scholarly journals Non-coding RNA Activated by DNA Damage: Review of Its Roles in the Carcinogenesis

Author(s):  
Soudeh Ghafouri-Fard ◽  
Tahereh Azimi ◽  
Bashdar Mahmud Hussen ◽  
Atefe Abak ◽  
Mohammad Taheri ◽  
...  

Long intergenic non-coding RNA 00657 (LINC00657) or “non-coding RNA activated by DNA damage” (NORAD) is an extremely conserved and copious long non-coding RNA (lncRNA). This transcript has pivotal role in the preservation of genome integrity. Several researches have appraised the role of NORAD in the evolution of human cancers with most of them indicating an oncogenic role for this lncRNA. Several miRNAs such as miR-199a-3p, miR-608, miR−155−5p, miR-590-3p, miR-495-3p, miR-608, miR-202-5p, miR-125a-3p, miR-144-3p, miR−202−5p, and miR-30a-5p have been recognized as targets of NORAD in different cancer cell lines. In addition, NORAD has interactions with cancer-related pathways, particularly STAT, TGF-β, Akt/mTOR, and PI3K/AKT pathway. Over-expression of NORAD has been related with poor clinical outcome of patients with diverse types of neoplasms. Collectively, NORAD is a prospective marker and target for combating cancer.

2020 ◽  
Author(s):  
Hao Ji ◽  
Juan Wang ◽  
Binbin Lu ◽  
Juan Li ◽  
Jing Zhou ◽  
...  

Abstract Background: LncRNA can regulate gene at various levels such as apparent genetics, alternative splicing, and regulation of mRNA degradation. However, the molecular mechanism of LncRNA in cholangiocarcinoma is still unclear. This deserves further exploration. Methods:We investigated the expression of AGAP2-AS1 in 32 CCA tissues and two CCA cell lines. We found a LncRNA AGAP2-AS1 which induced by SP1 has not been reported in CCA, and Knockdown and overexpression were used to investigate the biological role of AGAP2-AS1 in vitro. CHIP and RIP were performed to verify the putative targets of AGAP2-AS1. Results:AGAP2-AS1 was significantly upregulated in CCA tumor tissues.SP1 induced AGAP2-AS1 plays an important role in tumorigenesis. AGAP2-AS1 knockdown significantly inhibited proliferation and caused apoptosis in CCA cells. In addition, we demonstrated that AGAP2-AS1 promotes the proliferation of CCA. Conclusions: We conclude that the long non-coding RNA AGAP2-AS1 plays a role in promoting the proliferation of cholangiocarcinoma.


Cancers ◽  
2020 ◽  
Vol 12 (1) ◽  
pp. 235 ◽  
Author(s):  
Simona Pellecchia ◽  
Romina Sepe ◽  
Myriam Decaussin-Petrucci ◽  
Cristina Ivan ◽  
Masayoshi Shimizu ◽  
...  

Anaplastic thyroid carcinoma (ATC) represents one the most aggressive neoplasias in humans, and, nowadays, limited advances have been made to extend the survival and reduce the mortality of ATC. Thus, the identification of molecular mechanism underlying its progression is needed. Here, we evaluated the long non-coding RNA (lncRNA) expression profile of nine ATC in comparison with five normal thyroid tissues by a lncRNA microarray. By this analysis, we identified 19 upregulated and 28 downregulated lncRNAs with a fold change >1.1 or <−1.1 and p-value < 0.05, in ATC samples. Some of them were subsequently validated by qRT-PCR. Then, we investigated the role of the lncRNA Prader Willi/Angelman region RNA5 (PAR5), drastically and specifically downregulated in ATC. The restoration of PAR5 reduces proliferation and migration rates of ATC-derived cell lines indicating that its downregulation contributes to thyroid cancer progression. Our results suggest that PAR5 exerts its anti-oncogenic role by impairing Enhancer of Zeste Homolog 2 (EZH2) oncogenic activity since we demonstrated that PAR5 interacts with it in thyroid cancer cell lines, reducing EZH2 protein levels and its binding on the E-cadherin promoter, relieving E-cadherin from the negative regulation by EZH2. Consistently, EZH2 is overexpressed in ATC, but not in differentiated thyroid carcinomas. The results reported here define a tumor suppressor role for PAR5 in undifferentiated thyroid neoplasias, further highlighting the pivotal role of lncRNAs in thyroid carcinogenesis.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3586-3586
Author(s):  
Ada Congrains ◽  
João Agostinho Machado-Neto ◽  
Flávia Adolfo Corrocher ◽  
Renata Giardini Rosa ◽  
Fernanda Soares Niemann ◽  
...  

Abstract Aberrant regulation of the WNT signaling pathway is a signature in numerous human cancers. Lymphoid enhancer-binding factor-1 (LEF1) is an important transcription factor downstream of this pathway. LEF1 over-expression induces AML in mice and plays a critical role in hematopoietic cell differentiation (Petropoulos et al JME 2008). Reduction of LEF1 expression through the progression of myelodysplastic syndrome has been reported and further supports the relevance of this gene in the disease pathogenesis (Pellagatti et al Br J Haematol. 2009). Our previous work using microarray technology revealed a decreased expression of a long non-coding RNA antisense to LEF1 (LEF1-AS) in MDS patients (Baratti et al BMC Medical Genomics 2010). Mounting evidence suggests that long non-coding transcripts play important roles in the epigenetic regulation of coding genes. In this context it is not surprising that long non-coding RNAs are emerging as key players in disease development and progression. Non-coding expression overlapping coding genes is very common and several examples of local regulation have been described in the literature. Here we investigate for the first time the role of LEF1 antisense long non-coding in hematopoiesis and demonstrated its contribution in the regulation of the LEF1 locus in a leukemic cell line. To explore a possible role of LEF1-AS in differentiation, we evaluated the expression pattern of LEF1-AS through erythroid cell differentiation using qRT-PCR. CD34+ HSC cells from 6 healthy donors were induced to differentiate into erythrocytes by addition of erythropoietin during 12 days. We observed that LEF1-AS is modulated during erythroid differentiation. It was significantly down-regulated during the first stages of differentiation from CD34+ HSC to erythroblast (from collection day 6 to day 8 after addition of erythropoietin, 78% mean reduction, P<0.0001) and it was up-regulated at the end-point of collection, day 12 (not significant). Lef1 coding gene displayed a similar expression pattern, consistent with previous reports of Lef1 expression during erythroid maturation (Edmaier et al Leukemia 2014). To explore a possible regulatory role of LEF1-AS, we cloned and over-expressed the transcript in KG1 CD34+ leukemia cell line. Transient over-expression of Lef1-AS led to a significant up-regulation of Lef1 gene (22% increase, P<0.05). We also observed an increase in cell viability (19% increase P<0.05), measured by MTT, which is consistent with the up-regulation of LEF1, a pro-proliferative and anti-apoptotic transcription factor. Our preliminary results from over-expressing LEF1-AS in CD34+ HSCs suggest a similar regulatory effect of LEF1-AS upon its coding counterpart, LEF1. Since aberrant expression of LEF1 is known to disrupt normal differentiation of CD34+ cells, LEF1-AS could potentially affect differentiation through the modulation of LEF1 coding gene. Our results reveal LEF1-AS transcript as a novel player in hematopoiesis and hematologic malignancy. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Hao Ji ◽  
Juan Wang ◽  
Binbin Lu ◽  
Juan Li ◽  
Jing Zhou ◽  
...  

Abstract Background: LncRNA can regulate gene at various levels such as apparent genetics, alternative splicing, and regulation of mRNA degradation.However, the molecular mechanism of LncRNA in cholangiocarcinoma is still unclear.This deserves further exploration.Methods: We investigated the expression of AGAP2-AS1 in 32 CCA tissues and two CCA cell lines. We found a LncRNA AGAP2-AS1 which induced by SP1 has not been reported in cholangiocarcinoma and studied itKnockdown and overexpression were used to investigate the biological role of AGAP2-AS1 in vitro. CHIP and RIP were performed to verify the putative targets of AGAP2-AS1.Results: AGAP2-AS1 was significantly upregulated in 32 CCA tumor tissues.SP1 induced AGAP2-AS1 plays an important role in tumorigenesis.AGAP2-AS1 knockdown significantly inhibited proliferation and caused apoptosis in CCA cells. In addition, we demonstrated that AGAP2-AS1 acts as an oncogene in CCA.Conclusions: We conclude that the long non-coding RNA AGAP2-AS1 plays a role in promoting the proliferation of cholangiocarcinoma.


2017 ◽  
Vol 45 (1) ◽  
pp. 131-147 ◽  
Author(s):  
Yana Gao ◽  
Hai Yu ◽  
Yunhui Liu ◽  
Xiaobai Liu ◽  
Jian Zheng ◽  
...  

Background/Aims: Vasculogenic mimicry (VM) has been reported to be a novel glioma neovascularization process. Anti-VM therapy provides new insight into glioma clinical management. In this study, we revealed the role of the long non-coding RNA HOXA cluster antisense RNA 2 (HOXA-AS2) in malignant glioma behaviors and VM formation. Methods: Quantitative real-time PCR was performed to determine the expression levels of HOXA-AS2 in glioma samples and glioblastoma cell lines. CD34-periodic acid-Schiff dual-staining was performed to assess VM in glioma samples. CCK-8, transwell, and Matrigel tube formation assays were performed to measure the effects of HOXA-AS2 knockdown on cell viability, migration, invasion, and VM tube formation, respectively. RNA immunoprecipitation, dual-luciferase reporter and Western blot assays were performed to explore the molecular mechanisms underlying the functions of HOXS-AS2 in glioblastoma cells. A nude mouse xenograft model was used to investigate the role of HOXA-AS2 in xenograft glioma growth and VM density. Student’s t-tests, one-way ANOVAs followed by Bonferroni posthoc tests, and chi-square tests were used for the statistical analyses. Results: HOXA-AS2 was upregulated in glioma samples and cell lines and was positively correlated with VM. HOXA-AS2 knockdown attenuated cell viability, migration, invasion, and VM formation in glioma cells and inhibited the expression of vascular endothelial-cadherin (VE-cadherin), as well as the expression and activity of matrix metalloproteinase matrix metalloproteinase (MMP)-2 and MMP-9. miR-373 was downregulated in glioma samples and cell lines and suppressed malignancy in glioblastoma cells. HOXA-AS2 bound to miR-373 and negatively regulated its expression. Epidermal growth factor receptor (EGFR), a target of miR-373, increased the expression levels of VE-cadherin, as well as the expression and activity levels of MMP-2 and MMP-9, via activating phosphatidylinositol 3-kinase/serine/threonine kinase pathways. HOXA-AS2 knockdown combined with miR-373 overexpression yielded optimal tumor suppressive effects and the lowest VM density in vivo. Conclusion: HOXA-AS2 knockdown inhibited malignant glioma behaviors and VM formation via the miR-373/EGFR axis.


2021 ◽  
Vol 27 (1) ◽  
Author(s):  
Hao Ji ◽  
Juan Wang ◽  
Binbin Lu ◽  
Juan Li ◽  
Jing Zhou ◽  
...  

Abstract Background LncRNA can regulate gene at various levels such as apparent genetics, alternative splicing, and regulation of mRNA degradation. However, the molecular mechanism of LncRNA in cholangiocarcinoma is still unclear. This deserves further exploration. Methods We investigated the expression of AGAP2-AS1 in 32 CCA tissues and two CCA cell lines. We found a LncRNA AGAP2-AS1 which induced by SP1 has not been reported in CCA, and Knockdown and overexpression were used to investigate the biological role of AGAP2-AS1 in vitro. CHIP and RIP were performed to verify the putative targets of AGAP2-AS1. Results AGAP2-AS1 was significantly upregulated in CCA tumor tissues. SP1 induced AGAP2-AS1 plays an important role in tumorigenesis. AGAP2-AS1 knockdown significantly inhibited proliferation and caused apoptosis in CCA cells. In addition, we demonstrated that AGAP2-AS1 promotes the proliferation of CCA. Conclusions We conclude that the long non-coding RNA AGAP2-AS1 plays a role in promoting the proliferation of cholangiocarcinoma.


2020 ◽  
Vol 64 (5) ◽  
pp. 721-735 ◽  
Author(s):  
Ruth F. Ketley ◽  
Monika Gullerova

Abstract The mechanisms by which RNA acts in the DNA damage response (DDR), specifically in the repair of DNA double-strand breaks (DSBs), are emerging as multifaceted and complex. Different RNA species, including but not limited to; microRNA (miRNA), long non-coding RNA (lncRNA), RNA:DNA hybrid structures, the recently identified damage-induced lncRNA (dilncRNA), damage-responsive transcripts (DARTs), and DNA damage-dependent small RNAs (DDRNAs), have been shown to play integral roles in the DSB response. The diverse properties of these RNAs, such as sequence, structure, and binding partners, enable them to fulfil a variety of functions in different cellular contexts. Additionally, RNA can be modified post-transcriptionally, a process which is regulated in response to cellular stressors such as DNA damage. Many of these mechanisms are not yet understood and the literature contradictory, reflecting the complexity and expansive nature of the roles of RNA in the DDR. However, it is clear that RNA is pivotal in ensuring the maintenance of genome integrity. In this review, we will discuss and summarise recent evidence which highlights the roles of these various RNAs in preserving genomic integrity, with a particular focus on the emerging role of RNA in the DSB repair response.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2115-2115
Author(s):  
Beatriz Martin-Antonio ◽  
Guillermo Suñe ◽  
Amer Najjar ◽  
Lorena Perez-Amill ◽  
Maria Velasco-de Andres ◽  
...  

Abstract Natural Killer (NK) cells are antitumoral, antiviral and antimicrobial cells. The first antitumor mechanism described for NK cells was the "missing self" recognition which happens between the inhibitory "Killer Immunoglobulin-like Receptors" (KIRs) on NK cells and the HLA-I present in all nucleated cells. Thus, down-regulation or absence of HLA-I in tumor cells leads to "missing self" recognition activating NK cells. As a consequence, NK cells deliver Granzyme-B activating apoptotic cell death. However, NK cells can mediate Granzyme-B and Caspase-independent cell death against tumor cells expressing HLA-I, such as multiple myeloma (MM) cells. The cytotoxic mediators mediating non-apoptotic cell death remain unknown. Mechanisms mediating NK cell antimicrobial activity have been less studied, being recognized the role of Granulysin mediating Caspase independent cell death. Cord blood (CB) derived NK cells (CB-NK) is a clinically applicable strategy for the generation of highly functional NK cells which can be used to treat MM and potentially other haematological malignancies. We have demonstrated that CB-NK cytotoxicity against MM is Granzyme B and Caspase-independent. This CB-NK cytotoxicity is transmissible among MM cells, meaning that the initial MM recipient cells (termed primary MM cells or 1ºMM cells)-which formed direct contact with CB-NK are able to transfer lipid-protein vesicles to neighbouring unexposed MM cells (termed secondary MM cells or 2ºMM cells) causing a transmissible cytotoxicity to 2ºMM cells, which have not been in direct contact with CB-NK. We aimed to determine the key cytotoxic mediators transferred from CB-NK to 1ºMM and afterwards to 2ºMM cells that mediate secondary cytotoxicity. To do that, we performed TRANS-SILAC proteomics to determine proteome trafficking between CB-NK and MM cells. TRANS-SILAC proteomics demonstrated increased cell-cell communication between MM cells after CB-NK exposure, with secondary communication between MM cells to transfer the previously received cytotoxic CB-NK material. This secondary transfer represents a dilution of the CB-NK cytotoxic material. At the same time, MM cells transferred their own proteome to neighboring MM cells. Histones were among the CB-NK transferred proteins to 1ºMM, and 2ºMM cells. We selected the Histone H2AZ variant 1 (H2AZ) for validation. Time lapse in vitro confocal microscopy with CB-NK over-expressing H2AZ confirmed dynamic transfer of H2AZ from CB-NK to MM cells. Transfer occurred through vesicles and large intercellular structures. Extracellular DNA staining confirmed that these structures with H2AZ were accompanied by DNA. At the extracellular level, Histones exhibit significant antimicrobial activity, being the main antimicrobial effectors of neutrophils. The multiple positive charges and hydrophobic residues present in Histones allow them to bind to and invade negatively-charged cell membranes which are present in both microbial pathogens and tumor cells and interact with the DNA. We determined the cytotoxic role of H2AZ. H2AZ over-expression caused Caspase independent cell death in four different MM cell lines (p<0.0001). No differences were found on K562 cells. H2AZ over-expression increased DNA damage in MM cell lines. Furthermore, MM cell lines over-expressing H2AZ transferred to neighboring MM cells both H2AZ and the DNA damage. H2AZ transfer and DNA damage transfer also translated into a transmissible cytotoxicity between MM cells, as MM cells over-expressing H2AZ transferred cytotoxicity to neighboring MM cells who received this H2AZ. H2AZ involvement in CB-NK cytotoxicity against MM cells was demonstrated as knock-down and over-expression of H2AZ in CB-NK reduced and increased, respectively, CB-NK cytotoxicity against MM cell lines (p<0.05). For K562, no impact was observed confirming again a different CB-NK cytotoxicity mechanism for MM cell lines. Knock-down and over-expression of H2AZ in CB-NK reduced and increased, respectively, CB-NK antimicrobial activity against Candida albicans and Escherichia coli, demonstrating the antimicrobial role of H2AZ. Last, the antitumoral and antimicrobial properties of the recombinant protein H2AZ against MM cell lines and against bacteria were also demonstrated. Taken together, we demonstrate for first time the antitumoral and antimicrobial role of CB-NK Histones, revealing new therapeutic targets Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Si-wei Li ◽  
Guo-liang Pi ◽  
Yong Zeng ◽  
Chang-li Ruan ◽  
Xiao-song He ◽  
...  

Abstract Objective: Radioresistance may result in nasopharyngeal carcinoma (NPC) radiation failure. To comprehend the concrete mechanism in NPC radioresistance, this work was initiated from long non-coding RNA (lncRNA) promoter of CDKN1A antisense DNA damage activated RNA (PANDAR), accompanied by sirtuin 1 (SIRT1), Ku70, and phosphatidylinositol 3 kinase (PI3K)/Akt pathway.Methods: NPC cancer tissues and normal tissues were harvested and NPC cancer tissues were specified into radioresistant and radiosensitive types. The connection between PANDA expression with NPC radioresistance, clinicopathological traits and prognosis was tested. Radioresistant CNE2-IR and 5-8F-IR cells were induced and transfected with depleted PANDAR or SIRT1 to identify their roles in cell proliferation, cycle distribution, apoptosis, SIRT1, Ku70 and PI3K/Akt pathway. PANDA and SIRT1 expression in CNE2-IR and 5-8F-IR cells were tested.Results: PANDA was elevated in NPC tissues and radioresistant tissues relative to normal tissues and radiosensitive tissues. Raised PANDA was connected with NPC radioresistance and unsatisfactory prognosis. CNE2-IR and 5-8F-IR cells expressed up-regulated PANDA and SIRT1. Down-regulating PANDA or SIRT1 inhibited radioresistant NPC cell proliferation, decreased SIRT1 and Ku70, and inactivated PI3K/Akt pathway.Conclusion: This work has clued that depleting PANDA decreases SIRT1 recruitment to inactivate Ku70 deacetylation-mediated PI3K/Akt pathway, thereby promoting NPC radiosensitivity.


2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Wei Zuo ◽  
Wei Zhang ◽  
Fei Xu ◽  
Jing Zhou ◽  
Wei Bai

Abstract Background Long non-coding RNAs (lncRNAs) are a family of non-protein-coding RNAs, which have the ability to influence the chemo-resistance of lung adenocarcinoma (LAC). In this study, we explored the mechanism by which LINC00485 competitively binds to microRNA-195 (miR-195) in the regulation of the chemotherapy sensitivity in LAC by regulating checkpoint kinase 1 (CHEK1). Methods Microarray analysis was used to screen out LAC related genes, and interaction between CHEK1 and miR-195, as well as that between miR-195 and LINC00485, was further confirmed by RNA-pull down and RIP. LINC00485 expression in LAC cells (A549 and H1299) was determined. The cells were then introduced with miR-195, anta-miR-195, LINC00485 or si-LINC00485 to identify the role of miR-195 and LINC00485 in LAC through evaluating the expression of CHEK1, CHEK1, Bax, Bcl-2, VEGF and HIF-1α in LAC cells by either RT-qPCR or Western blot analysis. After being treated with different concentration of cisplatin, cell proliferation, colony formation and apoptosis were assessed. Results LINC00485 acted as a competitive endogenous RNA against miR-195, and miR-195 directly targeted CHEK1. The expression of LINC00485 was higher in LAC cells. The down-regulation of LINC00485 or the up-regulation of miR-195 decreased the expression of CHEK1, Bcl-2, VEGF and HIF-1α, while also increasing the expression of Bax. Moreover, the over-expression of miR-195, or the silencing of LINC00485 enhanced the sensitivity of LAC cells to cisplatin, thereby promoting the apoptosis of LAC cells while suppressing the proliferation. Conclusion LINC00485 competitively binds to miR-195 to elevate CHEK1 expression in LAC cells, suggesting that LINC00485 is a novel direction for therapeutic strategies of LAC.


Sign in / Sign up

Export Citation Format

Share Document