knockdown of said lncRNA or upregulating miR-181a-5p, which is negatively correlated with ANRIL, may change that effect on colon cancer cells, resulting in radiosensitivity enhancement [44].
In osteosarcoma cells and tissues, LINC00210 is significantly elevated and correlated with enhanced radioresistance. On the other hand, LINC00210 knockdown inhibits colony formation ability, decreases cell viability, and significantly induces apoptosis by inhibiting levels of cyclin D1 and Bcl-2, as well as overexpressing p21 and Bax. The abovementioned lncRNA is negatively correlated with miR-342-3p, which may reverse all effects of LINC00210 up- or downregulation. Additionally, LINC00210 is positively correlated with GFRA1, which when downregulated, also increases the radiosensitivity of osteosarcoma cells [45].
A different kind of IR-induced programmed cell death is pyroptosis—an inflammatory process that reduces the proliferation and invasiveness of cancer cells. It has been proven that it is regulated by lncRNA NEAT1 in CRC cell lines through the miR-448/GSDME axis. The expression level of the studied lncRNA was upregulated in tumor tissue and increased further after IR in a time-dependent manner, promoting GSDME-mediated pyroptosis. Activation of this type of cell death leads to an increase in the radioresistance of tumor cells [46].
The process of IR-promoted autophagy could protect in detrimental conditions, provided that it is not extensive, then it could activate apoptosis. Jiang et al. conducted an in silico analysis of autophagy-related lncRNAs in lung adenocarcinoma (LUAD). They discovered molecular signatures consisting of lncRNAs TMPO-AS1 and BIRC5, which were both overexpressed in patients’ samples. These molecules’ co-expression has been correlated with the advanced stage of the disease and unfavorable outcomes, suggesting the emergence of interesting future therapeutic targets [47]. A study by Gao et al. showed that lncRNA TP53TG1 is upregulated in glioma cell lines, and its expression level tends to rise after radiation. Moreover, its high levels potentiated tumor progression and radioresistance via autophagy activation. The authors indicated that lncRNA depletion reduced tumor growth and promoted apoptosis through the TP53TG1/miR-524-5p/RAB5A axis, improving radiotherapy effectiveness [48]. Another lncRNA involved in the activation of autophagy processes in glioma is linc-RA1. Zheng et al. proved that lincRNA stabilized the level of H2B K120 monoubiquitination (H2Bub1) via binding with H2B, subsequently reducing the interaction between H2Bub1 and ubiquitin-specific protease 44 (USP44). The described axis leads to autophagy inhibition and radioresistance progression [49]. The above underlines the still ambiguous role of autophagy in the process of cancer treatment.
Hepatocellular carcinoma upregulated long non-coding RNA (HULC) is significantly overexpressed in PCa cells. Its downregulation causes notably higher cell radiosensitivity. HULC downregulation also upregulated Bax and active-caspase 3 and downregulated PCNA and cyclinD1, suggesting that this lncRNA depletion increased the apoptosis rate among PCa cells after IR exposure. HULC knockdown also causes G0/G1 cell cycle arrest, while its overexpression arrests cells at the S phase. Additionally, HULC downregulation elevated phosphorylated levels of Beclin-1 and promoted autophagy through inhibition of the mTOR pathway [50].
Cancer susceptibility 19 (CASC19) lncRNA strongly contributes to the radioresistance of NPC cells by promoting autophagy through the AMPK/mTOR pathway. CASC19 is overexpressed in NPC cells and is associated with radioresistance. However, its knockdown enhances radiosensitivity and decreases autophagy via the AMPK/mTOR pathway. Inhibition of the studied lncRNA expression caused higher levels of IR-induced DNA damage, elevated IR-induced apoptosis with PARP1, and enhancement of cleaved caspase-3. This indicates that silencing CASC19 disturbed the protective effect of autophagy on IR-induced apoptosis. The abovementioned lncRNA contributes to the radioresistance of NPC cells by promoting autophagy and inhibiting apoptosis through the AMPK/mTOR signaling pathway [51].

2.3. Reactive Oxygen Species (ROS)

Ionizing radiation causes ROS generation and subsequently oxidative stress. Additionally, the process of tumor development itself generates significant amounts of these reactive molecules due to an increase in DNA mutations, high genome instability, and cell proliferation. Wang et al. determined that novel lncRNA AL033381.2 may exert its oncogenic function in hepatocellular carcinoma (HCC) through interacting with the PRKRA protein and targeting genes related to oxidative stress. Experiments carried out on mice with HCC xenografts treated with complexes combining nanoparticles and AL033381.2 siRNA showed anticancer effects leading to a reduction of tumor volume and weight. This lncRNA has the potential to become a therapeutic target in the future [52].
One of the factors promoting cancer-related oxidative stress is hypoxia—an oxygen deficiency resulting from intensive cell proliferation. This phenomenon is a characteristic feature of non-small cell lung cancer. It has been proven that lncRNA PVT1 regulates HIF1α by sponging miR-199a-5p in NSCLC cell lines. Moreover, it can be a potential therapeutic target supporting the fight against hypoxia and thus IR resistance [53].
Another lncRNA modulating HIF1α expression levels is HOTAIR. Studies carried out on CC cell lines indicated that high levels of this lncRNA were associated with developing resistance to radiotherapy. Interestingly, IR tends to diminish HOTAIR and HIF1α levels in cells, which results in decreasing cell viability and tumor growth, as well as inhibiting apoptosis. This implies that induced reduction of studied lncRNA expression accompanied by radiation treatment could bring satisfactory results in CC therapy [54].

2.4. DNA Damages and Repair Mechanisms

Radiotherapy induces cell death predominantly via promoting DNA double-strand breaks (DSBs).
A study by Wang et al. indicated that lncRNA LINC01134 is significantly overexpressed in HCC and affects the response to IR by regulating DNA damage repair mechanisms. This molecule acts as a molecular sponge sequestering miR-342-3p and recruiting insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) to modulate mitogen-activated protein Kinase 1 (MAPK1) expression. LINC01134 depletion resulted in a higher DNA damage rate, tumor growth inhibition, and radioresistance reduction in HCC cells [55].
Another lncRNA playing a crucial role in modulating DNA repair mechanisms is LINC-PINT. Experiments carried out on NPC cells showed that this lncRNA might be a radiosensitizing tumor suppressor reducing these types of molecular pathways through the ATM/ATR-Chk1/Chk2 signaling axis. Additionally, LINC-PINT interacts with DNA-dependent kinase proteins (DNA-PKcs), inhibiting DNA damage response cascades and mediating cell cycle arrest predominantly in the G2 phase, in which the cell is the most vulnerable to IR [56].
LINC00312 is significantly downregulated in NPC and correlates with poor radiotherapy efficacy. On the other hand, LINC00312 overexpression results in decreased cell viability, impaired colony formation ability, cell cycle arrest in the G0/G1 phase, and better overall survival of NPC patients. Its overexpression enhances the sensitivity of xenograft tumors to radiation in vitro.
Studied lncRNA contributes to the NPC cells’ radioresistance through impairing DNA damage repair. LINC00312 inhibits the recruitment of DNA-PKc to Ku80 in response to DNA double-strand breaks. Overexpression of LINC00312 combined with exposure to irradiation decreased levels of p-ATM, p-ATR, p-Chk1, and p-Chk2, which play a crucial role in DNA damage checkpoint control and tumor inhibition. All of these results show that LINC00312 affects radiosensitivity by regulating the DNA damage repair pathway [57].
Yao et al. observed that the ANKHD1/MALAT1/YAP1 feedback loop induces radioresistance of CRC cells through regulating processes of DNA-damage repair, probably via the YAP1/AKT axis. They have proven that the knockdown of ANKHD1 and/or lncRNA MALAT1 significantly improves radiotherapy efficacy [58].
A study by Feng et al. described the STAT1/LINC00504/CPEB2/TAF15 interaction axis, which modulates the BC cells’ response to radiotherapy. The examined lncRNA could affect DNA damage through its influence on ATM/ATR activation. LINC00504 depletion weakened the survival rate of BC cells, which was further enhanced by applied IR, suggesting its therapeutic potential [59]. Moreover, Wang et al. discovered different lncRNA, whose downregulation could significantly improve BC patients’ response to radiotherapy. LINC02582 combines with ubiquitin-specific peptidase 7 (USP7) to deubiquitinate and stabilize checkpoint kinase 1 (CHK1), affecting DNA damage response, which in consequence leads to radioresistance. It has been proven that miR-200c, a known radiosensitizer of BC, directly regulates this lncRNA expression. Induced overexpression of said miRNA suppressed DNA repair mechanisms and elevated γ-H2AX levels after IR. Survival analysis corroborated the favorable impact of high miR-200c expression levels on patients’ outcomes, implying the importance of further studying the LINC02582 mechanism of action and its possible future application in treatment [60].
HOTAIR is significantly upregulated in many cancer types, including BC tissue. Its overexpression is strictly correlated with cells’ radioresistance and shows a poor prognosis for patients. Silencing HOTAIR caused significantly reduced cell proliferation and colony formation ability [61,62]. Additionally, HOTAIR upregulates HSPA1A, a well-known stress-inducible oncogene in irradiated BC cells. On the other hand, the examined lncRNA is negatively correlated with miR-449b-5p, which has the ability to reverse all effects of HOTAIR and HSPA1A. Taking this together, HOTAIR is capable of enhancing the growth of BC tumors under IR-induced stress through the miR-449b-5p/HSPA1A signaling pathway [61]. According to the results from another study considering HOTAIR and BC, alongside overexpressed HOTAIR, expression of DNA damage repair factors, such as Ku70, Ku80, DNA-PKcs, and ATM, was significantly increased. Additionally, higher HOTAIR expression induced recruitment of EHZ2 to the specific target gene c-MYC. In addition, the above lncRNA knockdown in breast cancer cells also resulted in a decrease in the number of cells in phase S, causing a significant increase in apoptosis [62].
In the triple-negative BC (TNBC), the process of DSB repair is regulated by lncRNA in the non-homologous end joining (NHEJ) pathway 1 (LINP1). This lncRNA plays the role of a scaffold that promotes interaction between Ku80 and DNA-PKcs, modulating the non-homologous end joining (NHEJ) pathway. Interestingly, Zhang et al. have also indicated that LINP1 expression could be regulated by EGF signaling, and in particular its activation of the RAS-MEK-JNK pathway, as well as the p53 pathway, which is mediated by the miR-29. Depletion of the studied lncRNA expression level might diminish DSB repair activity and lead to tumor radiosensitization [63]. LINP1 has a significant impact not only on TNBC biology but also affects CC development. In this type of cancer, this lncRNA also augments radioresistance via modulating the NHEJ pathway. Wang et al. determined that LINP1 undergoes IR-induced translocation from cytosol to the nucleus, where it interacts with the aforementioned proteins—Ku80 and PKcs. LINP1 silencing with subsequent IR leads to delayed DNA repair mechanisms, promoted cell apoptosis, and increased radiosensitivity in CC cell lines [64]. The above suggests promising therapeutic or prognostic potential of the said lncRNA [63,64].
Analyses carried out on glioma cell lines indicated that lncRNA SNHG18 plays a notable role in modulating cancer response to radiotherapy. Its depletion caused increased γ-H2AX and cleaved caspase-3 levels, affecting the DNA damage response. Additionally, SNHG18 expression was negatively correlated with semaphorin 5a (Sema5A) levels, which potentiated radioresistance. These results seem to underline the importance of further SNHG18-related studies [65].
Dynamin 3 opposite strand (DNM3OS) is highly expressed in ESCC cell lines as well as in tumor tissue compared to normal, matched tissues. The decreased expression of DNM3OS causes significantly increased radiosensitivity in vitro and in vivo. This lncRNA regulates cellular DNA damage response in ESCC. Its inhibition enhances irradiation-induced DNA damage and attenuates DNA repair response. The downregulation of the studied lncRNA resulted in elevated expression of the DNA damage markers γH2AX and PARP and decreased expression of DNA repair proteins such as p-ATM, Rad50, p-CHK2, Ku80, MRE11, NBS1, DNA-PKcs, and p53 in response to irradiation. Interestingly, cancer-associated fibroblasts (CAFs) promote DNM3OS expression in a PDGFβ/PDGFRβ/FOXO1 signaling pathway-dependent manner in ESCC, resulting in enhancing radioresistance [66].
The lncRNA CYTOR is highly correlated with the radioresistance of NSCLC cells. CYTOR is significantly overexpressed in NSCLC cell lines and tissues and is responsible for a poor prognosis of patients. Silencing the said lncRNA enhances radiosensitivity in NSCLC cells, weakening colony formation ability and expressing high expression of γH2AX. CYTOR knockdown also enhances the radiosensitivity of xenograft tumors in mice. The studied lncRNA also sponges miR-206, which can reverse all CYTOR effects on cells. However, miR-206 regulates PTMA, which has a similar impact on NSCLC cells as CYTOR, enhancing the radioresistance of NSCLC cells [67].

2.5. Changes in Cellular Phenotype

A study by Zhu et al. discovered that lncRNA RBM5-AS1 is upregulated in radioresistant medulloblastoma (MB) cell lines and implements its oncogenic function through stabilizing the sirtuin 6 (SIRT6) protein. This interaction has been associated with the self-renewal of medulloblastoma cancer stem cells (CSCs). Knockdown inhibited viability, tumor growth, and expression of a known stemness marker—CD133, simultaneously improving IR efficacy through promoting DNA damage and apoptosis [68]. A different lncRNA linked with modulating CSC phenotypes and activity is MALAT1. Experiments carried out on NPC cell lines determined that the MALAT1/miR-1/slug axis affects cancer radioresistance. Additionally, depletion of the said lncRNA inhibited cell proliferation and invasion, which emphasizes its therapeutic potential [69].
Moreover, Lin et al. reported that lncRNA NEAT1 regulates the phenotype of the TNBC cells through interaction with NAD(P)H:quinone oxidoreductase 1 (NQO1). Induced reduction in lncRNA expression level potentiated radiosensitivity, inhibited proliferative ability and CSC activity, and diminished expression levels of known stemness genes—BMI1, Oct4, and Sox2. These results suggest that NOQ1 bioactivatable compounds could be used as a potential therapeutic target inhibiting radioresistance [70].
Ye et al. described a complex interaction axis consisting of FEZF1-AS1/miR-107/ZNF312B, which promotes the progression of pancreatic ductal adenocarcinoma (PDAC). In addition, it facilitates in cancer cells the Warburg effect—a unique metabolic phenotype with increased glycolysis and reduced oxidative phosphorylation despite oxygen availability. Downregulation of lncRNA FEZF1-AS1 caused inhibition of tumor growth, probably through modulation of the apoptosis and in the G1-S checkpoint [71]. The discovered regulatory network provided an interesting new strategy that could help reduce radioresistance, which, as implied by Liu et al., can be mediated by glycolysis [72]. During their studies on cutaneous malignant melanoma (CMM), the authors presented the LINC00518/miR-33a-3p/HIF1α negative feedback loop, which caused metabolic changes and promoted cells’ malignant phenotypes, consequently decreasing radiotherapy effectiveness [72].
A recent study by Yi et al. identified lncRNA PTPRG-AS1/miR-194-3p/PRC1 regulatory circuitry that modulates radiosensitivity and metastatic phenotype of NPC cell lines. Knockdown of lncRNA or induced overexpression of miRNA prevented cell migration, invasion, and growth, improving response to the treatment [73]. Likewise, a type of lncRNA called CYTOR in NSCLC acts as a molecular sponge binding miR-195 and modulating tumors’ malignant phenotypes. Silencing of this lncRNA diminished proliferation, migration, and invasion of cancer cells, leading to enhancement of IR-induced therapeutic effects [74]. Another lncRNA affecting tumors’ metastatic ability is HOTAIR. Yang et al. discovered that its knockdown in CRC cell lines reduces proliferation and subsequently inhibits cell invasion, as well as promoting apoptosis in an IR-mediated manner [75]. Furthermore, in CC cell lines, this lncRNA is also upregulated and exerts an oncogenic function. HOTAIR enhanced cell migration, invasion, and proliferation, by reducing p21 protein levels. Moreover, its high expression causes suppression of apoptosis [76]. The HOTAIR’s significant influence on the development of a metastatic phenotype implies its therapeutic potential [75,76].

3. Potential Use of lncRNAs as Biomarkers

Zhang et al. identified a three-lncRNA-based signature that showed significant prognostic accuracy in ESCC patients who had undergone neoadjuvant chemoradiotherapy (nCRT). This molecular model consisting of SCAT1, PRKAG2-AS1, and FLG-AS1 can accurately predict which patients will benefit from nCRT and obtain a complete pathological response. Such a signature is suitable for long-term treatment evaluation and may be the first step to individualized therapy of ESCC [77]. A study by Li et al. indicated that highly expressed lncRNA Rpph1 is a promising diagnostic biomarker of esophageal carcinoma. Its level has been associated with T stage, N stage, and clinical stage, as well as prognosis of patients’s outcome. Experiments carried out on cell lines proved that Rpph1 silencing can suppress tumor development and enhance radiosensitivity, underlining the tremendous potential of this molecule [78]. Many studies indicated that different lncRNA molecules have the potential ability to be applied as biomarkers in oncology and personalized radiotherapy.
A good biomarker should be simply obtained from diverse sources, it should be of high quantity and quality and the measurement methods should not be complicated. lncRNAs may be found in various sources such as tissues, cell lines, peripheral blood, serum, plasma, saliva, urine, exosomes, and FFPET [16,79,80,81,82,83,84]. However, not all are present in every type of biological material. For instance, MALAT1, MEG3, HULC, HOTAIR, UCA1, and NEAT1 were all previously found in OSCC tissues but in the saliva of the same patients, only MALAT1 and HOTAIR were detectable [16,83]. lncRNAs are also easily extracted from these samples; despite the lack of dedicated commercial kits for lncRNAs extraction, it can be done with a standard TRIzol method or ready-to-use commercial kits for RNA extraction. There is a lack of standardized extraction methods for lncRNAs, as well as storage recommendations or even reference genes for qRT-PCR [16]. However, some lncRNAs are more stable than miRNAs. Their half-life is approximately 16 h and they are resistant to RNase A digestion and room temperature incubation [16,84]. Most lncRNAs are present at low copy numbers, but the addition of polyA tails and annealing anchor dT adapters before cDNA synthesis solves the problem. This solution also enhances the specificity and sensitivity of lncRNA quantification [16,85]. Additionally, lncRNAs may be examined using many different methods such as immunoprecipitation, in situ hybridization, Au-NP assay (gold nanoparticle-based), Northern blot, high resolution melting (HRM), microarrays, next generation sequencing (NGS), and PCR methods (real-time PCR, droplet PCR) [16,79,80,86,87,88]. The choice of the procedure depends on the study (screening or specific detection), type of material, and costs [16]. lncRNAs show properties of good biomarkers for being easy to obtain, specific, and easy to measure. Even small challenges in working with lncRNAs are simple to overcome, making them very promising biomarkers.
Finding the appropriate biomarker of radiosensitivity/radioresistance or a molecule that modulates the response to this type of treatment may improve the process of selecting an accurate irradiation regimen. This type of personalized approach might significantly reduce the adverse effects associated with IR. It is well known that complications such as fibrosis in surrounding normal tissues, immune toxicity, and inflammation can substantially decrease the quality of patients’ life [89,90]. Designing a panel of biomarkers dedicated to individual cancers would provide the opportunity to propose to individuals sensitive to radiation a therapy that is less intensive than the standard treatment scheme. On the other hand, people with radiation-resistant tumors could benefit from a higher IR dose, more frequent applications, or, e.g., linear energy transfer (LET) radiation [91]. This type of therapy effectively eradicates cancer with the lowest biologically effective dose and the maximum reduction in toxicity. A study by Niemantsverdriet et al. proved that cell lines treated with high-LET radiation had significantly lower survival and higher apoptosis rates [91]. The method is not generally available because it is more expensive and complex than conventional therapy. The use of specific biomarkers would enable the process of qualifying patients for treatment strategies that will be the most beneficial.
Examples of known lncRNAs with diagnostic potential in cancers are summarized in Figure 2 and Table 1.
Table 1. Known lncRNAs affecting cell radiotherapy response.
Table 1. Known lncRNAs affecting cell radiotherapy response.
Gene NameCancer TypeExpressionImpact on RadiotherapyTargetsReference
AFAP-AS1Breast cancer (BC)UpregulatedAFAP-AS1 overexpression enhances radioresistance of BC cells (promoted cell proliferation, invasion, tumor growth, inhibits apoptosis)Wnt/β-catenin[92]
AGAP2-AS1Lung cancerUpregulatedPromotes the immunologic function after IRmiR-296/NOTCH2[93]
AHIFGlioblastoma (GBM)UpregulatedAHIF knockdown enhances radiosensitivityBax/Bcl-2[94]
ANRILColon cancerUpregulatedANRIL suppress radiosensitivity by binding to miR-181a-5p and reversing functions of chitooligosaccharides (COS)miR-181a-5p/
chitooligosaccharides (COS)
[44]
BLACAT1Head and neck squamous cell carcinoma (HNSCC)UpregulatedBLACAT1 knockdown enhances radiosensitivity of HNSCC cellsPSEN1[95]
CASC19Nasopharyngeal carcinoma (NPC)UpregulatedCASC19 contributes to the radioresistance of NPC cells by promotion of autophagy and inhibition of apoptosis through AMPK/mTOR signaling pathwayPARP1/AMPK/mTOR[51]
CASC2Non-small cell lung cancer (NSCLC)DownregulatedCASC2 overexpression induces radiosensitivity of NSCLC cellsPERK/CHOP[96]
Papillary Thyroid Cancer (PTC)DownregulatedLow expression of CASC2 causes high IR resistance of PTC cells; overexpression of CASC2 results in higher IR sensitivity of PTC cells (induced cell viability and inhibited post-IR apoptosis); CASC2 enhances radiosensitivity in PTC by sponging miR-155miR-155[97]
CCAT1Breast cancer (BC)UpregulatedDownregulation of CCAT1 enhances radiosensitivity through miR148b negative regulation (decreased colony formation rate, promoted apoptosis)miR-148b/miR-218/ZFX[92,98]
Non-small cell lung cancer (NSCLC)UpregulatedHigher CCAT1 expression correlates with higher radioresistance of NSCLC cells; downregulation of CCAT1 can improve the radiosensitivity of NSCLC cells by mediating cell cycle arrest, DNA damage, and apoptosisMAPK/MAPK1/ERK/
MEK
[33]
CCAT2Esophageal carcinoma (EC)UpregulatedCCAT2 knockdown results in radiosensitivity enhancement of EC cells (induced apoptosis); overexpressed CCAT2 causes EC cells to gain radioresistant features through inhibiting apoptosis via miR-145/p70S6K1 signaling pathways and by activating the Akt/ERK/p70S6K1 signaling pathwaysmiR-145/p70S6K1/p53/
c-Myc/Akt signaling pathway
[99]
CRNDEOvarian cancerUpregulatedCRNDE silencing resulted in enhanced radiosensitivity, inhibited clone formation and tumor growth in miceNo data[28]
CYTORNon-small cell lung cancer (NSCLC)UpregulatedSilencing CYTOR results in enhanced radiosensitivity of NSCLC cells (weak colony formation, high levels of H2AX); CYTOR binds to miR-206, silencing it and causing upregulation of PTMA, resulting in radioresistancemiR-206/PTMA[67]
Suppresses radiosensitivity through regulating malignant phenotypesmiR-195[74]
DGCR5Laryngeal carcinoma (LC)UpregulatedKnockdown could sensitize tumor cells to radiation through modulating miR-195miR-195[37]
Dio3osHead and neck squamous cell carcinoma (HNSCC)Downregulated after IRNo dataMYH7B/SRCAP/
HELZ2/NOS1/CROCC/CEP250/LPP/ABI2/
HERC2/RTEL1/
SMC1A1/HERC1/GAS7/NOTCH2/PKD1/
CFLAR/FAT3/FAT2/
CELSR3/CBL/NCOR2
[100]
DNM3OSEsophageal squamous cell carcinoma (ESCC)UpregulatedDecreased DNM3OS cause significantly increased radiosensitivity in vitro and in vivoCancer associated fibroblasts (CAFs)/PDGFB/
PDGFRB/FOXO1
[66]
GAS5Thyroid carcinoma (TC)DownregulatedGAS5 overexpression enhances radiosensitivitymiR-362-5p/SMG1[101]
Cervical cancer (CC)DownregulatedGAS5 overexpression enhances CC cells radiosensitivitymiR-106b/IER3[102]
Breast cancer (BC)Downregulated after IR further decrease in expression levelInduced overexpression reduces miR-21 expression leading to radiosensitizationmiR-21[31]
Esophageal squamous cell carcinoma (ESCC)DownregulatedSimultaneous upregulation with miR-21 knockdown improves radiosensitivity after IRmiR-21/RECK[41]
Prostate cancer (PCa)DownregulatedArtificially elevated level enhances α-solanine-induced radiosensitivitymiR18a[30]
Non-small cell lung cancer (NSCLC)DownregulatedEnhances radiosensitivitymiR-135b[103]
H19Cardiac carcinomaUpregulatedH19 knockdown resulted in enhanced radiosensitivity of cardiac carcinoma cellsmiR-130a-3p/miR-17-5p[104]
Hepatocellular carcinoma (HCC)DownregulatedH19 overexpression enhances radiosensitivity of HCC cells through the miR-193a-3p/PSEN1 axis (promoted apoptosis, inhibited DNA double-strand break repair)miR-193a-3p/PSEN1[105]
HAR1AHead and neck squamous cell carcinoma (HNSCC)Downregulated after IRNo dataRANBP2/LPP/ABI2/
HELZ/PHC3/HERC1/
MTO5A/FZD3/
CTNNA3/CBL/BMPR2/FAT3/CFLAR
[100]
HAR1BHead and neck squamous cell carcinoma (HNSCC)Downregulated after IRNo dataLPP/ABI2/RSF1/HERC2/HELZ/FZD3/CFLAR/
FAT3/FRK/FER/PDK1/
FAT1
[100]
HCP5Esophageal carcinoma
(EC)
UpregulatedKnockdown of HCP5 enhances radiosensitivity trough modulating the Akt signaling pathwaymiR-216a-3p/PDK1[26]
HMMR-AS1Glioblastoma (GBM)UpregulatedKnockdown may suppress and radiosensitize the tumormay regulate ERK1/2 by altering HMMR expression[34]
HOTAIRColorectal cancer (CRC)Upregulated; increases after IR in dose-dependent mannerInhibits radiotherapy efficacy through regulating miR-93/ATG12-mediated autophagymiR-93/ATG[39]
Colorectal cancer (CRC)UpregulatedDownregulation of HOTAIR enhanced radiosensitivity via reducing cell proliferation and invasivenessNo data[75]
Head and neck squamous cell carcinoma (HNSCC)Upregulated after IRHigher expression of HOTAIR is correlated with a higher resistance to radiotherapy in colon and breast cancer cell lines; high expression of HOTAIR is connected with the EMT process, maintaining of cancer initiating cells, and aggressive types of HNSCCLPP/ABI2/NOS1/
CFLAR/REL/FAT3/
PDK1/FZD3/SMAD2/
FRK/SRCAP/WNT2B/
CBL
[100]
Cervical cancer (CC)UpregulatedLeads to tumor radioresistance via increasing HIF1α expressionHIF1α[54]
Cervical cancer (CC)UpregulatedPromotes radioresistance through p21 inhibitionp21[76]
Breast cancer (BC)UpregulatedDownregulation of HOTAIR enhanced radiosensitivitymiR-218[106]
Breast cancer (BC)UpregulatedOverexpression of HOTAIR results in higher radioresistance of BC cells; HOTAIR overexpression enhances the cell proliferation and growth under irradiation stress; HOTAIR knockdown resulted in increased apoptosis and a reduced number of BC cells in the S phase of a cell cycle; its expression is positively correlated with DNA damage repair factorsHSPA1A/
miR-449b-5p/EZH2/PRC2/EED/
SUZ12/Ku70/Ku80/
DNA-Pk/ATM
[61,62]
HULCProstate cancer (PCa)Upregulated after IRHULC knockdown enhances sensitivity of PCa cell to IR; cell apoptosis and proliferation induced by IR are enhanced by HULC knockdown and decreased by HULC overexpressionBax/PCNA/cyclinD1/
caspase-3/Beclin-1/
p-4E-BP1
[50]
LINC00210OsteosarcomaUpregulatedKnockdown of LINC00210 results in enhanced radiosensitivity (after IR: decreased cell viability, induced apoptosis, inhibited levels of CyclinD1 and Bcl-2, increased levels of p21 and Bax); regulates radiosensitivity through the miR-342-3p/GFRA1 axismiR-342-3p/GFRA1/
CyclinD1/Bcl-2/p21/Bax
[45]
LINC00312Nasopharyngeal carcinomaDownregulatedOverexpression suppresses radiotherapy resistanceRAD50/MRE11/NBS1/
Ku80
[57]
LINC00473Esophageal squamous cell carcinoma (ESCC)UpregulatedReduces radiotherapy effectiveness increasing cancer proliferative abilitymiR-497-5p/CDC25A, miR-374a-5p/SPIN1[35,36]
Head and neck squamous cell carcinoma (HNSCC)UpregulatedLINC00473 knockdown enhances radiosensitivity of HNSCC cellsBax/Bcl2/Wnt/β-catenin pathway[107]
LINC00504Breast cancer (BC)UpregulatedDecreases cell radiosensitivity by regulating CPEB2 expressionTAF15/CPEB2[59]
LINC00511Thyroid carcinoma
(TC)
UpregulatedPotentiates resistance to radiotherapy by modulating the TAF1/JAK2/STAT3 axisTAF1/JAK2/STAT3[108]
Breast cancer (BC)UpregulatedLINC00511 knockdown enhances radiosensitivity (restricts cell proliferation, promotes apoptosis, and inhibits tumor growth)STXBP4/miR-185[92]
LINC00518Cutaneous malignant melanoma (CMM)UpregulatedPotentiates radioresistance through enhancing glycolytic metabolismmiR-33a-3p/HIF1α/
LDHA
[98]
LINC00630Colorectal cancer (CRC)UpregulatedSilencing could increase radiosensitivity by epigenetically repress BEX1 expressionEZH2/BEX1[38]
LINC00963Breast cancer (BC)UpregulatedHighly expressed LINC00963 causes BC cells to enhance radioresistance; its silencing results in an increase of radiosensitivity (restrains cell proliferation, impairs colony formation and tumor growth, arrest cells at the G0/G1 phase, stimulates apoptosis); LINC00963 induced radiosensitivity through the miR-324-3p/ACK1 axismiR-324-3p/ACK1/
CDK6/p27/CyclinD1
[109]
LINC01123GliomaUpregulatedEnhances radioresistance by creating the LINC01123/miR-151a/CENPB axismiR-151a/CENPB[110]
LINC01134Hepatocellular carcinoma (HC)UpregulatedAugments resistance to radiotherapy via modulating the MAPK1 signaling pathwaymiR-342-3p/IGF2BP2/
MAPK1
[55]
LINC01447Low-grade GliomaUpregulatedLINC01447 inhibition results in radiosensitivity enhancement in low-grade glioma cells (decreased cell viability, inhibited colony formation, increased apoptosis)No data[111]
LINC01977Non-small cell lung cancer (NSCLC)UpregulatedLINC01977 inhibition results in enhanced radiosensitivity in NSCLC cells (reduced colony formation, higher expression of H2AX)No data[112]
LINC02582Breast cancer (BC)UpregulatedPromotes radioresistance via the USP7/CHK1 signaling axisUSP7/CHK1[60]
LINC-PINTNasopharyngeal carcinoma (NPC)DownregulatedArtificial upregulation potentiates radiosensitivity through an increase in apoptosis rateATM/ATR-Chk1/Chk2 pathway and DNA-PKcs[56]
linc-RA1GliomaUpregulatedStrengthens radioresistance via inhibiting autophagy activationH2Bub1/USP44[49]
lincRNA-p21Colorectal cancer (CRC)Downregulated IR cause further decrease in expression levelEnhances radiosensitivity after IR through activating pro-apoptotic mechanismsWnt/β-catenin/c-myc and cyclin D1 axis, Noxa[40,113]
LINP1Cervical cancer (CC)UpregulatedAugments radioresistance via enhancing dsDNA break repair through the NHEJ pathwayKu80, DNA-PKcs[64]
Triple negative breast cancer (TNBC)[63]
LUCAT1Breast cancer (BC)UpregulatedLUCAT1 knockdown results in enhanced radiosensitivity of BC cells through the miR-181a-5p/KLF6/KLF15 axis (reduced cell proliferation, migration, viability, and invasion)miR-181a-5p/KLF6/
KLF15
[114]
MAGI2-AS3Esophageal squamous cell carcinoma (ESCC)DownregulatedMAGI2-AS3 silencing strengthens resistance of ESCC cells to IR in vivoHOXB7/EZH2/
miR-374b-5p/
CCDC19/miR-15b-5p
[43]
MALAT1Colorectal cancer (CRC)UpregulatedSilencing may increase radiosensitivity through the YAP1/AKT axisANKHD1/YAP1/AKT[58]
Nasopharyngeal carcinoma (NPC)Downregulation strengthens IR effectsmiR-1/slug[69]
High-risk HPV-positive cervical cancer (HR-HPV+ CC)Increases radioresistance through negatively regulating miR-145miR-145[32]
MEG3Thyroid carcinoma
(TC)
DownregulatedMEG3 overexpression results in higher TC cells radiosensitivity (inhibited proliferation, promoted apoptosis, and DNA damage) through miR-182 spongingmiR-182[115]
NEAT1Colorectal cancer (CRC)Upregulated; increases after IR in time-dependent mannerAugments radioresistance by promoting IR-induced pyroptosismiR-448/GSDME[46]
Triple negative breast cancer (TNBC)UpregulatedKnockdown improves cell sensitivity to radiation via positive regulation of NQO1NQO1/miR-218[70,92]
Nasopharyngeal carcinoma (NPC)UpregulatedNEAT1 downregulation sensitizes NPC cells to radiationmiR-204/ZEB1[116]
Hepatocellular carcinoma (HCC)UpregulatedNEAT1_2 down-regulation enhances radiosensitivity of HCC cellsmiR-101-3p/WEE1[117]
NKILALaryngeal carcinoma (LC)DownregulatedOverexpression of NKILA reduces radioresistance of LC cells by inhibiting p65 nuclear translocation (suppresses cell viability, DNA synthesis capability, and migration ability)p65[118]
OIP5-AS1Colorectal cancer (CRC)DownregulatedOverexpressed OIP5-AS1 impedes cell viability, promotes radio-induced apoptosis, and enhances radiosensitivity of CRC cells through the miR-369-3p/DYRK1A axismiR-369-3p/DYRK1A[119]
PCAT1Non-small cell lung cancer (NSCLC)UpregulatedInhibition of PCAT1/SOX2 together with radiation promotes IR-induced anti-tumor immune responsesSOX2/cGAS/STING[120]
Cervical cancer (CC)UpregulatedPCAT1 knockdown enhances radiosensitivity of CC cells through regulating the miR-128/GOLM1 axis (inhibited cell proliferation, migration, and invasion)miR-128/GOLM1[121]
PCAT6Breast cancer (BC)UpregulatedPCAT6 knockdown results in enhanced radiosensitivity (reduced cell proliferation, promoted apoptosis)TPD52/miR-185-5p[92]
PTPRG-AS1Non-small cell lung cancer (NSCLC)UpregulatedDiminishes radiotherapy efficacy by modulating miR-200c-3p/TCF4miR-200c-3p/TCF4[122]
Nasopharyngeal carcinoma (NPC)Silencing leads to significant improvement of radiosensitivitymiR-194-3p/PRC1[73]
PTENP1Head and neck squamous cell carcinoma (HNSCC)Upregulated after IRInhibition of miR-21 causes cell radiosensitivity by increasing the PTEN protein expression in HNSCCmiR-21[100]
PVT1Non-small cell lung cancer (NSCLC)Upregulated; the highest levels reached under hypoxiaInduced downregulation could weaken radioresistance by reducing hypoxiaHIF1α/miR-199a-5p[53]
Nasopharyngeal carcinoma (NPC)UpregulatedInduces radioresistance; knockdown of PVT1 enhances the radiosensitivity of NPC cell linesNo data[42]
RBM5-AS1Medulloblastoma (MB)Upregulated; the highest levels in radioresistant medulloblastoma cellsPromotes radioresistance and cancer stemnessSIRT6[68]
SBF2-AS1Non-small cell lung cancer (NSCLC)UpregulatedDownregulation of SBF2-AS1 enhances NSCLC cells’ radiosensitivity through the miR-302a/MBNL3 axis (inhibited cell proliferation, enhanced apoptosis, reduced tumor growth in mice)miR-302a/MBNL3[123]
SLC25A21-AS1Gastric cancer (GC)DownregulatedOverexpression of SLC25A21-AS1 enhances the radiosensitivity and inhibits the malignant behaviors of GC cells by upregulating the miR-15a-5p/SNCG axismiR-15a-5p/SNCG[124]
SNHG5Head and neck squamous cell carcinoma (HNSCC)Downregulated after IRNo dataLPP/ABI2/HELZ/
RANBP2/CEP250/
CDK6/HERC1/PHC3/
MYO5A/FZD3/CFLAR/FAT3/FAT1/FRK/
SMAD2/BMPR2/PTEN/ZMAT3/MDM4/FAT2/APC/FER
[100]
SNHG7Thyroid carcinoma
(TC)
UpregulatedInduced depletion may diminish radioactive iodine resistance through the PI3K/Akt pathwaymiR-9-5p/DPP4/PI3K/
Akt
[25]
SNHG18GliomaUpregulatedPotentiates radioresistance via modulating levels of DNA damage response proteinsSema5A[65]
TP53TG1GliomaUpregulatedInduced downregulation could lead to radiation-mediated cancer growth inhibitionmiR-524-5p/RAB5A[48]
TP73-AS1Hepatocellular carcinoma (HCC)UpregulatedTP73-AS1 knockdown results in radiosensitivity enhancement of HCC cells through the PTEN/Akt signaling pathway (reduced proliferation, reduced colony formation ability, and induced apoptosis)PTEN[125]
TTN-AS1Large intestine cancerUpregulatedTTN-AS1 knockdown resulted in radiosensitivity enhancement in large intestine cancer cellsmiR-134-5p/PAK3[126]
UCA1Prostate cancer (PCa)UpregulatedUCA1 knockdown enhances radiosensitivity of PCa cellsmiR-331-3p/EIF4G1[127]
XISTNeuroblastoma (NB)UpregulatedHigher XIST expression is correlated with higher NB cells’ radioresistance; its silencing results in inhibition of cell cycle progression, cell proliferation, colony formation, and enhanced post-IR apoptosis rate; it regulates IT through the miR-375/L1CAM axismiR-375/L1CAM[128]

4. Conclusions and Future Directions

Over time, long non-coding RNAs, despite their lack of protein-coding ability, have proven to be essential molecules in carcinogenesis but also the subsequent functioning of cancer cells. Their expression directly affects mRNAs, miRNAs, and other lncRNAs. Moreover, they have the ability to regulate transcription factors and chromatin modifications, as well as influence the chromatin condensation state. Due to little knowledge about them and the continuous analysis of their activity in particular cell types, the molecular mechanisms of tumorigenesis are still being explored. However, there are hopes that they will be important diagnostic tools in the future, including in assessing the success of radiotherapy, which is the most common treatment method among cancer patients. lncRNAs regulate the crucial processes implicated in radioresistance/radiosensitivity. It is known that these molecules are implicated in DNA damage response and repair mechanisms, regulation of HIF1α and ROS levels, mediation of cell death, autophagy, cell cycle, and proliferation. Many reports indicated potential uses of lncRNAs in the personalization of radiotherapy, but these studies were carried out on small groups of patients and were not randomized. A great opportunity is seen in the analysis of data from the Cancer Genome Atlas (TCGA) project and the use of AI. Big data analysis gives the possibility of selecting proper lncRNAs, which can be used for validation based on a larger group of patients. This approach potentially will give the ability to clinically use lncRNA-based panels for personalization of radiotherapy.

Author Contributions

Conceptualization, T.K. and K.L.; methodology, J.K.-M.; K.G. and T.K; investigation, J.K.-M., K.G., A.P., T.K. and M.P.; data curation, J.K.-M., K.G., A.P. and T.K.; writing—original draft preparation, J.K.-M. and K.G.; writing—review and editing, J.K.-M., K.G., A.P., T.K., M.P., A.T. and R.B.; Visualization, K.G. and T.K.; Supervision, T.K. and K.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Greater Poland Cancer Center (internal grant No.: 11/2021(247)) and supported by Adam Mickiewicz University, Poznań, Poland; J.K-M.was supported by the grant PRELUDIUM 20 from the National Science Center, Poland, allocated on the basis of the decision No.: 2021/41/N/NZ5/02966; K.G. has received a scholarship writing this manuscript from the European Union POWER PhD program.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data are available online with common access.

Acknowledgments

We would like to thank all people from the Greater Poland Cancer Center and Adam Mickiewicz University for helping and supporting our work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cabrera-Licona, A.; Pérez-Añorve, I.X.; Flores-Fortis, M.; Moral-Hernández, O.D.; González-de la Rosa, C.H.; Suárez-Sánchez, R.; Chávez-Saldaña, M.; Aréchaga-Ocampo, E. Deciphering the epigenetic network in cancer radioresistance. Radiother. Oncol. 2021, 159, 48–59. [Google Scholar] [CrossRef]
  2. Smits, K.M.; Melotte, V.; Niessen, H.E.; Dubois, L.; Oberije, C.; Troost, E.G.; Starmans, M.H.; Boutros, P.C.; Vooijs, M.; van Engeland, M.; et al. Epigenetics in radiotherapy: Where are we heading? Radiother. Oncol. 2014, 111, 168–177. [Google Scholar] [CrossRef]
  3. Story, M.D.; Durante, M. Radiogenomics. Med. Phys. 2018, 45, e1111–e1122. [Google Scholar] [CrossRef] [PubMed]
  4. Ouellette, M.M.; Zhou, S.; Yan, Y. Cell Signaling Pathways That Promote Radioresistance of Cancer Cells. Diagnostics 2022, 12, 656. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, Y.; Han, Y.; Jin, Y.; He, Q.; Wang, Z. The Advances in Epigenetics for Cancer Radiotherapy. Int. J. Mol. Sci. 2022, 23, 5654. [Google Scholar] [CrossRef] [PubMed]
  6. Chan, J.J.; Tay, Y. Noncoding RNA:RNA Regulatory Networks in Cancer. Int. J. Mol. Sci. 2018, 19, 1310. [Google Scholar] [CrossRef]
  7. Podralska, M.; Ciesielska, S.; Kluiver, J.; van den Berg, A.; Dzikiewicz-Krawczyk, A.; Slezak-Prochazka, I. Non-Coding RNAs in Cancer Radiosensitivity: MicroRNAs and lncRNAs as Regulators of Radiation-Induced Signaling Pathways. Cancers 2020, 12, 1662. [Google Scholar] [CrossRef]
  8. Schofield, P.N.; Kulka, U.; Tapio, S.; Grosche, B. Big data in radiation biology and epidemiology; an overview of the historical and contemporary landscape of data and biomaterial archives. Int. J. Radiat. Biol. 2019, 95, 861–878. [Google Scholar] [CrossRef]
  9. De Mey, S.; Dufait, I.; De Ridder, M. Radioresistance of Human Cancers: Clinical Implications of Genetic Expression Signatures. Front. Oncol. 2021, 11, 761901. [Google Scholar] [CrossRef]
  10. Coates, J.; Souhami, L.; El Naqa, I. Big Data Analytics for Prostate Radiotherapy. Front. Oncol. 2016, 6, 149. [Google Scholar] [CrossRef] [Green Version]
  11. Kozłowska, J.; Kolenda, T.; Poter, P.; Sobocińska, J.; Guglas, K.; Stasiak, M.; Bliźniak, R.; Teresiak, A.; Lamperska, K. Long Intergenic Non-Coding RNAs in HNSCC: From “Junk DNA” to Important Prognostic Factor. Cancers 2021, 13, 2949. [Google Scholar] [CrossRef] [PubMed]
  12. Stasiak, M.; Kolenda, T.; Kozłowska-Masłoń, J.; Sobocińska, J.; Poter, P.; Guglas, K.; Paszkowska, A.; Bliźniak, R.; Teresiak, A.; Kazimierczak, U.; et al. The World of Pseudogenes: New Diagnostic and Therapeutic Targets in Cancers or Still Mystery Molecules? Life 2021, 11, 1354. [Google Scholar] [CrossRef] [PubMed]
  13. Mercer, T.R.; Dinger, M.E.; Mattick, J.S. Long non-coding RNAs: Insights into functions. Nat. Rev. Genet. 2009, 10, 155–159. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, L.L. Linking Long Noncoding RNA Localization and Function. Trends Biochem. Sci. 2016, 41, 761–772. [Google Scholar] [CrossRef]
  15. Ransohoff, J.D.; Wei, Y.; Khavari, P.A. The functions and unique features of long intergenic non-coding RNA. Nat. Rev. Mol. Cell Biol. 2018, 19, 143–157. [Google Scholar] [CrossRef]
  16. Guglas, K.; Bogaczyńska, M.; Kolenda, T.; Ryś, M.; Teresiak, A.; Bliźniak, R.; Łasińska, I.; Mackiewicz, J.; Lamperska, K. lncRNA in HNSCC: Challenges and potential. Contemp. Oncol. 2017, 21, 259–266. [Google Scholar] [CrossRef]
  17. Kelley, D.; Rinn, J. Transposable elements reveal a stem cell-specific class of long noncoding RNAs. Genome Biol. 2012, 13, R107. [Google Scholar] [CrossRef]
  18. Hoffmann, M.J.; Dehn, J.; Droop, J.; Niegisch, G.; Niedworok, C.; Szarvas, T.; Schulz, W.A. Truncated Isoforms of lncRNA ANRIL Are Overexpressed in Bladder Cancer, But Do Not Contribute to Repression of INK4 Tumor Suppressors. Noncoding RNA 2015, 1, 266–284. [Google Scholar] [CrossRef]
  19. Smith, K.N.; Miller, S.C.; Varani, G.; Calabrese, J.M.; Magnuson, T. Multimodal Long Noncoding RNA Interaction Networks: Control Panels for Cell Fate Specification. Genetics 2019, 213, 1093–1110. [Google Scholar] [CrossRef]
  20. Li, Y.; Ge, C.; Feng, G.; Xiao, H.; Dong, J.; Zhu, C.; Jiang, M.; Cui, M.; Fan, S. Low dose irradiation facilitates hepatocellular carcinoma genesis involving HULC. Mol. Carcinog. 2018, 57, 926–935. [Google Scholar] [CrossRef]
  21. Aryankalayil, M.J.; Chopra, S.; Levin, J.; Eke, I.; Makinde, A.; Das, S.; Shankavaram, U.; Vanpouille-Box, C.; Demaria, S.; Coleman, C.N. Radiation-Induced Long Noncoding RNAs in a Mouse Model after Whole-Body Irradiation. Radiat. Res. 2018, 189, 251–263. [Google Scholar] [CrossRef] [PubMed]
  22. Wei, J.; Wang, B.; Wang, H.; Meng, L.; Zhao, Q.; Li, X.; Xin, Y.; Jiang, X. Radiation-Induced Normal Tissue Damage: Oxidative Stress and Epigenetic Mechanisms. Oxid. Med. Cell. Longev. 2019, 2019, 3010342. [Google Scholar] [CrossRef] [PubMed]
  23. Zhu, J.; Chen, S.; Yang, B.; Mao, W.; Yang, X.; Cai, J. Molecular mechanisms of lncRNAs in regulating cancer cell radiosensitivity. Biosci. Rep. 2019, 39, BSR20190590. [Google Scholar] [CrossRef]
  24. Zhou, J.M.; Liang, R.; Zhu, S.Y.; Wang, H.; Zou, M.; Zou, W.J.; Nie, S.L. LncRNA WWC2-AS1 functions AS a novel competing endogenous RNA in the regulation of FGF2 expression by sponging miR-16 in radiation-induced intestinal fibrosis. BMC Cancer 2019, 19, 647. [Google Scholar] [CrossRef] [PubMed]
  25. Chen, W.; Yu, J.; Xie, R.; Zhou, T.; Xiong, C.; Zhang, S.; Zhong, M. Roles of the SNHG7/microRNA-9-5p/DPP4 ceRNA network in the growth and 131I resistance of thyroid carcinoma cells through PI3K/Akt activation. Oncol. Rep. 2021, 45, 3. [Google Scholar] [CrossRef] [PubMed]
  26. Guo, Y.; Wang, L.; Yang, H.; Ding, N. Knockdown long non-coding RNA HCP5 enhances the radiosensitivity of esophageal carcinoma by modulating AKT signaling activation. Bioengineered 2022, 13, 884–893. [Google Scholar] [CrossRef]
  27. Wang, Y.Q.; Huang, G.; Chen, J.; Cao, H.; Xu, W.T. LncRNA SNHG6 promotes breast cancer progression and epithelial-mesenchymal transition via miR-543/LAMC1 axis. Breast Cancer Res. Treat. 2021, 188, 1–14. [Google Scholar] [CrossRef]
  28. Yang, W.; Li, X.; Zhao, L.; Zhao, F. Reversal of Radiotherapy Resistance of Ovarian Cancer Cell Strain CAOV3/R by Targeting lncRNA CRNDE. J. Healthc. Eng. 2021, 2021, 8556965. [Google Scholar] [CrossRef]
  29. Fotouhi Ghiam, A.; Taeb, S.; Huang, X.; Huang, V.; Ray, J.; Scarcello, S.; Hoey, C.; Jahangiri, S.; Fokas, E.; Loblaw, A.; et al. Long non-coding RNA urothelial carcinoma associated 1 (UCA1) mediates radiation response in prostate cancer. Oncotarget 2017, 8, 4668–4689. [Google Scholar] [CrossRef]
  30. Yang, J.; Hao, T.; Sun, J.; Wei, P.; Zhang, H. Long noncoding RNA GAS5 modulates α-Solanine-induced radiosensitivity by negatively regulating miR-18a in human prostate cancer cells. Biomed. Pharmacother. 2019, 112, 108656. [Google Scholar] [CrossRef]
  31. Ma, Y.; Yu, L.; Yan, W.; Qiu, L.; Zhang, J.; Jia, X. lncRNA GAS5 Sensitizes Breast Cancer Cells to Ionizing Radiation by Inhibiting DNA Repair. Biomed. Res. Int. 2022, 2022, 1987519. [Google Scholar] [CrossRef] [PubMed]
  32. Lu, H.; He, Y.; Lin, L.; Qi, Z.; Ma, L.; Li, L.; Su, Y. Long non-coding RNA MALAT1 modulates radiosensitivity of HR-HPV+ cervical cancer via sponging miR-145. Tumour Biol. 2016, 37, 1683–1691. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Z. LncRNA CCAT1 downregulation increases the radiosensitivity of non-small cell lung cancer cells. Kaohsiung J. Med. Sci. 2021, 37, 654–663. [Google Scholar] [CrossRef] [PubMed]
  34. Li, J.; Ji, X.; Wang, H. Targeting Long Noncoding RNA HMMR-AS1 Suppresses and Radiosensitizes Glioblastoma. Neoplasia 2018, 20, 456–466. [Google Scholar] [CrossRef]
  35. Liu, W.H.; Qiao, H.Y.; Xu, J.; Wang, W.Q.; Wu, Y.L.; Wu, X. LINC00473 contributes to the radioresistance of esophageal squamous cell carcinoma by regulating microRNA-497-5p and cell division cycle 25A. Int. J. Mol. Med. 2020, 46, 571–582. [Google Scholar] [CrossRef]
  36. Chen, W.; Zhang, Y.; Wang, H.; Pan, T.; Zhang, Y.; Li, C. LINC00473/miR-374a-5p regulates esophageal squamous cell carcinoma via targeting SPIN1 to weaken the effect of radiotherapy. J. Cell. Biochem. 2019, 120, 14562–14572. [Google Scholar] [CrossRef]
  37. Tang, T.; Shan, G.; Zeng, F. Knockdown of DGCR5 enhances the radiosensitivity of human laryngeal carcinoma cells via inducing miR-195. J. Cell. Physiol. 2019, 234, 12918–12925. [Google Scholar] [CrossRef]
  38. Liu, F.; Huang, W.; Hong, J.; Cai, C.; Zhang, W.; Zhang, J.; Kang, Z. Long noncoding RNA LINC00630 promotes radio-resistance by regulating BEX1 gene methylation in colorectal cancer cells. IUBMB Life 2020, 72, 1404–1414. [Google Scholar] [CrossRef]
  39. Liu, Y.; Chen, X.; Chen, X.; Liu, J.; Gu, H.; Fan, R.; Ge, H. Long non-coding RNA HOTAIR knockdown enhances radiosensitivity through regulating microRNA-93/ATG12 axis in colorectal cancer. Cell Death Dis. 2020, 11, 175. [Google Scholar] [CrossRef]
  40. Chen, L.; Yuan, D.; Yang, Y.; Ren, M. LincRNA-p21 enhances the sensitivity of radiotherapy for gastric cancer by targeting the β-catenin signaling pathway. J. Cell. Biochem. 2019, 120, 6178–6187. [Google Scholar] [CrossRef]
  41. Lin, J.; Liu, Z.; Liao, S.; Li, E.; Wu, X.; Zeng, W. Elevation of long non-coding RNA GAS5 and knockdown of microRNA-21 up-regulate RECK expression to enhance esophageal squamous cell carcinoma cell radio-sensitivity after radiotherapy. Genomics 2020, 112, 2173–2185. [Google Scholar] [CrossRef] [PubMed]
  42. He, Y.; Jing, Y.; Wei, F.; Tang, Y.; Yang, L.; Luo, J.; Yang, P.; Ni, Q.; Pang, J.; Liao, Q.; et al. Long non-coding RNA PVT1 predicts poor prognosis and induces radioresistance by regulating DNA repair and cell apoptosis in nasopharyngeal carcinoma. Cell Death Dis. 2018, 9, 235. [Google Scholar] [CrossRef] [PubMed]
  43. Cheng, W.; Shi, X.; Lin, M.; Yao, Q.; Ma, J.; Li, J. LncRNA MAGI2-AS3 Overexpression Sensitizes Esophageal Cancer Cells to Irradiation Through Down-Regulation of HOXB7 via EZH2. Front. Cell Dev. Biol. 2020, 8, 552822. [Google Scholar] [CrossRef] [PubMed]
  44. Sun, C.; Shen, C.; Zhang, Y.; Hu, C. LncRNA ANRIL negatively regulated chitooligosaccharide-induced radiosensitivity in colon cancer cells by sponging miR-181a-5p. Adv. Clin. Exp. Med. 2021, 30, 55–65. [Google Scholar] [CrossRef]
  45. He, P.; Xu, Y.Q.; Wang, Z.J.; Sheng, B. LncRNA LINC00210 regulated radiosensitivity of osteosarcoma cells via miR-342-3p/GFRA1 axis. J. Clin. Lab. Anal. 2020, 34, e23540. [Google Scholar] [CrossRef]
  46. Su, F.; Duan, J.; Zhu, J.; Fu, H.; Zheng, X.; Ge, C. Long non-coding RNA nuclear paraspeckle assembly transcript 1 regulates ionizing radiation-induced pyroptosis via microRNA-448/gasdermin E in colorectal cancer cells. Int. J. Oncol. 2021, 59, 79. [Google Scholar] [CrossRef]
  47. Jiang, A.; Liu, N.; Bai, S.; Wang, J.; Gao, H.; Zheng, X.; Fu, X.; Ren, M.; Zhang, X.; Tian, T.; et al. Identification and validation of an autophagy-related long non-coding RNA signature as a prognostic biomarker for patients with lung adenocarcinoma. J. Thorac. Dis. 2021, 13, 720–734. [Google Scholar] [CrossRef]
  48. Gao, W.; Qiao, M.; Luo, K. Long Noncoding RNA TP53TG1 Contributes to Radioresistance of Glioma Cells Via miR-524-5p/RAB5A Axis. Cancer Biother. Radiopharm. 2021, 36, 600–612. [Google Scholar] [CrossRef]
  49. Zheng, J.; Wang, B.; Zheng, R.; Zhang, J.; Huang, C.; Zheng, R.; Huang, Z.; Qiu, W.; Liu, M.; Yang, K.; et al. Linc-RA1 inhibits autophagy and promotes radioresistance by preventing H2Bub1/USP44 combination in glioma cells. Cell Death Dis. 2020, 11, 758. [Google Scholar] [CrossRef]
  50. Chen, C.; Wang, K.; Wang, Q.; Wang, X. LncRNA HULC mediates radioresistance via autophagy in prostate cancer cells. Braz. J. Med. Biol. Res. 2018, 51, e7080. [Google Scholar] [CrossRef] [Green Version]
  51. Liu, H.; Zheng, W.; Chen, Q.; Zhou, Y.; Pan, Y.; Zhang, J.; Bai, Y.; Shao, C. lncRNA CASC19 Contributes to Radioresistance of Nasopharyngeal Carcinoma by Promoting Autophagy via AMPK-mTOR Pathway. Int. J. Mol. Sci. 2021, 22, 1407. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, F.; Zhu, L.; Xue, Q.; Tang, C.; Tang, W.; Zhang, N.; Dai, C.; Chen, Z. Novel lncRNA AL033381.2 Promotes Hepatocellular Carcinoma Progression by Upregulating PRKRA Expression. Oxid. Med. Cell. Longev. 2022, 2022, 1125932. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, C.; Han, C.; Zhang, Y.; Liu, F. LncRNA PVT1 regulate expression of HIF1α via functioning as ceRNA for miR-199a-5p in non-small cell lung cancer under hypoxia. Mol. Med. Rep. 2018, 17, 1105–1110. [Google Scholar] [CrossRef] [PubMed]
  54. Li, N.; Meng, D.D.; Gao, L.; Xu, Y.; Liu, P.J.; Tian, Y.W.; Yi, Z.Y.; Zhang, Y.; Tie, X.J.; Xu, Z.Q. Overexpression of HOTAIR leads to radioresistance of human cervical cancer via promoting HIF-1α expression. Radiat. Oncol. 2018, 13, 210. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, Z.; Wang, X.; Rong, Z.; Dai, L.; Qin, C.; Wang, S.; Geng, W. LncRNA LINC01134 Contributes to Radioresistance in Hepatocellular Carcinoma by Regulating DNA Damage Response viaMAPK Signaling Pathway. Front. Pharmacol. 2022, 12, 791889. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, Y.H.; Guo, Z.; An, L.; Zhou, Y.; Xu, H.; Xiong, J.; Liu, Z.Q.; Chen, X.P.; Zhou, H.H.; Li, X.; et al. LINC-PINT impedes DNA repair and enhances radiotherapeutic response by targeting DNA-PKcs in nasopharyngeal cancer. Cell Death Dis. 2021, 12, 454. [Google Scholar] [CrossRef] [PubMed]
  57. Guo, Z.; Wang, Y.H.; Xu, H.; Yuan, C.S.; Zhou, H.H.; Huang, W.H.; Wang, H.; Zhang, W. LncRNA linc00312 suppresses radiotherapy resistance by targeting DNA-PKcs and impairing DNA damage repair in nasopharyngeal carcinoma. Cell Death Dis. 2021, 12, 69. [Google Scholar] [CrossRef]
  58. Yao, P.A.; Wu, Y.; Zhao, K.; Li, Y.; Cao, J.; Xing, C. The feedback loop of ANKHD1/lncRNA MALAT1/YAP1 strengthens the radioresistance of CRC by activating YAP1/AKT signaling. Cell Death Dis. 2022, 13, 103. [Google Scholar] [CrossRef]
  59. Feng, J.; Li, Y.; Zhu, L.; Zhao, Q.; Li, D.; Li, Y.; Wu, T. STAT1 mediated long non-coding RNA LINC00504 influences radio-sensitivity of breast cancer via binding to TAF15 and stabilizing CPEB2 expression. Cancer Biol. Ther. 2021, 22, 630–639. [Google Scholar] [CrossRef]
  60. Wang, B.; Zheng, J.; Li, R.; Tian, Y.; Lin, J.; Liang, Y.; Sun, Q.; Xu, A.; Zheng, R.; Liu, M.; et al. Long noncoding RNA LINC02582 acts downstream of miR-200c to promote radioresistance through CHK1 in breast cancer cells. Cell Death Dis. 2019, 10, 764. [Google Scholar] [CrossRef] [Green Version]
  61. Zhang, S.; Wang, B.; Xiao, H.; Dong, J.; Li, Y.; Zhu, C.; Jin, Y.; Li, H.; Cui, M.; Fan, S. LncRNA HOTAIR enhances breast cancer radioresistance through facilitating HSPA1A expression via sequestering miR-449b-5p. Thorac. Cancer 2020, 11, 1801–1816. [Google Scholar] [CrossRef] [PubMed]
  62. Qian, L.; Fei, Q.; Zhang, H.; Qiu, M.; Zhang, B.; Wang, Q.; Yu, Y.; Guo, C.; Ren, Y.; Mei, M.; et al. lncRNA HOTAIR Promotes DNA Repair and Radioresistance of Breast Cancer via EZH2. DNA Cell Biol. 2020, 39, 2166–2173. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, Y.; He, Q.; Hu, Z.; Feng, Y.; Fan, L.; Tang, Z.; Yuan, J.; Shan, W.; Li, C.; Hu, X.; et al. Long noncoding RNA LINP1 regulates repair of DNA double-strand breaks in triple-negative breast cancer. Nat. Struct Mol. Biol. 2016, 23, 522–530. [Google Scholar] [CrossRef]
  64. Wang, X.; Liu, H.; Shi, L.; Yu, X.; Gu, Y.; Sun, X. LINP1 facilitates DNA damage repair through non-homologous end joining (NHEJ) pathway and subsequently decreases the sensitivity of cervical cancer cells to ionizing radiation. Cell Cycle 2018, 17, 439–447. [Google Scholar] [CrossRef] [PubMed]
  65. Zheng, R.; Yao, Q.; Ren, C.; Liu, Y.; Yang, H.; Xie, G.; Du, S.; Yang, K.; Yuan, Y. Upregulation of Long Noncoding RNA Small Nucleolar RNA Host Gene 18 Promotes Radioresistance of Glioma by Repressing Semaphorin 5A. Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, 877–887. [Google Scholar] [CrossRef]
  66. Zhang, H.; Hua, Y.; Jiang, Z.; Yue, J.; Shi, M.; Zhen, X.; Zhang, X.; Yang, L.; Zhou, R.; Wu, S. Cancer-associated Fibroblast-promoted LncRNA DNM3OS Confers Radioresistance by Regulating DNA Damage Response in Esophageal Squamous Cell Carcinoma. Clin. Cancer Res. 2019, 25, 1989–2000. [Google Scholar] [CrossRef]
  67. Jiang, G.; Yu, H.; Li, Z.; Zhang, F. lncRNA cytoskeleton regulator reduces non-small cell lung cancer radiosensitivity by downregulating miRNA-206 and activating prothymosin α. Int. J. Oncol. 2021, 59, 88. [Google Scholar] [CrossRef]
  68. Zhu, C.; Li, K.; Jiang, M.; Chen, S. RBM5-AS1 promotes radioresistance in medulloblastoma through stabilization of SIRT6 protein. Acta Neuropathol. Commun. 2021, 9, 123. [Google Scholar] [CrossRef]
  69. Jin, C.; Yan, B.; Lu, Q.; Lin, Y.; Ma, L. The role of MALAT1/miR-1/slug axis on radioresistance in nasopharyngeal carcinoma. Tumour Biol. 2016, 37, 4025–4033. [Google Scholar] [CrossRef]
  70. Lin, L.C.; Lee, H.T.; Chien, P.J.; Huang, Y.H.; Chang, M.Y.; Lee, Y.C.; Chang, W.W. NAD(P)H:quinone oxidoreductase 1 determines radiosensitivity of triple negative breast cancer cells and is controlled by long non-coding RNA NEAT1. Int. J. Med. Sci. 2020, 17, 2214–2224. [Google Scholar] [CrossRef]
  71. Ye, H.; Zhou, Q.; Zheng, S.; Li, G.; Lin, Q.; Ye, L.; Wang, Y.; Wei, L.; Zhao, X.; Li, W.; et al. FEZF1-AS1/miR-107/ZNF312B axis facilitates progression and Warburg effect in pancreatic ductal adenocarcinoma. Cell Death Dis. 2018, 9, 34. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, Y.; He, D.; Xiao, M.; Zhu, Y.; Zhou, J.; Cao, K. Long noncoding RNA LINC00518 induces radioresistance by regulating glycolysis through an miR-33a-3p/HIF-1α negative feedback loop in melanoma. Cell Death Dis. 2021, 12, 245. [Google Scholar] [CrossRef] [PubMed]
  73. Yi, L.; Ouyang, L.; Wang, S.; Li, S.S.; Yang, X.M. Long noncoding RNA PTPRG-AS1 acts as a microRNA-194-3p sponge to regulate radiosensitivity and metastasis of nasopharyngeal carcinoma cells via PRC1. J Cell Physiol. 2019, 234, 19088–19102. [Google Scholar] [CrossRef]
  74. Zhang, J.; Li, W. Long noncoding RNA CYTOR sponges miR-195 to modulate proliferation, migration, invasion and radiosensitivity in nonsmall cell lung cancer cells. Biosci. Rep. 2018, 38, BSR20181599. [Google Scholar] [CrossRef] [PubMed]
  75. Yang, X.D.; Xu, H.T.; Xu, X.H.; Ru, G.; Liu, W.; Zhu, J.J.; Wu, Y.Y.; Zhao, K.; Wu, Y.; Xing, C.G.; et al. Knockdown of long non-coding RNA HOTAIR inhibits proliferation and invasiveness and improves radiosensitivity in colorectal cancer. Oncol. Rep. 2016, 35, 479–487. [Google Scholar] [CrossRef]
  76. Jing, L.; Yuan, W.; Ruofan, D.; Jinjin, Y.; Haifeng, Q. HOTAIR enhanced aggressive biological behaviors and induced radio-resistance via inhibiting p21 in cervical cancer. Tumour Biol. 2015, 36, 3611–3619. [Google Scholar] [CrossRef]
  77. Zhang, C.; Zhang, Z.; Zhang, G.; Xue, L.; Yang, H.; Luo, Y.; Zheng, X.; Zhang, Y.; Yuan, Y.; Lei, R.; et al. A three-lncRNA signature of pretreatment biopsies predicts pathological response and outcome in esophageal squamous cell carcinoma with neoadjuvant chemoradiotherapy. Clin. Transl. Med. 2020, 10, e156. [Google Scholar] [CrossRef]
  78. Li, Z.Y.; Li, H.F.; Zhang, Y.Y.; Zhang, X.L.; Wang, B.; Liu, J.T. Value of long non-coding RNA Rpph1 in esophageal cancer and its effect on cancer cell sensitivity to radiotherapy. World J. Gastroenterol. 2020, 26, 1775–1791. [Google Scholar] [CrossRef]
  79. Eissa, S.; Matboli, M.; Essawy, N.O.; Shehta, M.; Kotb, Y.M. Rapid detection of urinary long non-coding RNA urothelial carcinoma associated one using a PCR-free nanoparticle-based assay. Biomarkers 2015, 20, 212–217. [Google Scholar] [CrossRef]
  80. Zhang, H.; Zhao, L.; Wang, Y.X.; Xi, M.; Liu, S.L.; Luo, L.L. Long non-coding RNA HOTTIP is correlated with progression and prognosis in tongue squamous cell carcinoma. Tumour Biol. 2015, 36, 8805–8809. [Google Scholar] [CrossRef]
  81. Gezer, U.; Özgür, E.; Cetinkaya, M.; Isin, M.; Dalay, N. Long non-coding RNAs with low expression levels in cells are enriched in secreted exosomes. Cell Biol. Int. 2014, 38, 1076–1079. [Google Scholar] [CrossRef] [PubMed]
  82. Fayda, M.; Isin, M.; Tambas, M.; Guveli, M.; Meral, R.; Altun, M.; Sahin, D.; Ozkan, G.; Sanli, Y.; Isin, H.; et al. Do circulating long non-coding RNAs (lncRNAs) (LincRNA-p21, GAS 5, HOTAIR) predict the treatment response in patients with head and neck cancer treated with chemoradiotherapy? Tumour Biol. 2016, 37, 3969–3978. [Google Scholar] [CrossRef] [PubMed]
  83. Tang, H.; Wu, Z.; Zhang, J.; Su, B. Salivary lncRNA as a potential marker for oral squamous cell carcinoma diagnosis. Mol. Med. Rep. 2013, 7, 761–766. [Google Scholar] [CrossRef] [PubMed]
  84. Zhou, X.; Yin, C.; Dang, Y.; Ye, F.; Zhang, G. Identification of the long non-coding RNA H19 in plasma as a novel biomarker for diagnosis of gastric cancer. Sci. Rep. 2015, 5, 11516. [Google Scholar] [CrossRef]
  85. Fang, Z.; Zhang, S.; Wang, Y.; Shen, S.; Wang, F.; Hao, Y.; Li, Y.; Zhang, B.; Zhou, Y.; Yang, H.; et al. Long non-coding RNA MALAT-1 modulates metastatic potential of tongue squamous cell carcinomas partially through the regulation of small proline rich proteins. BMC Cancer 2016, 16, 706. [Google Scholar] [CrossRef]
  86. Feng, Y.; Hu, X.; Zhang, Y.; Zhang, D.; Li, C.; Zhang, L. Methods for the study of long noncoding RNA in cancer cell signaling. Methods Mol. Biol. 2014, 1165, 115–143. [Google Scholar]
  87. Wojdacz, T.K.; Dobrovic, A.; Algar, E.M. Rapid detection of methylation change at H19 in human imprinting disorders using methylation-sensitive high-resolution melting. Hum. Mutat. 2008, 29, 1255–1260. [Google Scholar] [CrossRef]
  88. Dodd, D.W.; Gagnon, K.T.; Corey, D.R. Digital quantitation of potential therapeutic target RNAs. Nucleic Acid Ther. 2013, 23, 188–194. [Google Scholar] [CrossRef]
  89. Chen, H.H.W.; Kuo, M.T. Improving radiotherapy in cancer treatment: Promises and challenges. Oncotarget 2017, 8, 62742–62758. [Google Scholar] [CrossRef]
  90. Ree, A.H.; Redalen, K.R. Personalized radiotherapy: Concepts, biomarkers and trial design. Br. J. Radiol. 2015, 88, 20150009. [Google Scholar] [CrossRef]
  91. Niemantsverdriet, M.; van Goethem, M.J.; Bron, R.; Hogewerf, W.; Brandenburg, S.; Langendijk, J.A.; van Luijk, P.; Coppes, R.P. High and low LET radiation differentially induce normal tissue damage signals. Int. J. Radiat. Oncol. Biol. Phys. 2012, 83, 1291–1297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Li, Z.; Wang, F.; Zhu, Y.; Guo, T.; Lin, M. Long Noncoding RNAs Regulate the Radioresistance of Breast Cancer. Anal. Cell. Pathol. 2021, 2021, 9005073. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, F.; Sang, Y.; Chen, D.; Wu, X.; Wang, X.; Yang, W.; Chen, Y. M2 macrophage-derived exosomal long non-coding RNA AGAP2-AS1 enhances radiotherapy immunity in lung cancer by reducing microRNA-296 and elevating NOTCH2. Cell Death Dis. 2021, 12, 467. [Google Scholar] [CrossRef] [PubMed]
  94. Liao, K.; Ma, X.; Chen, B.; Lu, X.; Hu, Y.; Lin, Y.; Huang, R.; Qiu, Y. Upregulated AHIF-mediated radioresistance in glioblastoma. Biochem. Biophys. Res. Commun. 2019, 509, 617–623. [Google Scholar] [CrossRef] [PubMed]
  95. Gou, C.; Han, P.; Li, J.; Gao, L.; Ji, X.; Dong, F.; Su, Q.; Zhang, Y.; Liu, X. Knockdown of lncRNA BLACAT1 enhances radiosensitivity of head and neck squamous cell carcinoma cells by regulating PSEN1. Br. J. Radiol. 2020, 93, 20190154. [Google Scholar] [CrossRef]
  96. Ding, Z.; Kang, J.; Yang, Y. Long non-coding RNA CASC2 enhances irradiation-induced endoplasmic reticulum stress in NSCLC cells through PERK signaling. 3 Biotech 2020, 10, 449. [Google Scholar] [CrossRef]
  97. Tao, L.; Tian, P.; Yang, L.; Guo, X. lncRNA CASC2 Enhances Sensitivity in Papillary Thyroid Cancer by Sponging miR-155. Biomed. Res. Int. 2020, 2020, 7183629. [Google Scholar] [CrossRef]
  98. Lai, Y.; Chen, Y.; Lin, Y.; Ye, L. Down-regulation of LncRNA CCAT1 enhances radiosensitivity via regulating miR-148b in breast cancer. Cell Biol. Int. 2018, 42, 227–236. [Google Scholar] [CrossRef]
  99. Wang, M.; Wang, L.; He, X.; Zhang, J.; Zhu, Z.; Zhang, M.; Li, X. lncRNA CCAT2 promotes radiotherapy resistance for human esophageal carcinoma cells via the miR-145/p70S6K1 and p53 pathway. Int. J. Oncol. 2020, 56, 327–336. [Google Scholar] [CrossRef]
  100. Guglas, K.; Kolenda, T.; Teresiak, A.; Kopczyńska, M.; Łasińska, I.; Mackiewicz, J.; Mackiewicz, A.; Lamperska, K. lncRNA Expression after Irradiation and Chemoexposure of HNSCC Cell Lines. Noncoding RNA 2018, 4, 33. [Google Scholar] [CrossRef]
  101. Li, L.; Lin, X.; Xu, P.; Jiao, Y.; Fu, P. LncRNA GAS5 sponges miR-362-5p to promote sensitivity of thyroid cancer cells to 131 I by upregulating SMG1. IUBMB Life 2020, 72, 2420–2431. [Google Scholar] [CrossRef] [PubMed]
  102. Gao, J.; Liu, L.; Li, G.; Cai, M.; Tan, C.; Han, X.; Han, L. LncRNA GAS5 confers the radio sensitivity of cervical cancer cells via regulating miR-106b/IER3 axis. Int. J. Biol. Macromol. 2019, 126, 994–1001. [Google Scholar] [CrossRef] [PubMed]
  103. Xue, Y.; Ni, T.; Jiang, Y.; Li, Y. Long Noncoding RNA GAS5 Inhibits Tumorigenesis and Enhances Radiosensitivity by Suppressing miR-135b Expression in Non-Small Cell Lung Cancer. Oncol. Res. 2017, 25, 1305–1316. [Google Scholar] [CrossRef] [PubMed]
  104. Jia, J.; Zhang, X.; Zhan, D.; Li, J.; Li, Z.; Li, H.; Qian, J. LncRNA H19 interacted with miR-130a-3p and miR-17-5p to modify radio-resistance and chemo-sensitivity of cardiac carcinoma cells. Cancer Med. 2019, 8, 1604–1618. [Google Scholar] [CrossRef] [PubMed]
  105. Ma, H.; Yuan, L.; Li, W.; Xu, K.; Yang, L. The LncRNA H19/miR-193a-3p axis modifies the radio-resistance and chemotherapeutic tolerance of hepatocellular carcinoma cells by targeting PSEN1. J. Cell. Biochem. 2018, 119, 8325–8335. [Google Scholar] [CrossRef]
  106. Hu, X.; Ding, D.; Zhang, J.; Cui, J. Knockdown of lncRNA HOTAIR sensitizes breast cancer cells to ionizing radiation through activating miR-218. Biosci Rep. 2019, 39, BSR20181038. [Google Scholar] [CrossRef]
  107. Han, P.B.; Ji, X.J.; Zhang, M.; Gao, L.Y. Upregulation of lncRNA LINC00473 promotes radioresistance of HNSCC cells through activating Wnt/β-catenin signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7305–7313. [Google Scholar]
  108. Chen, Y.; Bao, C.; Zhang, X.; Lin, X.; Fu, Y. Knockdown of LINC00511 promotes radiosensitivity of thyroid carcinoma cells via suppressing JAK2/STAT3 signaling pathway. Cancer Biol. Ther. 2019, 20, 1249–1257. [Google Scholar] [CrossRef]
  109. Zhang, N.; Zeng, X.; Sun, C.; Guo, H.; Wang, T.; Wei, L.; Zhang, Y.; Zhao, J.; Ma, X. LncRNA LINC00963 Promotes Tumorigenesis and Radioresistance in Breast Cancer by Sponging miR-324-3p and Inducing ACK1 Expression. Mol. Ther. Nucleic Acids 2019, 18, 871–881. [Google Scholar] [CrossRef]
  110. Tian, W.; Zhang, Y.; Liu, H.; Jin, H.; Sun, T. LINC01123 potentially correlates with radioresistance in glioma through the miR-151a/CENPB axis. Neuropathology 2022, 42, 3–15. [Google Scholar] [CrossRef]
  111. Lin, W.; Huang, Z.; Xu, Y.; Chen, X.; Chen, T.; Ye, Y.; Ding, J.; Chen, Z.; Chen, L.; Qiu, X.; et al. A three-lncRNA signature predicts clinical outcomes in low-grade glioma patients after radiotherapy. Aging 2020, 12, 9188–9204. [Google Scholar] [CrossRef] [PubMed]
  112. Song, J.; Zhang, S.; Sun, Y.; Gu, J.; Ye, Z.; Sun, X.; Tang, Q. A Radioresponse-Related lncRNA Biomarker Signature for Risk Classification and Prognosis Prediction in Non-Small-Cell Lung Cancer. J. Oncol. 2021, 2021, 4338838. [Google Scholar] [CrossRef] [PubMed]
  113. Ogunwobi, O.O.; Mahmood, F.; Akingboye, A. Biomarkers in Colorectal Cancer: Current Research and Future Prospects. Int. J. Mol. Sci. 2020, 21, 5311. [Google Scholar] [CrossRef] [PubMed]
  114. Liu, Y.; Cheng, T.; Du, Y.; Hu, X.; Xia, W. LncRNA LUCAT1/miR-181a-5p axis promotes proliferation and invasion of breast cancer via targeting KLF6 and KLF15. BMC Mol. Cell Biol. 2020, 21, 69. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, Y.; Yue, P.; Zhou, T.; Zhang, F.; Wang, H.; Chen, X. LncRNA MEG3 enhances 131I sensitivity in thyroid carcinoma via sponging miR-182. Biomed. Pharmacother. 2018, 105, 1232–1239. [Google Scholar] [CrossRef] [PubMed]
  116. Lu, Y.; Li, T.; Wei, G.; Liu, L.; Chen, Q.; Xu, L.; Zhang, K.; Zeng, D.; Liao, R. The long non-coding RNA NEAT1 regulates epithelial to mesenchymal transition and radioresistance in through miR-204/ZEB1 axis in nasopharyngeal carcinoma. Tumour Biol. 2016, 37, 11733–11741. [Google Scholar] [CrossRef]
  117. Chen, X.; Zhang, N. Downregulation of lncRNA NEAT1_2 radiosensitizes hepatocellular carcinoma cells through regulation of miR-101-3p/WEE1 axis. Cell Biol. Int. 2019, 43, 44–55. [Google Scholar] [CrossRef]
  118. Yang, T.; Li, S.; Liu, J.; Yin, D.; Yang, X.; Tang, Q. lncRNA-NKILA/NF-κB feedback loop modulates laryngeal cancer cell proliferation, invasion, and radioresistance. Cancer Med. 2018, 7, 2048–2063. [Google Scholar] [CrossRef]
  119. Zou, Y.; Yao, S.; Chen, X.; Liu, D.; Wang, J.; Yuan, X.; Rao, J.; Xiong, H.; Yu, S.; Yuan, X.; et al. LncRNA OIP5-AS1 regulates radioresistance by targeting DYRK1A through miR-369-3p in colorectal cancer cells. Eur. J. Cell Biol. 2018, 97, 369–378. [Google Scholar] [CrossRef]
  120. Gao, Y.; Zhang, N.; Zeng, Z.; Wu, Q.; Jiang, X.; Li, S.; Sun, W.; Zhang, J.; Li, Y.; Li, J.; et al. LncRNA PCAT1 activates SOX2 and suppresses radioimmune responses via regulating cGAS/STING signalling in non-small cell lung cancer. Clin. Transl. Med. 2022, 12, e792. [Google Scholar] [CrossRef]
  121. Ge, X.; Gu, Y.; Li, D.; Jiang, M.; Zhao, S.; Li, Z.; Liu, S. Knockdown of lncRNA PCAT1 Enhances Radiosensitivity of Cervical Cancer by Regulating miR-128/GOLM1 Axis. OncoTargets Ther. 2020, 13, 10373–10385. [Google Scholar] [CrossRef] [PubMed]
  122. Ma, Q.; Niu, R.; Huang, W.; Da, L.; Tang, Y.; Jiang, D.; Xi, Y.; Zhang, C. Long Noncoding RNA PTPRG Antisense RNA 1 Reduces Radiosensitivity of Nonsmall Cell Lung Cancer Cells Via Regulating MiR-200c-3p/TCF4. Technol Cancer Res. Treat. 2020, 19, 1533033820942615. [Google Scholar] [CrossRef] [PubMed]
  123. Yu, Z.; Wang, G.; Zhang, C.; Liu, Y.; Chen, W.; Wang, H.; Liu, H. LncRNA SBF2-AS1 affects the radiosensitivity of non-small cell lung cancer via modulating microRNA-302a/MBNL3 axis. Cell Cycle 2020, 19, 300–316. [Google Scholar] [CrossRef]
  124. Wang, L.; Wang, Z.; Wang, L. Long Noncoding RNA Solute Carrier Family 25 Member 21 Antisense RNA 1 Inhibits Cell Malignant Behaviors and Enhances Radiosensitivity of Gastric Cancer Cells by Upregulating Synuclein Gamma Expression. Tohoku J. Exp. Med. 2022, 257, 225–239. [Google Scholar] [CrossRef] [PubMed]
  125. Song, W.; Zhang, J.; Xia, Q.; Sun, M. Down-regulated lncRNA TP73-AS1 reduces radioresistance in hepatocellular carcinoma via the PTEN/Akt signaling pathway. Cell Cycle 2019, 18, 3177–3188. [Google Scholar] [CrossRef]
  126. Zuo, Z.; Ji, S.; He, L.; Zhang, Y.; Peng, Z.; Han, J. LncRNA TTN-AS1/miR-134-5p/PAK3 axis regulates the radiosensitivity of human large intestine cancer cells through the P21 pathway and AKT/GSK-3β/β-catenin pathway. Cell Biol. Int. 2020, 44, 2284–2292. [Google Scholar] [CrossRef] [PubMed]
  127. Hu, M.; Yang, J. Down-regulation of lncRNA UCA1 enhances radiosensitivity in prostate cancer by suppressing EIF4G1 expression via sponging miR-331-3p. Cancer Cell Int. 2020, 20, 449. [Google Scholar] [CrossRef]
  128. Yang, H.; Zhang, X.; Zhao, Y.; Sun, G.; Zhang, J.; Gao, Y.; Liu, Q.; Zhang, W.; Zhu, H. Downregulation of lncRNA XIST Represses Tumor Growth and Boosts Radiosensitivity of Neuroblastoma via Modulation of the miR-375/L1CAM Axis. Neurochem. Res. 2020, 45, 2679–2690. [Google Scholar] [CrossRef]
Figure 1. lncRNAs in different processes associated with radioresponse, including: DNA damages and repair mechanism, cell cycle and proliferation, cell death and autophagy, regulation of reactive oxygen species (ROS), and changes in cellular phenotype.
Figure 1. lncRNAs in different processes associated with radioresponse, including: DNA damages and repair mechanism, cell cycle and proliferation, cell death and autophagy, regulation of reactive oxygen species (ROS), and changes in cellular phenotype.
Jpm 12 01605 g001
Figure 2. lncRNAs with diagnostic potential in personalized radiotherapy in different types of human cancers.
Figure 2. lncRNAs with diagnostic potential in personalized radiotherapy in different types of human cancers.
Jpm 12 01605 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kozłowska-Masłoń, J.; Guglas, K.; Paszkowska, A.; Kolenda, T.; Podralska, M.; Teresiak, A.; Bliźniak, R.; Lamperska, K. Radio-lncRNAs: Biological Function and Potential Use as Biomarkers for Personalized Oncology. J. Pers. Med. 2022, 12, 1605. https://doi.org/10.3390/jpm12101605

AMA Style

Kozłowska-Masłoń J, Guglas K, Paszkowska A, Kolenda T, Podralska M, Teresiak A, Bliźniak R, Lamperska K. Radio-lncRNAs: Biological Function and Potential Use as Biomarkers for Personalized Oncology. Journal of Personalized Medicine. 2022; 12(10):1605. https://doi.org/10.3390/jpm12101605

Chicago/Turabian Style

Kozłowska-Masłoń, Joanna, Kacper Guglas, Anna Paszkowska, Tomasz Kolenda, Marta Podralska, Anna Teresiak, Renata Bliźniak, and Katarzyna Lamperska. 2022. "Radio-lncRNAs: Biological Function and Potential Use as Biomarkers for Personalized Oncology" Journal of Personalized Medicine 12, no. 10: 1605. https://doi.org/10.3390/jpm12101605

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

Article Metrics

Back to TopTop