Open Access

Interplay between cyclooxygenase‑2 and microRNAs in cancer (Review)

  • Authors:
    • Zexiong Gong
    • Weiguo Huang
    • Baiyun Wang
    • Na Liang
    • Songkai Long
    • Wanjun Li
    • Qier Zhou
  • View Affiliations

  • Published online on: March 11, 2021     https://doi.org/10.3892/mmr.2021.11986
  • Article Number: 347
  • Copyright: © Gong et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tumor‑associated inflammation and aberrantly expressed biomarkers have been demonstrated to play crucial roles in the cancer microenvironment. Cyclooxygenase‑2 (COX‑2), a prominent inflammatory factor, is highly expressed in tumor cells and contributes to tumor growth, recurrence and metastasis. Overexpression of COX‑2 may occur at both transcriptional and post‑transcriptional levels. Thus, an improved understanding of the regulatory mechanisms of COX‑2 can facilitate the development of novel antitumor therapies. MicroRNAs (miRNAs) are a group of small non‑coding RNAs that act as translation repressors of target mRNAs, and play vital roles in regulating cancer development and progression. The present review discusses the association between miRNAs and COX‑2 expression in different types of cancer. Understanding the regulatory role of miRNAs in COX‑2 post‑transcription can provide novel insight for suppressing COX‑2 expression via gene silencing mechanisms, which offer new perspectives and future directions for the development of novel COX‑2 selective inhibitors based on miRNAs.

Introduction

Cancer is the second leading cause of mortality worldwide, with 18.1 million new cases and 9.6 million mortalities reported in 2018 (1). Previous studies have demonstrated that there is a causal association between inflammation and carcinogenesis (2,3). In addition, inflammation notably contributes to tumor growth, progression, metastasis, recurrence and treatment resistance (2). Cyclooxygenase (COX) is classified into three isozymes: COX-1, COX-2 and COX-3 (3). COX-1 is predominantly expressed in most tissues, such as in blood vessels, stomach and kidney, and acts as a housekeeping enzyme during cellular homeostasis (2,3). COX-2 is constitutively expressed in certain pathological processes and is required to produce prostaglandin E2 (PGE2), an inflammatory mediator expressed in different types of cancer (2). COX-3 is predominantly expressed in the spinal cord and brain (4). Upregulated COX-2 expression is observed at inflammation sites that predispose to cancer development (5). However, COX-2 is expressed at relatively low levels in normal tissues adjacent to tumor tissues (6). COX-2 is considered an important therapeutic target for several diseases, including cancer, autoimmune diseases and gastric inflammation (Fig. 1). Given that COX-2 can exert pleiotropic effects on cancer development, the role of COX-2 in tumor growth and metastasis has been investigated using COX-2-specific microRNAs (miRNAs/miRs) (2,4).

Over the past years, miRNAs have been of great interest in cancer research due to their prominent role as multiple gene regulators. miRNAs are involved in every phase of malignant, premalignant and inflammatory processes, including cytokine production and immunity activation (715). miRNAs are a group of small non-coding RNAs that control mRNA stability and translation, and also regulate transcription (7). Upon complementary binding to the 3′-untralsated region (UTR) of the target mRNA, miRNAs can translationally inhibit and degrade mRNAs, which in turn decreases protein expression (7). Several studies have reported that a single miRNA can bind to >200 target genes with diverse functions, and up to one third of human mRNAs are regulated by miRNAs (8,9). Currently, several miRNAs have been identified in humans, which are expressed in a tissue-dependent manner (10). Active RNA-induced silencing complex utilizes the guide strands to cleave the target mRNAs, thereby inducing translational repression or degradation (11) and affects various cellular processes (12). miRNAs are important regulators of target genes responsible for cell proliferation, apoptosis, and/or differentiation (13). Several miRNAs, such as miR-101, have been reported to play key roles in cancer development and progression (14). Abnormal expression of miRNAs has been observed in different types of human tumors, and each tumor has a distinct miRNA signature (15). Recent studies suggest that dysregulated miRNA expression may exert detrimental effects on cell survival, particularly in cancer cells (9,13).

The present review discusses the role of COX-2 in cancer by targeting upregulated or downregulated miRNAs to provide insight on the future of molecular cancer therapy.

Roles of COX-2 in cancer

In addition to cancer cells, the networks of vascular cells, lymphatic endothelial cells, immune cells, stromal cells, endothelial cells and cancer-associated fibroblasts are alternatives but basic choices for tumor cells to invade and survive (16). Tumor-associated inflammation and aberrantly expressed biomarkers have been demonstrated to play crucial roles in the cancer microenvironment. COX-2 is released by macrophage type II cells, cancer-associated fibroblasts and tumor cells to the cancer microenvironment (16). In a healthy state, COX-2 is involved in the maintenance of cellular homeostasis; however, when homeostasis is perturbed by certain diseases, it may respond to homeostatic dysregulation and lead to the development of cancer (17). Several factors affect COX-2 expression (18), and its overexpression may be explained by the mechanisms of transcriptional and/or post-transcriptional regulation (19). The experimental results of cells and animal models have demonstrated that overexpression of COX-2 can inhibit tumor cell apoptosis, enhance cellular adhesion to achieve an invasive phenotype and promote tumor-induced angiogenesis (20,21). These theories have been confirmed in different types of tumors, including gastric (4), lung (22), pancreas (23), bladder (24), head and neck (25) and breast cancer (26). The present review discusses the effect of overexpressing COX-2 on the regulation of tumor growth and carcinogenesis.

COX-2 upregulates survivin expression to decrease apoptosis of tumor cells (27). The COX-2/PGE2 axis increases the expression levels of the pro-angiogenic proteins, surviving (28) and B-cell lymphoma 2 (29), while suppressing the transcriptional activity of caspase-3 (30). Increasing evidence support the role of COX-2 in promoting tumor cell proliferation (3135). COX-2 can promote the proliferation of cancer cells by regulating aromatase gene expression, activating neutrophils and inducing stromal cancer-associated fibroblasts (32). Furthermore, upregulated COX-2/PGE2 expression induces the levels of aromatase-catalyzed estrogen and aromatase cytochrome P450 in a paracrine manner, resulting in uncontrolled epithelial cell proliferation (33). COX-2 recruits macrophages and neutrophils to consecutively sustain proliferative signaling in cancer cells (34). Cancer-associated fibroblasts also confer a chronic proliferative signal in cancer cells (35). In addition, alterations in cell adhesion molecules are crucial for the proliferation of cancer cells. Downregulated E-cadherin expression is associated with upregulated COX-2 expression (36). Notably, COX-2 can promote cancer cell metastasis in the liver (37) and brain (38). Epithelial-to-mesenchymal transition (EMT) is an inducer of cancer invasiveness (36), and COX-2 promotes EMT by upregulating miR-526b expression (39). In addition, COX-2 induces β1-integrin and membrane proteases-like matriptase, which are involved in tumor cell invasion (40).

Tumors use multiple mechanisms to avoid recognition and destruction by the immune system (4143). In addition to tumor development and progression, COX-2 has the potential to alter the phenotype of tumor cells into an immunosuppressed milieu (41), in favor of cancer cell activation (42). In addition, COX-2/PGE2 released from tumor cells into this milieu impairs the immune responses against tumor-associated antigens by impairing cytotoxic T lymphocytes (CTLs) effector functions and causing CTLs exhaustion (43). Macrophage type 2 cells, by releasing COX-2, are involved in tumor angiogenesis, invasion and metastasis (44). In addition, COX-2 can modulate the actions of the immune system by constitutively upregulating indoleamine 2,3-dioxygenase 1 expression in human tumor cells (45). However, COX-2 knockdown significantly suppresses the degree of differentiation in genetically modified mice bearing cutaneous cancer (46) and esophageal cancer (47). Notably, the lack of differentiation is an important hallmark of cancer cells (17). A recent study demonstrated that COX-2 can initiate the formation of aggressive cancer cells from tumor-prone stem cells in mouse skin, and is involved in the occurrence and progression of epithelial cancer cells (17).

Based on previous studies, COX-2 is considered an inducer of different types of cancer, which exerts multiple functions (3145) (Fig. 2). Thus, it is essential to assess the effects of COX-2 on the tumor microenvironment to implement effective prevention measures for cancer. Notably, an improved understanding of the regulatory mechanisms of COX-2 is required to facilitate the development of novel antitumor therapies, particularly with the concomitant use of other chemotherapeutic agents.

Expression of miRNAs in cancer

Cancer is a complex, multifactorial disease characterized by uncontrolled proliferation of abnormal cells, mainly due to oncogenes or tumor suppressor genes (48). Recent studies have highlighted the importance of miRNAs in the development and progression of cancer, and deregulated miRNA expression has been observed in different types of cancer, including hepatocellular carcinoma, gastric cancer and colorectal cancer (9,13). A significant association has been reported between miRNAs and cancer incidence (48). miRNAs bind to their target oncogene or tumor suppressor gene (13). As a target of oncogenes, miR-21 is highly expressed in different types of cancer cells (49,50) and is strongly associated with immune-inflammatory responses (51). Conversely, as a target of tumor suppressor genes, miR-101 inhibits the development of cancer and is typically downregulated in tumor cells (52,53). However, some miRNAs are often cancer specific, whereby the miRNA is overexpressed in a specific type of tumor but is suppressed in other types of cancer (54).

Dysregulated expression of miRNAs is associated with increased cancer incidence, which are considered oncomiRs or anti-oncomiRs (48). Downregulated or upregulated expression of miRNAs can regulate carcinogenesis and affect cell proliferation by interfering with cell cycle regulators (48). During tumorigenesis, miRNAs control programmed cell death in cancer cells, which in turn affects the survival of these cells (7). It is speculated that upregulated expression of miRNAs may inhibit different tumor suppressor genes in cancer cells, while downregulated expression of miRNAs may suppress oncogenic transformation in healthy tissues. In addition, epigenetic mechanisms, such as DNA methylation and histone modifications, can modulate the expression of miRNAs (48). Loss of transcription factor, extragenic suppression and gene deletion can also inhibit the expression of tumor suppressive miRNAs in cancer cells (55). However, whether abnormal miRNA expression can induce the development of cancer, or whether it is a consequence of this pathological state remains largely unknown.

From the therapeutic point of view, miRNAs exhibit unique features of multi-target and effective regulation, which holds a great promise for the development of novel antitumor drugs. miRNAs are small and stable, and are not easily degradable by endogenous ribonuclease when extracted from blood and feces. This allows them to be used as promising biomarkers for early diagnosis and prognosis, with potential reflection of treatment outcome (5658). miRNAs may be therapeutically targeted in vivo (56). For example, miR-9 serves as a promising non-invasive marker for patients with breast cancer, which is detectable in blood, urine and bile samples (57,58). A meta-analysis revealed that overexpression of miR-125b predicts poor prognosis in patients with non-small cell lung cancer (NSCLC) and prostate cancer (59), suggesting that miR-125b acts as a potential biomarker for predicting poor clinical outcomes in patients with cancer.

Post-transcriptional COX-2 regulation is mediated by miRNAs

Several intracellular pathways are responsible for the increase/decrease in COX-2 protein expression in cancer cells. For example, miR-101 negatively modulates COX-2 protein expression, which in turn decreases the proliferative ability of cancer cells (60). Studies have demonstrated that the COX-2 gene comprises several putative miRNA binding sites, and its expression is associated with miRNA-mediated translational repression (53,60) (Table I). Previous studies have reported that miRNAs can bind to the 3′-UTR of the COX-2 gene, which in turn decreases its expression (60). In addition, miR-101 exerts suppressive effects on different types of cancer cells, and its expression is downregulated in glioblastoma (61), esophageal squamous cell carcinoma (62), lung cancer (63) and gastric cancer (64). Similarly, miR-101 re-expression inhibits angiogenesis and cell proliferation of aggressive endometrial carcinoma via COX-2 activation (53). Given that miR-101 has low or null toxicity (60), it can serve as a novel class of COX-2 selective inhibitor for the treatment and prevention of cancer.

Table I.

COX-2 regulation mediated by miRNAs.

Table I.

COX-2 regulation mediated by miRNAs.

miRNATarget gene/pathway/proteinRegulation of COX-2Types of cancerFunction(Refs.)
miR-101COX-2 mRNADownregulatedProstate cancerProliferation (−)(60)
Growth (−)
Endometrial carcinomaAngiogenesis (−)(53)
miR-143-5pCOX-2 mRNADownregulatedGCGrowth (−)(69)
miR-144COX-2 mRNADownregulated Proliferation (−)(70)
Apoptosis (+)
miR-30a-5pCOX-2 mRNADownregulated Growth (−)(71)
Migration (−)
miR-137COX-2 mRNADownregulated Proliferation (−)(72)
Migration (−)
miR-1297COX-2 mRNADownregulatedCRCGrowth (−)(73)
Migration (−)
Invasion (−)
miR-216a-3pCOX-2 mRNADownregulated Growth (−)(74)
Proliferation (−)
miR16COX-2 mRNADownregulatedHCCProliferation (−)(77)
HUR Apoptosis (+)
miR-136COX-2 mRNADownregulated Proliferation (−)(78)
Migration (−)
Invasion (−)
miR-146aCOX-2DownregulatedLung cancer-(80)
miR-26bCOX-2 mRNADownregulated Proliferation (−)(81)
Migration (−)
Invasion (−)
miR-144-3pWT1DDownregulated Proliferation (−)(83)
miR-221/222PTENUpregulatedBreast cancerProliferation (+)(84)
Migration (+)
Invasion (+)
miR-27aZBTB10-protein pathwayUpregulatedOvarian epithelial cancerAngiogenesis (+)(87)
miR-128COX-2 mRNADownregulatedGliomaProliferation (−)(88)
Invasion (−)
miR-26bCOX-2 mRNADownregulated Migration (−)(89)
Invasion (−)
miR-137COX-2 mRNADownregulatedRBProliferation (−)(91)
Migration (−)
miR-143COX-2 mRNADownregulatedBladder cancerProliferation (−)(92)
Migration (−)
miR-203COX-2 mRNADownregulatedLaryngeal carcinomaProliferation (−)(93)

[i] COX-2, cyclooxygenase-2; miRNA/miR, microRNA; HUR, Human Antigen R; WT1, Wilms' tumor 1; PTEN, phosphatase and tensin homolog; GC, gastric cancer; CRC, colorectal cancer; HCC, hepatocellular carcinoma; RB, retinoblastoma; -, unknown; (−), inhibition; (+), promotion.

Gastrointestinal tumors

Increasing evidence suggest that COX-2 plays an important role in gastrointestinal tumors (6572). miRNAs exert either oncogenic or tumor suppressive roles in gastrointestinal tumors by regulating their target genes (66,67). A previous study reported that miR-143 expression is markedly downregulated in gastric cancer (GC), which is positively associated with GC progression (68). Further analysis revealed that miR-143 can bind to the 3′-UTR of COX-2, and COX-2 protein expression was downregulated following transfection with miR-143 (69). Furthermore, the results of a dual-luciferase reporter assay demonstrated that miR-144 directly targets and suppresses COX-2 expression, thus inhibiting the proliferation of GC cells (70). Taken together, these findings suggest that miR-143 and miR-144 may serve as potential diagnostic biomarkers and therapeutic targets for patients with GC. In addition, both in vivo and in vitro experimental results have demonstrated that miR-30a-3p inhibits the proliferation and migration of Helicobacter pylori-infected GC cells by targeting COX-2 mRNA (71). Furthermore, miR-137 suppresses COX-2 expression, and upregulated expression may decrease the aggressive properties of cancer cells (72). Collectively, these findings suggest a strong association between miR-137, miR-143, miR-144, miR-101, miR-30a-3p and COX-2 expression in GC, which can help further understand the role of miRNAs in GC by targeting COX-2.

Colorectal cancer (CRC) is one of the most common human malignancies (73). miR-1297 has been demonstrated to downregulate COX-2 expression, and its expression is markedly lower in CRC tissues compared with normal adjacent tissues (73). miR-1297 directly binds to the 3′-UTR of COX-2, and decreased COX-2 expression has been observed in HCT116 and LOVO cells overexpressing miR-1297 (73). In addition, low miR-216a-3p expression markedly enhances the proliferation of CRC cells. Reverse transcription-quantitative PCR and western blot analyses and dual-luciferase reporter assays have demonstrated that miR-216a-3p regulates COX-2 expression by directly targeting its 3′-UTR (74). Further analysis revealed that miR-216a-3p can suppress COX-2 expression in CRC cells (74). A previous study has demonstrated that miR-155 promotes COX-2 expression during inflammation, whereas its downregulation diminishes carcinogenesis (75). Several miRNAs, such as miR-26, miR-155, miR-101 and miR-1297, are involved in the regulation of COX-2 during carcinogenesis (7375). Thus, these miRNAs may serve as promising targets for inhibiting COX-2 expression in CRC.

In most cases, chronic liver inflammation and the inflammation-associated microenvironment can promote the initiation and progression of hepatocellular carcinoma (HCC) (76). The COX-2/PGE2 pathway plays an essential role in mediating the pathophysiology of liver diseases, including cirrhosis and HCC (76). A previous study demonstrated that miR-16 directly silences COX-2 expression in HCC cells and indirectly through downregulation of human antigen R (77). In addition, miR-16 suppresses cell proliferation and induces cell apoptosis in HCC cell lines by downregulating COX-2 expression (77). Notably, there is no significant association between miR-101 and COX-2 expression in HCC. This may be due to the tumor tissue-specific expression of miRNAs. The latest report indicates that miR-136 expression is markedly downregulated in HCC cells and tissues, and negatively associated with COX-2 mRNA expression (78). Taken together, these results suggest that miR-136 plays a key role in regulating HCC cell proliferation and metastasis by targeting COX-2.

Lung cancer

Chronic inflammation serves a key role in the pathogenesis of lung cancer (79). It has been reported that overexpression of COX-2 promotes the initiation and progression of NSCLC and other types of lung tumors (79). A previous study has demonstrated that miR-146a negatively regulates the production of cytokines and chemokines from lung cancer cells, and downregulates COX-2 expression by destabilizing its mRNA (80). In addition, miR-26b can inhibit the proliferation, migration and invasion of lung cancer cells by directly targeting COX-2 (81,82). When one of the signaling pathways is truncated or suppressed, the others may enrich their functions to maintain a stable molecular dynamic. The latest research has demonstrated that COX-2 protein expression markedly increases following suppression of the IL-1β/miR-144-3p/WT1D signaling pathway via transfection with miR-144-3p mimic (83).

Gynecological cancer types

Aberrant angiogenesis is associated with cancer progression and metastasis, and is mediated by miR-101. Upregulated miR-101 expression can slow tumor growth, the effects of which are reversed following downregulation of miR-101 expression (53). A previous study reported that miR-101 regulates abnormal angiogenesis in endometrial cancer via COX-2 (53). COX-2 expression has been observed in nearly 40% of patients with primary breast cancer, at both pre-invasive and invasive stages of the disease. In addition, COX-2 expression is significantly associated with breast cancer progression (84,85). Through downregulation of phosphatase and tensin homolog deleted on chromosome 10, miR-221/222 induces AKT phosphorylation and subsequently activates the COX-2 gene, which increases COX-2 expression in cancer cells (84). Thus, miR-221/222 induces tumor growth and maintains breast cancer stem-like characteristics by upregulating COX-2 expression (84). Notably, overexpression of COX-2 upregulates the expression levels of miR-526b and miR-655 in breast cancer cell lines (85). Furthermore, miR-526b expression is upregulated via the COX-2 and EP4 pathways in high-grade primary breast tumors (39). According to the gonadotropin theory, ovarian cancer commonly occurs in postmenopausal women, mainly due to the high levels of follicle-stimulating hormone and luteinizing hormone resulting from the negative feedback of estrogen (86). Follicle-stimulating hormone increases miR-27a expression, which in turn increases the expression levels of COX-2, survivin and vascular endothelial growth factor via the ZBTB10-specificity protein pathway (87).

Other types of cancer

Glioma is the most common type of brain tumor. Downregulated miR-128/26b expression and upregulated COX-2 expression have been detected in glioma tissues (88,89). Transfection with miR-128 and miR-26b mimics decreases the luciferase activity associated with the 3′-UTR of COX-2, and miR-128 notably decreases the stability of COX-2 mRNA. Conversely, transfection with miR-128 inhibitor markedly increases COX-2 mRNA expression, as well as the protein expression levels of ki67 and MMP9, while promoting the growth of glioma tumors (88,89). The direct association between COX-2 and miR-128/26b has been verified by publicly available data (88,89). Retinoblastoma is the most common type of eye cancer, which accounts for high mortality rates in young children (90). Bioinformatic analysis has demonstrated that miR-137 negatively modulates COX-2 expression and PGE2 production in retinoblastoma cells (91). Targeted COX-2 knockdown inhibits the invasion and increases the proliferation of retinoblastoma cells, while inhibiting the synthesis of PGE2 (91). Bladder cancer is a common malignancy of the urinary tract and the fifth most diagnosed cancer type in Western countries (92). The latest report suggests that miR-143 negatively regulates COX-2 expression in bladder carcinoma T24 cells (93). In addition, it has been reported that the role of miR-143 in tumor growth and migration is mainly due to its involvement in the COX-2 pathway (93). Laryngeal cancer is the most common type of cancer in the head and neck area (94). A previous study revealed that miR-203 expression was notably downregulated in laryngeal squamous cell carcinoma tissues (94). Collectively, this finding suggest that miR-203 acts as a tumor suppressor in laryngeal squamous cell carcinoma, partially by regulating COX-2 expression.

Upregulated expression of miRNAs by COX-2 selective inhibitor

Currently, there are three methods used to inhibit COX-2 expression, post-transcriptional control, inhibitory transcription factors and COX-2 inhibitors. Celecoxib was the first COX-2 inhibitor approved by the FDA. This drug has been used for over 20 years as an anti-inflammatory, analgesic and antipyretic agent (95). Given the role of inflammation in carcinogenesis, celecoxib has gained a novel opportunity for its application (95). The antitumor and chemoprevention effects of celecoxib on colon carcinogenesis were first demonstrated in rats (96), and later in different in vivo experimental models (97).

Celecoxib inhibits the migration, invasion and EMT of bladder cancer cells, partially by regulating the miR-145/TGFBR2/Smad3 pathway (98). In addition, the concomitant use of celecoxib and miR-145 mimic notably inhibits the migration and invasion of bladder cancer cells (98). The results of other experiments also suggest that celecoxib increases miR-146a expression in high-risk human papillomavirus (HPV) (99). Another miRNA investigated in this experiment was miR-150. miR-150 is positively mediated by NF-kB (99), a common transcription factor expressed in HPV-related cancer types (100). In this experiment, celecoxib also downregulated the NF-kB pathway via miR-150. Taken together, these findings suggest that the antitumor effect of celecoxib against HPV-induced lesions is partly mediated by upregulating miR-146a and miR-150 expression (101). Furthermore, miRNA microarray analysis has demonstrated that miR-29c expression is markedly higher in GC tissues compared with normal gastric mucosa, and celecoxib can promote miR-29c expression in GC cells (102). In addition, Mcl-1 is a target of miR-29, which encodes Bcl-2-like antiapoptotic proteins (103). A previous study reported that miR-29 regulates cell apoptosis by targeting Mcl-1 (103). Celecoxib increases miR-29c expression and inhibits its target oncogene, Mcl-1, resulting in the apoptosis of GC cells (102). In addition, miR526b/miR655 expression is significantly higher in breast tumors, and the interplay between COX-2 and hypoxia has been demonstrated to promote tumor aggression (104). Previous studies have demonstrated that celecoxib regulates hypoxia-enhanced function in breast cancer cells by downregulating miR526b/miR655 expression (104,105).

Parecoxib is another important selective COX-2 inhibitor, with high postoperative pain control and less side effects (106). Treatment with parecoxib has exhibited a promising anticancer role in different types of human cancer (106,107). The latest research suggests that parecoxib inhibits the proliferation, migration and invasion of glioblastoma cells by upregulating miR-29c expression (108).

Despite the extensive use of COX-2 inhibitors in the treatment of cancer, their application is limited due to the associated adverse events. The most reported side effect of celecoxib is the increased frequency of cardiovascular disorders following its long-term use (109). Considering that post-transcriptional control may exert a more appreciable effect, it has attracted great interest. Although the expression patterns of miRNAs are not yet fully understood in human cells, their functional roles represent one of the most exciting topics for elucidating the molecular mechanisms underlying the therapeutic effects of COX-2 inhibitors (Table II).

Table II.

Effect of COX-2 selective inhibitors in suppressing cancer by regulating miRNA expression.

Table II.

Effect of COX-2 selective inhibitors in suppressing cancer by regulating miRNA expression.

COX-2 selective inhibitorModelTarget miRNARegulation of miRNAEffect(Refs.)
CelecoxibBladder cancer cellsmiR-145UpregulatedMigration (−)(98)
Invasion (−)
MousemiR-150Upregulated Tumor-associated(101)
inflammation (−)
Gastric cancer cellsmiR-29cUpregulatedApoptosis (+)(102)
Breast cancer cellsmiR-256bDownregulatedMigration (−)(104)
miR-655 Invasion (−)
ParecoxibGlioblastoma cellsmiR-29cUpregulatedProliferation (−)(108)
Migration (−)
Invasion (−)

[i] miRNA/miR, microRNA; COX-2, cyclooxygenase-2; (−), inhibition; (+), promotion.

Conclusions

Based on the current literature regarding miRNA-mediated COX-2 regulation, a novel COX-2 selective inhibitor may be developed with miRNAs. It was suggested that anti-COX-2 miRNAs may serve as novel targets for the treatment of cancer. In addition, small interfering (si)RNAs, instead of miRNAs, may also be used to inhibit COX-2 expression via similar molecular mechanisms (97). However, off-target effects may exist due to mRNA destabilization by identical siRNA sequences, thus suppressing other mRNAs with partial complementarity (96). Taken together, the results discussed here suggest that miRNA-based strategies hold great promise for inhibiting COX-2 expression, and thus may be used to treat cancer types overexpressing COX-2.

Acknowledgements

Not applicable.

Funding

The present review was supported by the Chinese National Science Foundation (grant no. 81172210) and the China Postdoctoral Science Foundation (grant no. 2012M521528).

Availability of data and materials

Not applicable.

Authors' contributions

WGH and BYW conceived the present review. ZXG performed the literature review and revised the manuscript for important intellectual content. NL prepared the figures. SKL, WJL and QZ interpreted the table data. ZXG and BYW confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Piotrowski I, Kulcenty K and Suchorska W: Interplay between inflammation and cancer. Rep Pract Oncol Radiother. 25:422–427. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Beeghly-Fadiel A, Wilson AJ, Keene S, Ramahi M, Xu S, Marnett LJ, Fadare O, Crispens MA and Khabele D: Differential cyclooxygenase expression levels and survival associations in type I and type II ovarian tumors. J Ovarian Res. 11:172018. View Article : Google Scholar : PubMed/NCBI

4 

Ayiomamitis GD, Notas G, Vasilakaki T, Tsavari A, Vederaki S, Theodosopoulos T, Kouroumalis E and Zaravinos A: Understanding the Interplay between COX-2 and hTERT in colorectal cancer using a multi-omics analysis. Cancers (Basel). 11:15362019. View Article : Google Scholar

5 

Pollock JK, Greene LM, Nathwani SM, Kinsella P, O'Boyle NM, Meegan MJ and Zisterer DM: Involvement of NF-κB in mediating the anti-tumour effects of combretastatins in T cells. Invest New Drugs. 36:523–535. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Gurram B, Zhang S, Li M, Li H, Xie Y, Cui H, Du J, Fan J, Wang J and Peng X: Celecoxib conjugated fluorescent probe for identification and discrimination of cyclooxygenase-2 enzyme in cancer cells. Anal Chem. 90:5187–5193. 2018. View Article : Google Scholar : PubMed/NCBI

7 

O'Brien J, Hayder H, Zayed Y and Peng C: Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne). 9:4022018. View Article : Google Scholar : PubMed/NCBI

8 

Esquela-Kerscher A and Slack FJ: Oncomirs microRNAs with a role in cancer. Nat Rev Cancer. 6:259–269. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Sun X, Ge X, Xu Z and Chen D: Identification of circular RNA-microRNA-messenger RNA regulatory network in hepatocellular carcinoma by integrated analysis. J Gastroenterol Hepatol. 35:157–164. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Griffiths-Jones S, Saini HK, van Dongen S and Enright AJ: miRBase: Tools for microRNA genomics. Nucleic Acids Res. 36:D154–D158. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Kim B, Jeong K and Kim VN: Genome-wide mapping of DROSHA cleavage sites on primary MicroRNAs and noncanonical substrates. Mol Cell. 66:258–269.e5. 2017. View Article : Google Scholar : PubMed/NCBI

12 

He L and Hannon GJ: MicroRNAs: Small RNAs with a big role in gene regulation. Nat Rev Genet. 5:522–531. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Babaei K, Shams S, Keymoradzadeh A, Vahidi S, Hamami P, Khaksar R, Norollahi SE and Samadani AA: An insight of microRNAs performance in carcinogenesis and tumorigenesis; an overview of cancer therapy. Life Sci. 240:1170772020. View Article : Google Scholar : PubMed/NCBI

14 

Calin GA and Croce CM: MicroRNA signatures in human cancers. Nat Rev Cancer. 6:857–866. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Lei X, Lei Y, Li JK, Du WX, Li RG, Yang J, Li J, Li F and Tan HB: Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 470:126–133. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Moon H, White AC and Borowsky AD: New insights into the functions of Cox-2 in skin and esophageal malignancies. Exp Mol Med. 52:538–547. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Han F, Ren J, Zhang J, Sun Y, Ma F, Liu Z, Yu H, Jia J and Li W: JMJD2B is required for Helicobacter pylori-induced gastric carcinogenesis via regulating COX-2 expression. Oncotarget. 7:38626–38637. 2016. View Article : Google Scholar : PubMed/NCBI

19 

Liu Y, Borchert GL, Surazynski A and Phang JM: Proline oxidase, a p53-induced gene, targets COX-2/PGE2 signaling to induce apoptosis and inhibit tumor growth in colorectal cancers. Oncogene. 27:6729–6737. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Hashemi Goradel N, Najafi M, Salehi E, Farhood B and Mortezaee K: Cyclooxygenase-2 in cancer: A review. J Cell Physiol. 234:5683–5699. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Montezuma MAP, Fonseca FP, Benites BM, Soares CD, do Amaral-Silva GK, de Almeida OP, Soares FA, Pagano RL and Fregnani ER: COX-2 as a determinant of lower disease-free survival for patients affected by ameloblastoma. Pathol Res Pract. 214:907–913. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Han L, Fang S, Li G, Wang M and Yu R: Total flavonoids suppress lung cancer growth via the COX-2-mediated Wnt/β-catenin signaling pathway. Oncol Lett. 19:1824–1830. 2020.PubMed/NCBI

23 

Conejo-Garcia JR: Breaking barriers for T cells by targeting the EPHA2/TGF-β/COX-2 axis in pancreatic cancer. J Clin Invest. 129:3521–3523. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Bourn J, Pandey S, Uddin J, Marnett L and Cekanova M: Detection of tyrosine kinase inhibitors-induced COX-2 expression in bladder cancer by fluorocoxib A. Oncotarget. 10:5168–5180. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Zhu Y, Shi C, Zeng L, Liu G, Jiang W, Zhang X, Chen S, Guo J, Jian X, Ouyang J, et al: High COX-2 expression in cancer-associated fibiroblasts contributes to poor survival and promotes migration and invasiveness in nasopharyngeal carcinoma. Mol Carcinog. 59:265–280. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Peng Y, Wang Y, Tang N, Sun D, Lan Y, Yu Z, Zhao X, Feng L, Zhang B, Jin L, et al: Andrographolide inhibits breast cancer through suppressing COX-2 expression and angiogenesis via inactivation of p300 signaling and VEGF pathway. J Exp Clin Cancer Res. 37:2482018. View Article : Google Scholar : PubMed/NCBI

27 

Yang Y, Zhu J, Gou H, Cao D, Jiang M and Hou M: Clinical significance of Cox-2, Survivin and Bcl-2 expression in hepatocellular carcinoma (HCC). Med Oncol. 28:796–803. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Garrido MP, Hurtado I, Valenzuela-Valderrama M, Salvatierra R, Hernández A, Vega M, Selman A, Quest AFG and Romero C: NGF-enhanced vasculogenic properties of epithelial ovarian cancer cells is reduced by inhibition of the COX-2/PGE2 signaling Axis. Cancers (Basel). 11:19702019. View Article : Google Scholar

29 

Hosseini F, Mahdian-Shakib A, Jadidi-Niaragh F, Enderami SE, Mohammadi H, Hemmatzadeh M, Mohammed HA, Anissian A, Kokhaei P, Mirshafiey A and Hassannia H: Anti-inflammatory and anti-tumor effects of α-l-guluronic acid (G2013) on cancer-related inflammation in a murine breast cancer model. Biomed Pharmacother. 98:793–800. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Janakiraman H, House RP, Talwar S, Courtney SM, Hazard ES, Hardiman G, Mehrotra S, Howe PH, Gangaraju V and Palanisamy V: Repression of caspase-3 and RNA-binding protein HuR cleavage by cyclooxygenase-2 promotes drug resistance in oral squamous cell carcinoma. Oncogene. 36:3137–3148. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Raj V, Bhadauria AS, Singh AK, Kumar U, Rai A, Keshari AK, Kumar P, Kumar D, Maity B, Nath S, et al: Novel 1,3,4-thiadiazoles inhibit colorectal cancer via blockade of IL-6/COX-2 mediated JAK2/STAT3 signals as evidenced through data-based mathematical modeling. Cytokine. 118:144–159. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Krishnamachary B, Stasinopoulos I, Kakkad S, Penet MF, Jacob D, Wildes F, Mironchik Y, Pathak AP, Solaiyappan M and Bhujwalla ZM: Breast cancer cell cyclooxygenase-2 expression alters extracellular matrix structure and function and numbers of cancer associated fibroblasts. Oncotarget. 8:17981–17994. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Esbona K, Yi Y, Saha S, Yu M, Van Doorn RR, Conklin MW, Graham DS, Wisinski KB, Ponik SM, Eliceiri KW, et al: The presence of cyclooxygenase 2, tumor-associated macrophages, and collagen alignment as prognostic markers for invasive breast carcinoma patients. Am J Pathol. 188:559–573. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Esbona K, Inman D, Saha S, Jeffery J, Schedin P, Wilke L and Keely P: COX-2 modulates mammary tumor progression in response to collagen density. Breast Cancer Res. 18:352016. View Article : Google Scholar : PubMed/NCBI

35 

Hull MA, Cuthbert RJ, Ko CWS, Scott DJ, Cartwright EJ, Hawcroft G, Perry SL, Ingram N, Carr IM, Markham AF, et al: Paracrine cyclooxygenase-2 activity by macrophages drives colorectal adenoma progression in the ApcMin/+ mouse model of intestinal tumorigenesis. Sci Rep. 7:60742017. View Article : Google Scholar : PubMed/NCBI

36 

Watanabe Y, Imanishi Y, Ozawa H, Sakamoto K, Fujii R, Shigetomi S, Habu N, Otsuka K, Sato Y, Sekimizu M, et al: Selective EP2 and Cox-2 inhibition suppresses cell migration by reversing epithelial-to-mesenchymal transition and Cox-2 overexpression and E-cadherin downregulation are implicated in neck metastasis of hypopharyngeal cancer. Am J Transl Res. 12:1096–1113. 2020.PubMed/NCBI

37 

Sorski L, Melamed R, Matzner P, Lavon H, Shaashua L, Rosenne E and Ben-Eliyahu S: Reducing liver metastases of colon cancer in the context of extensive and minor surgeries through beta-adrenoceptors blockade and COX2 inhibition. Brain Behav Immun. 58:91–98. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Soto MS, O'Brien ER, Andreou K, Scrace SF, Zakaria R, Jenkinson MD, O'Neill E and Sibson NR: Disruption of tumour-host communication by downregulation of LFA-1 reduces COX-2 and e-NOS expression and inhibits brain metastasis growth. Oncotarget. 7:52375–52391. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Majumder M, Landman E, Liu L, Hess D and Lala PK: COX-2 elevates oncogenic miR-526b in breast cancer by EP4 activation. Mol Cancer Res. 13:1022–1033. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Pan J, Yang Q, Shao J, Zhang L, Ma J, Wang Y, Jiang BH, Leng J and Bai X: Cyclooxygenase-2 induced β1-integrin expression in NSCLC and promoted cell invasion via the EP1/MAPK/E2F-1/FoxC2 signal pathway. Sci Rep. 6:338232016. View Article : Google Scholar : PubMed/NCBI

41 

Lang S, Picu A, Hofmann T, Andratschke M, Mack B, Moosmann A, Gires O, Tiwari S and Zeidler R: COX-inhibitors relieve the immunosuppressive effect of tumor cells and improve functions of immune effectors. Int J Immunopathol Pharmacol. 19:409–419. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Höing B, Kanaan O, Altenhoff P, Petri R, Thangavelu K, Schlüter A, Lang S, Bankfalvi A and Brandau S: Stromal versus tumoral inflammation differentially contribute to metastasis and poor survival in laryngeal squamous cell carcinoma. Oncotarget. 9:8415–8426. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Mortezaee K: Immune escape: A critical hallmark in solid tumors. Life Sci. 258:1181102020. View Article : Google Scholar : PubMed/NCBI

44 

Miao J, Lu X, Hu Y, Piao C, Wu X, Liu X, Huang C, Wang Y, Li D and Liu J: Prostaglandin E 2 and PD-1 mediated inhibition of antitumor CTL responses in the human tumor microenvironment. Oncotarget. 8:89802–89810. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Hennequart M, Pilotte L, Cane S, Hoffmann D, Stroobant V, Plaen E and Van den Eynde BJ: Constitutive IDO1 expression in human tumors is driven by cyclooxygenase-2 and mediates intrinsic immune resistance. Cancer Immunol Res. 5:695–709. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Moon H, Kim D, Donahue LR and White AC: Phenotypic plasticity of cutaneous squamous cell carcinoma mediated by cyclooxygenase-2. J Invest Dermatol. 140:1665–1669, e1665. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Moon H, Zhu J, Donahue LR, Choi E and White AC: Krt5+/Krt15+ foregut basal progenitors give rise to cyclooxygenase-2-dependent tumours in response to gastric acid stress. Nat Commun. 10:22252019. View Article : Google Scholar : PubMed/NCBI

48 

Xiang Y, Tian Q, Guan L and Niu SS: The dual role of miR-186 in cancers: Oncomir battling with tumor suppressor miRNA. Front Oncol. 10:2332020. View Article : Google Scholar : PubMed/NCBI

49 

Bai J, Xu J, Zhao J and Zhang R: LncRNA NBR2 suppresses migration and invasion of colorectal cancer cells by downregulating miRNA-21. Hum Cell. 33:98–103. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Zhang W, Chen J and He G, Xu W and He G: Impact of mirna-21 on survival prognosis in patients with pancreatic cancer: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 99:e220452020. View Article : Google Scholar : PubMed/NCBI

51 

Bascuñán KA, Pérez-Bravo F, Gaudioso G, Vaira V, Roncoroni L, Elli L, Monguzzi E and Araya M: A miRNA-based blood and mucosal approach for detecting and monitoring celiac disease. Dig Dis Sci. 65:1982–1991. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Irimie AI, Braicu C, Sonea L, Zimta AA, Cojocneanu-Petric R, Tonchev K, Mehterov N, Diudea D, Buduru S and Berindan-Neagoe I: A looking-glass of non-coding RNAs in oral cancer. Int J Mol Sci. 18:26202017. View Article : Google Scholar

53 

Liu Y, Li H, Zhao C and Jia H: MicroRNA-101 inhibits angiogenesis via COX-2 in endometrial carcinoma. Mol Cell Biochem. 448:61–69. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Pop-Bica C, Pintea S, Cojocneanu-Petric R, Del Sal G, Piazza S, Wu ZH, Alencar AJ, Lossos IS, Berindan-Neagoe I and Calin GA: MiR-181 family-specific behavior in different cancers: a meta-analysis view. Cancer Metastasis Rev. 37:17–32. 2018. View Article : Google Scholar : PubMed/NCBI

55 

Ruan K, Fang X and Ouyang G: MicroRNAs: Novel regulators in the hallmarks of human cancer. Cancer Lett. 285:116–126. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Villadsen SB, Bramsen JB, Ostenfeld MS, Wiklund ED, Fristrup N, Gao S, Hansen TB, Jensen TI, Borre M, Ørntoft TF, et al: The miR-143/-145 cluster regulates plasminogen activator inhibitor-1 in bladder cancer. Br J Cancer. 106:366–374. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Aldebasi YH, Rahmani AH, Khan AA and Aly SM: The effect of vascular endothelial growth factor in the progression of bladder cancer and diabetic retinopathy. Int J Clin Exp Med. 6:239–251. 2013.PubMed/NCBI

58 

Li X, Zeng Z, Wang J, Wu Y, Chen W, Zheng L, Xi T, Wang A and Lu Y: MicroRNA-9 and breast cancer. Biomed Pharmacother. 122:1096872020. View Article : Google Scholar : PubMed/NCBI

59 

Stiegelbauer V, Perakis S, Deutsch A, Ling H, Gerger A and Pichler M: MicroRNAs as novel predictive biomarkers and therapeutic targets in colorectal cancer. World J Gastroenterol. 20:11727–11735. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Hao Y, Gu X, Zhao Y, Greene S, Sha W, Smoot DT, Califano J, Wu TC and Pang X: Enforced expression of miR-101 inhibits prostate cancer cell growth by modulating the COX-2 pathway in vivo. Cancer Prev Res (Phila). 4:1073–1083. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Smits M, Nilsson J, Mir SE, van der Stoop PM, Hulleman E, Niers JM, de Witt Hamer PC, Marquez VE, Cloos J, Krichevsky AM, et al: miR-101 is down-regulated in glioblastoma resulting in EZH2-induced proliferation, migration, and angiogenesis. Oncotarget. 1:710–720. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Shao Y, Li P, Zhu ST, Yue JP, Ji XJ, He Z, Ma D, Wang L, Wang YJ, Zong Y, et al: Cyclooxygenase-2, a potential therapeutic target, is regulated by miR-101 in esophageal squamous cell carcinoma. PLoS One. 10:e01406422015. View Article : Google Scholar : PubMed/NCBI

63 

Lv P, Zhang P, Li X and Chen Y: Micro ribonucleic acid (RNA)-101 inhibits cell proliferation and invasion of lung cancer by regulating cyclooxygenase-2. Thorac Cancer. 6:778–784. 2015. View Article : Google Scholar : PubMed/NCBI

64 

He XP, Shao Y, Li XL, Xu W, Chen GS, Sun HH, Xu HC, Xu X, Tang D, Zheng XF, et al: Downregulation of miR-101 in gastric cancer correlates with cyclooxygenase-2 overexpression and tumor growth. FEBS J. 279:4201–4212. 2012. View Article : Google Scholar : PubMed/NCBI

65 

Nagaraju GP and El-Rayes BF: Cyclooxygenase-2 in gastrointestinal malignancies. Cancer. 125:1221–1227. 2019. View Article : Google Scholar : PubMed/NCBI

66 

Wang J, Ding Y, Wu Y and Wang X: Identification of the complex regulatory relationships related to gastric cancer from lncRNA-miRNA-mRNA network. J Cell Biochem. 121:876–887. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Liu G and Li B: Role of miRNA in transformation from normal tissue to colorectal adenoma and cancer. J Cancer Res Ther. 15:278–285. 2019.PubMed/NCBI

68 

Takagi T, Iio A, Nakagawa Y, Naoe T, Tanigawa N and Akao Y: Decreased expression of microRNA-143 and −145 in human gastric cancers. Oncology. 77:12–21. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Wu XL, Cheng B, Li PY, Huang HJ, Zhao Q, Dan ZL, Tian DA and Zhang P: MicroRNA-143 suppresses gastric cancer cell growth and induces apoptosis by targeting COX-2. World J Gastroenterol. 19:7758–7765. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Yao Q, Gu A, Wang Z and Xue Y: MicroRNA-144 functions as a tumor suppressor in gastric cancer by targeting cyclooxygenase-2. Exp Ther Med. 15:3088–3095. 2018.PubMed/NCBI

71 

Liu X, Ji Q, Zhang C, Liu X, Liu Y, Liu N, Sui H, Zhou L, Wang S and Li Q: miR-30a acts as a tumor suppressor by double-targeting COX-2 and BCL9 in H. pylori gastric cancer models. Sci Rep. 7:71132017. View Article : Google Scholar : PubMed/NCBI

72 

Cheng Y, Li Y, Liu D, Zhang R and Zhang J: miR-137 effects on gastric carcinogenesis are mediated by targeting Cox-2-activated PI3K/AKT signaling pathway. FEBS Lett. 588:3274–3281. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Chen P, Wang BL, Pan BS and Guo W: MiR-1297 regulates the growth, migration and invasion of colorectal cancer cells by targeting cyclo-oxygenase-2. Asian Pac J Cancer Prev. 15:9185–9190. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Wang D, Li Y, Zhang C, Li X and Yu J: MiR-216a-3p inhibits colorectal cancer cell proliferation through direct targeting COX-2 and ALOX5. J Cell Biochem. 119:1755–1766. 2018. View Article : Google Scholar : PubMed/NCBI

75 

Chakraborty C, Sharma AR, Sharma G and Lee SS: The interplay among miRNAs, major cytokines, and cancer-related inflammation. Mol Ther Nucleic Acids. 20:606–620. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Yang YM, Kim SY and Seki E: Inflammation and liver cancer: Molecular mechanisms and therapeutic targets. Semin Liver Dis. 39:26–42. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Agra Andrieu N, Motiño O, Mayoral R, Llorente Izquierdo C, Fernández-Alvarez A, Boscá L, Casado M and Martín-Sanz P: Cyclooxygenase-2 is a target of microRNA-16 in human hepatoma cells. PLoS One. 7:e509352012. View Article : Google Scholar : PubMed/NCBI

78 

Jia H, Wang H, Yao Y, Wang C and Li P: miR-136 inhibits malignant progression of hepatocellular carcinoma cells by targeting cyclooxygenase 2. Oncol Res. 26:967–976. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Li J, Lu X, Zou X, Jiang Y, Yao J, Liu H, Ni B and Ma H: COX-2 rs5275 and rs689466 polymorphism and risk of lung cancer: A PRISMA-compliant meta-analysis. Medicine (Baltimore). 97:e118592018. View Article : Google Scholar : PubMed/NCBI

80 

Zago M, Rico de Souza A, Hecht E, Rousseau S, Hamid Q, Eidelman DH and Baglole CJ: The NF-κB family member RelB regulates microRNA miR-146a to suppress cigarette smoke-induced COX-2 protein expression in lung fibroblasts. Toxicol Lett. 226:107–116. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Xia M, Duan ML, Tong JH and Xu JG: MiR-26b suppresses tumor cell proliferation, migration and invasion by directly targeting COX-2 in lung cancer. Eur Rev Med Pharmacol Sci. 19:4728–4737. 2015.PubMed/NCBI

82 

Kwon Y, Kim Y, Eom S, Kim M, Park D, Kim H, Noh K, Lee H, Lee YS, Choe J, et al: MicroRNA-26a/-26b-COX-2-MIP-2 loop regulates allergic inflammation and allergic inflammation-promoted enhanced tumorigenic and metastatic potential of cancer cells. J Biol Chem. 290:14245–14266. 2015. View Article : Google Scholar : PubMed/NCBI

83 

Wu C, Li X, Zhang D, Xu B, Hu W, Zheng X, Zhu D, Zhou Q, Jiang J and Wu C: IL-1β-mediated Up-regulation of WT1D via miR-144-3p and their synergistic effect with NF-κB/COX-2/HIF-1α pathway on cell proliferation in LUAD. Cell Physiol Biochem. 48:2493–2502. 2018. View Article : Google Scholar : PubMed/NCBI

84 

Li B, Lu Y, Yu L, Han X, Wang H, Mao J, Shen J, Wang B, Tang J, Li C and Song B: miR-221/222 promote cancer stem-like cell properties and tumor growth of breast cancer via targeting PTEN and sustained Akt/NF-κB/COX-2 activation. Chem Biol Interact. 277:33–42. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Majumder M, Dunn L, Liu L, Hasan A, Vincent K, Brackstone M, Hess D and Lala PK: COX-2 induces oncogenic micro RNA miR655 in human breast cancer. Sci Rep. 8:3272018. View Article : Google Scholar : PubMed/NCBI

86 

Liao H, Zhou Q, Gu Y, Duan T and Feng Y: Luteinizing hormone facilitates angiogenesis in ovarian epithelial tumor cells and metformin inhibits the effect through the mTOR signaling pathway. Oncol Rep. 27:1873–1878. 2012.PubMed/NCBI

87 

Lai Y, Zhang X, Zhang Z, Shu Y, Luo X, Yang Y, Wang X, Yang G, Li L and Feng Y: The microRNA-27a: ZBTB10-specificity protein pathway is involved in follicle stimulating hormone-induced VEGF, Cox2 and survivin expression in ovarian epithelial cancer cells. Int J Oncol. 42:776–784. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Lin Y and Wu Z: MicroRNA-128 inhibits proliferation and invasion of glioma cells by targeting COX-2. Gene. 658:63–69. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Chen ZG, Zheng CY, Cai WQ, Li DW, Ye FY, Zhou J, Wu R and Yang K: miR-26b mimic inhibits glioma proliferation in vitro and in vivo suppressing COX-2 expression. Oncol Res. 27:147–155. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Shields CL and Shields JA: Retinoblastoma management: Advances in enucleation, intravenous chemoreduction, and intra-arterial chemotherapy. Curr Opin Ophthalmol. 21:203–212. 2010. View Article : Google Scholar : PubMed/NCBI

91 

Zhang J, He J and Zhang L: The down-regulation of microRNA-137 contributes to the up-regulation of retinoblastoma cell proliferation and invasion by regulating COX-2/PGE2 signaling. Biomed Pharmacother. 106:35–42. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Yu Q, Zhang K, Wang X, Liu X and Zhang Z: Expression of transcription factors snail, slug, and twist in human bladder carcinoma. J Exp Clin Cancer Res. 29:1192010. View Article : Google Scholar : PubMed/NCBI

93 

Song T, Zhang X, Wang C, Wu Y, Dong J, Gao J, Cai W and Hong B: Expression of miR-143 reduces growth and migration of human bladder carcinoma cells by targeting cyclooxygenase-2. Asian Pac J Cancer Prev. 12:9292011.PubMed/NCBI

94 

Xu L, Shen B, Chen T and Dong P: miR-203 is involved in the laryngeal carcinoma pathogenesis via targeting VEGFA and Cox-2. Onco Targets Ther. 9:4629–4637. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Tołoczko-Iwaniuk N, Dziemiańczyk-Pakieła D, Nowaszewska BK, Celińska-Janowicz K and Miltyk W: Celecoxib in cancer therapy and prevention-review. Curr Drug Targets. 20:302–315. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Jackson AL, Bartz SR, Schelter J, Kobayashi SV, Burchard J, Mao M, Li B, Cavet G and Linsley PS: Expression profiling reveals off-target gene regulation by RNAi. Nat Biotechnol. 21:635–637. 2003. View Article : Google Scholar : PubMed/NCBI

97 

Strillacci A, Griffoni C, Valerii MC, Lazzarini G, Tomasi V and Spisni E: RNAi-based strategies for cyclooxygenase-2 inhibition in cancer. J Biomed Biotechnol. 2010:8280452010. View Article : Google Scholar : PubMed/NCBI

98 

Liu X, Wu Y, Zhou Z, Huang M, Deng W, Wang Y, Zhou X, Chen L, Li Y, Zeng T, et al: Celecoxib inhibits the epithelial-to-mesenchymal transition in bladder cancer via the miRNA-145/TGFBR2/Smad3 axis. Int J Mol Med. 44:683–693. 2019.PubMed/NCBI

99 

Ghose J and Bhattacharyya NP: Transcriptional regulation of microRNA-100, −146a, and −150 genes by p53 and NFκB p65/RelA in mouse striatal STHdh(Q7)/Hdh(Q7) cells and human cervical carcinoma HeLa cells. RNA Biol. 12:457–477. 2015. View Article : Google Scholar : PubMed/NCBI

100 

DA Costa RM, Bastos MM, Medeiros R and Oliveira PA: The NFkB signaling pathway in papillomavirus-induced lesions: Friend or foe? Anticancer Res. 36:2073–2083. 2016.PubMed/NCBI

101 

DA Costa RMG, Araújo R, Santos JMO, Fernandes M, Neto T, Sousa H, Ribeiro J, Bastos MMSM, Oliveira PA, Carmo D, et al: Regulation of miRNA-146a and miRNA-150 Levels by celecoxib in premalignant lesions of K14-HPV16 mice. Anticancer Res. 37:2913–2918. 2017.PubMed/NCBI

102 

Saito Y, Suzuki H, Imaeda H, Matsuzaki J, Hirata K, Tsugawa H, Hibino S, Kanai Y, Saito H and Hibi T: The tumor suppressor microRNA-29c is downregulated and restored by celecoxib in human gastric cancer cells. Int J Cancer. 132:1751–1760. 2013. View Article : Google Scholar : PubMed/NCBI

103 

Mott JL, Kobayashi S, Bronk SF and Gores GJ: mir-29 regulates Mcl-1 protein expression and apoptosis. Oncogene. 26:6133–6140. 2007. View Article : Google Scholar : PubMed/NCBI

104 

Hunter S, Nault B, Ugwuagbo KC, Maiti S and Majumder M: Chemicall induced hypoxia enhances miRNA functions in breast cancer. Cancers (Basel). 12:20082020. View Article : Google Scholar

105 

Najafi M, Farhood B, Mortezaee K, Kharazinejad E, Majidpoor J and Ahadi R: Hypoxia in solid tumors: A key promoter of cancer stem cell (CSC) resistance. J Cancer Res Clin Oncol. 146:19–31. 2020. View Article : Google Scholar : PubMed/NCBI

106 

Xiong W, Li WH, Jiang YX, Liu S, Ai YQ, Liu R, Chang L, Zhang M, Wang XL, Bai H, et al: Parecoxib: An enhancer of radiation therapy for colorectal cancer. Asian Pac J Cancer Prev. 16:627–633. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Zagani R, Hamzaoui N, Cacheux W, de Reyniès A, Terris B, Chaussade S, Romagnolo B, Perret C and Lamarque D: Cyclooxygenase-2 inhibitors down-regulate osteopontin and Nr4A2-new therapeutic targets for colorectal cancers. Gastroenterology. 137:1358–1366.e1-3. 2009. View Article : Google Scholar : PubMed/NCBI

108 

Li LY, Xiao J, Liu Q and Xia K: Parecoxib inhibits glioblastoma cell proliferation, migration and invasion by up-regulating miRNA-29c. Biol Open. 6:311–316. 2016. View Article : Google Scholar

109 

Nissen SE, Yeomans ND, Solomon DH, Lüscher TF, Libby P, Husni ME, Graham DY, Borer JS, Wisniewski LM, Wolski KE, et al: Cardiovascular safety of celecoxib, naproxen, or ibuprofen for arthritis. N Engl J Med. 375:2519–2529. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 23 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gong Z, Huang W, Wang B, Liang N, Long S, Li W and Zhou Q: Interplay between cyclooxygenase‑2 and microRNAs in cancer (Review). Mol Med Rep 23: 347, 2021
APA
Gong, Z., Huang, W., Wang, B., Liang, N., Long, S., Li, W., & Zhou, Q. (2021). Interplay between cyclooxygenase‑2 and microRNAs in cancer (Review). Molecular Medicine Reports, 23, 347. https://doi.org/10.3892/mmr.2021.11986
MLA
Gong, Z., Huang, W., Wang, B., Liang, N., Long, S., Li, W., Zhou, Q."Interplay between cyclooxygenase‑2 and microRNAs in cancer (Review)". Molecular Medicine Reports 23.5 (2021): 347.
Chicago
Gong, Z., Huang, W., Wang, B., Liang, N., Long, S., Li, W., Zhou, Q."Interplay between cyclooxygenase‑2 and microRNAs in cancer (Review)". Molecular Medicine Reports 23, no. 5 (2021): 347. https://doi.org/10.3892/mmr.2021.11986