Skip to main content

miR-939, as an important regulator in various cancers pathogenesis, has diagnostic, prognostic, and therapeutic values: a review

Abstract

Background

MicroRNAs (miRNAs or miRs) are highly conserved non-coding RNAs with a short length (18–24 nucleotides) that directly bind to a complementary sequence within 3′-untranslated regions of their target mRNAs and regulate gene expression, post-transcriptionally. They play crucial roles in diverse biological processes, including cell proliferation, apoptosis, and differentiation. In the context of cancer, miRNAs are key regulators of growth, angiogenesis, metastasis, and drug resistance.

Main body

This review primarily focuses on miR-939 and its expanding roles and target genes in cancer pathogenesis. It compiles findings from various investigations. MiRNAs, due to their dysregulated expression in tumor environments, hold potential as cancer biomarkers. Several studies have highlighted the dysregulation of miR-939 expression in human cancers.

Conclusion

Our study highlights the potential of miR-939 as a valuable target in cancer diagnosis, prognosis, and treatment. The aberrant expression of miR-939, along with other miRNAs, underscores their significance in advancing our understanding of cancer biology and their promise in personalized cancer care.

Introduction

Despite the advanced diagnosis, monitoring, and management methods, the mortality rate due to various cancers is still high, as cases are diagnosed at the late disease stages. Hence, access to biomarkers that can provide valuable information about disease status is urgently needed for managing cancers.

MicroRNAs (also called miRNAs or miRs) are short and endogenous non-coding RNAs that range in size from 18 to 24 nucleotides. Due to their crucial roles in several cellular functions and almost all biological processes, they have been introduced as promising biomarkers in different cancer types [1, 2]. It is estimated that the expression of 30% of human genes is controlled by miRNAs [3]. MiRNAs do not have any role in encoding proteins independently. However, they recognize the 3′-untranslated regions (UTR) of messenger RNA (mRNA) via a complementary 6–8 nucleotide sequence, affect mRNA stability, and inhibit protein translation processes [4, 5]. Thus, miRNAs are associated with cellular homeostasis pathways such as proliferation, differentiation, and apoptosis by crosstalking with mRNA expression [2, 6, 7]. Due to the undeniable effects of miRNAs on various human biological processes, their close relation to various cancers is not out of mind [8]. Several studies have shown miRNA dysregulation in cancer cells and tumor microenvironments [9]. The protective and promoting effects of miRNAs in different cancers could appear when they inhibit the expression of oncogenes and tumor suppressor genes, respectively (Fig. 1) [1, 10]. Besides, miRNAs play essential roles in regulating anti-tumor responses of the immune system by controlling immune checkpoints [11,12,13].

Fig. 1
figure 1

The role of miR-939 in various cancers. Dysregulation of miR-939 reveals its oncogenic (red background) or suppressive (blue background) role in various cancers. Abbreviations: APC2, APC regulator of WNT signaling pathway 2; ARHGAP4, Rho GTPase‐activating protein 4; BCL-XL, B cell lymphoma-extra large; BCYRN1, brain cytoplasmic RNA 1; ESR1, estrogen receptor 1; GPD1, glycerol-3-phosphate dehydrogenase 1; HDGF, hepatoma-derived growth factor; IGF-1R, insulin-like growth factor type 1 receptor; JUNB, JunB proto-oncogene; LDLR, low-density lipoprotein receptor; LIMK2, LIM domain kinase 2; MID1IP1, MID1 interacting protein 1; miR, microRNAs; NO, nitric oxide; NOS2, nitric oxide synthase 2; NSCLC, non-small cell lung cancer; SLC34A2, solute carrier family 34 member 2; THRSP, thyroid hormone responsive spot 14; TIMP2, tissue inhibitor of metalloproteinases 2; TNBC, triple‐negative breast cancer; VE-cadherin, vascular endothelial-cadherin; ↓, decrease; ↑, increase

A widely reported miRNA, miR-939, has gained significant attention recently due to its unique role in the development of various malignancies, including hepatocellular carcinoma [14], gastric [15], ovarian [16], lung [17], colorectal [18], pancreatic [19], and prostate [20] cancers. Numerous studies have emphasized the remarkable role of miR-939 in the oncogenic process as indicated by its association with advanced stages, altered cell proliferation, high invasion, and metastatic potential [17, 19,20,21,22]. In the current study, the pathogenic mechanisms of miR-939 in various cancers have been discussed in detail. We have also reviewed the diagnostic, prognostic, and therapeutic values of miR-939 in association with other molecules and genes involved in the pathogenesis of cancers.

MiRNA-939 and various cancers

Pancreatic cancer

Pancreatic cancer is the 12th most common and the 6th most lethal cancer, according to GLOBOCAN 2020 [23, 24]. The high metastatic capacity and poor prognosis of pancreatic tumors have introduced them as a critical global burden [25]. The 5-year survival rate of patients with pancreatic cancer in the American population is only 10%, and 80–85% of cases are incurable. It is predicted that its burden will keep increasing and will become the second etiology of cancer mortality in the USA during the next 20 to 30 years [26, 27]. Therefore, recognizing new molecular targets involved in the progression of pancreatic cancer is essential to improve diagnosis, prognosis, and managing disease, and decreasing mortality.

Recent studies have indicated the role of Rho GTPase-activating proteins (RhoGAPs), such as Rho GTPase‐activating protein 4 (ARHGAP4), as tumor repressors in human cancers, particularly pancreatic cancer, where they play an important role in angiogenesis, aggressive behaviors, staging, and outcomes of tumors [28, 29]. In this regard, miR-939 negatively regulates the expression of ARHGAP4 by binding to the 3′-UTR of the mRNA [19]. Because of the anti-tumorgenesis role of ARHGAP4, positive impacts of miR-939 are expected in the progression of pancreatic neoplasms. The results of a study have shown elevated levels of miR-939 in tumor tissues compared to normal mucosa. Also, they have reported positive correlations between the expression levels of miR-939 and poor prognosis and a low survival rate in patients with pancreatic cancer. Besides, the role of miR-939 in cell migration and invasion has been confirmed by stimulating pancreatic cells with miR-939 and comparing them with the control group [19].

Hepatocellular carcinoma

Globally, tumors originating from hepatic tissue are among the six most prevalent cancers and the fourth cause of malignancy-related mortality [30]. More than three-fourths of primary liver cancers appear as hepatocellular carcinoma (HCC), which severely burdens healthcare organizations [31]. Although clear improvements have been made in diagnosing, monitoring, and managing cancers, new cases and deaths related to HCC are still growing worldwide due to late diagnosis [30]. The prognosis of HCC is closely dependent on the diagnostic, monitoring, and management procedures [32]. Identifying and targeting effective molecules in disease progression could significantly reduce the HCC burden.

Following this goal, we investigate miR-939’s role as a potential diagnostic and therapeutic target in HCC based on previous reports. Findings have reported a higher level of miR-939 expression in HCC tissue compared to healthy adjuvant mucosa. It is also found that low expression of miR-939 is associated with a good prognosis and high survival rate in HCC patients [21]. Chen et al. employed an imitator or inhibitor to investigate the role of miR-939 in HCC progression. The miR-939 inhibitor suppressed the translation of epithelial-mesenchymal transition (EMT)-related proteins in LM3 cells, while the miR-939 imitator induced the cell invasion. In terms of the mechanism of action, miR-939 downregulates the estrogen receptor 1 (ESR1) expression, an HCC tumor suppressor gene, by modulating protein functions in the cytoplasm and gene expression in the nucleus [21, 33, 34]. It has been shown that the elevated level of ESR1 expression in patients with HCC leads to better disease outcomes. Downregulation of ESR1, on the other hand, is linked to increased EMT and invasiveness of LM3 cells [21]. Besides, the correlation between miR-939 and HCC has been substantiated in another study. Fornari et al. reported that the level of miR-939 expression in HCC-positive cirrhotic patients is significantly higher compared to HCC-negative cases [14]. Overall, focusing on the miR-939 role would be an innovative strategy for managing HCC.

Prostate cancer

Following lung and colorectal tumors, prostate malignancies are in third place among the most diagnosed cancers worldwide (7.3% of all patients) [23]. They are the most frequently diagnosed cancers in European and USA male populations [35]. In recent decades, significant development has occurred in the early detection and treatment of prostate neoplasms. However, hoped-for improvements in the long-term prognosis have yet to materialize [36, 37]. Although several tumor-suppressing and activating genes that play antithetical roles in disease progression have been identified, the molecular pathways contributing to disease progression are not entirely understood [38, 39]. To date, prostate-specific antigen (PSA) serum level is the primary biomarker for diagnosing prostate cancer. Nonetheless, due to low specificity and limitations in early disease detection, researchers have recently focused on discovering new biomarkers [40, 41].

One of the novel molecules is miR-939, which has been shown to play a protective role against various aspects of prostate tumorigenesis [20, 42]. It is reported that the expression of miR-939 in prostate tumor specimens is regulated lower than in non-malignant prostate tissue [20]. Besides, a meta-analysis that studied the expression level of 37 miRNAs suggested lower expression of miR-939 in recurrent prostate cancer samples compared to non-recurrent ones [43]. Also, it has been found that silencing the expression of miR-939 is closely associated with poor outcomes and lower survival in patients with prostate cancer. Conversely, the proliferation of prostate cancer cells is suppressed by the overexpression of miR-939 [20]. The upregulation of miR-939 in prostate malignant tissue is correlated with decreased prefiltration capacity, colony constitution, migration and invasion, and enhanced apoptosis of tumor cells in vitro [20]. Inversely, hepatoma-derived growth factor (HDGF) is expressed at a higher level in malignant cell lines and directly confronts the antineoplastic activity of miR-939. Bioinformatics analysis and luciferase reporter assays have confirmed the direct binding of miR-939 to the 3′-UTR of the HDGF gene, which subsequently downregulates the expression of HDGF-related mRNAs and proteins [20]. The results of another study demonstrated that the overexpression of brain cytoplasmic RNA 1 (BCYRN1) is related to poor prognosis and the metastatic ability of prostate malignant cell lines by downregulating miR-939 expression [42]. Overall, these findings reveal the diagnostic and prognostic roles of miR-939 in prostate tumors.

Glioma

Gliomas are CNS neoplasms that originate from the brain and spinal cord glial cells and have the most frequent malignant picture among all central nervous system cancers in young adults [44]. According to the classification published by the World Health Organization (WHO), 2nd-grade and 3rd-grade gliomas are classified as diffuse lower-grade gliomas (LGGs), and 4th-grade gliomas are characterized as glioblastomas (GBMs) [45]. High-grade gliomas, such as GBM and anaplastic astrocytoma, are responsible for both adult and pediatric glioma-related deaths [46, 47]. The global incidence rate of glioma has significantly grown in recent years [48]. Meanwhile, GBM is the most common type of glioma (57.7%), diagnosed in 3.23 individuals out of 100,000 Americans [49].

Surgery is still the main strategy for treatment, despite the deployment of new methods in the management of glioma patients, such as radiotherapy, chemotherapy, and immunotherapy [50, 51]. Although there are combinations of diagnostic and therapeutic procedures for glioma, none have a good prognosis and do not reduce disease recurrence [52, 53]. Thus, discovering novel molecular targets with the capacity to become prognostic and therapeutic biomarkers is one of the urgent needs in glioma management.

MiRNAs are potential targets for this purpose due to their regulatory role in cancer. Findings have shown the upregulation of miR-939 in glioma cell lines [22]. Besides, some glioma characteristics, including tumor size and grading (based on the WHO definition), are significantly correlated with miR-939 expression. Also, proliferation, colony constitution, invasion, and migration of malignant cells are observable in miR-939 overexpression [22]. As a result, the findings suggested that miR-939 plays tumorigenic roles in the pathophysiology of gliomas and is associated with poor outcomes [22]. Bioinformatic analysis and luciferase assay have confirmed that tissue inhibitor of metalloproteinases 2 (TIMP2), introduced as a tumor inhibitor gene in glioma, is a target gene for miR-939. MiR-939 binds to the 3′-UTR of TIMP2 mRNA and reduces its translation [22]. Hence, miR-939 could be a prognostic and therapeutic biomarker in glioma.

Lung cancer

Lung tumors are the leading cause of cancer-related mortality (18% of total deaths) and the second most common cancer worldwide [23]. Nearly 85% of lung malignancies belong to the non-small cell lung cancer (NSCLC) subgroup, accounting for most lung cancer deaths [54, 55]. NSCLC consists of three subtypes: adenocarcinoma (ADC), squamous cell carcinoma (SCC), and large cell carcinoma (LCC) [56]. Despite the molecular and histological dissimilarity of NSCLC subtypes, the treatment strategy was almost similar until now [57]. However, recent clinical trials suggest different methods for managing NSCLC subtypes to meet the best goals and reduce side effects [57, 58]. On the other hand, small cell lung cancer (SCLC), which accounts for only 15% of all lung tumor cases, is more malignant and invasive and has a lower survival rate than NSCLC [59, 60]. Despite the development of new lung cancer treatment methods, the 5-year survival rate remains less than 10% mainly because of the late diagnosis [61]. Therefore, access to helpful biomarkers for diagnosing, monitoring, and managing patients with lung cancer would improve long-term outcomes.

Recently, miR-939 was reported as an oncogenic micro-RNA in lung cancers [17, 62, 63]. A higher level of miR-939 expression has been identified in lung cancer tissues than in normal lung mucosa [17]. Also, a comparative study between lung cancer patients with high and low expression levels of miR-939 has shown a positive correlation of this miRNA with metastatic abilities and advanced tumor grading (TNM stages) [17]. This study revealed that transfection of miR-939 emulators into tumor cells stimulated cell proliferation, migration, and invasion. On the other hand, NSCLC cell lines showed a repressed proliferative capacity after miR-939 inhibitor transfection [62]. Furthermore, miR-939 is an independent poor prognostic factor in lung cancer patients, whereas its low expression is related to higher survival [17].

In ADC, the most frequent subtype of NSCLC responsible for more than half of all lung cancer cases [56], miR-939 could be an early diagnostic factor [64]. It is reported that miR-939 is positively regulated in most patients with ADC compared to the same sex and age control group, whereas miR-939 expression levels change with each ADC stage. Its expression decreases in stage 3 compared to stages 1 and 2 and rises again at stage 4 [63]. The results of a study indicated the regulatory role of miR-939 on TIMP Metallopeptidase Inhibitor 2 (TIMP2) expression in lung cancer. Accordingly, TIMP2, the tissue inhibitor of matrix metalloproteinase (MMPs), is an NSCLC-related gene that correlates with the expression level of miR-939. MiR-939 regulates TIMP2 expression at the translational level by binding to the 3′-UTR of TIMP2 mRNA [62]. The findings suggest that miR-939 plays an important role in lung cancer and its subtypes. Hence, miR-939 may be a potential target in early detection, predicting outcomes, and treating lung cancer. Further studies are needed to evaluate its function, accurately.

Ovarian cancer

Ovarian cancer (OC) is the fifth most frequent cause of mortality among women's malignancies and accounts for the most annual deaths of gynecologic cancers in developed societies [65]. Epithelial ovarian cancer (EOC) is the most common OCs, with about a 90% incidence rate [65]. Despite significant progress in therapeutic modalities, OC is diagnosed in an advanced stage (stages III-IV according to the International Federation of Gynecology and Obstetrics (FIGO) staging system), with a 5-year survival rate of less than 30% [66]. This late detection results from poor screening methods [66]. Hence, recognizing efficacious diagnostic and prognostic factors is essential in OC.

Ying and colleagues found that the expression of miR-939 in OC cell lines is regulated at a higher level compared to normal adjuvant ovarian tissue [67]. Also, it has been shown that proliferative, colony-forming, and cell growth abilities enhance after the transfection of OC cells with miR-939 mimics. Thus, miR-939 positively regulates tumorigenesis in OC and can be one of the novel biomarkers for diagnoses and management. Studies confirmed a negative association between miR-939 expression levels and APC2, a tumor suppressor gene, via suppression of the Wnt/ β-catenin signaling pathway [67]. Activation of the wnt/β-catenin pathway modulates extensive cell proliferation and differentiation [68,69,70] via upregulating cyclin D1, c-MYC, and TCF genes in several human cancers [71,72,73,74,75]. Consequently, miR-939 overexpression, as an oncogene, leads to more protein production from the cyclin D1 and c-MYC genes [67]. Accordingly, a luciferase reporter assay has demonstrated that miR-939 binds the 3′-UTR site of the APC2 mRNA and suppresses its translation. On the other hand, inhibiting miR-939 has a positive effect on APC2 protein translation [67].

The miR-939 also acts as a link between platelet microparticles (PMPs) and EMT. A study has shown that the expression level of miR-939 is elevated in the PMPs. Also, a correlation is reported between miR-939 and the expression level of EMT-associated molecules, including vimentin, E-cadherin, and claudin [16]. PMPs are small components derived from the stimulation of platelets with thrombin and play a crucial role in OC oncogenicity by boosting tumor cells' proliferation and migration capability. In addition, PMPs induce metastatic behaviors (cell proliferation and migration) in the EOC via modulating EMT-related molecules [16]. In this regard, miR-939 stimulates OC development and metastatic behaviors by stimulating vimentin expression and inhibiting endothelial (E)-cadherin and claudin expression [76, 77]. It is reported that miR-939 could be a predictive biomarker for choosing the most efficient treatment strategy. By tissue biopsy or using ascites, miR-939 expression levels could be measured, and the proper decision about immediate surgery would be made [78]. Overall, miR-939 might be a potential target for the diagnosis, monitoring, and treatment of ovarian cancer.

Colorectal cancer

Colorectal cancer (CRC), with a 1.9 million incidence rate and a 0.9 million mortality rate in 2020, is placed in the 3rd rank of the most frequent cancers and the 2nd rank of the most fatal cancers worldwide. Global statistics in 2020 showed that CRC was responsible for 10% and 9.4% of all new cancer cases and cancer-associated mortality, respectively. The CRC burden is predicted to keep increasing in further years and reach 3.2 million new patients in 2040. This growth rate results from the industrialization of human lifestyles and the developing and aging population [79, 80]. Although early CRC diagnosis plays a fundamental role in suppressing metastasis and limiting mortality and morbidity, unfortunately, CRC manifestation is revealed only in the late stages. Because of this, more than half of CRC patients die due to long-distance metastasis, particularly metastasis to liver tissue [81]. Thus, introducing novel biomarkers to relieve the CRC burden is urgent.

MiR-939 can be a molecular target for this purpose, and its role is reported in CRC progression. Zhang et al. discovered suppressor functions for miR-939 in CRC. In this regard, there was an inverse association between miR-939 levels and long intergenic non-protein coding RNA 460 (LINC00460) for regulating LIM domain kinase 2 (LIMK2) expression. Whereas miR-939 suppresses the expression of LIMK2 mRNA via binding to its specific sequence, LIN00460 upregulates the expression of LIMK2 by sponging miR-939. LIMK2 promotes the migration and invasion of cancer cells and the poor prognosis of CRC. Hence, miR-939 and LIN00460 act as suppressors and oncogenes in CRC pathogenesis, respectively [18]. Long noncoding RNA HEIH (LncRNA-HEIH) is another miR-939-related oncogenic LncRNA that promotes the viability, proliferation, and invasion of CRC cells by inducing B cell lymphoma-extra large (Bcl-xL) expression. This LncRNA acts as a sponge for miR-939 and competes with the organizing nuclear factor-κB (NF-κB)-miR-939 complex. Following this, NF-κB targets the Bcl-xL promoter and upregulates its expression [82].

In colon cancer (stage T2-3N0), a study showed a negative correlation between the miR-939 level and “shorter distant metastasis-free survival (DMFS)” [83]. Another study did not demonstrate a significant difference in the expression levels of miR-939 between colon adenocarcinoma patients responding and non-responding to chemotherapy [84]. Therefore, miR-939 has no diagnostic value for determining chemoresistant colon cancer cases [84], whereas it has a prognostic value in the early stages of colon cancer [83].

Gastric cancer

In 2020, gastric cancer (GC) accounted for 5.6% (6th rank) of global cancer incidence and 7.7% (3rd rank) of cancer mortality [23]. In different regions, the incidence rate varies by up to 15–20-fold [85]. For example, GC is not among the ten most common cancers by occurrence or death in the US population [86]. But eastern randomized trials elucidated 30 to 40% higher GC surveillance compared to Western ones [87, 88]. Up until now, the primary strategies for treatment have been surgical resection and palliative chemotherapy [89]. The prognosis of GC patients is still unfavorable despite the implications of novel therapeutic approaches, and chemotherapy resistance and metastatic behaviors are the main etiologies of mortality in patients. Nevertheless, GC-related molecular pathways are not clearly understood [90,91,92]. Hence, investigating key molecules related to GC would be critical in determining potential targets for early diagnosis and management of the disease.

A study found a drastically reduced expression level of miR-939 in GC tissues and cell lines compared to normal tissues and cell lines. Besides, a study of patients with stage I-III GC cancer found that local relapse or distant metastasis is associated with a decrease in miR-939 expression in GC tissue [15]. Hence, miR-939 has been introduced as a prognostic biomarker for predicting GC patients’ responses to chemotherapy. Findings show that complete or partial response to therapy in GC patients is associated with upregulation of miR-939, whereas downregulation of miR-939 is common in the no response and progressive groups [15]. Accordingly, a novel therapeutic strategy is applying miR-939 and 5-fluorouracil (Fu) simultaneously, which have a synergic effect on the apoptosis or necrosis of GC cells and inhibit clonogenicity. In the first 48 h, miR-939 sensitizes GC cells to 5-Fu, a commonly used chemotherapeutic agent in GC patients. In addition, study findings showed that miR-939 silencing enhanced oncogenesis and cell proliferation of GC cell lines in vitro [15].

Regarding the mechanism of action, solute carrier family 34 member 2 (SLC34A2) is a direct target gene for miR-939 that negatively correlates with it [15]. Upregulation of miR-939 significantly downregulates expression of SLC34A2 mRNA by binding to the 3′-UTR site and inhibits MEK1/2 phosphorylation and Raf-1 level, resulting in the suppression of proliferation, migration, and invasion in cells [93]. The elevated expression of miR-939 in patients with GC improves survival and reduces tumor recurrence rate. While SLC34A2 upregulation is associated with unsatisfactory survival and increased tumor recurrence. GC patients with an elevated level of miR-939 and a reduced level of SLC34A2 present the best outcomes and survival rates [15]. Overall, miR-939 could be an oncosuppressor biomarker with promising prognostic and therapeutic value in patients with GC.

Breast cancer

Breast cancer (BC) is the most common form of cancer worldwide [23] and the first cause of death among all cancers in women 20 to 50 years old [65]. BC mortality rate varies greatly from the East to Europe and the USA [94]. This difference results from various environmental factors and lifestyles, which have been demonstrated by increased mortality and incidence rates during migration from safe BC regions to areas with higher mortality rates [95]. Metastasis and relapse, as the primary cause of malignancy-associated death, occur in 10 to 20% of the European population with BC after initial relief [96, 97]. The most aggressive BC subtype, triple‐negative breast cancer (TNBC), comprises 15–20% of all BC cases. The negative expression of three receptors, including the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2), is the unique specification of TNBC [98, 99]. The first incidental rank of BC among all tumors, recurrences, and metastatic behaviors emphasizes the necessity of recognizing potential molecular targets for early diagnosis, monitoring, and effective therapy.

In this regard, the expression of miR-939 in BC cells is regulated at a higher level than in healthy breast tissues, and it is a frequent finding in the highly invasive BC subtype (TNBC) [100]. Furthermore, it has been discovered that the synergistic effects of miR-939 upregulation and lymph node involvement increase the risk of TNBC relapse by approximately sixfold. More findings have shown that miR-939 exerts a significant role in TNBC migration [100] through blood vessels instead of lymphatic metastasis [101]. In silico analysis determined that miR-939 targets vascular endothelial (VE)-cadherin mRNA (3UTR), which regulates the junction between endothelial cells and controls vessel permeability. In confirmation, miR-939 directly reduced 40 to 60% of VE-cadherin protein levels in HUVEC, but no changes in the level of VE-cadherin mRNA have been seen. This suggests that miR-939 inhibits the translation of VE-cadherin mRNA instead of mRNA degradation. A decline in VE-cadherin protein levels after miR-939 overexpression weakens the monolayer integrity of endothelial cells in the vascular system. As a result, it opens some "gates" to malignant cells in blood vessels that facilitate hematogenous metastasis. Hence, elevating miR-939 in TNBC is correlated with poor disease-free survival [100].

Interestingly, a recent study has suggested the downregulation of miR-939 in all BC subtypes (including TNBC) compared to non-malignant mammary controls [102]. In this study, nitric oxide synthase (NOS)2/NOS3 expression and, as a result, high NO production (an immunosuppressor and cancer inducer) were negatively correlated with miR-939 expression in TNBC cells. These findings suggested that miR‐939 is a tumor suppressor in TNBC by hindering the NOS2/NO axis. Transfection of miR-939 into MDA‐MB‐231 cells (a type of TNBC cell line) significantly depresses NOS2-induced NO production, but not NOS3. Besides, miR-939 upregulation in TNBC tissues induced cell apoptosis and attenuated cell migration and colony formation. Recently, it has been found that lncRNA HEIH leads to tumor progression by increasing the expression of NOS2, PDL1, and MICA/B and decreasing the expression of NKG2D ligands in cancer cells. The sponging of miR-939 has been introduced as a probable mechanism for the lncRNA HEIH acts in TNBC [102]. Overall, these paradoxical results express the indigence of further studies about the role of miR-939 in BC and its subtypes.

Osteosarcoma

Osteosarcoma (OS), the most frequent bone malignancy, is more prevalent among children and adolescents [103]. Long bone metaphysis, including the distal femur (43%), proximal tibia (23%), and proximal humor (10%), are the major origins of OS [104]. To our knowledge, OS's annual incidence and mortality rate have developed over recent years [105, 106]. Combining systemic chemotherapy and extensive surgical resection is the most helpful therapeutic technique [107,108,109], which has resulted in a 60–70% improvement in the 5-year survival rate of patients with non-metastatic osteosarcoma [110]. However, the long-term outcome is still unsatisfactory, and this multistage therapy could survive only 11–30% of patients with distant metastasis [111, 112]. Hence, improving the prognosis of OS patients would be impossible unless the pathophysiological pathways underlying OS tumorigenicity were understood.

MiR-939 is one of the novel biomarkers whose role has been determined in OS pathophysiology. Zhao et al. reported a decreased expression level of miR-939 in OS tissue compared to paired adjacent tissue. They also indicated that OS patients with a low level of miR-939 present advanced clinical stages and more distant metastases than patients with a higher level. An in vitro study confirmed that invasive abilities in OS are associated with miR-939 expression level [113]. In this regard, the proliferation, viability, metastatic, and migratory behaviors of cancer cells were suppressed by transfection and overexpression of miR-939. MiR-939 was also transfected into nude mice to determine its impact in vivo. Results indicated that the tumor weight and size were inversely related to the expression level of miR-939 [113]. In addition, miR-939 injection into tumor xenografts downregulated the expression of insulin-like growth factor type 1 receptor (IGF-1R), p-PI3K, and p-Akt compared with the control group. Bioinformatic analysis and luciferase assays introduce IGF-1R as a direct target gene for miR-939, which is highly expressed in OS cell lines [113]. MiR-939 binds to the IGF-1R gene’s 3′-UTR sequence and inhibits IGF-1R-associated mRNA expression and protein translation. The results indicate that the overexpression of IGF-1R increases proliferation and migration and decreases apoptosis of cancer cells in OS samples by activating the PI3K/Akt axis [113]. Hence, miR-939 has a prognostic value in osteosarcoma cases, and its downregulation is associated with a poor prognosis and a reduced overall survival rate.

Sebaceous gland carcinoma of the eyelid

Sebaceous gland carcinoma (SGC) is a malignant tumor type of the sebaceous gland that occurs in the eyelid. SGC placed in the 2nd and 3rd (or 4th) rank of the most prevalent malignant tumors arising from the eyelid in some Asian and European countries, respectively [114, 115]. Two main therapeutic strategies are extensive resection and elective radiotherapy. However, delayed disease diagnosis decreases these therapies' impacts and enhances morbidity and mortality in patients with SGC [116]. Meanwhile, late diagnosis of SGC is correlated with aggressive behaviors such as invasion into lymph nodes and other organs, leading to a nearly 6–9% mortality rate [117,118,119] and attenuating long-term prognosis and survival [116]. Therefore, determining molecular targets for early diagnosis and more effective treatment of SCG is urgently needed to improve patient prognosis.

The leading etiology of SGC development is abnormal lipid metabolism and accumulation in the cytoplasm of the meibomian and Zeis glands, the primary sites of SGC initiation [120]; hence, investigating lipid metabolism pathways can provide good clues about effective molecules in the SGC. It has been shown that the upregulation of miR-939 (plus 15 other miRNAs) followed by the downregulation of 516 mRNAs is associated with the loss of lipid metabolism function and is enriched in cholesterol biosynthesis pathways. Therefore, miR-939 can be a potential diagnostic and therapeutic target.

Thyroid hormone responsive spot 14 (THRSP), MID1 interacting protein 1 (MID1IP1), low-density lipoprotein receptor (LDLR), and glycerol-3-phosphate dehydrogenase 1 (GPD1), which control fatty acid, cholesterol, and triglyceride synthesis, are all linked to miR-939 expression. Overall, miR-939 is involved in tumorigenesis of the sebaceous gland through its role in ectopic lipid metabolism [120]. However, the results mentioned above were derived from bioinformatics analysis. Further molecular research is warranted to substantiate the existing evidence.

Pediatric anaplastic large cell lymphoma

One rare subtype of peripheral T cell lymphoma is anaplastic large cell lymphoma (ALCL), which is more common among children and young adults than in older patients [121]. Based on the WHO categorization, ALCL consists of two subgroups, ALCL with upregulation of anaplastic lymphoma kinase (ALK+ ALCL) and ALCL with downregulation of ALK expression (ALK ALCL) [122]. Most pediatrics with ALCL are ALK+ (90%), which is characterized by t(2;5)(p23;q35) chromosomal translocation and overexpression of nucleophosmin/ALK (NPM-ALK) fusion protein [123]. ALK+ ALCL reveals more aggression and growth abilities compared to the ALK ALCL subgroup. Consequently, the relapse of the ALK+ ALCL is approximately sixfold in comparison with ALK ALCL patients (5 months versus 30 months) [123]. It is shown that ALK expression and its signaling cascade are associated with the ALCL carcinogenic pathways and pathogenesis [124,125,126]. To determine the molecular pathways correlated with ALK+ ALCL invasive behavior, various miRNAs were introduced as potential targets. The differences in miR-939 levels have been reported between ALK-positive and ALK-negative cell lines. The results have also suggested a notable upregulation of miR-939 in ALK-negative tumor cells. Moreover, it has been demonstrated that miR-939 up-modulation prominently inhibits the invasion, migration, clonogenicity, and growth capacity of entire ALCL malignant cells. However, miR-939 has shown no control over NPM-ALK expression. Regarding the mechanism of action, JUNB is a target gene for miR-939 in ALK+ ALCL cell lines, associated with increased cell proliferation. MiR-939 binds to the 3′-UTR of JUNB mRNA and regulates JUNB-related protein levels. Platelet-derived growth factor receptor B (PDGFRB) is a direct transcriptional target for JUNB, contributing to ALCL oncogenicity and lymphomagenesis. PDGFR inhibitors, when combined with ALK, decrease the relapse rates of ALCL cases [127]. Finally, the findings introduced miR-939 as a tumor-suppressor gene in ALCL via suppression of the JUNB/PDGFRB axis.

Prospects and challenges

MiR-939 has immense influence over a range of cellular functions, not only in the normal metabolism of healthy cells but also in the tumorigenic process of malignant tissues. The accumulated results reveal that miR-939 is a critical regulator of cellular processes such as proliferation, differentiation, development, and death, especially in cancer. Although we mentioned the role of miR-939 in the pathogenicity of many malignancies, the following challenges remain to be solved in further investigations. (1) Only 12 variant types of cancer have been evaluated to date, and the precise function of miR-939 in many other cancers remains unclear. (2) Additionally, among these 12 types, certain cancers such as glioma require further evaluation and the provision of stronger evidence. (3) Our evidence about this microRNA, compared to the older ones, needs to be revised, and there are still several fields to be investigated. (4) The cellular interaction between miR-939 and other molecular pathways needs more clarity in different cancer types. (5) Regulating the expression level of miR-939 could be a consequence or a cause of oncogenesis; hence, finding the exact fact requires more evaluation.

Conclusion

In the final word, we provided a comprehensive review of the miR-939 dysregulation in twelve types of cancer, as summarized in Table 1. By targeting various signaling pathways and processes, miR-939 plays roles in tumor cell proliferation or growth, EMT, apoptosis rate, invasive or metastatic capacities, and resistance to therapeutic methods. Hence, miR-939 can be a potential target for cancer identification, patient surveillance, and long-term outcomes. Numerous investigations exploring the association between miR-939 and tumor figures have identified either tumor suppressor or promoter effects of miR-939 in cancers. Despite the revelation of predictor diagnostic and prognostic value of miR-939 in recent studies, we sincerely hope this review provides more passion for further investigations with larger sample sizes about the clinical potential of miR-939 in cancers.

Table 1 The expression of miR-939 and target genes in various cancers

Availability of data and materials

Not applicable.

Abbreviations

ADC:

Adenocarcinoma

Akt:

AKT serine/threonine kinase 1

ALCL:

Anaplastic large cell lymphoma

ALK:

Anaplastic lymphoma kinase

APC2:

APC regulator of WNT signaling pathway 2

ARHGAP4:

Rho GTPase‐activating protein 4

BC:

Breast cancer

BCL-XL:

B cell lymphoma-extra large

BCYRN1:

Brain cytoplasmic RNA 1

c-MYC:

MYC proto-oncogene

CRC:

Colorectal cancer

DMFS:

Distant metastasis-free survival

E-cadherin:

Endothelial-cadherin

EMT:

Epithelial‑mesenchymal transition

EOC:

Epithelial ovarian cancer

ER:

Estrogen receptor

ESR1:

Estrogen receptor 1

5-FU:

5-Fluorouracil

GBM:

Glioblastoma

GC:

Gastric cancer

GPD1:

Glycerol-3-phosphate dehydrogenase 1

HCC:

Hepatocellular carcinoma

HDGF:

Hepatoma-derived growth factor

HER2:

Human epidermal growth factor receptor-2

IGF-1R:

Insulin-like growth factor type 1 receptor

JUNB:

JunB proto-oncogene

LCC:

Large cell carcinoma

LDLR:

Low-density lipoprotein receptor

LGG:

Lower-grade glioma

LIMK2:

LIM domain kinase 2

LINC00460:

Long intergenic non-protein coding RNA 460

LncRNA-HEIH:

Hepatocellular carcinoma upregulated EZH2-associated long non-coding RNA

MEK:

Mitogen-activated protein kinase

MID1IP1:

MID1 interacting protein 1

miR:

MicroRNAs

MMPs:

Metalloproteinase

mRNA:

Messenger RNA

NF-κB:

Nuclear factor-κB

NO:

Nitric oxide

NOS2:

Nitric oxide synthase 2

NSCLC:

Non-small cell lung cancer

OC:

Ovarian cancer

Onc:

Oncogenic

OS:

Osteosarcoma

PI3K:

Phosphatidylinositol 3-kinase

PMP:

Platelet microparticles

PR:

Progesterone receptor

PSA:

Prostate-specific antigen

Raf-1:

Raf-1 proto-oncogene (serine/threonine kinase)

SCC:

Squamous cell carcinoma

SCLC:

Small cell lung cancer

SGC:

Sebaceous gland carcinoma

SLC34A2:

Solute carrier family 34 member 2

THRSP:

Thyroid hormone responsive spot 14

TIMP2:

Tissue inhibitor of metalloproteinases 2

TNBC:

Triple‐negative breast cancer

TS:

Tumor suppressor

UTR:

Untranslated region

VE-cadherin:

Vascular endothelial-cadherin

WHO:

World Health Organization

References

  1. Calin GA, Croce CM. MicroRNA signatures in human cancers. Nat Rev Cancer. 2006;6(11):857–66.

    Article  CAS  PubMed  Google Scholar 

  2. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116(2):281–97.

    Article  CAS  PubMed  Google Scholar 

  3. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120(1):15–20.

    Article  CAS  PubMed  Google Scholar 

  4. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75(5):843–54.

    Article  CAS  PubMed  Google Scholar 

  5. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136(2):215–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. MacFarlane L-A, Murphy PR. MicroRNA: biogenesis, function and role in cancer. Curr Genomics. 2010;11(7):537–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hill M, Tran N. miRNA interplay: Mechanisms and consequences in cancer. Dis Model Mech. 2021;14(4):dmm047662.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Volinia S, Calin GA, Liu C-G, Ambs S, Cimmino A, Petrocca F, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc Natl Acad Sci. 2006;103(7):2257–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Ventura A, Jacks T. MicroRNAs and cancer: short RNAs go a long way. Cell. 2009;136(4):586–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Omar HA, El-Serafi AT, Hersi F, Arafa ESA, Zaher DM, Madkour M, et al. Immunomodulatory MicroRNAs in cancer: targeting immune checkpoints and the tumor microenvironment. FEBS J. 2019;286(18):3540–57.

    Article  CAS  PubMed  Google Scholar 

  12. Pastina P, Nardone V, Croci S, Battaglia G, Vanni F, Bellan C, et al. Anti-cancer activity of dose-fractioned mPE+/− bevacizumab regimen is paralleled by immune-modulation in advanced squamous NSLC patients. J Thorac Dis. 2017;9(9):3123.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Mearini E, Poli G, Cochetti G, Boni A, Egidi MG, Brancorsini S. Expression of urinary miRNAs targeting NLRs inflammasomes in bladder cancer. Onco Targets Ther. 2017;10:2665.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Fornari F, Ferracin M, Trerè D, Milazzo M, Marinelli S, Galassi M, et al. Circulating microRNAs, miR-939, miR-595, miR-519d and miR-494, identify cirrhotic patients with HCC. PLoS One. 2015;10(10):e0141448.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Zhang J-X, Xu Y, Gao Y, Chen C, Zheng Z-S, Yun M, et al. Decreased expression of miR-939 contributes to chemoresistance and metastasis of gastric cancer via dysregulation of SLC34A2 and Raf/MEK/ERK pathway. Mol Cancer. 2017;16(1):1–15.

    Article  Google Scholar 

  16. Tang M, Jiang L, Lin Y, Wu X, Wang K, He Q, et al. Platelet microparticle-mediated transfer of miR-939 to epithelial ovarian cancer cells promotes epithelial to mesenchymal transition. Oncotarget. 2017;8(57):97464.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Han X, Du C, Chen Y, Zhong X, Wang F, Wang J, et al. Overexpression of miR-939-3p predicts poor prognosis and promotes progression in lung cancer. Cancer Biomark. 2019;25(4):325–32.

    Article  CAS  PubMed  Google Scholar 

  18. Zhang Y, Liu X, Li Q, Zhang Y. lncRNA LINC00460 promoted colorectal cancer cells metastasis via miR-939-5p sponging. Cancer Manag Res. 2019;11:1779.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Shen Y, Chen G, Gao H, Li Y, Zhuang L, Meng Z, et al. miR-939-5p contributes to the migration and invasion of pancreatic cancer by targeting ARHGAP4. Onco Targets Ther. 2020;13:389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Situ J, Zhang H, Jin Z, Li K, Mao Y, Huang W. MicroRNA-939 directly targets HDGF to inhibit the aggressiveness of prostate cancer via deactivation of the WNT/β-catenin pathway. Onco Targets Ther. 2020;13:4257.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chen F, Ni X, Chen L, Wang X, Xu J. miR-939-3p promotes epithelial-mesenchymal transition and may be used as a prognostic marker in hepatocellular carcinoma. Oncol Lett. 2020;19(4):2727–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zheng H, Zhao H, Ye G. Overexpression of microRNA-939-5p contributes to cell proliferation and associates poor prognosis in glioma. NeuroMol Med. 2021;23(4):531–9.

    Article  Google Scholar 

  23. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  24. Cancer IAfRo. Pancreas. Global cancer observatory: World Health Organization. 2020. https://gco.iarc.fr/today/explore.

  25. Maisonneuve P, Lowenfels AB. Epidemiology of pancreatic cancer: an update. Dig Dis. 2010;28(4–5):645–56.

    Article  PubMed  Google Scholar 

  26. Viale PH. The American Cancer Society’s facts & figures: 2020 edition. J Adv Pract Oncol. 2020;11(2):135.

    PubMed  PubMed Central  Google Scholar 

  27. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30.

    Article  PubMed  Google Scholar 

  28. Shen Y, Xu L, Ning Z, Liu L, Lin J, Chen H, et al. ARHGAP4 regulates the cell migration and invasion of pancreatic cancer by the HDAC2/β-catenin signaling pathway. Carcinogenesis. 2019;40(11):1405–14.

    Article  CAS  PubMed  Google Scholar 

  29. Shen Y, Chen G, Zhuang L, Xu L, Lin J, Liu L. ARHGAP4 mediates the Warburg effect in pancreatic cancer through the mTOR and HIF-1α signaling pathways. Onco Targets Ther. 2019;12:5003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang H, Naghavi M, Allen C, Barber RM, Bhutta ZA, Carter A, et al. Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015. The lancet. 2016;388(10053):1459–544.

    Article  Google Scholar 

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

    Article  PubMed  Google Scholar 

  32. Singal AG, Lampertico P, Nahon P. Epidemiology and surveillance for hepatocellular carcinoma: New trends. J Hepatol. 2020;72(2):250–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hishida M, Nomoto S, Inokawa Y, Hayashi M, Kanda M, Okamura Y, et al. Estrogen receptor 1 gene as a tumor suppressor gene in hepatocellular carcinoma detected by triple-combination array analysis. Int J Oncol. 2013;43(1):88–94.

    Article  CAS  PubMed  Google Scholar 

  34. Bjornstrom L, Sjoberg M. Mechanisms of estrogen receptor signaling: convergence of genomic and nongenomic actions on target genes. Mol Endocrinol. 2005;19(4):833–42.

    Article  PubMed  Google Scholar 

  35. Egidi MG, Cochetti G, Serva MR, Guelfi G, Zampini D, Mechelli L, et al. Circulating microRNAs and kallikreins before and after radical prostatectomy: are they really prostate cancer markers? BioMed Res Int. 2013;2013:241780.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Barry MJ, Simmons LH. Prevention of prostate cancer morbidity and mortality: primary prevention and early detection. Med Clin. 2017;101(4):787–806.

    Google Scholar 

  37. Miyake H, Fujisawa M. Prognostic prediction following radical prostatectomy for prostate cancer using conventional as well as molecular biological approaches. Int J Urol. 2013;20(3):301–11.

    Article  PubMed  Google Scholar 

  38. Antognelli C, Mezzasoma L, Fettucciari K, Mearini E, Talesa VN. Role of glyoxalase I in the proliferation and apoptosis control of human LNCaP and PC3 prostate cancer cells. Prostate. 2013;73(2):121–32.

    Article  CAS  PubMed  Google Scholar 

  39. Antognelli C, Mezzasoma L, Mearini E, Talesa VN. Glyoxalase 1–419C> A variant is associated with oxidative stress: Implications in prostate cancer progression. PLoS One. 2013;8(9):e74014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Guelfi G, Cochetti G, Stefanetti V, Zampini D, Diverio S, Boni A, et al. Next generation sequencing of urine exfoliated cells: an approach of prostate cancer microRNAs research. Sci Rep. 2018;8(1):1–8.

    Article  CAS  Google Scholar 

  41. Kanwal R, Plaga AR, Liu X, Shukla GC, Gupta S. MicroRNAs in prostate cancer: Functional role as biomarkers. Cancer Lett. 2017;407:9–20.

    Article  CAS  PubMed  Google Scholar 

  42. Huo W, Qi F, Wang K. Long non-coding RNA BCYRN1 promotes prostate cancer progression via elevation of HDAC11. Oncol Rep. 2020;44(3):1233–45.

    Article  CAS  PubMed  Google Scholar 

  43. Pashaei E, Pashaei E, Ahmady M, Ozen M, Aydin N. Meta-analysis of miRNA expression profiles for prostate cancer recurrence following radical prostatectomy. PLoS One. 2017;12(6):e0179543.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Weller M, Wick W, Aldape K, Brada M, Berger M, Pfister SM, et al. Glioma. Nat Rev Dis Primers. 2015;1(1):1–18.

    Article  Google Scholar 

  45. Zhang H, Wang R, Yu Y, Liu J, Luo T, Fan F. Glioblastoma treatment modalities besides surgery. J Cancer. 2019;10(20):4793.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wen PY, Reardon DA. Progress in glioma diagnosis, classification and treatment. Nat Rev Neurol. 2016;12(2):69–70.

    Article  CAS  PubMed  Google Scholar 

  47. Reardon DA, Wen PY. Unravelling tumour heterogeneity—implications for therapy. Nat Rev Clin Oncol. 2015;12(2):69–70.

    Article  CAS  PubMed  Google Scholar 

  48. Rees J, Watt H, Jäger HR, Benton C, Tozer D, Tofts P, et al. Volumes and growth rates of untreated adult low-grade gliomas indicate risk of early malignant transformation. Eur J Radiol. 2009;72(1):54–64.

    Article  PubMed  Google Scholar 

  49. Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2013–2017. Neuro-oncology. 2020;22(Supplement_1):iv1–96.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Cheung N-KV, Dyer MA. Neuroblastoma: developmental biology, cancer genomics and immunotherapy. Nat Rev Cancer. 2013;13(6):397–411.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hafazalla K, Sahgal A, Jaja B, Perry JR, Das S. Procarbazine, CCNU and vincristine (PCV) versus temozolomide chemotherapy for patients with low-grade glioma: a systematic review. Oncotarget. 2018;9(72):33623.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Sun J, Shi H, Lai N, Liao K, Zhang S, Lu X. Overexpression of microRNA-155 predicts poor prognosis in glioma patients. Med Oncol. 2014;31(4):1–5.

    Article  Google Scholar 

  53. Nieder C, Adam M, Molls M, Grosu AL. Therapeutic options for recurrent high-grade glioma in adult patients: recent advances. Crit Rev Oncol Hematol. 2006;60(3):181–93.

    Article  PubMed  Google Scholar 

  54. Chen W, Zuo T, Zheng R, Zeng H, Zhang S, He J. Lung cancer incidence and mortality in China in 2013. Zhonghua zhong liu za zhi [Chinese journal of oncology]. 2017;39(10):795–800.

    CAS  PubMed  Google Scholar 

  55. Morgensztern D, Ng SH, Gao F, Govindan R. Trends in stage distribution for patients with non-small cell lung cancer: a National Cancer Database survey. J Thorac Oncol. 2010;5(1):29–33.

    Article  PubMed  Google Scholar 

  56. Cagle PT, Allen TC, Dacic S, Beasley MB, Borczuk AC, Chirieac LR, et al. Revolution in lung cancer: new challenges for the surgical pathologist. Arch Pathol Lab Med. 2011;135(1):110–6.

    Article  PubMed  Google Scholar 

  57. Langer CJ, Besse B, Gualberto A, Brambilla E, Soria J-C. The evolving role of histology in the management of advanced non–small-cell lung cancer. J Clin Oncol. 2010;28(36):5311–20.

    Article  PubMed  Google Scholar 

  58. Wilkerson MD, Yin X, Walter V, Zhao N, Cabanski CR, Hayward MC, et al. Differential pathogenesis of lung adenocarcinoma subtypes involving sequence mutations, copy number, chromosomal instability, and methylation. PLoS One. 2012;7(5):e36530.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Babadi AS, Kiani A, Mortaz E, Taghavi K, Khosravi A, Marjani M, et al. Serum interleukin-27 level in different clinical stages of lung cancer. Open Access Maced J Med Sci. 2019;7(1):45.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Oronsky B, Reid TR, Oronsky A, Carter CA. What’s new in SCLC? A review Neoplasia. 2017;19(10):842–7.

    Article  CAS  PubMed  Google Scholar 

  61. de Cos JS, González MAS, Montero MV, Calvo MCP, Vicente MJM, Valle MH. Non-small cell lung cancer and silent brain metastasis: Survival and prognostic factors. Lung Cancer. 2009;63(1):140–5.

    Article  Google Scholar 

  62. Chen A, Liu S, Lu X, Wei L, Chen Y. Inhibition of microRNA-939 suppresses the development of human non-small cell lung cancer via the upregulation of tissue inhibitor of metalloproteinases 2. Mol Med Rep. 2018;18(6):4831–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Rani S, Gately K, Crown J, O’Byrne K, O’Driscoll L. Global analysis of serum microRNAs as potential biomarkers for lung adenocarcinoma. Cancer Biol Ther. 2013;14(12):1104–12.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Ma R, Wang C, Wang J, Wang D, Xu J. miRNA–mRNA interaction network in non-small cell lung cancer. Interdiscip Sci. 2016;8(3):209–19.

    Article  CAS  PubMed  Google Scholar 

  65. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69(1):7–34.

    Article  PubMed  Google Scholar 

  66. Davis AN, Afshar-Kharghan V, Sood AK, editors. Platelet effects on ovarian cancer. Semin Oncol. 2014;41(3):378–84.

  67. Ying X, Li-ya Q, Feng Z, Yin W, Ji-hong L. MiR-939 promotes the proliferation of human ovarian cancer cells by repressing APC2 expression. Biomed Pharmacother. 2015;71:64–9.

    Article  CAS  PubMed  Google Scholar 

  68. Ochoa-Hernández AB, Juárez-Vázquez CI, Rosales-Reynoso MA, Barros-Núñez P. Wnt-β-catenina signaling pathway and its relationship with cancer. Cir Cir. 2012;80(4):389–98.

    PubMed  Google Scholar 

  69. Kim KH, Seol HJ, Kim EH, Rheey J, Jin HJ, Lee Y, et al. Wnt/β-catenin signaling is a key downstream mediator of MET signaling in glioblastoma stem cells. Neuro Oncol. 2013;15(2):161–71.

    Article  CAS  PubMed  Google Scholar 

  70. Hsieh I-S, Chang K-C, Tsai Y-T, Ke J-Y, Lu P-J, Lee K-H, et al. MicroRNA-320 suppresses the stem cell-like characteristics of prostate cancer cells by downregulating the Wnt/beta-catenin signaling pathway. Carcinogenesis. 2013;34(3):530–8.

    Article  CAS  PubMed  Google Scholar 

  71. He T-C, Sparks AB, Rago C, Hermeking H, Zawel L, Da Costa LT, et al. Identification of c-MYC as a target of the APC pathway. Science. 1998;281(5382):1509–12.

    Article  CAS  PubMed  Google Scholar 

  72. Kolligs FT, Bommer G, Göke B. Wnt/beta-catenin/tcf signaling: a critical pathway in gastrointestinal tumorigenesis. Digestion. 2002;66(3):131–44.

    Article  CAS  PubMed  Google Scholar 

  73. Nagayama M, Iwamoto M, Hargett A, Kamiya N, Tamamura Y, Young B, et al. Wnt/β-catenin signaling regulates cranial base development and growth. J Dent Res. 2008;87(3):244–9.

    Article  CAS  PubMed  Google Scholar 

  74. Huang F-I, Chen Y-L, Chang C-N, Yuan R-H, Jeng Y-M. Hepatocyte growth factor activates Wnt pathway by transcriptional activation of LEF1 to facilitate tumor invasion. Carcinogenesis. 2012;33(6):1142–8.

    Article  CAS  PubMed  Google Scholar 

  75. Planutiene M, Planutis K, Holcombe RF. Lymphoid enhancer-binding factor 1, a representative of vertebrate-specific Lef1/Tcf1 sub-family, is a Wnt-beta-catenin pathway target gene in human endothelial cells which regulates matrix metalloproteinase-2 expression and promotes endothelial cell invasion. Vascular Cell. 2011;3(1):1–10.

    Article  Google Scholar 

  76. Che J, Yang Y, Xiao J, Zhao P, Yan B, Dong S, et al. Decreased expression of claudin-3 is associated with a poor prognosis and EMT in completely resected squamous cell lung carcinoma. Tumor Biology. 2015;36(8):6559–68.

    Article  CAS  PubMed  Google Scholar 

  77. Zeisberg M, Neilson EG. Biomarkers for epithelial-mesenchymal transitions. J Clin Investig. 2009;119(6):1429–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Prahm KP, Høgdall CK, Karlsen MA, Christensen IJ, Novotny GW, Høgdall E. MicroRNA characteristics in epithelial ovarian cancer. PLoS One. 2021;16(6):e0252401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Murphy N, Moreno V, Hughes DJ, Vodicka L, Vodicka P, Aglago EK, et al. Lifestyle and dietary environmental factors in colorectal cancer susceptibility. Mol Aspects Med. 2019;69:2–9.

    Article  PubMed  Google Scholar 

  80. Keum N, Giovannucci E. Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies. Nat Rev Gastroenterol Hepatol. 2019;16(12):713–32.

    Article  PubMed  Google Scholar 

  81. Manfredi S, Lepage C, Hatem C, Coatmeur O, Faivre J, Bouvier A-M. Epidemiology and management of liver metastases from colorectal cancer. Ann Surg. 2006;244(2):254.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Cui C, Zhai D, Cai L, Duan Q, Xie L, Yu J. Long noncoding RNA HEIH promotes colorectal cancer tumorigenesis via counteracting miR-939Mediated transcriptional repression of Bcl-xL. Cancer Res Treat. 2018;50(3):992–1008.

    Article  CAS  PubMed  Google Scholar 

  83. Bobowicz M, Skrzypski M, Czapiewski P, Marczyk M, Maciejewska A, Jankowski M, et al. Prognostic value of 5-microRNA based signature in T2–T3N0 colon cancer. Clin Exp Metas. 2016;33(8):765–73.

    Article  CAS  Google Scholar 

  84. Hu J, Xu Y, Cai S. Specific microRNAs as novel biomarkers for combination chemotherapy resistance detection of colon adenocarcinoma. Eur J Med Res. 2015;20(1):1–9.

    Article  Google Scholar 

  85. Sitarz R, Skierucha M, Mielko J, Offerhaus GJA, Maciejewski R, Polkowski WP. Gastric cancer: epidemiology, prevention, classification, and treatment. Cancer Manag Res. 2018;10:239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Society TAC. Cancer Statistics Center June 16, 2020 . Available from: https://www.cancerstatisticscenter.cancer.org.

  87. Badgwell B. Multimodality therapy of localized gastric adenocarcinoma. J Natl Compr Canc Netw. 2016;14(10):1321–7.

    Article  CAS  PubMed  Google Scholar 

  88. Badgwell B, Das P, Ajani J. Treatment of localized gastric and gastroesophageal adenocarcinoma: the role of accurate staging and preoperative therapy. J Hematol Oncol. 2017;10(1):1–7.

    Article  Google Scholar 

  89. Elimova E, Ajani JA. Surgical resection first for localized gastric adenocarcinoma: are there adjuvant options? J Clin Oncol. 2015;33(28):3085–91.

    Article  CAS  PubMed  Google Scholar 

  90. Jácome AA, Coutinho AK, Lima EM, Andrade AC, Dos Santos JS. Personalized medicine in gastric cancer: Where are we and where are we going? World J Gastroenterol. 2016;22(3):1160.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Lippert TH, Ruoff H-J, Volm M. Intrinsic and acquired drug resistance in malignant tumors. Arzneimittelforschung. 2008;58(06):261–4.

    CAS  PubMed  Google Scholar 

  92. Szakács G, Paterson JK, Ludwig JA, Booth-Genthe C, Gottesman MM. Targeting multidrug resistance in cancer. Nat Rev Drug Discovery. 2006;5(3):219–34.

    Article  PubMed  Google Scholar 

  93. Johnson GL, Stuhlmiller TJ, Angus SP, Zawistowski JS, Graves LM. Molecular Pathways: Adaptive Kinome Reprogramming in Response to Targeted Inhibition of the BRAF–MEK–ERK Pathway in CancerAdaptive Response to Kinase Inhibition. Clin Cancer Res. 2014;20(10):2516–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Goding Sauer A, et al. Breast cancer statistics, 2019. CA Cancer J Clin. 2019;69(6):438–51.

    Article  PubMed  Google Scholar 

  95. Mousavi SM, Hemminki K. Cancer incidence, trends, and survival among immigrants to Sweden. Eur J Cancer Prev. 2015;24:S1–63.

    Article  PubMed  Google Scholar 

  96. Colzani E, Johansson A, Liljegren A, Foukakis T, Clements M, Adolfsson J, et al. Time-dependent risk of developing distant metastasis in breast cancer patients according to treatment, age and tumour characteristics. Br J Cancer. 2014;110(5):1378–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Purushotham A, Shamil E, Cariati M, Agbaje O, Muhidin A, Gillett C, et al. Age at diagnosis and distant metastasis in breast cancer–a surprising inverse relationship. Eur J Cancer. 2014;50(10):1697–705.

    Article  CAS  PubMed  Google Scholar 

  98. Yin L, Duan J-J, Bian X-W, Yu S-c. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 2020;22(1):1–13.

    Article  Google Scholar 

  99. Youness RA, Gad MZ. Long non-coding RNAs: functional regulatory players in breast cancer. Non-coding RNA research. 2019;4(1):36–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Di Modica M, Regondi V, Sandri M, Iorio MV, Zanetti A, Tagliabue E, et al. Breast cancer-secreted miR-939 downregulates VE-cadherin and destroys the barrier function of endothelial monolayers. Cancer Lett. 2017;384:94–100.

    Article  PubMed  Google Scholar 

  101. Yaman S, Gumuskaya B, Ozkan C, Aksoy S, Guler G, Altundag K. Lymphatic and capillary invasion patterns in triple negative breast cancer. Am Surg. 2012;78(11):1238–42.

    Article  PubMed  Google Scholar 

  102. Nafea H, Youness RA, Abou-Aisha K, Gad MZ. LncRNA HEIH/miR-939-5p interplay modulates triple-negative breast cancer progression through NOS2-induced nitric oxide production. J Cell Physiol. 2021;236(7):5362–72.

    Article  CAS  PubMed  Google Scholar 

  103. Ritter J, Bielack S. Osteosarcoma. Annals Oncol. 2010;21:vii320–5.

    Article  Google Scholar 

  104. Bielack SS, Kempf-Bielack B, Delling Gn, Exner GU, Flege S, Helmke K, et al. Prognostic factors in high-grade osteosarcoma of the extremities or trunk: an analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol. 2002;20(3):776–90.

    Article  PubMed  Google Scholar 

  105. Wang Y, Huang Y, Xiang P, Tian W. LncRNA expression and implication in osteosarcoma: a systematic review and meta-analysis. Onco Targets Ther. 2017;10:5355.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Wu G, Zhang M. A novel risk score model based on eight genes and a nomogram for predicting overall survival of patients with osteosarcoma. BMC Cancer. 2020;20(1):1–12.

    Article  Google Scholar 

  107. Luetke A, Meyers PA, Lewis I, Juergens H. Osteosarcoma treatment–where do we stand? A state of the art review. Cancer Treat Rev. 2014;40(4):523–32.

    Article  PubMed  Google Scholar 

  108. Jafari F, Javdansirat S, Sanaie S, Naseri A, Shamekh A, Rostamzadeh D, et al. Osteosarcoma: a comprehensive review of management and treatment strategies. Ann Diagn Pathol. 2020;49:151654.

    Article  PubMed  Google Scholar 

  109. Tian Z, Niu X, Yao W. Receptor tyrosine kinases in osteosarcoma treatment: which is the key target? Front Oncol. 2020;10:1642.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Shankar GM, Clarke MJ, Ailon T, Rhines LD, Patel SR, Sahgal A, et al. The role of revision surgery and adjuvant therapy following subtotal resection of osteosarcoma of the spine: a systematic review with meta-analysis. J Neurosurg Spine. 2017;27(1):97–104.

    Article  PubMed  Google Scholar 

  111. Smeland S, Bielack SS, Whelan J, Bernstein M, Hogendoorn P, Krailo MD, et al. Survival and prognosis with osteosarcoma: outcomes in more than 2000 patients in the EURAMOS-1 (European and American Osteosarcoma Study) cohort. Eur J Cancer. 2019;109:36–50.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Kager L, Zoubek A, Pötschger U, Kastner U, Flege S, Kempf-Bielack B, et al. Primary metastatic osteosarcoma: presentation and outcome of patients treated on neoadjuvant Cooperative Osteosarcoma Study Group protocols. J Clin Oncol. 2003;21(10):2011–8.

    Article  PubMed  Google Scholar 

  113. Zhao X, Li J, Yu D. MicroRNA-939-5p directly targets IGF-1R to inhibit the aggressive phenotypes of osteosarcoma through deactivating the PI3K/Akt pathway. Int J Mol Med. 2019;44(5):1833–43.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Cook BE Jr, Bartley GB. Treatment options and future prospects for the management of eyelid malignancies: an evidence-based update. Ophthalmology. 2001;108(11):2088–98.

    Article  PubMed  Google Scholar 

  115. Wang L, Shan Y, Dai X, You N, Shao J, Pan X, et al. Clinicopathological analysis of 5146 eyelid tumours and tumour-like lesions in an eye centre in South China, 2000–2018: a retrospective cohort study. BMJ Open. 2021;11(1):e041854.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Sekgololo JM, Chauke RF, Tshazi N. A rare giant extraocular, anterior chest wall sebaceous carcinoma. Int J Surg Case Rep. 2020;66:110–4.

    Article  PubMed  Google Scholar 

  117. Zürcher M, Hintschich CR, Garner A, Bunce C, Collin JRO. Sebaceous carcinoma of the eyelid: a clinicopathological study. Br J Ophthalmol. 1998;82(9):1049–55.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Muqit M, Roberts F, Lee W, Kemp E. Improved survival rates in sebaceous carcinoma of the eyelid. Eye. 2004;18(1):49–53.

    Article  CAS  PubMed  Google Scholar 

  119. Song A, Carter KD, Syed NA, Song J, Nerad JA. Sebaceous cell carcinoma of the ocular adnexa: clinical presentations, histopathology, and outcomes. Ophthalmic Plast Reconstr Surg. 2008;24(3):194–200.

    Article  PubMed  Google Scholar 

  120. Hirano T, Yunoki T, Furusawa Y, Tabuchi Y, Hayashi A. Bioinformatics analysis of the microRNA-mRNA network in sebaceous gland carcinoma of the eyelid. Mole Med Rep. 2021;23(1):1.

    Google Scholar 

  121. Turner SD, Lamant L, Kenner L, Brugieres L. Anaplastic large cell lymphoma in paediatric and young adult patients. Br J Haematol. 2016;173(4):560–72.

    Article  PubMed  Google Scholar 

  122. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. WHO Press: International agency for research on cancer; 2008.

  123. Pomari E, Basso G, Bresolin S, Pillon M, Carraro E, d’Amore E, et al. NPM-ALK expression levels identify two distinct subtypes of paediatric anaplastic large cell lymphoma. Leukemia. 2017;31(2):498–501.

    Article  CAS  PubMed  Google Scholar 

  124. Brugieres L, Le Deley M-C, Rosolen A, Williams D, Horibe K, Wrobel G, et al. Impact of the methotrexate administration dose on the need for intrathecal treatment in children and adolescents with anaplastic large-cell lymphoma: results of a randomized trial of the EICNHL Group. J Clin Oncol. 2009;27(6):897–903.

    Article  CAS  PubMed  Google Scholar 

  125. Le Deley M-C, Rosolen A, Williams DM, Horibe K, Wrobel G, Attarbaschi A, et al. Vinblastine in children and adolescents with high-risk anaplastic large-cell lymphoma: results of the randomized ALCL99-vinblastine trial. J Clin Oncol. 2010;28(25):3987–93.

    Article  PubMed  Google Scholar 

  126. Gascoyne RD, Aoun P, Wu D, Chhanabhai M, Skinnider BF, Greiner TC, et al. Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma. Blood. 1999;93(11):3913–21.

    Article  CAS  PubMed  Google Scholar 

  127. Garbin A, Lovisa F, Holmes AB, Damanti CC, Gallingani I, Carraro E, et al. miR-939 acts as tumor suppressor by modulating JUNB transcriptional activity in pediatric anaplastic large cell lymphoma. Haematologica. 2021;106(2):610.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This study was approved and supported by the Deputy of Research and Technology of Fasa University of Medical Sciences, Fasa, Iran, with the number 402293.

Funding

The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research received no specific grant.

Author information

Authors and Affiliations

Authors

Contributions

HK and PKA contributed to the search performance, manuscript writing, and primary revising. FD, AG, and PKE contributed to figure and table designing and manuscript editing. HF and RT designed the study and performed the final revision. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Hamed Fouladseresht or Reza Tabrizi.

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kouchaki, H., Kamyab, P., Darbeheshti, F. et al. miR-939, as an important regulator in various cancers pathogenesis, has diagnostic, prognostic, and therapeutic values: a review. J Egypt Natl Canc Inst 36, 16 (2024). https://doi.org/10.1186/s43046-024-00220-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43046-024-00220-8

Keywords