Skip to main content

Role of the long non-coding RNAs in regulation of Gemcitabine response in tumor cells

Abstract

Chemotherapy is widely used as one of the first line therapeutic methods in cancer patients. However, chemotherapeutic resistance is one of the most common problems in cancer patients, which leads to the therapeutic failure and tumor relapse. Considering the side effects of chemotherapy drugs in normal tissues, it is required to investigate the molecular mechanisms involved in drug resistance to improve the therapeutic strategies in cancer patients. Long non-coding RNAs (lncRNAs) have pivotal roles in regulation of cellular processes associated with drug resistance. LncRNAs deregulations have been frequently reported in a wide range of chemo-resistant tumors. Gemcitabine (GEM) as a nucleoside analog has a wide therapeutic application in different cancers. However, GEM resistance is considered as a therapeutic challenge. Considering the role of lncRNAs in the occurrence of GEM resistance, in the present review we discussed the molecular mechanisms of lncRNAs in regulation of GEM response among cancer patients. It has been reported that lncRNAs have mainly an oncogenic role as the inducers of GEM resistance through direct or indirect regulation of transcription factors, autophagy, polycomb complex, and signaling pathways such as PI3K/AKT, MAPK, WNT, JAK/STAT, and TGF-β. This review paves the way to present the lncRNAs as non-invasive markers to predict GEM response in cancer patients. Therefore, lncRNAs can be introduced as the efficient markers to reduce the possible chemotherapeutic side effects in GEM resistant cancer patients and define a suitable therapeutic strategy among these patients.

Background

Surgery, radiotherapy, and chemotherapy are the most common therapeutic methods in cancer patients that can be used alone or in combination. Among these strategies, chemotherapy has been considered as the first-line therapeutic method that is widely applied in many cancers. Chemotherapy approaches are based on the chemical anti-cancer substances that affect either tumor or normal cells. Consequently, this non-specific function causes a wide range of adverse effects in cancer patients [1]. The main challenge of chemotherapy is drug resistance that is responsible for approximately 90% of treatment failures and tumor relapses [2]. Tumor relapse can be observed in more than half of the Non–small cell lung carcinoma (NSCLC) patients following the chemotherapeutic treatment [3]. There was also 50–70% of tumor recurrence after chemotherapy in ovarian cancer patients [4]. Therefore, precise identification of molecular mechanisms involved in drug resistance is necessary to improve the chemotherapeutic efficacy. Genetic mutations, epigenetics alterations, drug efflux, DNA repair, epithelial–mesenchymal transition (EMT), and tumor microenvironment have been associated with drug responses in tumor cells [2, 5, 6]. It is noteworthy that more information about the molecular mechanisms of drug resistance can be useful to present the appropriate prognostic markers to minimize the tumor relapse and side effects. However, due to the heterogeneity among tumor cells and tissues, determining a better strategy will be challenging [7]. Gemcitabine (GEM) is a chemotherapy drug that has been approved for the treatment of late-stage pancreatic cancer (PC). However, it is currently used as an adjuvant therapy in various solid tumors. GEM acts as a deoxycytidine analog that prevents DNA synthesis, thereby promoting apoptosis in malignant tumor cells [8, 9]. Although, GEM is a common drug in cancer patients that significantly improves the overall survival, GEM resistance is still considered as a big challenge among a noticeable rate of cancer patients [10]. Different transcription factors, molecular mechanisms, signaling pathways, and metabolic enzymes are involved in GEM response [8]. Long non-coding RNAs (lncRNAs) are a group of the non-coding RNAs that modulate the transcription or translation of target genes [11]. They have pivotal functions in various cellular processes such as cell proliferation, differentiation, and migration through interaction with proteins, DNA, or RNA [12]. LncRNAs function as competing endogenous RNAs (ceRNAs) to affect miRNAs functions and their target genes. They can also directly bind with the DNA and transcription factors to repress and promote gene expressions, respectively [13]. Deregulation of lncRNAs is correlated with tumor initiation and progression as either oncogenes or tumor suppressors [14,15,16,17,18]. As the lncRNAs are extensively participated in various physiological processes and tumorigenesis, aberrant expression of lncRNAs can be associated with chemotherapy resistance [19]. LncRNAs induce the expression of genes that are associated with drug resistance via enhancing proliferation while reducing apoptosis in various cancers [20]. Role of lncRNAs in GEM resistance has been frequently reported in different cancers [21,22,23]. According to the presence of lncRNAs in blood samples, they can also be suggested as reliable non-invasive prognostic and diagnostic indicators in cancer patients. Therefore, in the present review we discussed the role of lncRNAs in regulation of GEM response to present them as the probable efficient non-invasive prognostic markers in cancer patients (Table 1).

Table 1 LncRNAs that are involved in Gemcitabine (GEM) response in tumor cells

PI3K/AKT and MAPK signaling pathways

PI3K/AKT is an important signaling pathway that can be activated by growth factors following the binding with receptor tyrosine kinases (RTKs). AKT is the most important downstream effector of PI3K that has pivotal roles in cellular metabolism, growth, and proliferation. Therefore, deregulation of PI3K/AKT pathway can be associated with drug resistance and tumor relapse [24]. LncRNAs have a key role in GEM response of tumor cells by regulation of PI3K/AKT signaling pathway (Fig. 1). Surgery, chemotherapy, and radiotherapy are the main therapeutic plans in osteosarcoma patients. A standard chemotherapy regimen including cisplatin, GEM, and doxorubicin can improve the 5-years survival rate of osteosarcoma patients [25]. However, drug resistance is still a big challenge for the effective treatment of osteosarcoma patients [26]. C-MET is a receptor tyrosine kinase that increases tumor progression in a wide range of tumors [27]. A study has reported that c-Met inhibition increased the sensitivity of osteosarcoma cells to cisplatin by PI3K/Akt suppression [28]. PVT1 activated the PI3K/AKT pathway via c-MET to increase the chemotherapy resistance of osteosarcoma cells. PVT1 had a pivotal role in the GEM resistance of osteosarcoma cells through miR-152 targeting to regulate the c-MET/PI3K pathway [29]. FGFR1 belongs to the RTK protein family that promotes MAPK and PI3K/Akt pathways [30]. FGFR1 activation has been associated with EMT process in several human cancers [31,32,33]. AGAP2-AS1 up regulation was correlated with advanced tumor stage and poor survival in CRC patients. AGAP2-AS1 sponged miR-497 to promote the growth and metastasis of CRC cells and GEM resistance via FGFR1 targeting [34]. YB1 as a highly conserved transcription factor is involved in regulation of a wide variety of biological processes [35]. The YB-1 phosphorylation by AKT triggers the mRNA translation [36]. PI3K/AKT over-activation is involved in GEM resistance. HIF1A-AS1 modulated the HIF1a expression in a glycolysis-dependent manner to promote GEM resistance in pancreatic tumor cells. HIF1A-AS1 improved the interaction between p-AKT and p-YB1 to induce YB1 phosphorylation. YB1 phosphorylation by AKT plays a pivotal role in the progression of GEM resistance in PC via HIF1a regulation by HIF1A-AS1. There was also a significant association between HIF1A-AS1/HIF1a up regulation and the poor prognosis in GEM-received PC patients [37].

Figure 1
figure 1

LncRNAs have a key role in GEM response of tumor cells by regulation of PI3K/AKT and MAPK/ERK signaling pathway. (Created with BioRender.com)

Drug resistance restricts the long-term therapeutic impacts of the cisplatin and GEM in bladder cancer treatment [38]. UCA1 induced cisplatin/GEM resistance in bladder tumor cells by CREB mediated miR-196a-5p regulation. UCA1 triggered the AKT pathway that resulted in CREB phosphorylation. UCA1 also increased cisplatin/GEM resistance via miR-196a-5p up regulation by CREB and p27Kip1 regulations in bladder cancer [39]. Although, GEM is an effective and widely prescribed therapy for NSCLC patients, GEM resistance has restricted its clinical application. Curcumin has been indicated to play a tumor suppressive role by regulation of the signaling pathways in various cancers [40, 41]. Curcumin promotes apoptosis and autophagy in lung tumor cells via STAT3 and PI3K/AKT pathways [42, 43]. PTEN as a negative regulator of PI3K/AKT pathway has a key role in various tumor associated phenotypes such as relapse and drug response. It has been shown that high concentrations of curcumin enhanced the apoptosis of GEM-resistant NSCLC cells. Curcumin up regulated the MEG3 that promoted the PTEN pathway in GEM-resistant NSCLC cells. Curcumin also suppressed the GEM-resistant NSCLC proliferation in the xenograft model [8]. The mTOR is the main downstream target of PI3K/Akt that induces cell growth and protein synthesis by activation of S6K and 4EBP1 [44]. LAT2 is a membrane transporter involved in mTOR activation to induce chemo resistance in PC cells [45]. GSTM3TV2 up regulated the LAT2 by competitively sponging let-7, resulting in increased GEM resistance in pancreatic tumor cells. GSTM3TV2 up regulation was associated with poor prognosis in PC patients [46]. BRCA2 prevents mutagenesis as a tumor-suppressor via regulating DNA double-strand break repair [47, 48]. GEM is a nucleoside analog that suppresses DNA replication in tumor cells. It has been found that the DLEU2L down regulated the Warburg effect modulators including GLUT1, LDHB, HK2, and PKM2 in PC cells. DLEU2L decreased ATP production and glucose uptake. It also inhibited the phosphorylation of AKT/mTOR, as well as S6K as downstream effectors. Moreover, DLEU2L inhibited GEM resistance in PC cells via miR-210-3p/BRCA2 axis [49].

Figure 2
figure 2

LncRNAs have an important function in GEM response of tumor cells by regulation of WNT and JAK/STAT signaling pathways. (Created with BioRender.com)

MAPK is a cascade of three kinases that can be activated by a variety of input signals such as cytokines, hormones, growth factors, and stress signals. It finally functions as a mitogen by ERK activation or stress response through JNK and p38. Therefore, MAPK/ERK has an oncogenic function by the induction of cell proliferation, migration, and drug resistance [50]. LncRNAs have a pivotal function in GEM response of tumor cells by regulation of MAPK/ERK signaling (Fig. 1). TUG1 up regulation has been shown in PDAC tissues in comparison with the normal margins. It promoted the GEM resistance in PDAC cells by ERK phosphorylation [51]. HDGF is a heparin-binding growth factor that promotes cell proliferation by the MAPK and PI3K pathways [52]. HCP5 up regulation was indicated in GEM-resistant PC tissues that were correlated with poor survival. HCP5 increased the GEM resistance by miR-214-3p/HDGF axis in PC cells [53].

Wnt/β-catenin, TGF-b, and JAK/STAT signaling pathways

WNT pathway is essential for cell proliferation, tissue homeostasis, and apoptosis. Wnt ligands bind to FZD/LRP receptors that promote β-catenin to enter the nucleus to stimulate Wnt target genes expression via binding with TCF/LEF transcription factors and BCL9L and PYGO2 co-activators [54, 55]. LncRNAs have an important function in GEM response of tumor cells by regulation of WNT signaling (Fig. 2). Solid tumors recruit autophagy to overcome hypoxia, ischemia, radiotherapy, and chemotherapy [56, 57]. ATG14 has an important role in the autophagosome formation [58]. PVT1 has been shown to promote GEM resistance in pancreatic tumor cells by increasing WNT and autophagic activities. PVT1 plays a critical role in modulating the GEM resistance of PC cells via the miR-619-5p/Pygo2 and ATG14 axes [59]. LINC00665 up regulation has been found in GEM-resistant cholangiocarcinoma (CCA) cells that was correlated with the prognosis and chemotherapy resistance of CCA patients. LINC00665 sponged miR-424-5p to regulate BCL9L expression and WNT activation. Silencing of the LINC00665 reduced GEM-induced EMT and stemness in resistant CCA cells by decreasing β-Catenin nucleus translocation and BCL9L down regulation [60]. There was CDKN2B-AS up regulation in bladder urothelial carcinoma (BUC) tissues that was correlated with higher grades. CDKN2B-AS increased the GEM resistance of BUC via Let-7 sponging to activate the CTNNB1 [61].

TGF-β signaling is a key pathway in regulation of cell proliferation, apoptosis, and drug resistance. It is initiated by the TGF-β binding with TβR receptors which phosphorylates and activates TβR. Then activated TβR phosphorylates the R-Smad and Smad2/3 to form a complex with Smad4. Subsequently, Smad complexes enter to the nucleus to regulate the TGF-β target genes [62]. Smad4/R-Smad is activated by the TGF-β receptor to modulate the expression of genes involved in angiogenesis and chemo resistance [63, 64]. Smad4 knockdown induced cetuximab-resistance via the MAPK pathway targeting. Smad4 also induced the angiogenesis in ovarian cancer and increased the tumor development [65]. Smad4 acts as a tumor suppressor and inhibits the proliferation of tumor cells via Smad4/R-Smad complex. SNHG16 over expression has been found in GEM-resistant PC cells. SNHG16 recruited the EZH2 to catalyze H3K27me3 and suppress the Smad4, resulted in promotion of the AKT-mediated GEM-resistant PC cells [66]. Cancer stem-like cells (CSCs) are a sub-population of chemo-resistant tumor cells that are responsible for tumor recurrence [67, 68]. NF90 is an RNA-binding protein that has a significant role in the stabilization, turnover, and translation of various mRNAs [69,70,71]. TGFβ1 as an essential cytokine that induces EMT is highly correlated with the CSCs features [72]. It has been found that CSCs were highly abundant in UBC upon GEM treatment. TGFβ1 was up regulated in GEM-treated UBC cells to deregulate the LET/NF90/miR-145 signaling cascade, resulting in increased chemo resistance. LET down regulation was associated with NF90 protein stability, decreased biogenesis of miR-145, and increased stemness markers. The miR-145 targeted KLF4 and HMGA2 in UBC cells to inhibit stemness of tumor cells [73].

Figure 3
figure 3

LncRNAs are involved in GEM response by regulation of structural proteins, autophagy, drug efflux, and cellular metabolism. (Created with BioRender.com)

The JAK/STAT signaling pathway is the main way to receive cytokines such as interleukins, interferons, and colony-stimulating factors. Therefore, it is involved in regulation of hematopoiesis, inflammation, and apoptosis. JAK/STAT signaling is initiated by cytokine binding to their receptors that recruits JAKs. Subsequently, activated JAKs promote the phosphorylation and dimerization of STATs that finally enter to the nucleus to regulate the JAK/STAT target genes. This pathway can be negatively regulated by the PIAS, SOCS, and PTPs. SOCS family inhibits the recruitment of STAT and JAK kinase activity [74]. LncRNAs have a pivotal role in GEM response of tumor cells by regulation of JAK/STAT signaling (Fig. 2). GAS5 suppressed GEM resistance in PC cells via miR-221/SOCS3 axis [75]. UCA1 suppressed SOCS3 via EZH2 recruitment in PC cells, thus promoting malignant traits and Gem resistance in pancreatic tumor cells [76].

Transcription factors

Transcription factors are mainly the final effectors of various signaling pathways to regulate the expression of genes involved in cellular responses toward the intra and extra cellular stimuli. Therefore, they can be also involved in drug response of tumor cells. It has been shown that lnRNAs can affect the Gem response in tumor cells by regulation of transcription factors. C-MYC is an oncogenic transcription factor involved in cellular metabolism, proliferation, and drug resistance [77]. PVT1 plays a pivotal role in cancer through co-overexpression with the Myc [78, 79]. It has been identified that GEM triggers PVT1 processing into miRNAs through modulating the activity of the microprocessor in PC cells. After GEM exposure, the miR-1207 was upregulated in PC cells and inhibited oncogenic signaling by MYC targeting. PVT1 processing was mediated by Drosha/DGCR8 and that GEM modulated miRNA processing via Drosha and DGCR8 targeting [80]. FOXO3 belongs to the Forkhead box transcription factors that is negatively regulated by PI3K/AKT and MAPK/ERK pathways [81, 82]. It promotes apoptosis by up regulation of pro-apoptotic factors such as Bim and FasL [83]. FOXO3 inactivation by AKT confers 5-FU resistance [84]. There was LINC01714 down regulation in CCA that was related with poor prognosis. LINC01714 inhibited the migration and aggressiveness of CCA tumor cells, while promoted GEM sensitivity via FOXO3 inhibition [85]. The Bromodomain-containing Protein 4 (BRD4) is a transcriptional regulator by binding to the acetylated histones that is involved in modulation of cell proliferation and invasion [86, 87]. BRD4 is an important DNA repair regulator, and its inhibition has antitumor effects on various cancers. It also promotes the chemo sensitivity to GEM treatment [87, 88]. p21 is a negative regulator of cell-cycle progression, causing CCNB1 degradation to maintain cell-cycle arrest during the G2/M phase [89, 90]. Chk1 is also a ser/thr kinase that regulates cell-cycle progression during the G2/M phase [91]. LINC00346 improved the pancreatic tumor cell proliferation and colony formation. LINC00346 down regulation induced G2/M cell-cycle arrest in pancreatic tumor cells via p21 expression and Chk1 phosphorylation. It sponged miR-188-3p to enhance PC growth and GEM resistance via BRD4 targeting [92]. The HOX family belongs to the homeobox genes that encode transcriptional regulators to control cell proliferation and differentiation [93]. There were HOTTIP up regulations in human PC tissues and cell lines, which promoted tumor cell proliferation, EMT, and invasion. HOTTIP also increased GEM resistance via HOXA13 targeting [94]. High mobility group (HMG) is a family of non-histone DNA-binding proteins that are participated in regulation of transcription, DNA repair, and nucleosome assembly [95]. It was discovered that ANRIL down regulation inhibited PC cell growth and invasion and decreased GEM resistance via miR-181a targeting to regulate HMGB1-induced cell autophagy. MiR-181a activated autophagy via reducing LC3 I/II and increasing Beclin1 [96].

Polycomb and RNA binding proteins

The majority of PC patients are detected at advanced stages, and there are limited surgical choices and a poor prognosis for these patients. Tumor recurrence and drug resistance are the most common causes of poor survival rate in PC patients [97]. GEM substitutes cytidine during DNA replication and blocks the production of deoxyribonucleotides to suppress pancreatic tumor cell growth [98]. The serine- and arginine-rich (SR) proteins belong to RNA-binding protein family, which regulates alternative splicing [99]. Serine/arginine-rich splicing factor 3 (SRSF3) is a SR protein family member that modulates cell senescence through detecting the alternative terminal exon [100, 101]. Messenger RNA metabolic processes from splicing to translation are regulated by m6A modification [102]. EZH2 as the catalytic member of Polycomb repressive complex 2 (PRC2) catalyzes the H3K27me3 to inhibit gene expression [103, 104]. LncRNAs are involved in GEM response by EZH2 promoter recruitment and transcriptional regulation of target genes (Fig. 3). There was SRSF3 up regulation in PC tissues that was correlated with GEM resistance and poor prognosis. SRSF3 modulated ANRIL exon inclusion, while improved exon skipping was observed by SRSF10 in PC cells. The m6A modification of ANRIL regulated its splicing in these cells. SRSF3 enhanced GEM resistance through ANRIL expression by constructing a complex with Ring1b and EZH2 that led to increased DNA homologous recombination repair [105]. HAT1 is a B-type histone acetyltransferase that regulates histone H4 N-terminus acetylation. Acetyl molecules can be present on cellular protein lysine residues as an epigenetic modulator [106]. Protein acetylation is a critical regulator of replication-dependent chromatin assembly, DNA damage repair, and gene expression [107, 108]. Mutated HAT1 enhances drug resistance in tumor cells. HAT1 improved liver tumor cell growth and caused cisplatin resistance [109]. EZH2 deregulation was correlated with GEM resistance via down regulation of p27Kip1 [110]. HAT1 silencing increased GEM sensitivity in PC cells via PVT1/EZH2 complex targeting. It also enhanced GEM resistance in PC cells by facilitating BRD4 binding to the PVT1 promoter and increasing PVT1 expression. Moreover, HAT1 protected EZH2 by inhibiting BRD4 from attaching to the N-terminal domain of EZH2 [111]. Cancer stem cells (CSCs) have a high self-renewal capacity that is associated with chemo resistance in PDAC tumors [112, 113]. It has been hypothesized that standard chemotherapy reduces the tumor mass by targeting the proliferating PDAC cells, while fails to target the CSCs that results in treatment failure [114]. Curcumin is a well-known suppressor of several oncogenes including ERK, AKT, and EZH2 in PDACs [115, 116]. PVT1 was up regulated in GEM-resistant PDAC cells. Curcumin also targeted CSCs and decreased the spheroid-forming capacity of GEM-resistant PDAC cells. Curcumin improved GEM sensitivity in pancreatic tumor cells via EZH2 blocking and its downstream target PVT1 [117]. GEM is a first-line chemotherapeutic agent that has been shown to enhance patient survival in un-resectable gallbladder cancer (GBC) patients [118, 119]. Nevertheless, only 36% of GBC patients benefit from GEM treatment that is related to the drug resistance [120]. NONO is a critical RNA binding protein in different types of cancers [121, 122]. NONO increased oxaliplatin sensitivity in CRC cells indicating that NONO has pivotal role in drug resistance of tumor cells [123]. There was SSTR5-AS1 up regulation in GBC tissues that was associated with poor OS in GBC patients. SSTR5-AS1 directly regulated the NONO protein, which resulted in increased GEM resistance in these patients [124].

Autophagy and cell cycle regulation

Autophagy is a critical biochemical process that preserves cellular homeostasis and survival by removing and recycling unneeded and damaged cellular materials and organelles. As a multidimensional catabolic process, autophagy has been preserved throughout the evolution. Drug resistance reduces the apoptotic response of tumor cells via abnormal cell autophagy [125,126,127]. Autophagy is a critical regulator of tumor development and cancer therapy via promoting cell survival [56]. PI3K pathway is one of the main regulators of autophagy in response to ROS levels. PI3Kα subunit is a suppressor of autophagy by AKT/mTORC2 axis under moderate ROS levels [128]. Regarding the role of autophagy in drug response, lncRNAs can be involved in autophagy mediated GEM response in tumor cells (Fig. 3). Inhibition of autophagy sensitizes the cancer cells to GEM, while decreasing the stemness of pancreatic cancer cells [129]. It has been shown that the SNHG14 enhanced PDAC cells progression and autophagy mediated GEM resistance via miR-101 sponging [130]. Neoadjuvant chemotherapy including capecitabine, bevacizumab, GEM, and taxanes, has improved the prognosis for patients with metastatic breast cancer [131]. GEM regulates autophagy via Beclin1, ATG16L1, and LC3, and promotes apoptosis in tumor cells via Bcl-2 and Bax [132,133,134,135]. Apoptosis is a key mechanism that controls tissue homeostasis, whereas autophagy is an essential biological process that eliminates damaged cellular components [136]. Linc-ROR suppressed histone H3 acetylation in the miR-34a promoter to reduce GEM-mediated apoptosis and autophagy, resulting in miR-34a down regulation. LincROR inhibition induced the LC3-II, Beclin1, and NOTCH1 expressions, while reduced p62 expression in breast tumor cells [137]. HOST2 down regulation reduced GEM resistance to promote apoptosis in pancreatic tumor cells [138]. Cyclin-dependent kinases (CDKs) have key roles in regulation of the cell cycle progression [139]. CDK14 regulates the cell cycle progression by interacting with CCND3 and CCNY. It also stimulates the WNT pathway by targeting its downstream proteins [140]. There were MSC-AS1 up regulations in PAAD and PDAC tissues that was directly correlated with CDK14 up regulation and poor prognosis. There was significant down regulation of miR-29b-3p in the PDAC tissues that was correlated with a poor outcome in PDAC patients. MSC-AS1/miR-29b-3p axis regulated CDK14-mediated cell proliferation and GEM-mediated apoptosis in PDAC cells [141].

ABC transporters and structural proteins

Structural proteins have not a direct role in transcriptional regulation. However, they have critical roles in regulation of cellular metabolism, membrane traffic, and cell migration. LncRNAs are also involved in GEM response by regulation of structural proteins, ABC transporters, and cellular metabolism (Fig. 3). Multidrug resistance-associated protein 1 (MRP1) belongs to the ABC transporter protein family that is primarily involved in the transport of multiple intracellular and extracellular complexes. As an efflux pump, it removes the chemotherapy drugs from tumor cells. MRP1 promotes the chemotherapeutic resistance in tumor cells via lowering intracellular drug concentration. It has been reported that there was GHET1 up regulation in BC that was associated with higher grades and muscle invasion. GHET1 enhanced the GEM resistance in BC via ABCC1 up regulation [142]. There was a dose-dependent association between FOXD2-AS1 up regulation and GEM-resistance of bladder tumor cells. FOXD2-AS1 enhanced the GEM-resistance of bladder cancer via miR-143/ABCC3 targeting [143].

A low intracellular reactive oxygen species (ROS) level is required to maintain the self-renewal of tumor cells [144]. GEM triggers the cell apoptosis via ROS production [145]. Chemo resistant tumor cells have a strong antioxidant system to regulate the excessive generation of ROS in order to survive under oxidative stress [146]. NRF2 is the major mediator of redox hemostasis that is typically up regulated in CSCs [147]. SKP1-Cul1-Rbx1 (SCFb-TRCP) E3 complex participates in the proteasomal degradation of NRF2 [148, 149]. SLC7A11-AS1 up regulation and its association with drug resistance was demonstrated in GEM-resistant PDAC cells. SLC7A11-AS1 preserved NRF2 via inhibiting ubiquitination mediated by SCFb-TRCP, which is essential to sustain the self-renewal and chemo resistance of PDAC cells. SLC7A11-AS1 up regulation was contributed with poor prognosis in PDAC patients [150]. USP44 as a deubiquitinase has pivotal roles in regulation of spindle assembly and anaphase onset by deubiquitination of CDC20 that is an inhibitor of APC/C. There was DNH-AS1 down regulation in GEM resistant PC cells which was associated with prognosis. The m6A methylase knockdown has been demonstrated to reduce the levels of DBH-AS expressions in PC cells. DNH-AS1 acted as a sponge for miR-3163 to up regulate the USP44 that was involved in the GEM absorption in tumor cells [151].

Twinfilin 1 (TWF1) as an inhibitor of the actin polymerization is involved in regulation of cell migration, drug sensitivity, and tumor progression [152, 153]. Inhibition of the SBF2-AS1 decreased the levels of TWF1 expressions via miR-142-3p sponging to increase GEM resistance in pancreatic tumor cells [154]. EMT is one of the pathophysiological cellular processes that is involved in regulation of embryogenesis, tumor metastasis, and drug resistance [155]. It is orchestrated by the up regulation of mesenchymal proteins such as CDH2 and VIM, while down regulation of CDH1 epithelial marker that finally changes the epithelial to mesenchymal phenotype to facilitate the tumor cell migration [156]. MVIH down regulation increased the expression of CDH1 while decreased the Vimentin expression, which induced GEM sensitivity in gastric tumor cells [157].

Conclusions

LncRNAs as the regulators of cellular mechanisms such as cell proliferation and apoptosis have a critical role in chemotherapeutic response. In the present review, we discussed the role of lncRNAs in GEM response. It has been shown that lncRNAs mainly induced GEM resistance through the regulation of transcription factors, autophagy, polycomb complex, and signaling pathways. Therefore, lncRNAs can be introduced as the non-invasive prognostic markers to predict the GEM response and improve the therapeutic strategy among cancer patients. Despite the clinical advantages of lncRNA as non-invasive diagnostic and prognostic markers in cancer patients, some limitations have slowed down the entry of these markers into the clinic. One of the upcoming limitations is the requirement to examine the levels of lncRNA expressions in the serum of cancer patients. Animal studies are also needed to introduce lncRNA as the prognostic markers and therapeutic targets. However, the majority of reports on the relationship between lncRNAs and GEM response are limited to the in-vitro studies and assessment of their expression levels in tumor tissues. Another existing limitation is the complexity of cellular processes affected by lncRNAs, which can be related to their ability to bind with DNA, RNA, and proteins that can affect a wide range of cellular processes. Therefore, detailed studies on the molecular mechanisms of lncRNAs can play a very important role in preclinical studies to pave the way to introduce lncRNAs as the reliable diagnostic and prognostic markers in the clinic.

Data Availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

BUC:

Bladder urothelial carcinoma

BRD4:

Bromodomain-containing Protein 4

CSCs:

Cancer stem cells

CCA:

Cholangiocarcinoma

ceRNAs:

Competing endogenous RNAs

CDKs:

Cyclin-dependent kinases

EMT:

Epithelial?mesenchymal transition

GBC:

Gallbladder cancer

GEM:

Gemcitabine

lncRNAs:

Long non-coding RNAs

MRP1:

Multidrug resistance-associated protein 1

NSCLC:

Non?small cell lung carcinoma

PC:

Pancreatic cancer

PRC2:

Polycomb repressive complex 2

ROS:

Reactive oxygen species

RTKs:

Receptor tyrosine kinases

SR:

Serine- and arginine-rich

SRSF3:

Serine/arginine-rich splicing factor 3

TWF1:

Twinfilin 1

References

  1. El-Hussein A, Manoto SL, Ombinda-Lemboumba S, Alrowaili ZA, Mthunzi-Kufa P. A review of chemotherapy and photodynamic therapy for lung cancer treatment. Anti-cancer agents in Medicinal Chemistry (formerly current Medicinal Chemistry-Anti-Cancer Agents). 2021;21(2):149–61.

  2. Emran TB, Shahriar A, Mahmud AR, Rahman T, Abir MH, Faijanur-Rob-Siddiquee M et al. Multidrug Resistance in Cancer: understanding Molecular Mechanisms, Immunoprevention, and therapeutic approaches. Front Oncol. 2022:2581.

  3. Uramoto H, Tanaka F. Recurrence after surgery in patients with NSCLC. Translational lung cancer research. 2014;3(4):242.

    PubMed  PubMed Central  Google Scholar 

  4. Castells M, Thibault B, Delord J-P, Couderc B. Implication of tumor microenvironment in chemoresistance: tumor-associated stromal cells protect tumor cells from cell death. Int J Mol Sci. 2012;13(8):9545–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zangouei AS, Moghbeli M. MicroRNAs as the critical regulators of cisplatin resistance in gastric tumor cells. Genes Environ. 2021;43(1):21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Moghbeli M. MicroRNAs as the critical regulators of cisplatin resistance in ovarian cancer cells. J Ovarian Res. 2021;14(1):127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mansoori B, Mohammadi A, Davudian S, Shirjang S, Baradaran B. The different mechanisms of cancer drug resistance: a brief review. Adv Pharm Bull. 2017;7(3):339.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gao L, Shao T, Zheng W, Ding J. Curcumin suppresses tumor growth of gemcitabine-resistant non-small cell lung cancer by regulating lncRNA-MEG3 and PTEN signaling. Clin translational oncology: official publication Federation Span Oncol Soc Natl Cancer Inst Mexico. 2021;23(7):1386–93.

    Article  CAS  Google Scholar 

  9. de Sousa Cavalcante L, Monteiro G. Gemcitabine: metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur J Pharmacol. 2014;741:8–16.

    Article  PubMed  Google Scholar 

  10. Burris Hr, Moore MJ, Andersen J, Green MR, Rothenberg ML, Modiano MR, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15(6):2403–13.

    Article  Google Scholar 

  11. Guttman M, Rinn JL. Modular regulatory principles of large non-coding RNAs. Nature. 2012;482(7385):339–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hamidi AA, Khalili-Tanha G, Nasrpour Navaei Z, Moghbeli M. Long non-coding RNAs as the critical regulators of epithelial mesenchymal transition in colorectal tumor cells: an overview. Cancer Cell Int. 2022;22(1):71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ghosal S, Das S, Chakrabarti J. Long noncoding RNAs: new players in the molecular mechanism for maintenance and differentiation of pluripotent stem cells. Stem Cells Dev. 2013;22(16):2240–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Guzel E, Okyay TM, Yalcinkaya B, Karacaoglu S, Gocmen M, Akcakuyu MH. Tumor suppressor and oncogenic role of long non-coding RNAs in cancer. North Clin Istanbul. 2020;7(1):81.

    Google Scholar 

  15. Kogo R, Shimamura T, Mimori K, Kawahara K, Imoto S, Sudo T, et al. Long noncoding RNA HOTAIR regulates polycomb-dependent chromatin modification and is associated with poor prognosis in colorectal cancers. Cancer Res. 2011;71(20):6320–6.

    Article  CAS  PubMed  Google Scholar 

  16. Russell MR, Penikis A, Oldridge DA, Alvarez-Dominguez JR, McDaniel L, Diamond M, et al. CASC15-S is a tumor suppressor lncRNA at the 6p22 Neuroblastoma susceptibility LocusCASC15 is a neuroblastoma suppressor gene. Cancer Res. 2015;75(15):3155–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Shahabi S, Kumaran V, Castillo J, Cong Z, Nandagopal G, Mullen DJ, et al. LINC00261 is an Epigenetically regulated tumor suppressor essential for activation of the DNA damage ResponseLINC00261, a tumor suppressor regulating DNA damage response. Cancer Res. 2019;79(12):3050–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rahmani Z, Mojarrad M, Moghbeli M. Long non-coding RNAs as the critical factors during tumor progressions among iranian population: an overview. Cell & bioscience. 2020;10:6.

    Article  Google Scholar 

  19. Khalili-Tanha G, Moghbeli M. Long non-coding RNAs as the critical regulators of doxorubicin resistance in tumor cells. Cell Mol Biol Lett. 2021;26(1):39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Malek E, Jagannathan S, Driscoll JJ. Correlation of long non-coding RNA expression with metastasis, drug resistance and clinical outcome in cancer. Oncotarget. 2014;5(18):8027–38.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Li Z, Zhao X, Zhou Y, Liu Y, Zhou Q, Ye H, et al. The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer. J translational Med. 2015;13(1):1–16.

    Article  Google Scholar 

  22. Sun Z-Y, Jian Y-K, Zhu H-Y, Li B. lncRNAPVT1 targets miR-152 to enhance chemoresistance of osteosarcoma to gemcitabine through activating c-MET/PI3K/AKT pathway. Pathology-Research and Practice. 2019;215(3):555–63.

    Article  CAS  PubMed  Google Scholar 

  23. Hong S, Yan Z, Song Y, Bi M, Li S. LncRNA AGAP2-AS1 augments cell viability and mobility, and confers gemcitabine resistance by inhibiting miR-497 in colorectal cancer. Aging. 2020;12(6):5183.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Navaei ZN, Khalili-Tanha G, Zangouei AS, Abbaszadegan MR, Moghbeli M. PI3K/AKT signaling pathway as a critical regulator of cisplatin response in tumor cells. Oncol Res. 2021;29(4):235–50.

    Article  PubMed  Google Scholar 

  25. Zhang CL, Zhu KP, Ma XL. Antisense lncRNA FOXC2-AS1 promotes doxorubicin resistance in osteosarcoma by increasing the expression of FOXC2. Cancer Lett. 2017;396:66–75.

    Article  CAS  PubMed  Google Scholar 

  26. Wang Y, Zhang L, Zheng X, Zhong W, Tian X, Yin B, et al. Long non-coding RNA LINC00161 sensitises osteosarcoma cells to cisplatin-induced apoptosis by regulating the mir-645-IFIT2 axis. Cancer Lett. 2016;382(2):137–46.

    Article  CAS  PubMed  Google Scholar 

  27. Wu H, Tao J, Li X, Zhang T, Zhao L, Wang Y, et al. MicroRNA-206 prevents the pathogenesis of hepatocellular carcinoma by modulating expression of met proto-oncogene and cyclin-dependent kinase 6 in mice. Hepatology (Baltimore MD). 2017;66(6):1952–67.

    Article  CAS  PubMed  Google Scholar 

  28. Wang K, Zhuang Y, Liu C, Li Y. Inhibition of c-Met activation sensitizes osteosarcoma cells to cisplatin via suppression of the PI3K-Akt signaling. Arch Biochem Biophys. 2012;526(1):38–43.

    Article  CAS  PubMed  Google Scholar 

  29. Sun ZY, Jian YK, Zhu HY, Li B. lncRNAPVT1 targets miR-152 to enhance chemoresistance of osteosarcoma to gemcitabine through activating c-MET/PI3K/AKT pathway. Pathol Res Pract. 2019;215(3):555–63.

    Article  CAS  PubMed  Google Scholar 

  30. Li J, Shi S, Srivastava SP, Kitada M, Nagai T, Nitta K, et al. FGFR1 is critical for the anti-endothelial mesenchymal transition effect of N-acetyl-seryl-aspartyl-lysyl-proline via induction of the MAP4K4 pathway. Cell Death Dis. 2017;8(8):e2965.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Schultheis AM, Bos M, Schmitz K, Wilsberg L, Binot E, Wolf J, et al. Fibroblast growth factor receptor 1 (FGFR1) amplification is a potential therapeutic target in small-cell lung cancer. Mod pathology: official J United States Can Acad Pathol Inc. 2014;27(2):214–21.

    Article  CAS  Google Scholar 

  32. Yang F, Zhang Y, Ressler SJ, Ittmann MM, Ayala GE, Dang TD, et al. FGFR1 is essential for prostate cancer progression and metastasis. Cancer Res. 2013;73(12):3716–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Murase H, Inokuchi M, Takagi Y, Kato K, Kojima K, Sugihara K. Prognostic significance of the co-overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer. Mol Clin Oncol. 2014;2(4):509–17.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Hong S, Yan Z, Song Y, Bi M, Li S. LncRNA AGAP2-AS1 augments cell viability and mobility, and confers gemcitabine resistance by inhibiting miR-497 in colorectal cancer. Aging. 2020;12(6):5183–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Lyabin DN, Eliseeva IA, Ovchinnikov LP. YB-1 protein: functions and regulation. Wiley interdisciplinary reviews RNA. 2014;5(1):95–110.

    Article  CAS  PubMed  Google Scholar 

  36. Evdokimova V, Ruzanov P, Anglesio MS, Sorokin AV, Ovchinnikov LP, Buckley J, et al. Akt-mediated YB-1 phosphorylation activates translation of silent mRNA species. Mol Cell Biol. 2006;26(1):277–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Xu F, Huang M, Chen Q, Niu Y, Hu Y, Hu P, et al. LncRNA HIF1A-AS1 promotes Gemcitabine Resistance of Pancreatic Cancer by enhancing glycolysis through modulating the AKT/YB1/HIF1α pathway. Cancer Res. 2021;81(22):5678–91.

    Article  CAS  PubMed  Google Scholar 

  38. Barocas DA, Clark PE. Bladder cancer. Curr Opin Oncol. 2008;20(3):307–14.

    Article  PubMed  Google Scholar 

  39. Pan J, Li X, Wu W, Xue M, Hou H, Zhai W, et al. Long non-coding RNA UCA1 promotes cisplatin/gemcitabine resistance through CREB modulating miR-196a-5p in bladder cancer cells. Cancer Lett. 2016;382(1):64–76.

    Article  CAS  PubMed  Google Scholar 

  40. Giordano A, Tommonaro G. Curcumin and Cancer. Nutrients. 2019;11(10).

  41. Willenbacher E, Khan SZ, Mujica SCA, Trapani D, Hussain S, Wolf D et al. Curcumin: New Insights into an ancient ingredient against Cancer. Int J Mol Sci. 2019;20(8).

  42. Feng C, Xia Y, Zou P, Shen M, Hu J, Ying S, et al. Curcumin analog L48H37 induces apoptosis through ROS-mediated endoplasmic reticulum stress and STAT3 pathways in human lung cancer cells. Mol Carcinog. 2017;56(7):1765–77.

    Article  CAS  PubMed  Google Scholar 

  43. Liu F, Gao S, Yang Y, Zhao X, Fan Y, Ma W, et al. Antitumor activity of curcumin by modulation of apoptosis and autophagy in human lung cancer A549 cells through inhibiting PI3K/Akt/mTOR pathway. Oncol Rep. 2018;39(3):1523–31.

    CAS  PubMed  Google Scholar 

  44. Rascio F, Spadaccino F, Rocchetti MT, Castellano G, Stallone G, Netti GS et al. The pathogenic role of PI3K/AKT pathway in Cancer Onset and Drug Resistance: an updated review. Cancers. 2021;13(16).

  45. Feng M, Xiong G, Cao Z, Yang G, Zheng S, Qiu J, et al. LAT2 regulates glutamine-dependent mTOR activation to promote glycolysis and chemoresistance in pancreatic cancer. J experimental Clin cancer research: CR. 2018;37(1):274.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Xiong G, Liu C, Yang G, Feng M, Xu J, Zhao F, et al. Long noncoding RNA GSTM3TV2 upregulates LAT2 and OLR1 by competitively sponging let-7 to promote gemcitabine resistance in pancreatic cancer. J Hematol Oncol. 2019;12(1):97.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Liu Y, West SC. Distinct functions of BRCA1 and BRCA2 in double-strand break repair. Breast cancer research: BCR. 2002;4(1):9–13.

    Article  CAS  PubMed  Google Scholar 

  48. Zámborszky J, Szikriszt B, Gervai JZ, Pipek O, Póti Á, Krzystanek M, et al. Loss of BRCA1 or BRCA2 markedly increases the rate of base substitution mutagenesis and has distinct effects on genomic deletions. Oncogene. 2017;36(6):746–55.

    Article  PubMed  Google Scholar 

  49. Xu F, Wu H, Xiong J, Peng T. Long non-coding RNA DLEU2L targets mir-210-3p to suppress Gemcitabine Resistance in Pancreatic Cancer cells via BRCA2 regulation. Front Mol Biosci. 2021;8:645365.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lee S, Rauch J, Kolch W. Targeting MAPK signaling in Cancer: mechanisms of Drug Resistance and Sensitivity. Int J Mol Sci. 2020;21(3).

  51. Yang F, Li X, Zhang L, Cheng L, Li X. LncRNA TUG1 promoted viability and associated with gemcitabine resistant in pancreatic ductal adenocarcinoma. J Pharmacol Sci. 2018;137(2):116–21.

    Article  CAS  PubMed  Google Scholar 

  52. Enomoto H, Nakamura H, Nishikawa H, Nishiguchi S, Iijima H. Hepatoma-Derived growth factor: an overview and its role as a potential therapeutic target molecule for Digestive Malignancies. Int J Mol Sci. 2020;21(12).

  53. Liu Y, Wang J, Dong L, Xia L, Zhu H, Li Z, et al. Long noncoding RNA HCP5 regulates pancreatic Cancer Gemcitabine (GEM) Resistance by sponging hsa-miR-214-3p To target HDGF. OncoTargets and therapy. 2019;12:8207–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Ferrarelli LK. Treating WNT-driven colorectal cancer. Sci (New York NY). 2017;356(6345):1346–8.

    Article  Google Scholar 

  55. Moghbeli M, Sadrizadeh A, Forghanifard MM, Mozaffari HM, Golmakani E, Abbaszadegan MR. Role of Msi1 and PYGO2 in esophageal squamous cell carcinoma depth of invasion. J cell communication Signal. 2016;10(1):49–53.

    Article  Google Scholar 

  56. Levy JMM, Towers CG, Thorburn A. Targeting autophagy in cancer. Nat Rev Cancer. 2017;17(9):528–42.

    Article  CAS  PubMed  Google Scholar 

  57. Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, et al. Fusobacterium nucleatum promotes Chemoresistance to Colorectal Cancer by modulating Autophagy. Cell. 2017;170(3):548–63e16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy. 2016;12(1):1-222.

  59. Zhou C, Yi C, Yi Y, Qin W, Yan Y, Dong X, et al. LncRNA PVT1 promotes gemcitabine resistance of pancreatic cancer via activating Wnt/β-catenin and autophagy pathway through modulating the miR-619-5p/Pygo2 and miR-619-5p/ATG14 axes. Mol Cancer. 2020;19(1):118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Lu M, Qin X, Zhou Y, Li G, Liu Z, Geng X, et al. Long non-coding RNA LINC00665 promotes gemcitabine resistance of Cholangiocarcinoma cells via regulating EMT and stemness properties through miR-424-5p/BCL9L axis. Cell Death Dis. 2021;12(1):72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Xie D, Zhang H, Shang C. Long non-coding RNA CDKN2B antisense RNA 1 gene inhibits Gemcitabine sensitivity in bladder urothelial carcinoma. J Cancer. 2018;9(12):2160–6.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Huang F, Chen YG. Regulation of TGF-beta receptor activity. Cell & bioscience. 2012;2:9.

    Article  CAS  Google Scholar 

  63. Wasserman I, Lee LH, Ogino S, Marco MR, Wu C, Chen X, et al. SMAD4 loss in Colorectal Cancer Patients correlates with recurrence, loss of Immune Infiltrate, and Chemoresistance. Clin cancer research: official J Am Association Cancer Res. 2019;25(6):1948–56.

    Article  CAS  Google Scholar 

  64. Schwarte-Waldhoff I, Schmiegel W. Smad4 transcriptional pathways and angiogenesis. Int J Gastrointest Cancer. 2002;31(1–3):47–59.

    Article  CAS  PubMed  Google Scholar 

  65. Wang J, Li Y, Zhou JH, Shen FR, Shi X, Chen YG. CircATRNL1 activates Smad4 signaling to inhibit angiogenesis and ovarian cancer metastasis via miR-378. Mol Oncol. 2021;15(4):1217–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Yu Y, Zou YF, Hong RQ, Chen WJ, Chen L, Chen WQ, et al. Long non-coding RNA SNHG16 decreased SMAD4 to induce gemcitabine resistance in pancreatic cancer via EZH2-mediated epigenetic modification. Kaohsiung J Med Sci. 2022;38(10):981–91.

    Article  CAS  PubMed  Google Scholar 

  67. McDermott SP, Wicha MS. Targeting breast cancer stem cells. Mol Oncol. 2010;4(5):404–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Moghbeli M, Mosannen Mozaffari H, Memar B, Forghanifard MM, Gholamin M, Abbaszadegan MR. Role of MAML1 in targeted therapy against the esophageal cancer stem cells. J translational Med. 2019;17(1):126.

    Article  Google Scholar 

  69. Kuwano Y, Pullmann R Jr, Marasa BS, Abdelmohsen K, Lee EK, Yang X, et al. NF90 selectively represses the translation of target mRNAs bearing an AU-rich signature motif. Nucleic Acids Res. 2010;38(1):225–38.

    Article  CAS  PubMed  Google Scholar 

  70. Shim J, Lim H, Karin JRY. Nuclear export of NF90 is required for interleukin-2 mRNA stabilization. Mol Cell. 2002;10(6):1331–44.

    Article  CAS  PubMed  Google Scholar 

  71. Vumbaca F, Phoenix KN, Rodriguez-Pinto D, Han DK, Claffey KP. Double-stranded RNA-binding protein regulates vascular endothelial growth factor mRNA stability, translation, and breast cancer angiogenesis. Mol Cell Biol. 2008;28(2):772–83.

    Article  CAS  PubMed  Google Scholar 

  72. Roussos ET, Keckesova Z, Haley JD, Epstein DM, Weinberg RA, Condeelis JS. AACR special conference on epithelial-mesenchymal transition and cancer progression and treatment. Cancer research. 2010;70(19):7360-4.

  73. Zhuang J, Shen L, Yang L, Huang X, Lu Q, Cui Y, et al. TGFβ1 promotes Gemcitabine Resistance through regulating the LncRNA-LET/NF90/miR-145 Signaling Axis in bladder Cancer. Theranostics. 2017;7(12):3053–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Hu X, Li J, Fu M, Zhao X, Wang W. The JAK/STAT signaling pathway: from bench to clinic. Signal Transduct Target therapy. 2021;6(1):402.

    Article  Google Scholar 

  75. Liu B, Wu S, Ma J, Yan S, Xiao Z, Wan L, et al. lncRNA GAS5 reverses EMT and tumor stem cell-mediated Gemcitabine Resistance and Metastasis by Targeting miR-221/SOCS3 in pancreatic Cancer. Mol therapy Nucleic acids. 2018;13:472–82.

    Article  CAS  Google Scholar 

  76. Chi Y, Xin H, Liu Z. Exosomal lncRNA UCA1 derived from pancreatic stellate cells promotes Gemcitabine Resistance in Pancreatic Cancer via the SOCS3/EZH2 Axis. Front Oncol. 2021;11:671082.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Pan XN, Chen JJ, Wang LX, Xiao RZ, Liu LL, Fang ZG, et al. Inhibition of c-Myc overcomes cytotoxic drug resistance in acute myeloid leukemia cells by promoting differentiation. PLoS ONE. 2014;9(8):e105381.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Carramusa L, Contino F, Ferro A, Minafra L, Perconti G, Giallongo A, et al. The PVT-1 oncogene is a myc protein target that is overexpressed in transformed cells. J Cell Physiol. 2007;213(2):511–8.

    Article  CAS  PubMed  Google Scholar 

  79. Tseng YY, Moriarity BS, Gong W, Akiyama R, Tiwari A, Kawakami H, et al. PVT1 dependence in cancer with MYC copy-number increase. Nature. 2014;512(7512):82–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. You L, Wang H, Yang G, Zhao F, Zhang J, Liu Z, et al. Gemcitabine exhibits a suppressive effect on pancreatic cancer cell growth by regulating processing of PVT1 to miR1207. Mol Oncol. 2018;12(12):2147–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Lam EW, Brosens JJ, Gomes AR, Koo CY. Forkhead box proteins: tuning forks for transcriptional harmony. Nat Rev Cancer. 2013;13(7):482–95.

    Article  CAS  PubMed  Google Scholar 

  82. Tzivion G, Dobson M, Ramakrishnan G. FoxO transcription factors; regulation by AKT and 14-3-3 proteins. Biochim Biophys Acta. 2011;1813(11):1938–45.

    Article  CAS  PubMed  Google Scholar 

  83. Myatt SS, Brosens JJ, Lam EW. Sense and sensitivity: FOXO and ROS in cancer development and treatment. Antioxid Redox Signal. 2011;14(4):675–87.

    Article  CAS  PubMed  Google Scholar 

  84. Wang H, Zhang L, Yang X, Jin Y, Pei S, Zhang D, et al. PUMA mediates the combinational therapy of 5-FU and NVP-BEZ235 in colon cancer. Oncotarget. 2015;6(16):14385–98.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Shen S, Wang J, Zheng B, Tao Y, Li M, Wang Y, et al. LINC01714 enhances Gemcitabine Sensitivity by modulating FOXO3 phosphorylation in Cholangiocarcinoma. Mol therapy Nucleic acids. 2020;19:446–57.

    Article  CAS  Google Scholar 

  86. Segatto M, Fittipaldi R, Pin F, Sartori R, Dae Ko K, Zare H, et al. Epigenetic targeting of bromodomain protein BRD4 counteracts cancer cachexia and prolongs survival. Nat Commun. 2017;8(1):1–16.

    Article  CAS  Google Scholar 

  87. Li X, Baek G, Ramanand SG, Sharp A, Gao Y, Yuan W, et al. BRD4 promotes DNA repair and mediates the formation of TMPRSS2-ERG gene rearrangements in prostate cancer. Cell Rep. 2018;22(3):796–808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Wang Y-H, Sui Y-N, Yan K, Wang L-S, Wang F, Zhou J-H. BRD4 promotes pancreatic ductal adenocarcinoma cell proliferation and enhances gemcitabine resistance. Oncol Rep. 2015;33(4):1699–706.

    Article  CAS  PubMed  Google Scholar 

  89. Deng T, Yan G, Song X, Xie L, Zhou Y, Li J, et al. Deubiquitylation and stabilization of p21 by USP11 is critical for cell-cycle progression and DNA damage responses. Proc Natl Acad Sci. 2018;115(18):4678–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Gillis LD, Leidal AM, Hill R, Lee PW. p21Cip1/WAF1 mediates cyclin B1 degradation in response to DNA damage. Cell Cycle. 2009;8(2):253–6.

    Article  CAS  PubMed  Google Scholar 

  91. Barker HE, Patel R, McLaughlin M, Schick U, Zaidi S, Nutting CM, et al. CHK1 inhibition Radiosensitizes Head and Neck Cancers to Paclitaxel-Based ChemoradiotherapyCHK1 inhibition Chemoradiosensitizes HNSCC cells. Mol Cancer Ther. 2016;15(9):2042–54.

    Article  CAS  PubMed  Google Scholar 

  92. Shi W, Zhang C, Ning Z, Hua Y, Li Y, Chen L, et al. Long non-coding RNA LINC00346 promotes pancreatic cancer growth and gemcitabine resistance by sponging mir-188-3p to derepress BRD4 expression. J experimental Clin cancer research: CR. 2019;38(1):60.

    Article  PubMed Central  Google Scholar 

  93. McGinnis W, Krumlauf R. Homeobox genes and axial patterning. Cell. 1992;68(2):283–302.

    Article  CAS  PubMed  Google Scholar 

  94. Li Z, Zhao X, Zhou Y, Liu Y, Zhou Q, Ye H, et al. The long non-coding RNA HOTTIP promotes progression and gemcitabine resistance by regulating HOXA13 in pancreatic cancer. J translational Med. 2015;13:84.

    Article  CAS  Google Scholar 

  95. Yuan S, Liu Z, Xu Z, Liu J, Zhang J. High mobility group box 1 (HMGB1): a pivotal regulator of hematopoietic malignancies. J Hematol Oncol. 2020;13(1):91.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Wang L, Bi R, Li L, Zhou K, Yin H. lncRNA ANRIL aggravates the chemoresistance of pancreatic cancer cells to gemcitabine by targeting inhibition of miR-181a and targeting HMGB1-induced autophagy. Aging. 2021;13(15):19272–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Wei MY, Shi S, Liang C, Meng QC, Hua J, Zhang YY, et al. The microbiota and microbiome in pancreatic cancer: more influential than expected. Mol Cancer. 2019;18(1):97.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Minami K, Shinsato Y, Yamamoto M, Takahashi H, Zhang S, Nishizawa Y, et al. Ribonucleotide reductase is an effective target to overcome gemcitabine resistance in gemcitabine-resistant pancreatic cancer cells with dual resistant factors. J Pharmacol Sci. 2015;127(3):319–25.

    Article  CAS  PubMed  Google Scholar 

  99. Jeong SSR, Proteins. Binders, regulators, and Connectors of RNA. Mol Cells. 2017;40(1):1–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Lou H, Neugebauer KM, Gagel RF, Berget SM. Regulation of alternative polyadenylation by U1 snRNPs and SRp20. Mol Cell Biol. 1998;18(9):4977–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Shen T, Li H, Song Y, Li L, Lin J, Wei G, et al. Alternative polyadenylation dependent function of splicing factor SRSF3 contributes to cellular senescence. Aging. 2019;11(5):1356–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Ma S, Chen C, Ji X, Liu J, Zhou Q, Wang G, et al. The interplay between m6A RNA methylation and noncoding RNA in cancer. J Hematol Oncol. 2019;12(1):121.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Cao R, Wang L, Wang H, Xia L, Erdjument-Bromage H, Tempst P, et al. Role of histone H3 lysine 27 methylation in polycomb-group silencing. Volume 298. New York, NY: Science; 2002. pp. 1039–43. 5595.

    Google Scholar 

  104. Cao R, Zhang Y. The functions of E(Z)/EZH2-mediated methylation of lysine 27 in histone H3. Curr Opin Genet Dev. 2004;14(2):155–64.

    Article  CAS  PubMed  Google Scholar 

  105. Wang ZW, Pan JJ, Hu JF, Zhang JQ, Huang L, Huang Y, et al. SRSF3-mediated regulation of N6-methyladenosine modification-related lncRNA ANRIL splicing promotes resistance of pancreatic cancer to gemcitabine. Cell Rep. 2022;39(6):110813.

    Article  CAS  PubMed  Google Scholar 

  106. Yang X, Li L, Liang J, Shi L, Yang J, Yi X, et al. Histone acetyltransferase 1 promotes homologous recombination in DNA repair by facilitating histone turnover. J Biol Chem. 2013;288(25):18271–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Audia JE, Campbell RM. Histone modifications and Cancer. Cold Spring Harb Perspect Biol. 2016;8(4):a019521.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Sadler AJ, Suliman BA, Yu L, Yuan X, Wang D, Irving AT, et al. The acetyltransferase HAT1 moderates the NF-κB response by regulating the transcription factor PLZF. Nat Commun. 2015;6:6795.

    Article  CAS  PubMed  Google Scholar 

  109. Jin X, Tian S, Li P. Histone acetyltransferase 1 promotes cell proliferation and induces Cisplatin Resistance in Hepatocellular Carcinoma. Oncol Res. 2017;25(6):939–46.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Ougolkov AV, Bilim VN, Billadeau DD. Regulation of pancreatic tumor cell proliferation and chemoresistance by the histone methyltransferase enhancer of zeste homologue 2. Clin cancer research: official J Am Association Cancer Res. 2008;14(21):6790–6.

    Article  CAS  Google Scholar 

  111. Sun Y, Ren D, Zhou Y, Shen J, Wu H, Jin X. Histone acetyltransferase 1 promotes gemcitabine resistance by regulating the PVT1/EZH2 complex in pancreatic cancer. Cell Death Dis. 2021;12(10):878.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Avan A, Crea F, Paolicchi E, Funel N, Galvani E, Marquez VE, et al. Molecular mechanisms involved in the synergistic interaction of the EZH2 inhibitor 3-deazaneplanocin A with gemcitabine in pancreatic cancer cells. Mol Cancer Ther. 2012;11(8):1735–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Sharma N, Nanta R, Sharma J, Gunewardena S, Singh KP, Shankar S, et al. PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget. 2015;6(31):32039–60.

    Article  PubMed  PubMed Central  Google Scholar 

  114. Sergeant G, Vankelecom H, Gremeaux L, Topal B. Role of cancer stem cells in pancreatic ductal adenocarcinoma. Nat reviews Clin Oncol. 2009;6(10):580–6.

    Article  Google Scholar 

  115. Bao B, Ali S, Ahmad A, Azmi AS, Li Y, Banerjee S, et al. Hypoxia-induced aggressiveness of pancreatic cancer cells is due to increased expression of VEGF, IL-6 and miR-21, which can be attenuated by CDF treatment. PLoS ONE. 2012;7(12):e50165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Lev-Ari S, Vexler A, Starr A, Ashkenazy-Voghera M, Greif J, Aderka D, et al. Curcumin augments gemcitabine cytotoxic effect on pancreatic adenocarcinoma cell lines. Cancer Invest. 2007;25(6):411–8.

    Article  CAS  PubMed  Google Scholar 

  117. Yoshida K, Toden S, Ravindranathan P, Han H, Goel A. Curcumin sensitizes pancreatic cancer cells to gemcitabine by attenuating PRC2 subunit EZH2, and the lncRNA PVT1 expression. Carcinogenesis. 2017;38(10):1036–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. You MS, Ryu JK, Choi YH, Choi JH, Huh G, Paik WH, et al. Therapeutic outcomes and prognostic factors in unresectable gallbladder cancer treated with gemcitabine plus cisplatin. BMC Cancer. 2019;19(1):10.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Valle J, Wasan H, Palmer DH, Cunningham D, Anthoney A, Maraveyas A, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362(14):1273–81.

    Article  CAS  PubMed  Google Scholar 

  120. Dutta U. Gallbladder cancer: can newer insights improve the outcome? J Gastroenterol Hepatol. 2012;27(4):642–53.

    Article  PubMed  Google Scholar 

  121. Clark J, Lu YJ, Sidhar SK, Parker C, Gill S, Smedley D, et al. Fusion of splicing factor genes PSF and NonO (p54nrb) to the TFE3 gene in papillary renal cell carcinoma. Oncogene. 1997;15(18):2233–9.

    Article  CAS  PubMed  Google Scholar 

  122. Yamamoto R, Osawa T, Sasaki Y, Yamamoto S, Anai M, Izumi K, et al. Overexpression of p54(nrb)/NONO induces differential EPHA6 splicing and contributes to castration-resistant prostate cancer growth. Oncotarget. 2018;9(12):10510–24.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Tsofack SP, Garand C, Sereduk C, Chow D, Aziz M, Guay D, et al. NONO and RALY proteins are required for YB-1 oxaliplatin induced resistance in colon adenocarcinoma cell lines. Mol Cancer. 2011;10:145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Xue Z, Yang B, Xu Q, Zhu X, Qin G. Long non-coding RNA SSTR5-AS1 facilitates gemcitabine resistance via stabilizing NONO in gallbladder carcinoma. Biochem Biophys Res Commun. 2020;522(4):952–9.

    Article  CAS  PubMed  Google Scholar 

  125. Liu Y, Tang J, Liu D, Zhang L, He Y, Li J, et al. Increased autophagy in EOC re-ascites cells can inhibit cell death and promote drug resistance. Cell Death Dis. 2018;9(4):419.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Lu Z, Xu N, He B, Pan C, Lan Y, Zhou H, et al. Inhibition of autophagy enhances the selective anti-cancer activity of tigecycline to overcome drug resistance in the treatment of chronic myeloid leukemia. J experimental Clin cancer research: CR. 2017;36(1):43.

    Article  PubMed Central  Google Scholar 

  127. Yang Y, Jiang C, Yang Y, Guo L, Huang J, Liu X et al. Silencing of LncRNA-HOTAIR decreases drug resistance of Non-Small Cell Lung Cancer cells by inactivating autophagy via suppressing the phosphorylation of ULK1. Biochemical and biophysical research communications. 2018;497(4):1003–10.

  128. Kma L, Baruah TJ. The interplay of ROS and the PI3K/Akt pathway in autophagy regulation. Biotechnol Appl Biochem. 2022;69(1):248–64.

    Article  CAS  PubMed  Google Scholar 

  129. Yang MC, Wang HC, Hou YC, Tung HL, Chiu TJ, Shan YS. Blockade of autophagy reduces pancreatic cancer stem cell activity and potentiates the tumoricidal effect of gemcitabine. Mol Cancer. 2015;14:179.

    Article  PubMed  PubMed Central  Google Scholar 

  130. Zhang X, Zhao P, Wang C, Xin B. SNHG14 enhances gemcitabine resistance by sponging miR-101 to stimulate cell autophagy in pancreatic cancer. Biochem Biophys Res Commun. 2019;510(4):508–14.

    Article  CAS  PubMed  Google Scholar 

  131. Bear HD, Tang G, Rastogi P, Geyer CE Jr, Robidoux A, Atkins JN, et al. Bevacizumab added to neoadjuvant chemotherapy for breast cancer. N Engl J Med. 2012;366(4):310–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Li X, Yan J, Wang L, Xiao F, Yang Y, Guo X, et al. Beclin1 inhibition promotes autophagy and decreases gemcitabine-induced apoptosis in Miapaca2 pancreatic cancer cells. Cancer Cell Int. 2013;13(1):26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Liu H, He Z, von Rütte T, Yousefi S, Hunger RE, Simon HU. Down-regulation of autophagy-related protein 5 (ATG5) contributes to the pathogenesis of early-stage cutaneous melanoma. Sci Transl Med. 2013;5(202):202ra123.

    Article  PubMed  Google Scholar 

  134. Cherra SJ 3rd, Kulich SM, Uechi G, Balasubramani M, Mountzouris J, Day BW, et al. Regulation of the autophagy protein LC3 by phosphorylation. J Cell Biol. 2010;190(4):533–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Ola MS, Nawaz M, Ahsan H. Role of Bcl-2 family proteins and caspases in the regulation of apoptosis. Mol Cell Biochem. 2011;351(1–2):41–58.

    Article  CAS  PubMed  Google Scholar 

  136. Di Gesualdo F, Capaccioli S, Lulli M. A pathophysiological view of the long non-coding RNA world. Oncotarget. 2014;5(22):10976–96.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Chen YM, Liu Y, Wei HY, Lv KZ, Fu PF. Large intergenic non-coding RNA-ROR reverses gemcitabine-induced autophagy and apoptosis in breast cancer cells. Oncotarget. 2016;7(37):59604–17.

    Article  PubMed  PubMed Central  Google Scholar 

  138. An N, Cheng D. The long noncoding RNA HOST2 promotes Gemcitabine Resistance in Human Pancreatic Cancer cells. Pathol Oncol research: POR. 2020;26(1):425–31.

    Article  CAS  Google Scholar 

  139. Sherr CJ, Roberts JM. Living with or without cyclins and cyclin-dependent kinases. Genes Dev. 2004;18(22):2699–711.

    Article  CAS  PubMed  Google Scholar 

  140. Duan C, Liu Y, Lu L, Cai R, Xue H, Mao X, et al. CDK14 contributes to reactive gliosis via Interaction with Cyclin Y in Rat Model of spinal cord Injury. J Mol neuroscience: MN. 2015;57(4):571–9.

    Article  CAS  Google Scholar 

  141. Sun Y, Wang P, Yang W, Shan Y, Zhang Q, Wu H. The role of lncRNA MSC-AS1/miR-29b-3p axis-mediated CDK14 modulation in pancreatic cancer proliferation and gemcitabine-induced apoptosis. Cancer Biol Ther. 2019;20(6):729–39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Li B, Xie D, Zhang H. Long non-coding RNA GHET1 contributes to chemotherapeutic resistance to Gemcitabine in bladder cancer. Cancer Chemother Pharmacol. 2019;84(1):187–94.

    Article  CAS  PubMed  Google Scholar 

  143. An Q, Zhou L, Xu N. Long noncoding RNA FOXD2-AS1 accelerates the gemcitabine-resistance of bladder cancer by sponging miR-143. Volume 103. Biomedicine & pharmacotherapy = Biomedecine &; 2018. pp. 415–20. pharmacotherapie.

  144. Diehn M, Cho RW, Lobo NA, Kalisky T, Dorie MJ, Kulp AN, et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature. 2009;458(7239):780–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Ju HQ, Gocho T, Aguilar M, Wu M, Zhuang ZN, Fu J, et al. Mechanisms of overcoming intrinsic resistance to Gemcitabine in Pancreatic Ductal Adenocarcinoma through the Redox Modulation. Mol Cancer Ther. 2015;14(3):788–98.

    Article  CAS  PubMed  Google Scholar 

  146. Shi X, Zhang Y, Zheng J, Pan J. Reactive oxygen species in cancer stem cells. Antioxid Redox Signal. 2012;16(11):1215–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Ryoo IG, Lee SH, Kwak MK. Redox modulating NRF2: a potential mediator of Cancer Stem Cell Resistance. Oxidative Med Cell Longev. 2016;2016:2428153.

    Article  Google Scholar 

  148. Furukawa M, Xiong Y. BTB protein Keap1 targets antioxidant transcription factor Nrf2 for ubiquitination by the Cullin 3-Roc1 ligase. Mol Cell Biol. 2005;25(1):162–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Chowdhry S, Zhang Y, McMahon M, Sutherland C, Cuadrado A, Hayes JD. Nrf2 is controlled by two distinct β-TrCP recognition motifs in its Neh6 domain, one of which can be modulated by GSK-3 activity. Oncogene. 2013;32(32):3765–81.

    Article  CAS  PubMed  Google Scholar 

  150. Yang Q, Li K, Huang X, Zhao C, Mei Y, Li X, et al. lncRNA SLC7A11-AS1 promotes Chemoresistance by blocking SCF(β-TRCP)-Mediated degradation of NRF2 in pancreatic Cancer. Mol therapy Nucleic acids. 2020;19:974–85.

    Article  CAS  Google Scholar 

  151. Ye X, Wang LP, Han C, Hu H, Ni CM, Qiao GL, et al. Increased m(6)a modification of lncRNA DBH-AS1 suppresses pancreatic cancer growth and gemcitabine resistance via the miR-3163/USP44 axis. Annals of translational medicine. 2022;10(6):304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Moseley JB, Okada K, Balcer HI, Kovar DR, Pollard TD, Goode BL. Twinfilin is an actin-filament-severing protein and promotes rapid turnover of actin structures in vivo. J Cell Sci. 2006;119(Pt 8):1547–57.

    Article  CAS  PubMed  Google Scholar 

  153. Bockhorn J, Dalton R, Nwachukwu C, Huang S, Prat A, Yee K, et al. MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat Commun. 2013;4:1393.

    Article  PubMed  Google Scholar 

  154. Hua YQ, Zhu YD, Xie GQ, Zhang K, Sheng J, Zhu ZF, et al. Long non-coding SBF2-AS1 acting as a competing endogenous RNA to sponge microRNA-142-3p to participate in gemcitabine resistance in pancreatic cancer via upregulating TWF1. Aging. 2019;11(20):8860–78.

    Article  CAS  PubMed  Google Scholar 

  155. Du B, Shim JS. Targeting epithelial-mesenchymal transition (EMT) to Overcome Drug Resistance in Cancer. Molecules. 2016;21(7).

  156. Forghanifard MM, Rad A, Farshchian M, Khaleghizadeh M, Gholamin M, Moghbeli M, et al. TWIST1 upregulates the MAGEA4 oncogene. Mol Carcinog. 2017;56(3):877–85.

    Article  CAS  PubMed  Google Scholar 

  157. Xu HF, Shi DM, Zhu XQ. A study of effect of lncRNA MVIH on sensitivity of gastric cancer cells to gemcitabine. Eur Rev Med Pharmacol Sci. 2019;23(21):9651–9.

    PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

AM, YS, HL, and FTG were involved in search strategy and drafting. MM designed, revised, structured, and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Meysam Moghbeli.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Maharati, A., Samsami, Y., Latifi, H. et al. Role of the long non-coding RNAs in regulation of Gemcitabine response in tumor cells. Cancer Cell Int 23, 168 (2023). https://doi.org/10.1186/s12935-023-03004-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-023-03004-7

Keywords