[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Next Article in Journal
Unveiling Secondary Mutations in Blended Phenotypes: Dual ERCC4 and OTOA Pathogenic Variants Through WES Analysis
Previous Article in Journal
Genome-Wide Identification and Characterization of GRAS Transcription Factor Family in Cultivated Hybrid Sugarcane ZZ1 (Saccharum officinarum) and Their Role in Development and Stress
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Critical Review on microRNAs as Prognostic Biomarkers in Laryngeal Carcinoma

by
Kristina S. Komitova
1,
Lyuben D. Dimitrov
1,
Gergana S. Stancheva
2,
Silva G. Kyurkchiyan
2,
Veronika Petkova
2,
Stoyan I. Dimitrov
1,
Silviya P. Skelina
3,
Radka P. Kaneva
2 and
Todor M. Popov
1,*
1
Department of ENT, Medical University, 1000 Sofia, Bulgaria
2
Molecular Medicine Center, Medical University, 1000 Sofia, Bulgaria
3
Department of Neurology, Medical University, 1000 Sofia, Bulgaria
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(24), 13468; https://doi.org/10.3390/ijms252413468
Submission received: 10 November 2024 / Revised: 7 December 2024 / Accepted: 11 December 2024 / Published: 16 December 2024
(This article belongs to the Special Issue The Role of RNAs in Cancers: Recent Advances)

Abstract

:
During the past decade, a vast number of studies were dedicated to unravelling the obscurities of non-coding RNAs in all fields of the medical sciences. A great amount of data has been accumulated, and consequently a natural need for organization and classification in all subfields arises. The aim of this review is to summarize all reports on microRNAs that were delineated as prognostic biomarkers in laryngeal carcinoma. Additionally, we attempt to allocate and organize these molecules according to their association with key pathways and oncogenes affected in laryngeal carcinoma. Finally, we critically analyze the common shortcomings and biases of the methodologies in some of the published papers in this area of research. A literature search was performed using the PubMed and MEDLINE databases with the keywords “laryngeal carcinoma” OR “laryngeal cancer” AND “microRNA” OR “miRNA” AND “prognostic marker” OR “prognosis”. Only research articles written in English were included, without any specific restrictions on study type. We have found 43 articles that report 39 microRNAs with prognostic value associated with laryngeal carcinoma, and all of them are summarized along with the major characteristics and methodology of the respective studies. A second layer of the review is structural analysis of the outlined microRNAs and their association with oncogenes and pathways connected with the cell cycle (p53, CCND1, CDKN2A/p16, E2F1), RTK/RAS/PI3K cascades (EGFR, PI3K, PTEN), cell differentiation (NOTCH, p63, FAT1), and cell death (FADD, TRAF3). Finally, we critically review common shortcomings in the methodology of the papers and their possible effect on their results.

1. Introduction

During the past decade, a vast number of studies were dedicated to unravelling the obscurities of non-coding RNAs in all fields of the medical sciences [1,2]. A great amount of data has been accumulated, and consequently a natural need for organization and classification in all subfields arises. The aim of this review is to summarize all reports on microRNAs that were delineated as prognostic biomarkers in laryngeal carcinoma. Additionally, we attempt to allocate and organize these molecules according to their association with key pathways and oncogenes affected in laryngeal carcinoma. Finally, we critically analyze the common shortcomings and biases of the methodologies in some of the published papers in this area of research.
MicroRNAs (miRNAs) are a class of small (around 22 nucleotides long), non-coding, single-stranded RNA molecules [3]. MiRNAs play a role in regulating gene expression of target mRNAs post-transcriptionally. They usually act as inhibitors by degrading or repressing the translation of targeted gene transcripts, which is achieved by guiding a multi-protein silencing complex to partially complementary sites on the mRNA of target genes, therefore leading to gene silencing [4,5]. However, in some cases miRNAs can also bind to target sequences in promoters and thus act as gene activators [6]. Due to the partial complementarity of the targeted sites on the mRNA, a single miRNA can take part in the regulation of many different mRNAs, but at the same time, different miRNAs can modulate an individual mRNA [4].
Since they can recognize a variety of mRNA transcripts, through their regulatory function miRNAs can interfere with many fundamental cellular processes, including division, proliferation, differentiation, development, apoptosis, angiogenesis, etc. Abnormal expressions of some miRNAs have been shown to be involved in different pathogenic processes, including carcinogenesis [6].
With the caveat that the size and direction of the regulatory effects of some miRNAs may vary and may depend on how they interact with the effects of other miRNAs, we could simplify and classify miRNAs into two main groups, depending on how they affect the development of tumors:
-
Tumor suppressor miRNAs, which negatively regulate the expression of oncogenes or genes that induce apoptosis. When downregulated, their suppressive function is decreased.
-
Oncogenic miRNAs, which activate oncogenes or silence tumor suppressor genes. Their overexpression facilitates oncogenesis [5].
Recently, extensive research has been carried out on the connection between miRNAs and different types of carcinomas, including LSCC (laryngeal squamous cell carcinoma). While dysregulated expression of miRNAs in LSCC tissues and patient blood as compared with normal adjacent tissues has been observed on many occasions and has been associated with diagnostic features, more and more studies have focused on the potential prognostic value of miRNAs’ expression changes, suggesting the usefulness of miRNAs as prognostic biomarkers for that specific type of cancer.

2. Methods

A literature search was performed using the PubMed and MEDLINE databases with the keywords “laryngeal carcinoma” OR “laryngeal cancer” AND “miRNA” OR “miRNA” AND “prognostic marker” OR “prognosis”. Only research articles written in English were included, without any specific restrictions on study type. Two authors independently screened the data sources, and data analysis was also carried out by both authors. The search strategy is outlined in Table 1.

3. MiRNAs as Prognostic Biomarkers in Laryngeal Carcinoma

We have found 43 articles that report 39 miRNAs with prognostic value associated with laryngeal carcinoma, all of which are summarized in Table 2 along with the major characteristics and methodology of the respective studies.

3.1. MiR-19a (Cluster 17-92), Chromosome 13

The miR-17/92 cluster (also known as ‘oncomiR-1’) is one of the best-studied miR clusters and is very often dysregulated in hematopoietic and solid cancers. It is located at chromosome 13. MiR- 19a is part of that cluster along with miRNA 17-5p, miRNA 18a, miRNA 20a, miRNA 19b-1, and miRNA 92a-1. Tian-Yi Wu et al. used quantitative real-time PCR to detect miR-19a expression in fresh frozen LSCC tissues of 83 patients with LSCC stage T1–T4 [8]. Patients with high miR-19a expression levels were shown to have significantly shorter overall survival than those with low miR-19a expression levels. Using in vitro functional studies and animal xenograft tumors, Tian-Yi Wu et al. assessed the effect of miR-19a on proliferation, apoptosis, and tumor growth. After a multivariate Cox analysis, they suggested miR-19a as an independent prognostic factor of overall survival rate of LSCC patients.

3.2. X-Chromosome-Located MiRs: miR-20b (Cluster 106a-363) and miR-766-5p

MiR-20b is a part of the tumor-related miR-106a/363 cluster, located at the X-chromosome. T.L. Pantazis et al. studied the potential prognostic value of miR-20b in a group of 105 patients diagnosed with T1–T4-stage LSCC using real-time qPCR methodology [9]. Kaplan–Meier survival analysis showed that negative miR-20b-5p expression status predicts favorable prognosis regarding DFS (disease-free survival) and OS (overall survival) of LSCC patients. Through N-stage stratification of the patient groups, it is demonstrated that the prognostic value of miR-20b-5p holds for node-negative patients, implying its possible independence from the extent to which the cancer has spread.
Another X-chromosome-located MiR is miR-766-5p. Fu Chen et al. examined miR-766-5p as a part of the circSHKBP1/miR-766-5p/HMGA2 axis in a cohort of 60 LSCC patients (stages I–IV) [48]. The results reveal that the miR-766-5p expression level is inversely proportional to circSHKBP1 expression, implying that circSHKBP1 serves as sponge for miR-766-5p. Furthermore, overall survival was found to be shorter in LSCC patients with decreased miR-766-5p expression levels.

3.3. MiR-21—The Classic miR Oncogene

MiR-21 has been reported to be overexpressed and also act as oncogenic miRNA in many human malignancies. Three studies reviewed the expression of miR-21 in patients with LSCC and assessed its association with patient survival. An Hu et al. (2014) evaluated miR-21 levels by quantitative real-time PCR in 46 fresh frozen samples of patients with laryngeal carcinoma stage T1–T4 [10]. It was demonstrated that the miR-21/miR-375 ratio is extremely sensitive and specific for disease prediction, and in addition, high miR-21 expression was linked to bad prognosis. Additionally, An Hu et al. (2015) demonstrated the potential of the MiR-21/miR-375 ratio as a prognostic factor in LSCC patients [11]. The relationship between MiR-21 and survival rates was also confirmed by S. Gao et al., who arrived at that conclusion after performing MiR array analysis as well as qRT-PCR in 236 serum samples stored at −80 °C, before RNA isolation [12].

3.4. MiR-23a, Cluster 23a-24-2, Chromosome 19, and miRs-26a/26b

MiR-23a is a component of the miR-23a/24-2/27a cluster. X.W. Zhang et al. described the role of miR-23a in tumor progression, proving its ability to promote cell migration and invasion in laryngeal carcinoma cell cultures when overexpressed [13]. Moreover, the association of increased miR-23a levels with worse five-year survival rates was evaluated by qRT-PCR in 52 pairs of LSCC and adjacent normal tissue and subsequent statistical analysis.
The same chromosome (chromosome 19) also carries miRs-26a/26b. According to D. Liu et al., miR-26a is upregulated and positively related to poor overall survival in human LSCC [14]. In addition to the expression of miR-26a in 56 fresh frozen samples, the authors also investigated the effects of sevoflurane in cell proliferation and apoptosis using cell lines and in vivo experiments. They came to the conclusion that sevoflurane induces apoptosis and affects EMT in laryngeal carcinoma via miR-26a targeting.
MiR-26b, similarly to miR-26a, is also reported to be highly expressed and OS- and DFS-related in LSCC tissues. S. Wang et al. aimed to elucidate the mechanisms underlying miR-26b promotion of cancer proliferation [15]. They found that miR-26b knockdown induced autophagy by targeting ULK2 and inactivated the PTEN/AKT pathway in Hep-2 cells, leading to inhibition of LSCC progression.

3.5. MiR-31 and miR-34

The expression of miR-31 was studied by H. Qiang et al. [16], using fresh frozen pairs from a heterogeneous cohort of patients, including predominantly laryngeal and hypopharyngeal SCC, and different cell lines served as material for the analysis. A significant association of low miR-31 levels with longer survival was discovered, along with a close correlation between high miR-31 and some clinicopathological features such as poor tumor differentiation, presence of metastasis, and advanced staging.
MiR-34s form a family of three members—miR-34a, miR-34b, and miR-34c—encoded on two different chromosomes (Chr1 and Chr11). They are often regarded as tumor suppressor miRs, and we found three studies proving their downregulation in laryngeal cancer. Massimo Re et al. (2015) detected the levels of miR-34c-5p in 90 LSCC FFPE blocks by reverse-transcription qPCR in a homogeneous group of T3–T4 LSCC patients and related lower levels to unfavorable DFS, OS, and high risk of recurrence [17]. The author and his team carried out further research on this relationship (Massimo Re et al. (2017)), which confirmed their initial results [18]. Z. Shen et al. (2012) described the ability of miR-34a to suppress cell proliferation by arresting cells at the G0/G1 phase, to target survival and last but not least to alter patient outcome [19].
Apart from the effects of quantitative changes, epigenetic changes in miR-34a and their outcomes have also been analyzed. Z. Shen et al. (2016) applied bisulfite pyrosequencing technology to measure the methylation levels of the miR-34a promoter [20]. Using Kaplan–Meier survival curves, they illustrated the correlation between promoter hypermethylation and decreased overall survival.
It should be noted, however, that miR-34s cannot be unanimously classified as tumor suppressor miRNAs. Conversely to the results outlined above, I. Piotrowski et al. observed upregulation of miR-34a-5p, miR-34b-5p, and miR-34c-5p in LSCC and reported that high expression of miR-34a-5p (HR = 3.466) and miR-34b-5p (HR = 6.761) was associated with a higher hazard and lower survival rate [21].

3.6. MiR-93, miR-210, and miR-144b

MiR-93-5p, miR-144-3p, and miR-210-3p are all strong predictors of recurrence-free survival.
This was shown by Popov et al. (2022) through examining their expression levels in LSCC samples using microarray profiling and RT-qPCR [22]. MiR-93-5p and miR-210-3p were clearly established as oncogenes, since their upregulation was related to significantly worse survival rates. On the other hand, miR-144-3p was demonstrated to be a tumor suppressor, as it appeared to be heavily downregulated in most cases.
This is consistent with the findings of S. Y. Zhang et al., who investigated the expression of miR-144-3p in laryngeal carcinoma and its in vitro and in vivo effects and found it to be a tumor suppressor which inhibits cellular migration, invasion, EMT, and cell growth [35]. S. Y. Zhang et al. also analyzed the expression of ETS-1, which miR-144-3p suppresses, and found that lower ETS-1 expression was linked to significantly higher survival rates.
Notably, Popov et al. discovered a paradoxical relationship between survival rates and the expression of miR-144-3p in that, despite its classification as a tumor suppressor, patients with relatively high expression levels had significantly worse survival rates when compared to patients with a more pronounced downregulation. How the expression levels of those three miRs related to each other was evaluated by Popov et al. by running a Spearman’s rank-order correlation, which revealed a very strong statistically significant positive correlation between miR-93-5p and miR-210-3p, as well as a strong association between miR-93-5p and miR-144-3p and between miR-144-3p and miR-210-3.

3.7. Chromosome-1-Located miRs: miR-9, miR-29c, miR-101, miR-200a, and miR-137

MiR-9 is located in Chr1 and has been reported to be overexpressed in different types of cancer. S. Wu et al. focused their study on the dysregulation of miR-9 expression and its potential use as a prognostic factor of outcomes in patients with laryngeal carcinoma [7]. Levels were detected to be significantly higher in tumor tissues from 103 patients diagnosed with LSCC compared with adjacent non-cancerous tissues. Overexpression was further linked to shorter OS. MiR-9 upregulation was found to be strongly correlated with some clinicopathological parameters such as tumor differentiation, lymph node metastasis, clinical stage, and thyroid cartilage invasion.
MiR-29c can be found in the same chromosome; however, its levels of expression in LSCC (unlike miR-9) are reported to be low. R. Fang et al. extracted miR-29c from formalin-fixed, FFPE tissue samples of 66 patients by quantitative real-time PCR and associated its decreased expression with worse overall survival, along with smoking index, tumor size, tumor site, tumor differentiation, T classification, TNM stage, and lymph node metastasis [44].
MiR-101 encoding genes are located at two different chromosomes (Chr1p31.3 and Chr9p24.1). M.H. Li et al. aimed to discover the role of miR-101 in LSCC cell proliferation, invasion, apoptosis, and cell cycle regulation and applied both in vitro and in vivo methods [31]. They determined miR-101 to be a downregulated tumor suppressor miR, correlated with advanced T stage, lymph node metastasis, and poor outcome. Furthermore, they identified CDK8 as its target gene, with miR-101 functioning as a CDK8, beta-catenin, and cyclin D1 suppressor.
The miR-200 family is composed of five members (miR-200a, miR-200b, miR-200c, miR-141, and miR-429). R. Cappellesso et al. studied the role of Slug and E-cadherin, which are distinctive markers of epithelial–mesenchymal transition, in LSCC patient outcome [45]. Additionally, using a quantitative reverse-transcription PCR assay to analyze the expression of miRs extracted from FFPE block samples, they assessed their potential to alter Slug gene regulation. Moreover, the analysis revealed that in cases with disease recurrence, the expression of miR-200a and miR200c is significantly lower.
MiR-137 is also placed on chromosome 1 (1p21.3). It has been described as taking part in cell cycle regulation via the G1/S-phase checkpoint. Unlike other authors, S. Langevin et al. explored not only the quantitative changes in miR levels, but also the methylation status of miR-137 promoter by methylation-specific polymerase chain reaction [46]. The samples used in this study are from patients with HNSCC; however, it is expressly clarified that results could be applied to all different sites (oral cavity, pharynx, and larynx) with no significant differences.

3.8. Chromosome-2-Located miRs: miR-1246, miR-149, and miR-375

The genes of miR-1246, miR-149, and miR-375 are encoded in chromosome 2.
Q. Huang et al. investigated miR-1246 levels in both laryngeal carcinoma tissues and patients’ blood, and expression was reported to be higher in cancerous versus control samples [27]. The study pointed out small extracellular vesicles (sEV) as the prevailing form in which extracellular miR-1246 is found in samples, rather than in its soluble form. In vitro analysis showed that lowering the levels of miR-1246 in sEV form leads to inhibition of tumorigenesis and cancer progression. The mechanism of action of sEV miR-1246 was related to CCNG2 targeting. A Kaplan–Meier curve was presented, suggesting miR-1246 upregulation as a potential predictor of unfavorable prognosis.
MiR-149 and miR-375 are both reported to be downregulated and associated with adverse outcome in LSCC patients. The data presented by Y. Xu et al. indicated correlation between decreased miR-149 levels and some clinicopathological factors, such as stage, differentiation, and presence of metastasis [37]. In vitro analysis in Hep-2 cell cultures shone light on the suppressive effects of ectopic miR-149 expression on cell proliferation. Correspondingly, An Hu et al. used formalin-fixed paraffin-embedded tissue samples from 46 LSCC patients in order to assess the levels of miR-375 and suggested an association of miR-375 depletion with supraglottic tumor site and alcohol consumption [10]. Interestingly, they look at miR-375 as a part of the combined miR-21/miR-375 ratio diagnostic biomarker, which was shown to be increased in LSCC. Another team, F. Dai et al., confirmed the correlation between miR-375 and CST1, one of its putative target genes, and used LSCC cell lines to explore the suppressive effects of miR-375 on cell viability and migration and its ability to promote apoptosis [42]. Being classified as a tumor-suppressive microRNA with strong association between its dysregulation and oncogenesis, several studies were conducted to examine the potential of miR-375 as a diagnostic and prognostic biomarker in different subsites of head and neck tumors. A significant role of miR-375 downregulation was shown in detection and long-term follow-up not only in LSCC, but also in oral carcinoma [51,52].

3.9. Chromosome-5-Located miRs: miR-146a, miR-143, miR-145, and miR-449a

Two research teams found miR-143 to be downregulated and related to poor overall survival in patients suffering from LSCC [33,34]. Both teams used series of in vitro and in vivo analyses to examine the impact of miR-143 expression on proliferation, invasion, and migration of LSCC cells and tumor growth. According to one of the teams, L. Han et al., miR-143-3p can be described as one of the MAGE-A9-related miRNAs, revealing an inverted correlation between miR and MAGE-9 protein levels [33]. The other team, F. Zhang et al., found that miR-143 directly targeted the k-Ras gene, suppressing tumor progression via the k Ras/Raf/MEK/ERK axis [34].
Increased levels of miR-146a and miR-449a were detected in HPV-negative fresh frozen LSCC samples by I. Piotrowski et al. [21]. Based on statistical analysis, the author and his team concluded that patients with high miR-146a expression levels had poorer outcomes. Surprisingly, low miR-449a levels predicted shorter survival. In LSCC, the combined expression of miR-449a-5p, miR-6510-3p, and miR-133a-5p showed good potential as a biomarker for tumor tissues, simultaneously boasting high sensitivity and high specificity.
Another miR that is derived from the same chromosome and can be classified as prognostic for patients with LSCC is miR-145-5p. W. Gao et al. assessed both the quantitative irregularities of miR-145-5p levels in cancerous tissues and the occurring epigenetic changes, such as promoter hypermethylation, leading to the aforementioned aberrant expression [36]. Further analysis indicated an inverse relationship between miR-145-5p and FSCN1 and pointed out the combined low miR-145-5p/high FSCN1 expression as an independent marker of poor patient survival.

3.10. Chromosome-9-Located miRs: miR-126 and miR-181a

MiR-126 levels were measured in LSCC plasma samples by X. Sun et al., who reported a significant association between miR-126 downregulation and low grade of tumor differentiation as well as unfavorable outcome [50]. Moreover, analysis showed an inverse relationship between miR-126 and Camsap1. Therefore, high Camsap1 expression leading to activation of the processes of formation and aggregation of microtubules was found to be inhibited by miR-126.
X. Zhao et al. described the role of miR-181a in laryngeal carcinoma progression as a part of the miR-181a/GATA6 axis [47]. Advanced T stage, positive cervical lymph node metastasis, and shorter overall survival were related to low miR-181a expression in LSCC tissue samples. Additional Hep-2 cell culture analyses helped the research team to evaluate the effect of miR-181a and GATA6 levels on cell proliferation, migration, invasion, and apoptosis. MiR-181a/SIRT1 is another axis known to be dysregulated in tumors of different types and sites [53]. X.M. Yu et al. used qRT-PCR and immunohistochemical methods to assess the levels of SIRT1 in samples from patients with laryngeal and hypopharyngeal carcinoma and found a statistically significant correlation between low SIRT1 levels and advanced clinical stage as well as positive lymph node metastasis [54].

3.11. Chromosome-14-Located miRs: miR-203 and miR-300

The downregulation of miR-203 was determined by L. Tian et al. to go hand in hand with a low grade of differentiation, advanced stages, positive lymph nodes, and, last but not least, poor five-year survival rate in patients with laryngeal squamous cell carcinoma [40]. A combination of in vitro and in vivo experiments revealed the ability of miR-203 to suppress LSCC cell proliferation, invasion, and tumor growth and more importantly to target the ASAP1 gene, to promote the E-cadherin expression, and to lower the CD44 (marker of cancer stem cells) levels.
Unlike miR-203, miR-300, located in the same chromosome, was reported by F.Y. He et al. to be notably upregulated in LSCC tissues and altered according to cervical lymph node status and TNM stage [41]. A Kaplan–Meier curve was used to illustrate that lower levels of miR-300 were linked to decreased OS.

3.12. Chromosome-17-Located miRs: miR-301a, miR-632, miR-10a, and miR-195

MiR-301a and miR-632, both located at chromosome 17, were observed to be significantly upregulated in laryngeal carcinoma samples. Y. Lu et al. conducted in vitro analysis showing that miR-301a knockdown resulted in suppressed proliferation, duplication, and colony formation along with activation of apoptosis, G0/G1 cell cycle arrest, and alteration of some of the most representative EMT markers: E-cadherin, N-cadherin, vimentin, MMP2, and MMP [25]. Y. Lu and their team identified Smad4 as a miR-301a target and outlined the combined high miR-301a-3p/low Smad4 status as an independent prognostic marker of adverse outcome. In contrast, Y.C. Cao et al. used blood samples to assess the correlation between increased miR-632 levels and low differentiation, positive lymph nodes, advanced stage, and poor OS and DFS [30].
Opposingly, miR-10a expression was found to be decreased in serum samples from patients suffering from LSCC. S. Gao et al. used an miR array method to detect miR-10a levels and further statistical analysis to explore their association with different clinicopathological factors and five-year survival rate [12]. The team examined miR-10a as a part of the combined miR-21/miR-10a marker and concluded that a higher ratio correlated to worse survival.
In two mutually complementary articles, both published in 2017, Y. Shuang et al. laid out their findings of a significant downregulation of miR-195 in LSCC tissues and linked its lowered expression to late T stage, positive lymph nodes, advanced clinical stage, distant metastasis, and poor prognosis [38,39]. MiR-195 possibly restrained growth, invasion, migration, and colony formation of tumor cells, induced G1 cell cycle arrest, and promoted apoptosis through targeting and suppressing DCUN1D1 and Bcl-2. A high miR-195/low DCUN1D1 expression combination was shown to imply longer overall survival and vice versa.

3.13. Chromosome-19-Located miRs: miR-196b and miR-519a

MiR-196b can be classified as oncogenic miRNA. Two studies reported its upregulation in LSCC tissues and suggested its potential as prognostic marker of poor outcome. M. Luo et al. described a statistically significant relationship between miR-196b levels and tumor stage and differentiation [23]. Furthermore, they identified PCDH-17 as a direct miR-196b target. On the other hand, X. Zhao et al. found that miR-196b and SOCS2 expressions were inversely correlated, and through that relationship, miR-196b boosted proliferation and invasion and suppressed apoptosis in LSCC cell lines [24].
The possible use of miR-519a, located at the same chromosome, as a marker of prognosis in laryngeal carcinoma was studied by Z. Shen et al. [43]. They used the reverse-transcription PCR method in 96 LSCC samples and observed that decreased miR-519a levels were correlated to worse disease-free survival. Moreover, LSCC cell cultures were used to observe the negative effects of miR-519a on cell growth and its ability to cause G2/M cell cycle arrest. HuR was found to be a miR-519a target gene. However, protein levels were regulated without an impact on mRNA levels.

3.14. MiR-1205 Chr8, miR-378c-5p Chr10, miR-125b-5p Chr11, miR-296-5p Chr20, and miR-155 Chr 21

MiR-1205 dysregulation and prognostic significance in laryngeal cancer was explored by P. Li et al. [49]. They reported a downregulation of miR-1205 in patients with LSCC and its association with clinicopathological markers, such as clinical stage, T stage, and lymph node metastasis. Remarkably, this article determined a strong association between miR-1205 and E2F1, an oncogene playing a pivotal role in laryngeal carcinogenesis. E2F1 was not only found to be a target of miR-1205, but also an inhibitor of its expression, through binding to its promotor.
In order to assess the expression of miR-378c-5p in laryngeal carcinoma, Y. Xu et al. used samples from patients stratified in three groups—patients with LSCC, patients with vocal cord polyposis, and healthy controls [26]. High levels of miR-378 were related to the presence of LSCC, low grade of differentiation, and advanced stage. Postoperative control samples were taken, which allowed the authors to observe miRNA-378 expression over time. A significant decline in expression was detected in both LSCC and VCP groups six months after the surgery, with miRNA-378 levels being significantly higher in patients with tumor recurrence. High miR-378c-5p expression was also linked to higher hazard and lower survival rate in laryngeal cancer by I. Piotrwski et al. [21].
F. Chen et al. published an article designating miR-125-5p as a tumor suppressor miR which leads to poor overall survival in LSCC patients when downregulated [32]. Cell cultures and animal xenograft models were used to determine the role of miR-125-5p as a cell growth and glycolysis inhibitor, a promoter of apoptosis and, last but not least, as a regulator of MAP3K9 levels.
The expression of miR-296-5p was studied in a small homogenous cohort of 34 patients suffering from early-stage LSCC, treated with radiotherapy only. D. Maia et al. observed a statistically significant correlation between increased miR-296 levels and both radioresistance and recurrence of the disease [28].
MiR-155, encoded in chromosome 21, was analyzed and reported to be higher in laryngeal carcinoma patients by X. Zhao et al. [29]. The analysis showed an association between its upregulation and advanced stages of the disease, low grade of differentiation, and positive lymph node status. Moreover, it implied a positive relationship between miR-155 and both YB-1 and c-Myb genes, and a Kaplan–Meier curve was used to illustrate the statistically significant relationship between the expression of YB-1 and the unfavorable prognosis of patient survival.

3.15. MiRNAs in Plasma

The main focus in this review has been set to be prognostic miRNA biomarkers in tumor tissue samples (fresh frozen FFPE, etc.), but for the sake of thoroughness, we are obliged to also briefly discuss plasma samples as sources of miRNAs for prognosis. A liquid biopsy is an emerging technology which consists in sampling non-solid body tissues (usually blood and saliva) and analyzing them for different circulating biomarkers, including microRNAs. Liquid biopsies could be an effective way of diagnosing and monitoring cancer because of how non-invasive, rapid, and repeatable they are [55]. Studies have already shown liquid biopsies to have an application for detecting oral cancer, as well as cancer located in other head and neck subsites [56], with circulating microRNAs functioning as biomarkers. The main reason liquid biopsies are still not widely used for screening is the poor result accuracy of the current technology when applied in a clinical setting. Even under academic conditions, problems with reporting and inconsistencies in collecting, storing, and processing samples compromises the results of studies in that area [57]. On some occasions, analysis of different types of samples reaches congruent conclusions. For example, as we already discussed in Section 3.3, both An Hu et al. (2014) [11], using fresh frozen samples, and S. Gao et al. [12], using serum samples, independently showed a relation between MiR-21 expression and LSCC survival rates. Another case, presented in Section 3.8 above, is that of Huang Q. et al. [27], who used a combination of sample types and proved that miR-1246 is overexpressed in both plasma sEV and LSCC tissues and that such overexpression is linked to worse prognosis. However, care should be taken when comparing miRNA expression in liquid biopsies and miRNA expression in tissue biopsies, as those should not necessarily be considered identical metrics. Before hypothesizing the predictive power of miR-21 serum levels in their article, S. Gao et al. [12] explicitly stated that relevant previous studies had focused almost exclusively on tissue samples and that, as of then, the link between circulating MiR-21 levels and LSCC prognosis had remained unknown.

4. Systematic Classification of miRs Exhibiting Prognostic Significance Based on Their Association with Pivotal Altered Pathways and Oncogenes in Laryngeal Carcinoma

We reviewed all available cancer studies that reported any molecular association between the most prominently affected oncogenes and pathways in laryngeal carcinoma [58] and the miRs that we have found to possess prognostic value in the literature. We have a summary of this deep analysis of the literature given in Table 3 along with the respective references. Additionally, we have prepared a Venn diagram to illustrate the collected data (Figure 1).

4.1. MiRs Demonstrating Prognostic Significance and Their Association with Oncogenes and Pathways Connected with the Cell Cycle: p53, CCND1, CDKN2A/p16, and E2F1

Out of the 43 (86%) referenced miRs with prognostic value, 37 are found to be in molecular association with p53 and its molecular pathway (Table 3). In most of the cases, we have found a sufficient number of papers for each miR reporting such a relationship. The percentage astoundingly corresponds to the percentage of p53 mutation—84%—found in patients with squamous cell carcinoma of the head and neck [58]. These findings suggest that most of the prognostic biomarkers are in regulatory co-dependency with the p53 oncogene. Only miRs miR-9, miR-20b, miR-196b, miR-301a, and miR-632 lack the data to be in a common regulatory network. Exclusively, a study from 2014 [59] reports that miR-9 is activated by HPV E6 in a p53-independent manner. We found that 79.5% (31 out of the 39 miRs) are reported to be in a regulatory dependence with the Cyclin D1 (CCND1) gene. MiR-19, miR-26a/b, miR-31, miR-196b, miR-10a, miR-203, and miR-300 are not reported in the literature to be associated with CCND1. Moreover, two studies confirm the functional 31 [111]/miR-10a [180]. Only 25.6% of the miRs that have prognostic value are reported to have common regulatory associations with CDKN2A/p16. A higher number of associated miRs is found when E2F1 (46%) is analyzed. This higher rate of association in comparison to p16 can be explained by the fact that E2F1 is involved in cell proliferation, migration, and cell cycle in both HPV-positive and HPV-negative tumors. Interestingly, we can see from Table 3 that only two miRs overlap in terms of regulatory networks for both CDKN2A/p16 and E2F1: miR-9 and miR-181a. Generally, we could sum up these findings as follows: (1) Almost all miRs with prognostic value are in regulatory association with cell cycle oncogenes (data lack only for miR-196b to be in any regulatory relationship with the cell cycle oncogenes). (2) Predominantly, biomarker miRs are associated with p53 and Cyclin D1. (3) According to available data, regulatory association of biomarker miRs with E2F1 excludes such connections with CDKN2A/p16 and vice versa—in terms of dependence, both oncogenes divide biomarker miRs in two distinct subgroups (chi-square, p < 0.05). Some of those differentiations are backed up with studies reporting regulatory independence—see Table 3 [82,112,144,186,239,260,283].

4.2. MicroRNAs Demonstrating Prognostic Significance and Their Association with Oncogenes and Pathways Connected with the RTK/RAS/PI3K Cascades: EGFR, PI3K, and PTEN

A very high percentage of the reviewed miRs—89.7% (35 out of 39)—were found to have regulatory links with the PTEN/PI3K cascade. Only miR-20b, miR-632, miR-200a, and miR-1205 lack the data to be in a common regulatory network. We have to acknowledge that the majority of dysregulated miRs with prognostic value are functionally connected in some way with the PTEN/PI3K pathway despite the fact that the percentage of genetic alterations in this cascade is within at least an order of magnitude less—ranging between 6% and 56% of the cases [58]. This is a major difference when comparing the same miRs and their relation to cell cycle oncogenes and pathways, where the percentages generally correspond. These findings would suggest that while not always genetically altered, the PTEN/PI3K cascade is phenotypically a key player for tumor progression and prognosis. Similar dissociation is evident with EGFR—only 15% of the HPV-negative patients and 6% of the HPV-positive patients have genetic alterations of EGFR [58], and a striking 64% of the dysregulated biomarker miRs from our list are found to have a regulatory effect on EGFR. These data indirectly suggest that EGFR and PI3K/PTEN pathways play a very significant role in laryngeal cancer progression and prognosis that extends far beyond the context of their genetic alterations’ frequency in these patients.

4.3. MiRs Demonstrating Prognostic Significance and Their Association with Oncogenes and Pathways Connected with Cell Differentiation: NOTCH, p63, and FAT1

Of the biomarker miRs, 59% were found be associated functionally with the NOTCH cascade and only 25.6% with p63. The frequency of genetic alterations in NOTCH in patients with squamous cell carcinoma revolve around 17% and 26%, whereas for p63, 19% and 28%, for HPV-negative and HPV-positive carcinoma, respectively [58]. Despite the very high rate of mutations in FAT1 (32%), we have found only four miRs from our list that are reported to have regulatory connections with this gene. We cannot assess whether the available data in the literature are just not sufficient or whether, at least from a prognostic point of view, this molecular domain does not have such a major role.

4.4. MiRs Demonstrating Prognostic Significance and Their Association with Oncogenes and Pathways Connected with Cell Death: FADD and TRAF3

We have looked into cell death domain by choosing FADD and TRAF3 as the most prominently genetically altered genes. In 32% of the HPV-negative head and neck cancers, FADD is found to carry driver mutations, whereas TRAF3 is genetically altered in 22% of the HPV-positive patients [58]. Despite these significant rates, we could find almost no data that could connect those two oncogenes with the list of miRs with prognostic value in laryngeal carcinoma. Only miR-146a is reported to be regulatorily associated with both FADD [136] and TRAF3 [137]. Additionally, miRs miR-101, miR-145, and miR-149 are connected functionally with FADD [197,234,242]. Similarly to FAT1, we cannot assess whether the available data in the literature are just not sufficient or whether this molecular domain does not carry such a weight from a prognostic standpoint.

5. Critical Remarks on Publications Reporting Prognostic Biomarkers

After a thorough analysis of the methodology of the publications that report miRs as prognostic markers in laryngeal carcinoma, we have found a number of major concerns that are present almost ubiquitously throughout the reviewed papers. The first very important shortcoming among the majority of the studies is the inclusion of a very wide range of stages in the studied cohort. From 43 articles that report miRs as prognostic biomarkers, only four have focused their research on a more homogenous group of patients [21,22,23,53], e.g., only advanced stage (stage T3/T4 or at least stage III/IV) or early-stage carcinoma. The vast majority of the papers included patients ranging across all the tumor stages (see Table 2). This is a predicament of bias in the design of the study, since it is a well-established fact that a great number of oncogenes/dysregulated molecules are consistently upregulated vs. their matching controls in a grade- and stage-dependent manner [310]. If we conscientiously take into account this significant bias, we cannot assess whether the degree of dysregulation of those miRs is not just a correlation with stage and we are measuring its stage-dependent prognostic value. From this perspective, one might conjecture that these dysregulated miRs may serve as biomarkers not solely for strict prognostic purposes, but rather as more generalized indicators of molecular tumor progression.
A critical consideration in survival analysis is the endpoint measurement, particularly in the context of Kaplan–Meier analysis. Focusing solely on overall survival metrics (69% of the reviewed papers) may obscure the significance of specific molecular signatures related to recurrence, particularly in smaller cohorts. To elucidate these molecular signatures effectively, authors should prioritize recurrence-free survival/disease-free survival as the primary endpoint. This approach will mitigate confounding factors associated with other causes of death within the studied population.
Finally, the standardization of treatment protocols within the studied cohort is a critical factor for accurately assessing the true value of a molecular signature in predicting survival outcomes. The presence of heterogeneous treatment protocols introduces additional non-molecular variables that may complicate the interpretation of results.

Regarding Other Reviews and Meta-Analyses on the Current Topic

Emerging research and accumulation of novel data on expression changes of different microRNAs in LSCC and their potential use as diagnostic and prognostic biomarkers are setting the stage for the development of new, more advanced methods for earlier detection and more efficient treatment, which warrants further efforts to organize, classify, and study that new data. There are already some research teams who have published systematic reviews and meta-analyses on prognostic microRNA biomarkers in LSCC [311,312], as well as authors who report the interrelation between microRNAs and some of the key pathways of signalization in laryngeal carcinoma [313]. In our study, we have searched all of the currently available literature and have listed all of the microRNAs for which a significant relationship between their expression level and the survival status of patients suffering from laryngeal carcinoma has been confirmed. We have attempted to organize and graphically illustrate their association with major signaling pathways and oncogenes, altered in LSCC. Last but not least, we have outlined some of the main shortcomings in the methodologies and study design characteristics of the cited articles, which must be taken into consideration when reading, analyzing, and interpreting their results.

6. Conclusions

MiRNA molecules constitute a field in molecular medicine that presents vast opportunities for finding quality biomarkers in cancer research. A wide range of miRNAs have been shown to correlate with various clinicopathological parameters, including tumor stage, lymph node metastasis, and patient survival. These miRNAs can act as oncogenes or tumor suppressors, and their dysregulation significantly impacts the progression of LSCC. Despite the significant number of miRNAs that are found to correlate with survival outcome in laryngeal carcinoma, validation in clinical practice and reproducibility of results are still not evident in the current published literature. Strict homogeneity of cohorts and meticulous standardizations of surgical/treatment protocols are required to sift out real prognostic biomarkers from more general indicators of molecular tumor progression. Reproducibility of results is a major necessity in order to progress from single-study results towards clinically significant molecular subtypization.

Funding

This study is financed by the European Union-NextGenerationEU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project BG-RRP-2.004-0004-C01, group 3.1.5, “Strategic research and innovation program for development of Medical University—Sofia”.

Acknowledgments

All authors provided substantial input into the conceptualization, drafting, and editing of this report. Each has given approval for the publication of the article.

Conflicts of Interest

The authors declare no competing interests.

Abbreviations

mRNAMessenger RNA
miRNA MicroRNA
miR MicroRNA (often used as a prefix for specific miRNAs)
LSCC Laryngeal Squamous Cell Carcinoma
DFS Disease-Free Survival
OS Overall Survival
FFPE Formalin-Fixed Paraffin-Embedded (FFPE)
qPCR Quantitative Polymerase Chain Reaction
HNSCC Head and Neck Squamous Cell Carcinoma
sEV Small Extracellular Vesicles
FADD Fas-Associated Death Domain
EMT Epithelial–Mesenchymal Transition
HR Hazard Ratio
TNM Tumor, Node, Metastasis (staging system)
p53 Tumor Protein p53 (a tumor suppressor)
PTEN Phosphatase and Tensin Homolog
PI3K Phosphoinositide 3-kinase
CCND1 Cyclin D1
CDKN2A Cyclin-Dependent Kinase Inhibitor 2A
/p16 A specific protein encoded by CDKN2A, often referred to as p16INK4a
EGFR Epidermal Growth Factor Receptor
NOTCH A family of genes that encodes transmembrane receptors
P63 Tumor Protein p63
FAT1 FAT Tumor Suppressor Protein
TRAF3 TNF Receptor-Associated Factor 3
E2F1 E2F Transcription Factor 1
circSHKBP1 Circular RNA SHKBP1
ETS-1 E26 Transformation-Specific Sequence 1
CDK8 Cyclin-Dependent Kinase 8
CCNG2 Cyclin G2
AKT Protein Kinase B

References

  1. Wu, H.H.; Leng, S.; Sergi, C.; Leng, R. How MicroRNAs Command the Battle against Cancer. Int. J. Mol. Sci. 2024, 25, 5865. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  2. Huang, C.; Azizi, P.; Vazirzadeh, M.; Aghaei-Zarch, S.M.; Aghaei-Zarch, F.; Ghanavi, J.; Farnia, P. Non-coding RNAs/DNMT3B axis in human cancers: From pathogenesis to clinical significance. J. Transl. Med. 2023, 21, 621. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Hibner, G.; Kimsa-Furdzik, M.; Francuz, T. Relevance of MicroRNAs as Potential Diagnostic and Prognostic Markers in Colorectal Cancer. Int. J. Mol. Sci. 2018, 19, 2944. [Google Scholar] [CrossRef] [PubMed]
  4. Lorenzo, F. Sempere Celebrating 25 Years of MicroRNA Research: From Discovery to Clinical Application. Int. J. Mol. Sci. 2019, 20, 1987. [Google Scholar] [CrossRef]
  5. Drobna, M.; Szarzynska-Zawadzka, B.; Daca-Roszak, P.; Kosmalska, M.; Jaksik, R.; Witt, M.; Dawidowska, M. Identification of Endogenous Control miRNAs for RT-qPCR in T-Cell Acute Lymphoblastic Leukemia. Int. J. Mol. Sci. 2018, 19, 2858. [Google Scholar] [CrossRef]
  6. Konstantinell, A.; Coucheron, D.H.; Sveinbjørnsson, B.; Moens, U. MicroRNAs as Potential Biomarkers in Merkel Cell Carcinoma. Int. J. Mol. Sci. 2018, 19, 1873. [Google Scholar] [CrossRef] [PubMed]
  7. Wu, S.; Jia, S.; Xu, P. MicroRNA-9 as a novel prognostic biomarker in human laryngeal squamous cell carcinoma. Int. J. Clin. Exp. Med. 2014, 7, 5523–5528. [Google Scholar] [PubMed] [PubMed Central]
  8. Wu, T.Y.; Zhang, T.H.; Qu, L.M.; Feng, J.P.; Tian, L.L.; Zhang, B.H.; Li, D.D.; Sun, Y.N.; Liu, M. MiR-19a is correlated with prognosis and apoptosis of laryngeal squamous cell carcinoma by regulating TIMP-2 expression. Int. J. Clin. Exp. Pathol. 2013, 7, 56–63. [Google Scholar] [PubMed] [PubMed Central]
  9. Pantazis, T.L.; Giotakis, A.I.; Karamagkiolas, S.; Giotakis, I.; Konstantoulakis, M.; Liakea, A.; Misiakos, E.P. Low expression of miR-20b-5p indicates favorable prognosis in laryngeal squamous cell carcinoma, especially in patients with non-infiltrated regional lymph nodes. Am. J. Otolaryngol. 2020, 41, 102563. [Google Scholar] [CrossRef] [PubMed]
  10. Hu, A.; Huang, J.J.; Xu, W.H.; Jin, X.J.; Li, J.P.; Tang, Y.J.; Huang, X.F.; Cui, H.J.; Sun, G.B. miR-21 and miR-375 microRNAs as candidate diagnostic biomarkers in squamous cell carcinoma of the larynx: Association with patient survival. Am. J. Transl. Res. 2014, 6, 604–613. [Google Scholar] [PubMed] [PubMed Central]
  11. Hu, A.; Huang, J.J.; Xu, W.H.; Jin, X.J.; Li, J.P.; Tang, Y.J.; Huang, X.F.; Cui, H.J.; Sun, G.B.; Li, R.L.; et al. MiR-21/miR-375 ratio is an independent prognostic factor in patients with laryngeal squamous cell carcinoma. Am. J. Cancer Res. 2015, 5, 1775–1785. [Google Scholar] [PubMed] [PubMed Central]
  12. Gao, S.; Xu, Q.; Zhou, Y.; Yi, Q. Serum levels of microRNA-21 and microRNA-10a can predict long-term prognosis in laryngeal cancer patients: A multicenter study. Transl. Cancer Res. 2020, 9, 3680–3690. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  13. Zhang, X.W.; Liu, N.; Chen, S.; Wang, Y.; Zhang, Z.X.; Sun, Y.Y.; Qiu, G.B.; Fu, W.N. High microRNA-23a expression in laryngeal squamous cell carcinoma is associated with poor patient prognosis. Diagn. Pathol. 2015, 10, 22. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  14. Liu, D.; Wan, L.; Gong, H.; Chen, S.; Kong, Y.; Xiao, B. Sevoflurane promotes the apoptosis of laryngeal squamous cell carcinoma in-vitro and inhibits its malignant progression via miR-26a/FOXO1 axis. Bioengineered 2021, 12, 6364–6376. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, S.; Guo, D.; Li, C. Downregulation of miRNA-26b inhibits cancer proliferation of laryngeal carcinoma through autophagy by targeting ULK2 and inactivation of the PTEN/AKT pathway. Oncol. Rep. 2017, 38, 1679–1687. [Google Scholar] [CrossRef] [PubMed]
  16. Qiang, H.; Zhan, X.; Wang, W.; Cheng, Z.; Ma, S.; Jiang, C. A Study on the Correlations of the miR-31 Expression with the Pathogenesis and Prognosis of Head and Neck Squamous Cell Carcinoma. Cancer Biother. Radiopharm. 2019, 34, 189–195. [Google Scholar] [CrossRef] [PubMed]
  17. Re, M.; Çeka, A.; Rubini, C.; Ferrante, L.; Zizzi, A.; Gioacchini, F.M.; Tulli, M.; Spazzafumo, L.; Sellari-Franceschini, S.; Procopio, A.D.; et al. MicroRNA-34c-5p is related to recurrence in laryngeal squamous cell carcinoma. Laryngoscope 2015, 125, E306–E312. [Google Scholar] [CrossRef] [PubMed]
  18. Re, M.; Magliulo, G.; Gioacchini, F.M.; Bajraktari, A.; Bertini, A.; Çeka, A.; Rubini, C.; Ferrante, L.; Procopio, A.D.; Olivieri, F. Expression Levels and Clinical Significance of miR-21-5p, miR-let-7a, and miR-34c-5p in Laryngeal Squamous Cell Carcinoma. Biomed. Res. Int. 2017, 2017, 3921258. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  19. Shen, Z.; Zhan, G.; Ye, D.; Ren, Y.; Cheng, L.; Wu, Z.; Guo, J. MicroRNA-34a affects the occurrence of laryngeal squamous cell carcinoma by targeting the antiapoptotic gene survivin. Med. Oncol. 2012, 29, 2473–2480. [Google Scholar] [CrossRef] [PubMed]
  20. Shen, Z.; Zhou, C.; Li, J.; Ye, D.; Li, Q.; Wang, J.; Cui, X.; Chen, X.; Bao, T.; Duan, S. Promoter hypermethylation of miR-34a contributes to the risk, progression, metastasis and poor survival of laryngeal squamous cell carcinoma. Gene 2016, 593, 272–276. [Google Scholar] [CrossRef] [PubMed]
  21. Piotrowski, I.; Zhu, X.; Saccon, T.D.; Ashiqueali, S.; Schneider, A.; de Carvalho Nunes, A.D.; Noureddine, S.; Sobecka, A.; Barczak, W.; Szewczyk, M.; et al. miRNAs as Biomarkers for Diagnosing and Predicting Survival of Head and Neck Squamous Cell Carcinoma Patients. Cancers 2021, 13, 3980. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Popov, T.M.; Stancheva, G.; Kyurkchiyan, S.G.; Petkova, V.; Panova, S.; Kaneva, R.P.; Popova, D.P. Global microRNA expression profile in laryngeal carcinoma unveils new prognostic biomarkers and novel insights into field cancerization. Sci. Rep. 2022, 12, 17051. [Google Scholar] [CrossRef] [PubMed]
  23. Luo, M.; Sun, G.; Sun, J.W. MiR-196b affects the progression and prognosis of human LSCC through targeting PCDH-17. Auris Nasus Larynx. 2019, 46, 583–592. [Google Scholar] [CrossRef] [PubMed]
  24. Zhao, X.; Zhang, W.; Ji, W. miR-196b is a prognostic factor of human laryngeal squamous cell carcinoma and promotes tumor progression by targeting SOCS2. Biochem. Biophys. Res. Commun. 2018, 501, 584–592. [Google Scholar] [CrossRef] [PubMed]
  25. Lu, Y.; Gao, W.; Zhang, C.; Wen, S.; Huangfu, H.; Kang, J.; Wang, B. Hsa-miR-301a-3p Acts as an Oncogene in Laryngeal Squamous Cell Carcinoma via Target Regulation of Smad4. J. Cancer 2015, 6, 1260–1275. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  26. Xu, Y.; Lin, Y.P.; Yang, D.; Zhang, G.; Zhou, H.F. Expression of serum microRNA-378 and its clinical significance in laryngeal squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 5137–5142. [Google Scholar] [PubMed]
  27. Huang, Q.; Hsueh, C.Y.; Guo, Y.; Wu, X.F.; Li, J.Y.; Zhou, L. Lack of miR-1246 in small extracellular vesicle blunts tumorigenesis of laryngeal carcinoma cells by regulating Cyclin G2. IUBMB Life 2020, 72, 1491–1503. [Google Scholar] [CrossRef] [PubMed]
  28. Maia, D.; de Carvalho, A.C.; Horst, M.A.; Carvalho, A.L.; Scapulatempo-Neto, C.; Vettore, A.L. Expression of miR-296-5p as predictive marker for radiotherapy resistance in early-stage laryngeal carcinoma. J. Transl. Med. 2015, 13, 262. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Zhao, X.; Zhang, W.; Ji, W. YB-1 promotes laryngeal squamous cell carcinoma progression by inducing miR-155 expression via c-Myb. Future Oncol. 2018, 14, 1579–1589. [Google Scholar] [CrossRef] [PubMed]
  30. Cao, Y.C.; Song, L.Q.; Xu, W.W.; Qi, J.J.; Wang, X.Y.; Su, Y. Serum miR-632 is a potential marker for the diagnosis and prognosis in laryngeal squamous cell carcinoma. Acta Otolaryngol. 2020, 140, 418–421. [Google Scholar] [CrossRef] [PubMed]
  31. Li, M.; Tian, L.; Ren, H.; Chen, X.; Wang, Y.; Ge, J.; Wu, S.; Sun, Y.; Liu, M.; Xiao, H. MicroRNA-101 is a potential prognostic indicator of laryngeal squamous cell carcinoma and modulates CDK8. J. Transl. Med. 2015, 13, 271. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  32. Chen, F.; Lao, Z.; Zhang, H.Y.; Wang, J.; Wang, S.Z. Elevation of miR-125b-5p is related to improved prognosis in laryngeal squamous cell carcinoma and inhibits the malignancy and glycometabolic disorder by targeting MAP3K9. Neoplasma 2022, 69, 550–559. [Google Scholar] [CrossRef] [PubMed]
  33. Han, L.; Tang, M.; Xu, X.; Jiang, B.; Wei, Y.; Qian, H.; Lu, X. MiR-143-3p suppresses cell proliferation, migration, and invasion by targeting Melanoma-Associated Antigen A9 in laryngeal squamous cell carcinoma. J. Cell Biochem. 2019, 120, 1245–1257. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, F.; Cao, H. MicroRNA 143 3p suppresses cell growth and invasion in laryngeal squamous cell carcinoma via targeting the k Ras/Raf/MEK/ERK signaling pathway. Int. J. Oncol. 2019, 54, 689–701. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, S.Y.; Lu, Z.M.; Lin, Y.F.; Chen, L.S.; Luo, X.N.; Song, X.H.; Chen, S.H.; Wu, Y.L. miR-144-3p, a tumor suppressive microRNA targeting ETS-1 in laryngeal squamous cell carcinoma. Oncotarget 2016, 7, 11637–11650. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  36. Gao, W.; Zhang, C.; Li, W.; Li, H.; Sang, J.; Zhao, Q.; Bo, Y.; Luo, H.; Zheng, X.; Lu, Y.; et al. Promoter Methylation-Regulated miR-145-5p Inhibits Laryngeal Squamous Cell Carcinoma Progression by Targeting FSCN1. Mol. Ther. 2019, 27, 365–379. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  37. Xu, Y.; Lin, Y.P.; Yang, D.; Zhang, G.; Zhou, H.F. Clinical Significance of miR-149 in the Survival of Patients with Laryngeal Squamous Cell Carcinoma. Biomed. Res. Int. 2016, 2016, 8561251. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Shuang, Y.; Li, C.; Zhou, X.; Huang, Y.; Zhang, L. MicroRNA-195 inhibits growth and invasion of laryngeal carcinoma cells by directly targeting DCUN1D1. Oncol. Rep. 2017, 38, 2155–2165. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  39. Shuang, Y.; Li, C.; Zhou, X.; Huang, Y.W.; Zhang, L. Expression of miR-195 in laryngeal squamous cell carcinoma and its effect on proliferation and apoptosis of Hep-2. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 3232–3238. [Google Scholar] [PubMed]
  40. Tian, L.; Li, M.; Ge, J.; Guo, Y.; Sun, Y.; Liu, M.; Xiao, H. MiR-203 is downregulated in laryngeal squamous cell carcinoma and can suppress proliferation and induce apoptosis of tumours. Tumour Biol. 2014, 35, 5953–5963. [Google Scholar] [CrossRef] [PubMed]
  41. He, F.Y.; Liu, H.J.; Guo, Q.; Sheng, J.L. Reduced miR-300 expression predicts poor prognosis in patients with laryngeal squamous cell carcinoma. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 760–764. [Google Scholar] [PubMed]
  42. Dai, F.; Xie, Z.; Yang, Q.; Zhong, Z.; Zhong, C.; Qiu, Y. MicroRNA-375 inhibits laryngeal squamous cell carcinoma progression via targeting CST1. Braz. J. Otorhinolaryngol. 2022, 88 (Suppl. S4), S108–S116. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  43. Shen, Z.; Zhan, G.; Deng, H.; Ren, Y.; Ye, D.; Xiao, B.; Guo, J. MicroRNA-519a demonstrates significant tumour suppressive activity in laryngeal squamous cells by targeting anti-carcinoma HuR gene. J. Laryngol. Otol. 2013, 127, 1194–1202. [Google Scholar] [CrossRef] [PubMed]
  44. Fang, R.; Huang, Y.; Xie, J.; Zhang, J.; Ji, X. Downregulation of miR-29c-3p is associated with a poor prognosis in patients with laryngeal squamous cell carcinoma. Diagn. Pathol. 2019, 14, 109. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  45. Cappellesso, R.; Marioni, G.; Crescenzi, M.; Giacomelli, L.; Guzzardo, V.; Mussato, A.; Staffieri, A.; Martini, A.; Blandamura, S.; Fassina, A. The prognostic role of the epithelial-mesenchymal transition markers E-cadherin and Slug in laryngeal squamous cell carcinoma. Histopathology 2015, 67, 491–500. [Google Scholar] [CrossRef] [PubMed]
  46. Langevin, S.M.; Stone, R.A.; Bunker, C.H.; Lyons-Weiler, M.A.; LaFramboise, W.A.; Kelly, L.; Seethala, R.R.; Grandis, J.R.; Sobol, R.W.; Taioli, E. MicroRNA-137 promoter methylation is associated with poorer overall survival in patients with squamous cell carcinoma of the head and neck. Cancer 2011, 117, 1454–1462. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Zhao, X.; Zhang, W.; Ji, W. miR-181a targets GATA6 to inhibit the progression of human laryngeal squamous cell carcinoma. Future Oncol. 2018, 14, 1741–1753. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, F.; Zhang, H.; Wang, J. Circular RNA CircSHKBP1 accelerates the proliferation, invasion, angiogenesis, and stem cell-like properties via modulation of microR-766-5p/high mobility group AT-hook 2 axis in laryngeal squamous cell carcinoma. Bioengineered 2022, 13, 11551–11563. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Li, P.; Lin, X.J.; Yang, Y.; Yang, A.K.; Di, J.M.; Jiang, Q.W.; Huang, J.R.; Yuan, M.L.; Xing, Z.H.; Wei, M.N.; et al. Reciprocal regulation of miR-1205 and E2F1 modulates progression of laryngeal squamous cell carcinoma. Cell Death Dis. 2019, 10, 916. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  50. Sun, X.; Wang, Z.M.; Song, Y.; Tai, X.H.; Ji, W.Y.; Gu, H. MicroRNA-126 modulates the tumor microenvironment by targeting calmodulin-regulated spectrin-associated protein 1 (Camsap1). Int. J. Oncol. 2014, 44, 1678–1684. [Google Scholar] [CrossRef] [PubMed]
  51. Crimi, S.; Falzone, L.; Gattuso, G.; Grillo, C.M.; Candido, S.; Bianchi, A.; Libra, M. Droplet Digital PCR Analysis of Liquid Biopsy Samples Unveils the Diagnostic Role of hsa-miR-133a-3p and hsa-miR-375-3p in Oral Cancer. Biology 2020, 9, 379. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  52. Tu, H.F.; Lin, L.H.; Chang, K.W.; Cheng, H.W.; Liu, C.J. Exploiting salivary miR-375 as a clinical biomarker of oral potentially malignant disorder. J. Dent. Sci. 2022, 17, 659–665. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Senousy, M.A.; Shaker, O.G.; Ayeldeen, G.; Radwan, A.F. Association of lncRNA MEG3 rs941576 polymorphism, expression profile, and its related targets with the risk of obesity-related colorectal cancer: Potential clinical insights. Sci. Rep. 2024, 14, 10271. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Yu, X.M.; Liu, Y.; Jin, T.; Liu, J.; Wang, J.; Ma, C.; Pan, X.L. The Expression of SIRT1 and DBC1 in Laryngeal and Hypopharyngeal Carcinomas. PLoS ONE 2013, 8, e66975. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  55. Gattuso, G.; Crimi, S.; Lavoro, A.; Rizzo, R.; Musumarra, G.; Gallo, S.; Facciponte, F.; Paratore, S.; Russo, A.; Bordonaro, R.; et al. Liquid Biopsy and Circulating Biomarkers for the Diagnosis of Precancerous and Cancerous Oral Lesions. Noncoding RNA 2022, 8, 60. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. Kabzinski, J.; Kucharska-Lusina, A.; Majsterek, I. RNA-Based Liquid Biopsy in Head and Neck Cancer. Cells 2023, 12, 1916. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Dharmawardana, N.; Ooi, E.H.; Woods, C.; Hussey, D. Circulating microRNAs in head and neck cancer: A scoping review of methods. Clin. Exp. Metastasis 2019, 36, 291–302. [Google Scholar] [CrossRef] [PubMed]
  58. Cancer Genome Atlas Network. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature 2015, 517, 576–582. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  59. Liu, W.; Gao, G.; Hu, X.; Wang, Y.; Schwarz, J.K.; Chen, J.J.; Grigsby, P.W.; Wang, X. Activation of miR-9 by human papillomavirus in cervical cancer. Oncotarget 2014, 5, 11620–11630. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  60. Zhu, W.J.; Huang, H.H.; Feng, Y.F.; Zhan, L.; Yang, J.; Zhu, L.; Wang, Q.Y.; Wei, B.; Wang, W.Y. Hypoxia-induced miR-9 expression promotes ovarian cancer progression via activating PI3K/AKT/mTOR/GSK3β signaling pathway. Neoplasma 2023, 70, 216–228. [Google Scholar] [CrossRef] [PubMed]
  61. Yao, P.; Ni, Y.; Liu, C. Long Non-Coding RNA 691 Regulated PTEN/PI3K/AKT Signaling Pathway in Osteosarcoma Through miRNA-9-5p. Onco Targets Ther. 2020, 13, 4597–4606. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Park, Y.R.; Lee, S.T.; Kim, S.L.; Zhu, S.M.; Lee, M.R.; Kim, S.H.; Kim, I.H.; Lee, S.O.; Seo, S.Y.; Kim, S.W. Down-regulation of miR-9 promotes epithelial mesenchymal transition via regulating anoctamin-1 (ANO1) in CRC cells. Cancer Genet. 2019, 231–232, 22–31. [Google Scholar] [CrossRef] [PubMed]
  63. Gao, S.; Wang, J.; Tian, S.; Luo, J. miR-9 depletion suppresses the proliferation of osteosarcoma cells by targeting p16. Int. J. Oncol. 2019, 54, 1921–1932. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  64. Gomez, G.G.; Volinia, S.; Croce, C.M.; Zanca, C.; Li, M.; Emnett, R.; Gutmann, D.H.; Brennan, C.W.; Furnari, F.B.; Cavenee, W.K. Suppression of microRNA-9 by mutant EGFR signaling upregulates FOXP1 to enhance glioblastoma tumorigenicity. Cancer Res. 2014, 74, 1429–1439. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Mohammadi-Yeganeh, S.; Mansouri, A.; Paryan, M. Targeting of miR9/NOTCH1 interaction reduces metastatic behavior in triple-negative breast cancer. Chem. Biol. Drug Des. 2015, 86, 1185–1191. [Google Scholar] [CrossRef] [PubMed]
  66. Zou, Z.; Chen, J.; Liu, A.; Zhou, X.; Song, Q.; Jia, C.; Chen, Z.; Lin, J.; Yang, C.; Li, M.; et al. mTORC2 promotes cell survival through c-Myc-dependent up-regulation of E2F1. J. Cell Biol. 2015, 211, 105–122. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Wang, X.; Wang, L.; Mo, Q.; Jia, A.; Dong, Y.; Wang, G. A positive feedback loop of p53/miR-19/TP53INP1 modulates pancreatic cancer cell proliferation and apoptosis. Oncol. Rep. 2016, 35, 518–523. [Google Scholar] [CrossRef] [PubMed]
  68. Zhang, G.F.; Zhong, J.M.; Lin, L.; Liu, Z.H. MiR-19 enhances pancreatic cancer progression by targeting PTEN through PI3K/AKT signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1098–1107. [Google Scholar] [CrossRef] [PubMed]
  69. Mavrakis, K.; Wolfe, A.; Oricchio, E.; Palomero, T.; De Keersmaecker, K.; McJunkin, K.; Zuber, J.; James, T.; Khan, A.A.; Leslie, C.S.; et al. Genome-wide RNA-mediated interference screen identifies miR-19 targets in Notch-induced T-cell acute lymphoblastic leukaemia. Nat. Cell Biol. 2010, 12, 372–379. [Google Scholar] [CrossRef]
  70. Mogilyansky, E.; Rigoutsos, I. The miR-17/92 cluster: A comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ. 2013, 20, 1603–1614. [Google Scholar] [CrossRef] [PubMed]
  71. Tai, L.; Huang, C.J.; Choo, K.B.; Cheong, S.K.; Kamarul, T. Oxidative Stress Down-Regulates MiR-20b-5p, MiR-106a-5p and E2F1 Expression to Suppress the G1/S Transition of the Cell Cycle in Multipotent Stromal Cells. Int. J. Med. Sci. 2020, 17, 457–470. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  72. Damodaran, M.; Chinambedu Dandapani, M.; SimonDuraiRaj; SandhyaSundaram; VenkatRamanan, S.; Ramachandran, I.; Venkatesan, V. Differentially expressed miR-20, miR-21, miR-100, miR-125a and miR-146a as a potential biomarker for prostate cancer. Mol. Biol. Rep. 2021, 48, 3349–3356. [Google Scholar] [CrossRef] [PubMed]
  73. Kim, S. Computational Model for Predicting the Relationship Between Micro-RNAs and Their Target Messenger RNAs in Breast and Colon Cancers. Cancer Inform. 2018, 17, 1176935118785145. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  74. Wu, Z.H.; Zhou, J.; Hu, G.H.; Liu, J.; Li, W.C.; Lai, X.H.; Liu, M. LncRNA CASC2 inhibits lung adenocarcinoma progression through forming feedback loop with miR-21/p53 axis. Kaohsiung J. Med. Sci. 2021, 37, 675–685. [Google Scholar] [CrossRef] [PubMed]
  75. da Silva, J.; da Costa, C.C.; de Farias Ramos, I.; Laus, A.C.; Sussuchi, L.; Reis, R.M.; Khayat, A.S.; Cavalli, L.R.; Pereira, S.R. Upregulated miRNAs on the TP53 and RB1 Binding Seedless Regions in High-Risk HPV-Associated Penile Cancer. Front. Genet. 2022, 13, 875939. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  76. Gao, L.; Ren, W.; Zhang, L.; Li, S.; Kong, X.; Zhang, H.; Dong, J.; Cai, G.; Jin, C.; Zheng, D.; et al. PTENp1, a natural sponge of miR-21, mediates PTEN expression to inhibit the proliferation of oral squamous cell carcinoma. Mol. Carcinog. 2017, 56, 1322–1334. [Google Scholar] [CrossRef] [PubMed]
  77. Bergez-Hernández, F.; Irigoyen-Arredondo, M.; Martínez-Camberos, A. A systematic review of mechanisms of PTEN gene down-regulation mediated by miRNA in prostate cancer. Heliyon 2024, 10, e34950. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  78. Li, G.; Yang, Y.; Xu, S.; He, M.; Zhang, Z. mir-21-5p inhibits the progression of human chondrosarcoma by regulating CCR7/STAT3/NF-κB pathway. Connect. Tissue Res. 2021, 62, 313–324. [Google Scholar] [CrossRef] [PubMed]
  79. Silva, G.; Aboussekhra, A. p16(INK4A) inhibits the pro-metastatic potentials of osteosarcoma cells through targeting the ERK pathway and TGF-β1. Mol. Carcinog. 2016, 55, 525–536. [Google Scholar] [CrossRef] [PubMed]
  80. Marin, I.; Ofek, E.; Bar, J.; Prisant, N.; Perelman, M.; Avivi, C.; Lavy-Shahaf, G.; Onn, A.; Katz, R.; Barshack, I. MiR-21, EGFR and PTEN in non-small cell lung cancer: An in situ hybridisation and immunohistochemistry study. J. Clin. Pathol. 2020, 73, 636–641. [Google Scholar] [CrossRef]
  81. Xiong, Y.; Zhang, Y.Y.; Wu, Y.Y.; Wang, X.D.; Wan, L.H.; Li, L.; Zhou, L.M. Correlation of over-expressions of miR-21 and Notch-1 in human colorectal cancer with clinical stages. Life Sci. 2014, 106, 19–24. [Google Scholar] [CrossRef] [PubMed]
  82. Dong, Z.; Ren, L.; Lin, L.; Li, J.; Huang, Y.; Li, J. Effect of microRNA-21 on multidrug resistance reversal in A549/DDP human lung cancer cells. Mol. Med. Rep. 2015, 11, 682–690. [Google Scholar] [CrossRef] [PubMed]
  83. Sun, Y.; Tao, C.; Huang, X.; He, H.; Shi, H.; Zhang, Q.; Wu, H. Metformin induces apoptosis of human hepatocellular carcinoma HepG2 cells by activating an AMPK/p53/miR-23a/FOXA1 pathway. Onco Targets Ther. 2016, 9, 2845–2853. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Ye, Y.; Wang, G.; Wang, G.; Zhuang, J.; He, S.; Song, Y.; Ni, J.; Xia, W.; Wang, J. The Oncogenic Role of Tribbles 1 in Hepatocellular Carcinoma Is Mediated by a Feedback Loop Involving microRNA-23a and p53. Front. Physiol. 2017, 8, 789. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  85. Chen, X.; Wu, W.; Jeong, J.H.; Rokavec, M.; Wei, R.; Feng, S.; Schroth, W.; Brauch, H.; Zhong, S.; Luo, J.-L. Cytokines-activated nuclear IKKα-FAT10 pathway induces breast cancer tamoxifen-resistance. Sci. China Life Sci. 2024, 67, 1413–1426. [Google Scholar] [CrossRef] [PubMed]
  86. Xing, Y.; Zha, W.J.; Li, X.M.; Li, H.; Gao, F.; Ye, T.; Du, W.Q.; Liu, Y.C. Circular RNA circ-Foxo3 inhibits esophageal squamous cell cancer progression via the miR-23a/PTEN axis. J. Cell Biochem. 2020, 121, 2595–2605. [Google Scholar] [CrossRef] [PubMed]
  87. Ma, Y.; Gao, J.; Guo, H. miR-23a-3p Regulates Runx2 to Inhibit the Proliferation and Metastasis of Oral Squamous Cell Carcinoma. J. Oncol. 2022, 2022, 8719542. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  88. Farhadi, E.; Zaker, F.; Safa, M.; Rezvani, M.R. miR-101 sensitizes K562 cell line to imatinib through Jak2 downregulation and inhibition of NF-κB target genes. Tumour Biol. 2016, 37, 14117–14128. [Google Scholar] [CrossRef] [PubMed]
  89. Fan, X.; Tao, S.; Li, Q.; Deng, B.; Tan, Q.Y.; Jin, H. The miR-23a/27a/24-2 cluster promotes postoperative progression of early-stage non-small cell lung cancer. Mol. Ther. Oncolytics. 2021, 24, 205–217. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  90. Ito, S.; Kamoto, Y.; Sakai, A.; Sasai, K.; Hayashi, T.; Toyooka, S.; Katayama, H. Unique circulating microRNAs in relation to EGFR mutation status in Japanese smoker male with lung adenocarcinoma. Oncotarget 2017, 8, 114685–114697. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  91. Guo, Y.; Tian, Y. MiR-26a inhibits proliferation and apoptosis of uveal melanoma cells via regulating p53/MDM2 pathway. J. BUON 2020, 25, 2476–2481. [Google Scholar] [PubMed]
  92. Jiang, F.Z.; He, Y.Y.; Wang, H.H.; Zhang, H.L.; Zhang, J.; Yan, X.F.; Wang, X.J.; Che, Q.; Ke, J.Q.; Chen, Z.; et al. Mutant p53 induces EZH2 expression and promotes epithelial-mesenchymal transition by disrupting p68-Drosha complex assembly and attenuating miR-26a processing. Oncotarget 2015, 6, 44660–44674. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  93. Batchu, R.B.; Gruzdyn, O.V.; Qazi, A.M.; Kaur, J.; Mahmud, E.M.; Weaver, D.W.; Gruber, S.A. Enhanced phosphorylation of p53 by microRNA-26a leading to growth inhibition of pancreatic cancer. Surgery 2015, 158, 981–986, discussion 986–987. [Google Scholar] [CrossRef] [PubMed]
  94. Coronel-Hernández, J.; López-Urrutia, E.; Contreras-Romero, C.; Delgado-Waldo, I.; Figueroa-González, G.; Campos-Parra, A.D.; Salgado-García, R.; Martínez-Gutierrez, A.; Rodríguez-Morales, M.; Jacobo-Herrera, N.; et al. Cell migration and proliferation are regulated by miR-26a in colorectal cancer via the PTEN-AKT axis. Cancer Cell Int. 2019, 19, 80. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  95. Zhao, W.T.; Lin, X.L.; Liu, Y.; Han, L.X.; Li, J.; Lin, T.Y.; Shi, J.W.; Wang, S.C.; Lian, M.; Chen, H.W.; et al. miR-26a promotes hepatocellular carcinoma invasion and metastasis by inhibiting PTEN and inhibits cell growth by repressing EZH2. Lab. Investig. 2019, 99, 1484–1500. [Google Scholar] [CrossRef] [PubMed]
  96. Xu, S.; Wang, T.; Yang, Z.; Li, Y.; Li, W.; Wang, T.; Wang, S.; Jia, L.; Zhang, S.; Li, S. miR-26a desensitizes non-small cell lung cancer cells to tyrosine kinase inhibitors by targeting PTPN13. Oncotarget 2016, 7, 45687–45701. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  97. Lu, J.; Song, G.; Tang, Q.; Yin, J.; Zou, C.; Zhao, Z.; Xie, X.; Xu, H.; Huang, G.; Wang, J.; et al. MiR-26a inhibits stem cell-like phenotype and tumor growth of osteosarcoma by targeting Jagged1. Oncogene 2017, 36, 231–241. [Google Scholar] [CrossRef]
  98. Yao, L.; Han, C.; Song, K.; Zhang, J.; Lim, K.; Wu, T. Omega-3 Polyunsaturated Fatty Acids Upregulate 15-PGDH Expression in Cholangiocarcinoma Cells by Inhibiting miR-26a/b Expression. Cancer Res. 2015, 75, 1388–1398. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  99. Zhang, Y.F.; Zhang, A.R.; Zhang, B.C.; Rao, Z.G.; Gao, J.F.; Lv, M.H.; Wu, Y.Y.; Wang, S.M.; Wang, R.Q.; Fang, D.C. MiR-26a regulates cell cycle and anoikis of human esophageal adenocarcinoma cells through Rb1-E2F1 signaling pathway. Mol. Biol. Rep. 2013, 40, 1711–1720. [Google Scholar] [CrossRef] [PubMed]
  100. Tang, G.; Zhao, H.; Xie, Z.; Wei, S.; Chen, G. Long non-coding RNA HAGLROS facilitates tumorigenesis and progression in hepatocellular carcinoma by sponging miR-26b-5p to up-regulate karyopherin α2 (KPNA2) and inactivate p53 signaling. Bioengineered 2022, 13, 7829–7846. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  101. Chen, E.; Li, E.; Liu, H.; Zhou, Y.; Wen, L.; Wang, J.; Wang, Y.; Ye, L.; Liang, T. miR-26b enhances the sensitivity of hepatocellular carcinoma to Doxorubicin via USP9X-dependent degradation of p53 and regulation of autophagy. Int. J. Biol. Sci. 2021, 17, 781–795. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  102. Gao, Z.; Ye, X.; Bordeaux, A.; Hettich, S.; Lin, S.; Han, F.; Jia, Y. miR-26b regulates cell proliferation and apoptosis of CD117+CD44+ ovarian cancer stem cells by targeting PTEN. Eur. J. Histochem. 2021, 65, 3186. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  103. Liang, S.; Guo, H.; Ma, K.; Li, X.; Wu, D.; Wang, Y.; Wang, W.; Zhang, S.; Cui, Y.; Liu, Y.; et al. A PLCB1-PI3K-AKT Signaling Axis Activates EMT to Promote Cholangiocarcinoma Progression. Cancer Res. 2021, 81, 5889–5903. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  104. De Robertis, M.; Loiacono, L.; Fusilli, C.; Poeta, M.L.; Mazza, T.; Sanchez, M.; Marchionni, L.; Signori, E.; Lamorte, G.; Vescovi, A.L.; et al. Dysregulation of EGFR Pathway in EphA2 Cell Subpopulation Significantly Associates with Poor Prognosis in Colorectal Cancer. Clin. Cancer Res. 2017, 23, 159–170. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  105. Zhang, K.; Zhang, B.; Bai, Y.; Dai, L. E2F1 promotes cancer cell sensitivity to cisplatin by regulating the cellular DNA damage response through miR-26b in esophageal squamous cell carcinoma. J. Cancer 2020, 11, 301–310. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  106. Creighton, C.J.; Fountain, M.D.; Yu, Z.; Nagaraja, A.K.; Zhu, H.; Khan, M.; Olokpa, E.; Zariff, A.; Gunaratne, P.H.; Matzuk, M.M.; et al. Molecular profiling uncovers a p53-associated role for microRNA-31 in inhibiting the proliferation of serous ovarian carcinomas and other cancers. Cancer Res. 2010, 70, 1906–1915. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  107. Zhu, C.; Wang, S.; Zheng, M.; Chen, Z.; Wang, G.; Ma, J.; Zhang, B.; Huang, W.; Sun, X.; Wang, C. miR-31-5p modulates cell progression in lung adenocarcinoma through TNS1/p53 axis. Strahlenther. Onkol. 2022, 198, 304–314. [Google Scholar] [CrossRef] [PubMed]
  108. Yan, G.; Lei, W. Role of ELK1 in regulating colorectal cancer progression: miR-31-5p/CDIP1 axis in CRC pathogenesis. PeerJ 2023, 11, e15602. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  109. Tian, T.; Hong, F.; Wang, Z.; Hu, J.; Chen, N.; Lv, L.; Yi, Q. HSD17B6 downregulation predicts poor prognosis and drives tumor progression via activating Akt signaling pathway in lung adenocarcinoma. Cell Death Discov. 2021, 7, 341. [Google Scholar] [CrossRef] [PubMed]
  110. Hou, C.; Sun, B.; Jiang, Y.; Zheng, J.; Yang, N.; Ji, C.; Liang, Z.; Shi, J.; Zhang, R.; Liu, Y.; et al. MicroRNA-31 inhibits lung adenocarcinoma stem-like cells via down-regulation of MET-PI3K-Akt signaling pathway. Anticancer Agents Med. Chem. 2016, 16, 501–518. [Google Scholar] [CrossRef] [PubMed]
  111. Sun, Y.; Jia, X.; Wang, M.; Deng, Y. Long noncoding RNA MIR31HG abrogates the availability of tumor suppressor microRNA-361 for the growth of osteosarcoma. Cancer Manag. Res. 2019, 11, 8055–8064. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  112. Rajbhandari, R.; McFarland, B.C.; Patel, A.; Gerigk, M.; Gray, G.K.; Fehling, S.C.; Bredel, M.; Berbari, N.F.; Kim, H.; Marks, M.P.; et al. Loss of tumor suppressive microRNA-31 enhances TRADD/NF-κB signaling in glioblastoma. Oncotarget 2015, 6, 17805–17816. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  113. Lu, W.C.; Kao, S.Y.; Yang, C.C.; Tu, H.F.; Wu, C.H.; Chang, K.W.; Lin, S.C. EGF up-regulates miR-31 through the C/EBPβ signal cascade in oral carcinoma. PLoS ONE 2014, 9, e108049. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  114. He, Z.; Zhong, Y.; Regmi, P.; Lv, T.; Ma, W.; Wang, J.; Liu, F.; Yang, S.; Zhong, Y.; Zhou, R.; et al. Exosomal long non-coding RNA TRPM2-AS promotes angiogenesis in gallbladder cancer through interacting with PABPC1 to activate NOTCH1 signaling pathway. Mol. Cancer 2024, 23, 65. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  115. Lin, P.C.; Chiu, Y.L.; Banerjee, S.; Park, K.; Mosquera, J.M.; Giannopoulou, E.; Alves, P.; Tewari, A.K.; Gerstein, M.B.; Beltran, H.; et al. Epigenetic repression of miR-31 disrupts androgen receptor homeostasis and contributes to prostate cancer progression. Cancer Res. 2013, 73, 1232–1244. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  116. Pan, W.; Chai, B.; Li, L.; Lu, Z.; Ma, Z. p53/MicroRNA-34 axis in cancer and beyond. Heliyon 2023, 9, e15155. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  117. Wu, X.; Cheng, Y.L.; Matthen, M.; Yoon, A.; Schwartz, G.K.; Bala, S.; Taylor, A.M.; Momen-Heravi, F. Down-regulation of the tumor suppressor miR-34a contributes to head and neck cancer by up-regulating the MET oncogene and modulating tumor immune evasion. J. Exp. Clin. Cancer Res. 2021, 40, 70. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  118. Li, Y.; Jin, L.; Rong, W.; Meng, F.; Wang, X.; Wang, S.; Yu, X. Expression and significance of miR-34 with PI3K.; AKT and mTOR proteins in colorectal adenocarcinoma tissues. Cell. Mol. Biol. 2022, 68, 57–62. [Google Scholar] [CrossRef] [PubMed]
  119. Wang, Y.; Lin, C.; Liu, Y. Molecular mechanism of miR-34b-5p and RNA binding protein HuR binding to lncRNA OIP5-AS1 in colon cancer cells. Cancer Gene Ther. 2022, 29, 612–624. [Google Scholar] [CrossRef] [PubMed]
  120. Achari, C.; Winslow, S.; Ceder, Y.; Larsson, C. Expression of miR-34c induces G2/M cell cycle arrest in breast cancer cells. BMC Cancer 2014, 14, 538. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  121. Hanafi, A.R.; Jayusman, A.M.; Alfasunu, S.; Sadewa, A.H.; Pramono, D.; Heriyanto, D.S.; Haryana, S.M. Serum MiRNA as Predictive and Prognosis Biomarker in Advanced Stage Non-small Cell Lung Cancer in Indonesia. Zhongguo Fei Ai Za Zhi 2020, 23, 321–332. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  122. Yu, X.; Zhang, W.; Ning, Q.; Luo, X. MicroRNA-34a inhibits human brain glioma cell growth by down-regulation of Notch1. J. Huazhong Univ. Sci.Technol. Med. Sci. 2012, 32, 370–374. [Google Scholar] [CrossRef] [PubMed]
  123. Lin, H.; Li, Z.; Chen, C.; Luo, X.; Xiao, J.; Dong, D.; Lu, Y.; Yang, B.; Wang, Z. Transcriptional and post-transcriptional mechanisms for oncogenic overexpression of ether à go-go K+ channel. PLoS ONE 2011, 6, e20362, Erratum in PLoS ONE 2011, 6. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  124. Chen, X.; Chen, S.; Xiu, Y.L.; Sun, K.X.; Zong, Z.H.; Zhao, Y. RhoC is a major target of microRNA-93-5P in epithelial ovarian carcinoma tumorigenesis and progression. Mol. Cancer 2015, 14, 31. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  125. de Conti, A.; Festa Ortega, J.; Tryndyak, V.; Dreval, K.; Salvador Moreno, F.; Rusyn, I.; Beland, F.A.; Pogribny, I.P. MicroRNA deregulation in nonalcoholic steatohepatitis-associated liver carcinogenesis. Oncotarget 2017, 8, 88517–88528. [Google Scholar] [CrossRef]
  126. Liu, M.X.; Liao, J.; Xie, M.; Gao, Z.K.; Wang, X.H.; Zhang, Y.; Shang, M.H.; Yin, L.H.; Pu, Y.P.; Liu, R. miR-93-5p Transferred by Exosomes Promotes the Proliferation of Esophageal Cancer Cells via Intercellular Communication by Targeting PTEN. Biomed. Environ. Sci. 2018, 31, 171–185. [Google Scholar] [CrossRef] [PubMed]
  127. Bao, C.; Chen, J.; Chen, D.; Lu, Y.; Lou, W.; Ding, B.; Xu, L.; Fan, W. MiR-93 suppresses tumorigenesis and enhances chemosensitivity of breast cancer via dual targeting E2F1 and CCND1. Cell Death Dis. 2020, 11, 618. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  128. Li, Y.; Wu, B.; Sun, R.; Zhao, M.; Li, N. miR-93-5p knockdown repressed hepatocellular carcinoma progression via increasing ERBB4 and TETs-dependent DNA demethylation. Autoimmunity 2021, 54, 547–560. [Google Scholar] [CrossRef] [PubMed]
  129. Noorolyai, S.; Baghbani, E.; Aghebati Maleki, L.; Baghbanzadeh Kojabad, A.; Shanehbansdi, D.; Khaze Shahgoli, V.; Mokhtarzadeh, A.; Baradaran, B. Restoration of miR-193a-5p and miR-146 a-5p Expression Induces G1 Arrest in Colorectal Cancer through Targeting of MDM2/p53. Adv. Pharm. Bull. 2020, 10, 130–134. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  130. Sathyanarayanan, A.; Chandrasekaran, K.S.; Karunagaran, D. microRNA-146a inhibits proliferation, migration and invasion of human cervical and colorectal cancer cells. Biochem. Biophys. Res. Commun. 2016, 480, 528–533. [Google Scholar] [CrossRef] [PubMed]
  131. Ma, W.; Zhao, X.; Liang, L.; Wang, G.; Li, Y.; Miao, X.; Zhao, Y. miR-146a and miR-146b promote proliferation, migration and invasion of follicular thyroid carcinoma via inhibition of ST8SIA4. Oncotarget 2017, 8, 28028–28041. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  132. Qiu, L.; Wang, J.; Chen, M.; Chen, F.; Tu, W. Exosomal microRNA-146a derived from mesenchymal stem cells increases the sensitivity of ovarian cancer cells to docetaxel and taxane via a LAMC2-mediated PI3K/Akt axis. Int. J. Mol. Med. 2020, 46, 609–620. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  133. Zhang, Y.; Ding, S.; Yang, J.; Chen, X.; Huang, W. Identification of miR-146a is Associated with the Aggressiveness and Suppresses Proliferation via Targeting CDKN2A in Breast Cancer. Pathol. Oncol. Res. 2020, 26, 245–251. [Google Scholar] [CrossRef] [PubMed]
  134. Afshar-Khamseh, R.; Javeri, A.; Taha, M.F. MiR-146a suppresses the expression of CXCR4 and alters survival, proliferation and migration rate in colorectal cancer cells. Tissue Cell 2021, 73, 101654. [Google Scholar] [CrossRef] [PubMed]
  135. Forloni, M.; Dogra, S.K.; Dong, Y.; Conte, D., Jr.; Ou, J.; Zhu, L.J.; Deng, A.; Mahalingam, M.; Green, M.R.; Wajapeyee, N. miR-146a promotes the initiation and progression of melanoma by activating Notch signaling. Elife 2014, 3, e01460. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  136. Sandhu, R.; Rein, J.; D’Arcy, M.; Herschkowitz, J.I.; Hoadley, K.A.; Troester, M.A. Overexpression of miR-146a in basal-like breast cancer cells confers enhanced tumorigenic potential in association with altered p53 status. Carcinogenesis 2014, 35, 2567–2575. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  137. Yan, Z.; Wen, J.X.; Cao, X.S.; Zhao, W.; Han, Y.L.; Wen, X.H.; Yan, L.; Zhang, M.; Wang, Y.F.; Hai, L.; et al. Tumor cell-derived exosomal microRNA-146a promotes non-small cell lung cancer cell invasion and proliferation by inhibiting M1 macrophage polarization. Ann. Transl. Med. 2022, 10, 1307. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  138. Neilsen, P.M.; Noll, J.E.; Mattiske, S.; Bracken, C.P.; Gregory, P.A.; Schulz, R.B.; Lim, S.P.; Kumar, R.; Suetani, R.J.; Goodall, G.J.; et al. Mutant p53 drives invasion in breast tumors through up-regulation of miR-155. Oncogene 2013, 32, 2992–3000. [Google Scholar] [CrossRef] [PubMed]
  139. Li, N.; Cui, T.; Guo, W.; Wang, D.; Mao, L. MiR-155-5p accelerates the metastasis of cervical cancer cell via targeting TP53INP1. Onco Targets Ther. 2019, 12, 3181–3196. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  140. Van Roosbroeck, K.; Fanini, F.; Setoyama, T.; Ivan, C.; Rodriguez-Aguayo, C.; Fuentes-Mattei, E.; Xiao, L.; Vannini, I.; Redis, R.S.; D’Abundo, L.; et al. Combining Anti-Mir-155 with Chemotherapy for the Treatment of Lung Cancers. Clin. Cancer Res. 2017, 23, 2891–2904. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  141. Zuo, W.N.; Zhu, H.; Li, L.P.; Jin, A.Y.; Wang, H.Q. MiR-155 promotes proliferation and inhibits apoptosis of nasopharyngeal carcinoma cells through targeting PTEN-PI3K/AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7935–7942. [Google Scholar] [CrossRef] [PubMed]
  142. Sun, J.F.; Zhang, D.; Gao, C.J.; Zhang, Y.W.; Dai, Q.S. Exosome-Mediated MiR-155 Transfer Contributes to Hepatocellular Carcinoma Cell Proliferation by Targeting PTEN. Med. Sci. Monit. Basic. Res. 2019, 25, 218–228. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  143. Gao, Y.; Xu, J.; Li, H.; Hu, Y.; Yu, G. Identification of Metastasis-Associated MicroRNAs in Metastatic Melanoma by miRNA Expression Profile and Experimental Validation. Front. Genet. 2021, 12, 663110. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  144. Kravets, O.; Burtyn, O.; Borikun, T.; Rossylna, O. The Study of Prognostic Value of microRNAs (miR-10b AND -155) and CDKN2A/P16INK4A in Oral Squamous Cell Carcinoma. Exp. Oncol. 2023, 45, 187–194. [Google Scholar] [CrossRef] [PubMed]
  145. Zhao, B.S.; Roundtree, I.A.; He, C. Post-transcriptional gene regulation by mRNA modifications. Nat. Rev. Mol. Cell Biol. 2017, 18, 31–42, Erratum in Nat. Rev. Mol. Cell Biol. 2018, 19, 808. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  146. Chen, L.; Yang, X.; Zhao, J.; Xiong, M.; Almaraihah, R.; Chen, Z.; Hou, T. Circ_0008532 promotes bladder cancer progression by regulation of the miR-155-5p/miR-330-5p/MTGR1 axis. J. Exp. Clin. Cancer Res. 2020, 39, 94. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  147. Liu, K.; Yao, H.; Lei, S.; Xiong, L.; Qi, H.; Qian, K.; Liu, J.; Wang, P.; Zhao, H. The miR-124-p63 feedback loop modulates colorectal cancer growth. Oncotarget 2017, 8, 29101–29115. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  148. Jiang, D.; Aguiar, R.C. MicroRNA-155 controls RB phosphorylation in normal and malignant B lymphocytes via the noncanonical TGF-β1/SMAD5 signaling module. Blood 2014, 123, 86–93. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  149. Li, N.A.; Wang, W.; Xu, B.; Gong, H. miR-196b regulates gastric cancer cell proliferation and invasion via PI3K/AKT/mTOR signaling pathway. Oncol. Lett. 2016, 11, 1745–1749. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  150. Bai, X.; Meng, L.; Sun, H.; Li, Z.; Zhang, X.; Hua, S. MicroRNA-196b Inhibits Cell Growth and Metastasis of Lung Cancer Cells by Targeting Runx2. Cell Physiol. Biochem. 2017, 43, 757–767. [Google Scholar] [CrossRef] [PubMed]
  151. Du, Y.; Wei, N.; Ma, R.; Jiang, S.; Song, D. A miR-210-3p regulon that controls the Warburg effect by modulating HIF-1α and p53 activity in triple-negative breast cancer. Cell Death Dis. 2020, 11, 731. [Google Scholar] [CrossRef]
  152. Islam, S.U.; Ahmed, M.B.; Sonn, J.K.; Jin, E.J.; Lee, Y.S. PRP4 Induces Epithelial-Mesenchymal Transition and Drug Resistance in Colon Cancer Cells via Activation of p53. Int. J. Mol. Sci. 2022, 23, 3092. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  153. Yang, F.; Yan, Y.; Yang, Y.; Hong, X.; Wang, M.; Yang, Z.; Liu, B.; Ye, L. MiR-210 in exosomes derived from CAFs promotes non-small cell lung cancer migration and invasion through PTEN/PI3K/AKT pathway. Cell. Signal. 2020, 73, 109675. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, L.; Song, Y.; Wang, H.; Liu, K.; Shao, Z.; Shang, Z. MiR-210-3p-EphrinA3-PI3K/AKT axis regulates the progression of oral cancer. J. Cell. Mol. Med. 2020, 24, 4011–4022. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  155. Evangelista, A.F.; Oliveira, R.J.; Silva, V.A.O.; Vieira, R.A.D.C.; Reis, R.M.; Marques, M.M.C. Integrated analysis of mRNA and miRNA profiles revealed the role of miR-193 and miR-210 as potential regulatory biomarkers in different molecular subtypes of breast cancer. BMC Cancer 2021, 21, 76. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  156. Tabibkhooei, A.; Izadpanahi, M.; Arab, A.; Zare-Mirzaei, A.; Minaeian, S.; Rostami, A.; Mohsenian, A. Profiling of novel circulating microRNAs as a non-invasive biomarker in diagnosis and follow-up of high and low-grade gliomas. Clin. Neurol. Neurosurg. 2020, 190, 105652. [Google Scholar] [CrossRef] [PubMed]
  157. Yoon, A.R.; Gao, R.; Kaul, Z.; Choi, I.K.; Ryu, J.; Noble, J.R.; Kato, Y.; Saito, S.; Hirano, T.; Ishii, T.; et al. MicroRNA-296 is enriched in cancer cells and downregulates p21WAF1 mRNA expression via interaction with its 3′ untranslated region. Nucleic Acids Res. 2011, 39, 8078–8091. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  158. Cheng, X.; Huang, Z.; Pan, A.; Long, D. ORLNC1 Suppresses Cell Growth in HER2-Positive Breast Cancer via miRNA-296 Sponging. Curr. Mol. Med. 2023, 23, 289–299. [Google Scholar] [CrossRef] [PubMed]
  159. Zhou, J.; Du, G.; Fu, H. miR-296-3p promotes the proliferation of glioblastoma cells by targeting ICA. T. Mol Med Rep. 2020, 21, 2151–2161. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  160. Zhang, F.; Sang, Y.; Chen, D.; Wu, X.; Wang, X.; Yang, W.; Chen, Y. M2 macrophage-derived exosomal long non-coding RNA AGAP2-AS1 enhances radiotherapy immunity in lung cancer by reducing microRNA-296 and elevating NOTCH2. Cell Death Dis. 2021, 12, 467. [Google Scholar] [CrossRef] [PubMed]
  161. Peng, L.N.; Shi, W.T.; Feng, H.R.; Wei, C.Y.; Yin, Q.N. Effect of miR-301a/PTEN pathway on the proliferation and apoptosis of cervical cancer. Innate Immun. 2019, 25, 217–223. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  162. Granda-Díaz, R.; Manterola, L.; Hermida-Prado, F.; Rodríguez, R.; Santos, L.; García-de-la-Fuente, V.; Fernández, M.T.; Corte-Torres, M.D.; Rodrigo, J.P.; Álvarez-Teijeiro, S.; et al. Targeting oncogenic functions of miR-301a in head and neck squamous cell carcinoma by PI3K/PTEN and MEK/ERK pathways. Biomed. Pharmacother. 2023, 161, 114512. [Google Scholar] [CrossRef] [PubMed]
  163. Su, H.; Wu, Y.; Fang, Y.; Shen, L.; Fei, Z.; Xie, C.; Chen, M. MicroRNA-301a targets WNT1 to suppress cell proliferation and migration and enhance radiosensitivity in esophageal cancer cells. Oncol. Rep. 2019, 41, 599–607. [Google Scholar] [CrossRef] [PubMed]
  164. Nam, R.K.; Benatar, T.; Wallis, C.J.; Amemiya, Y.; Yang, W.; Garbens, A.; Naeim, M.; Sherman, C.; Sugar, L.; Seth, A. MiR-301a regulates E-cadherin expression and is predictive of prostate cancer recurrence. Prostate 2016, 76, 869–884. [Google Scholar] [CrossRef] [PubMed]
  165. Yin, J.; Chen, D.; Luo, K.; Lu, M.; Gu, Y.; Zeng, S.; Chen, X.; Song, Y.; Zhang, Z.; Zheng, G.; et al. Cip2a/miR-301a feedback loop promotes cell proliferation and invasion of triple-negative breast cancer. J. Cancer 2019, 10, 5964–5974. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  166. Wang, J.; Li, Y.; Ma, Q.; Huang, J. miR-378 in combination with ultrasonic irradiation and SonoVue microbubbles transfection inhibits hepatoma cell growth. Mol. Med. Rep. 2020, 21, 2493–2501. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  167. Skrzypek, K.; Tertil, M.; Golda, S.; Ciesla, M.; Weglarczyk, K.; Collet, G.; Guichard, A.; Kozakowska, M.; Boczkowski, J.; Was, H.; et al. Interplay between heme oxygenase-1 and miR-378 affects non-small cell lung carcinoma growth, vascularization, and metastasis. Antioxid. Redox Signal. 2013, 19, 644–660. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  168. Cui, Z.; Bao, X.; Liu, Q.; Li, Q.; Huang, L.; Wang, H.; Jiao, K. MicroRNA-378-3p/5p represses proliferation and induces apoptosis of oral squamous carcinoma cells via targeting KLK4. Clin. Exp. Pharmacol. Physiol. 2020, 47, 713–724. [Google Scholar] [CrossRef] [PubMed]
  169. Weng, W.H.; Leung, W.H.; Pang, Y.J.; Hsu, H.H. Lauric acid can improve the sensitization of Cetuximab in KRAS/BRAF mutated colorectal cancer cells by retrievable microRNA-378 expression. Oncol. Rep. 2016, 35, 107–116. [Google Scholar] [CrossRef] [PubMed]
  170. Zhou, Z.X.; Zhang, Z.P.; Tao, Z.Z.; Tan, T.Z. miR-632 Promotes Laryngeal Carcinoma Cell Proliferation, Migration, and Invasion Through Negative Regulation of GSK3β. Oncol. Res. 2020, 28, 21–31. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  171. Liao, J.M.; Zhou, X.; Zhang, Y.; Lu, H. MiR-1246: A new link of the p53 family with cancer and Down syndrome. Cell Cycle 2012, 11, 2624–2630. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  172. Zhang, Q.; Cao, L.Y.; Cheng, S.J.; Zhang, A.M.; Jin, X.S.; Li, Y. p53-induced microRNA-1246 inhibits the cell growth of human hepatocellular carcinoma cells by targeting NFI.B. Oncol. Rep. 2015, 33, 1335–1341. [Google Scholar] [CrossRef] [PubMed]
  173. Li, J.; Zhang, Z.; Hu, J.; Wan, X.; Huang, W.; Zhang, H.; Jiang, N. MiR-1246 regulates the PI3K/AKT signaling pathway by targeting PIK3AP1 and inhibits thyroid cancer cell proliferation and tumor growth. Mol. Cell. Biochem. 2022, 477, 649–661. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  174. Chen, Y.; Chen, L.; Zhu, S.; Yang, H.; Ye, Z.; Wang, H.; Wu, H.; Wu, Y.; Sun, Q.; Liu, X.; et al. Exosomal derived miR-1246 from hydroquinone-transformed cells drives S phase accumulation arrest by targeting cyclin G2 in TK6 cells. Chem. Biol. Interact. 2024, 387, 110809. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, Q.Y.; Jiao, D.M.; Zhu, Y.; Hu, H.; Wang, J.; Tang, X.; Chen, J.; Yan, L. Identification of carcinogenic potential-associated molecular mechanisms in CD133(+) A549 cells based on microRNA profiles. Tumour Biol. 2016, 37, 521–530. [Google Scholar] [CrossRef] [PubMed]
  176. Vu, T.T.; Stölzel, F.; Wang, K.W.; Röllig, C.; Tursky, M.L.; Molloy, T.J.; Ma, D.D. miR-10a as a therapeutic target and predictive biomarker for MDM2 inhibition in acute myeloid leukemia. Leukemia 2021, 35, 1933–1948. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  177. Meng, X.; Peng, J.; Xie, X.; Yu, F.; Wang, W.; Pan, Q.; Jin, H.; Huang, X.; Yu, H.; Li, S.; et al. Roles of lncRNA LVBU in regulating urea cycle/polyamine synthesis axis to promote colorectal carcinoma progression. Oncogene 2022, 41, 4231–4243. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  178. Yu, T.; Liu, L.; Li, J.; Yan, M.; Lin, H.; Liu, Y.; Chu, D.; Tu, H.; Gu, A.; Yao, M. MiRNA-10a is upregulated in NSCLC and may promote cancer by targeting PTEN. Oncotarget 2015, 6, 30239–30250. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  179. Zeng, T.; Li, G. MicroRNA-10a enhances the metastatic potential of cervical cancer cells by targeting phosphatase and tensin homologue. Mol. Med. Rep. 2014, 10, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
  180. Ashmawy, A.M.; Elgeshy, K.M.; Abdel Salam, E.T.; Ghareeb, M.; Kobaisi, M.H.; Amin, H.A.A.; Sharawy, S.K.; Abdel Wahab, A.H.A. Crosstalk between liver-related microRNAs and Wnt/β-catenin pathway in hepatocellular carcinoma patients. Arab. J. Gastroenterol. 2017, 18, 144–150. [Google Scholar] [CrossRef] [PubMed]
  181. Wang, H.; Fu, L.; Wei, D.; Wang, B.; Zhang, C.; Zhu, T.; Ma, Z.; Li, Z.; Wu, Y.; Yu, G. MiR-29c-3p Suppresses the Migration, Invasion and Cell Cycle in Esophageal Carcinoma via CCNA2/p53 Axis. Front. Bioeng. Biotechnol. 2020, 8, 75. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  182. Chen, G.; Zhou, T.; Li, Y.; Yu, Z.; Sun, L. p53 target miR-29c-3p suppresses colon cancer cell invasion and migration through inhibition of PHLDB2. Biochem. Biophys. Res. Commun. 2017, 487, 90–95. [Google Scholar] [CrossRef] [PubMed]
  183. Zhan, S.; Wang, C.; Yin, F. MicroRNA-29c inhibits proliferation and promotes apoptosis in non-small cell lung cancer cells by targeting VEGFA. Mol. Med. Rep. 2018, 17, 6705–6710. [Google Scholar] [CrossRef] [PubMed]
  184. Wardana, T.; Oktriani, R.; Murjayanto, C.H.; Putri, D.U.; Anwar, S.L.; Aryandono, T.; Haryana, S.M. MicroRNA Gene Signature for Predicting Mechanisms in Nasopharyngeal Carcinoma: A Case Study on the Potential Application of Circulating Biomarkers. Microrna 2023, 12, 29–44. [Google Scholar] [CrossRef] [PubMed]
  185. Fan, Y.C.; Mei, P.J.; Chen, C.; Miao, F.A.; Zhang, H.; Li, Z.L. MiR-29c inhibits glioma cell proliferation, migration, invasion and angiogenesis. J. Neurooncol. 2013, 115, 179–188. [Google Scholar] [CrossRef] [PubMed]
  186. He, S.; Yang, S.; Niu, M.; Zhong, Y.; Dan, G.; Zhang, Y.; Ma, H.; Xiong, W.; Zhou, M.; Zhou, Y.; et al. HMG-box transcription factor 1: A positive regulator of the G1/S transition through the Cyclin-CDK-CDKI molecular network in nasopharyngeal carcinoma. Cell Death Dis. 2018, 9, 100. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  187. Wang, L.; Yu, T.; Li, W.; Li, M.; Zuo, Q.; Zou, Q.; Xiao, B. The miR-29c-KIAA1199 axis regulates gastric cancer migration by binding with WBP11 and PTP4A3. Oncogene 2019, 38, 3134–3150. [Google Scholar] [CrossRef] [PubMed]
  188. Ma, Z.; Zhu, T.; Wang, H.; Wang, B.; Fu, L.; Yu, G. E2F1 Reduces Sorafenib’s Sensitivity of Esophageal Carcinoma Cells via Modulating the miR-29c-3p/COL11A1 Signaling Axis. Curr. Mol. Pharmacol. 2024, 17, 36876834. [Google Scholar] [CrossRef] [PubMed]
  189. Wang, X.W.; Jiang, Y.H.; Ye, W.; Shao, C.F.; Xie, J.J.; Li, X. SIRT1 promotes the progression and chemoresistance of colorectal cancer through the p53/miR-101/KPNA3 axis. Cancer Biol. Ther. 2023, 24, 2235770. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  190. Park, J.; Cho, M.; Cho, J.; Kim, E.E.; Song, E.J. MicroRNA-101-3p Suppresses Cancer Cell Growth by Inhibiting the USP47-Induced Deubiquitination of RPL11. Cancers 2022, 14, 964. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  191. Lv, X.; Li, J.; Yang, B. Clinical effects of miR-101 on prognosis of hepatocellular carcinoma and carcinogenic mechanism of anti-miR-101. Oncol. Rep. 2016, 36, 2184–2192. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, L.; Yao, J.; Sun, H.; He, K.; Tong, D.; Song, T.; Huang, C. MicroRNA-101 suppresses progression of lung cancer through the PTEN/AKT signaling pathway by targeting DNA methyltransferase 3A. Oncol. Lett. 2017, 13, 329–338. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  193. Zhou, X.; Xia, Y.; Li, L.; Zhang, G. MiR-101 inhibits cell growth and tumorigenesis of Helicobacter pylori related gastric cancer by repression of SOCS2. Cancer Biol. Ther. 2015, 16, 160–169. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  194. Zhang, X.; Gao, D.; Fang, K.; Guo, Z.; Li, L. Med19 is targeted by miR-101-3p/miR-422a and promotes breast cancer progression by regulating the EGFR/MEK/ERK signaling pathway. Cancer Lett. 2019, 444, 105–115. [Google Scholar] [CrossRef] [PubMed]
  195. Wu, H.B.; Huang, S.S.; Lu, C.G.; Tian, S.D.; Chen, M. CircAPLP2 regulates the proliferation and metastasis of colorectal cancer by targeting miR-101-3p to activate the Notch signalling pathway. Am. J. Transl. Res. 2020, 12, 2554–2569. [Google Scholar] [PubMed] [PubMed Central]
  196. Ratovitski, E.A. Phospho-ΔNp63α-responsive microRNAs contribute to the regulation of necroptosis in squamous cell carcinoma upon cisplatin exposure. FEBS Lett. 2015, 589, 1352–1358. [Google Scholar] [CrossRef] [PubMed]
  197. Tao, L.; Xu, C.; Shen, W.; Tan, J.; Li, L.; Fan, M.; Sun, D.; Lai, Y.; Cheng, H. HIPK3 Inhibition by Exosomal hsa-miR-101-3p Is Related to Metabolic Reprogramming in Colorectal Cancer. Front. Oncol. 2022, 11, 758336. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  198. Chen, J.J.; Liu, S.X.; Chen, M.Z.; Zhao, Z.Y. Has-miR-125a and 125b are induced by treatment with cisplatin in nasopharyngeal carcinoma and inhibit apoptosis in a p53-dependent manner by targeting p53 mRNA. Mol. Med. Rep. 2015, 12, 3569–3574. [Google Scholar] [CrossRef] [PubMed]
  199. Shi, X.B.; Xue, L.; Ma, A.H.; Tepper, C.G.; Kung, H.J.; White, R.W. miR-125b promotes growth of prostate cancer xenograft tumor through targeting pro-apoptotic genes. Prostate 2011, 71, 538–549. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  200. Zhang, H.M.; Fan, T.T.; Li, W.; Li, X.X. Expressions and significances of TTF-1 and PTEN in early endometrial cancer. Eur. Rev. Med. Pharmacol. Sci. 2017, 21 (Suppl. S3), 20–26. [Google Scholar] [PubMed]
  201. Bu, Q.; You, F.; Pan, G.; Yuan, Q.; Cui, T.; Hao, L.; Zhang, J. MiR-125b inhibits anaplastic thyroid cancer cell migration and invasion by targeting PIK3CD. Biomed. Pharmacother. 2017, 88, 443–448. [Google Scholar] [CrossRef] [PubMed]
  202. Mei, L.L.; Wang, W.J.; Qiu, Y.T.; Xie, X.F.; Bai, J.; Shi, Z.Z. miR-125b-5p functions as a tumor suppressor gene partially by regulating HMGA2 in esophageal squamous cell carcinoma. PLoS ONE 2017, 12, e0185636. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  203. Wu, Q.; Liu, P.; Lao, G.; Liu, Y.; Zhang, W.; Ma, C. Comprehensive Analysis of circRNA-miRNA-mRNA Network in Cervical Squamous Cell Carcinoma by Integrated Analysis. Onco Targets Ther. 2020, 13, 8641–8650. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  204. Zhang, L.; Ge, Y.; Fuchs, E. miR-125b can enhance skin tumor initiation and promote malignant progression by repressing differentiation and prolonging cell survival. Genes. Dev. 2014, 28, 2532–2546. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  205. Gao, D.; Xiao, Z.; Li, H.P.; Han, D.H.; Zhang, Y.P. The mechanism study of miR-125b in occurrence and progression of multiple myeloma. Cancer Med. 2018, 7, 134–145. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  206. Boldrup, L.; Coates, P.J.; Wahlgren, M.; Laurell, G.; Nylander, K. Subsite-based alterations in miR-21, miR-125b, and miR-203 in squamous cell carcinoma of the oral cavity and correlation to important target proteins. J. Carcinog. 2012, 11, 18. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  207. Tomasetti, M.; Gaetani, S.; Monaco, F.; Neuzil, J.; Santarelli, L. Epigenetic Regulation of miRNA Expression in Malignant Mesothelioma: miRNAs as Biomarkers of Early Diagnosis and Therapy. Front. Oncol. 2019, 9, 1293. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  208. Bi, X.; Lv, X.; Liu, D.; Guo, H.; Yao, G.; Wang, L.; Liang, X.; Yang, Y. METTL3-mediated maturation of miR-126-5p promotes ovarian cancer progression via PTEN-mediated PI3K/Akt/mTOR pathway. Cancer Gene Ther. 2021, 28, 335–349. [Google Scholar] [CrossRef] [PubMed]
  209. Di Paolo, D.; Pontis, F.; Moro, M.; Centonze, G.; Bertolini, G.; Milione, M.; Mensah, M.; Segale, M.; Petraroia, I.; Borzi, C.; et al. Cotargeting of miR-126-3p and miR-221-3p inhibits PIK3R2 and PTEN.; reducing lung cancer growth and metastasis by blocking AKT and CXCR4 signalling. Mol. Oncol. 2021, 15, 2969–2988. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  210. Feng, Q.; Wang, D.; Guo, P.; Zhang, Z.; Feng, J. Long non-coding RNA HOTAIR promotes the progression of synovial sarcoma through microRNA-126/stromal cell-derived factor-1 regulation. Oncol. Lett. 2021, 21, 444. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  211. Li, X.; Lu, H.; Fan, G.; He, M.; Sun, Y.; Xu, K.; Shi, F. A novel interplay between HOTAIR and DNA methylation in osteosarcoma cells indicates a new therapeutic strategy. J. Cancer Res. Clin. Oncol. 2017, 143, 2189–2200. [Google Scholar] [CrossRef] [PubMed]
  212. Liu, R.; Gu, J.; Jiang, P.; Zheng, Y.; Liu, X.; Jiang, X.; Huang, E.; Xiong, S.; Xu, F.; Liu, G.; et al. DNMT1-microRNA126 epigenetic circuit contributes to esophageal squamous cell carcinoma growth via ADAM9-EGFR-AKT signaling. Clin. Cancer Res. 2015, 21, 854–863. [Google Scholar] [CrossRef] [PubMed]
  213. Dong, P.; Xiong, Y.; Hanley, S.J.B.; Yue, J.; Watari, H. Musashi-2, a novel oncoprotein promoting cervical cancer cell growth and invasion, is negatively regulated by p53-induced miR-143 and miR-107 activation. J. Exp. Clin. Cancer Res. 2017, 36, 150. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  214. Zhang, J.; Sun, Q.; Zhang, Z.; Ge, S.; Han, Z.-G.; Chen, W.-T. Loss of microRNA-143/145 disturbs cellular growth and apoptosis of human epithelial cancers by impairing the MDM2-p53 feedback loop. Oncogene 2013, 32, 61–69. [Google Scholar] [CrossRef] [PubMed]
  215. Ng, E.K.; Li, R.; Shin, V.Y.; Siu, J.M.; Ma, E.S.; Kwong, A. MicroRNA-143 is downregulated in breast cancer and regulates DNA methyltransferases 3A in breast cancer cells. Tumour Biol. 2014, 35, 2591–2598. [Google Scholar] [CrossRef] [PubMed]
  216. Zhou, J.H.; Yao, Z.X.; Zheng, Z.; Yang, J.; Wang, R.; Fu, S.J.; Pan, X.F.; Liu, Z.H.; Wu, K. G-MDSCs-Derived Exosomal miRNA-143-3p Promotes Proliferation via Targeting of ITM2B in Lung Cancer. Onco Targets Ther. 2020, 13, 9701–9719. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  217. Zhang, Q.; Feng, Y.; Liu, P.; Yang, J. MiR-143 inhibits cell proliferation and invasion by targeting DNMT3A in gastric cancer. Tumour Biol. 2017, 39, 1010428317711312. [Google Scholar] [CrossRef] [PubMed]
  218. Dong, Y.Z.; Hu, T. Effects of miR-143 overexpression on proliferation, apoptosis, EGFR and downstream signaling pathways in PC9/GR cell line. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 1709–1716. [Google Scholar] [CrossRef] [PubMed]
  219. Ngalame, N.N.; Makia, N.L.; Waalkes, M.P.; Tokar, E.J. Mitigation of arsenic-induced acquired cancer phenotype in prostate cancer stem cells by miR-143 restoration. Toxicol. Appl. Pharmacol. 2016, 312, 11–18. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  220. Gao, Z.; Zhang, P.; Xie, M.; Gao, H.; Yin, L.; Liu, R. miR-144/451 cluster plays an oncogenic role in esophageal cancer by inhibiting cell invasion. Cancer Cell Int. 2018, 18, 184. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  221. Ma, Y.; Zhang, H.; Li, G.; Hu, J.; Liu, X.; Lin, L. LncRNA ANRIL promotes cell growth, migration and invasion of hepatocellular carcinoma cells via sponging miR-144. Anticancer Drugs 2019, 30, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
  222. Zhang, L.Y.; Ho-Fun Lee, V.; Wong, A.M.; Kwong, D.L.; Zhu, Y.H.; Dong, S.S.; Kong, K.L.; Chen, J.; Tsao, S.W.; Guan, X.Y.; et al. MicroRNA-144 promotes cell proliferation, migration and invasion in nasopharyngeal carcinoma through repression of PTEN. Carcinogenesis 2013, 34, 454–463. [Google Scholar] [CrossRef] [PubMed]
  223. Liu, C.; Su, C.; Chen, Y.; Li, G. MiR-144-3p promotes the tumor growth and metastasis of papillary thyroid carcinoma by targeting paired box gene 8. Cancer Cell Int. 2018, 18, 54. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  224. Wang, P.; Yang, Z.; Ye, T.; Shao, F.; Li, J.; Sun, N.; He, J. lncTUG1/miR-144-3p affect the radiosensitivity of esophageal squamous cell carcinoma by competitively regulating c-MET. J. Exp. Clin. Cancer Res. 2020, 39, 7. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  225. Sureban, S.M.; May, R.; Mondalek, F.G.; Qu, D.; Ponnurangam, S.; Pantazis, P.; Anant, S.; Ramanujam, R.P.; Houchen, C.W. Nanoparticle-based delivery of siDCAMKL-1 increases microRNA-144 and inhibits colorectal cancer tumor growth via a Notch-1 dependent mechanism. J. Nanobiotechnol. 2011, 9, 40. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  226. Wang, B.; Wu, H.H.; Abuetabh, Y.; Leng, S.; Davidge, S.T.; Flores, E.R.; Eisenstat, D.D.; Leng, R. p63, a key regulator of Ago2, links to the microRNA-144 cluster. Cell Death Dis. 2022, 13, 397. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  227. Qiao, Y.; Wang, B.; Yan, Y.; Niu, L. Long noncoding RNA ST8SIA6-AS1 promotes cell proliferation and metastasis in triple-negative breast cancer by targeting miR-145-5p/CDCA3 to inactivate the p53/p21 signaling pathway. Environ. Toxicol. 2022, 37, 2398–2411. [Google Scholar] [CrossRef] [PubMed]
  228. Jodeiry Zaer, S.; Aghamaali, M.; Amini, M.; Doustvandi, M.A.; Hosseini, S.S.; Baradaran, B.; Najafi, S.; Baghay Esfandyari, Y.; Mokhtarzadeh, A. Cooperatively inhibition effect of miR-143-5p and miR-145-5p in tumorigenesis of glioblastoma cells through modulating AKT signaling pathway. Bioimpacts 2024, 14, 29913. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  229. Xin, Z.; Tong, Z.; Tan, J.; Liu, C. MicroRNA-145-5p aggravates cell apoptosis and oxidative stress in tongue squamous cell carcinoma. Exp. Ther. Med. 2021, 21, 373. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  230. Zhou, J.; Jin, S. Circ_0058063 Contributed to Oral Squamous Cell Carcinoma Development by Sponging miR-145 and Regulating PI3K/AKT Pathway. Mol. Biotechnol. 2023, 65, 2049–2060. [Google Scholar] [CrossRef] [PubMed]
  231. Mei, L.L.; Wang, W.J.; Qiu, Y.T.; Xie, X.F.; Bai, J.; Shi, Z.Z. miR-145-5p Suppresses Tumor Cell Migration, Invasion and Epithelial to Mesenchymal Transition by Regulating the Sp1/NF-κB Signaling Pathway in Esophageal Squamous Cell Carcinoma. Int. J. Mol. Sci. 2017, 18, 1833. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  232. Du, Y.; Li, J.; Xu, T.; Zhou, D.D.; Zhang, L.; Wang, X. MicroRNA-145 induces apoptosis of glioma cells by targeting BNIP3 and Notch signaling. Oncotarget 2017, 8, 61510–61527. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  233. Fujii, T.; Shimada, K.; Tatsumi, Y.; Hatakeyama, K.; Obayashi, C.; Fujimoto, K.; Konishi, N. microRNA-145 promotes differentiation in human urothelial carcinoma through down-regulation of syndecan-1. BMC Cancer 2015, 15, 818. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  234. Zheng, M.; Wu, Z.; Wu, A.; Huang, Z.; He, N.; Xie, X. MiR-145 promotes TNF-α-induced apoptosis by facilitating the formation of RIP1-FADDcaspase-8 complex in triple-negative breast cancer. Tumour Biol. 2016, 37, 8599–8607. [Google Scholar] [CrossRef] [PubMed]
  235. Liang, Y.; Hou, L.; Li, L.; Li, L.; Zhu, L.; Wang, Y.; Huang, X.; Hou, Y.; Zhu, D.; Zou, H.; et al. Dichloroacetate restores colorectal cancer chemosensitivity through the p53/miR-149-3p/PDK2-mediated glucose metabolic pathway. Oncogene 2020, 39, 469–485. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  236. Jin, L.; Hu, W.L.; Jiang, C.C.; Wang, J.X.; Han, C.C.; Chu, P.; Zhang, L.J.; Thorne, R.F.; Wilmott, J.; Scolyer, R.A.; et al. MicroRNA-149*, a p53-responsive microRNA functions as an oncogenic regulator in human melanoma. Proc. Natl. Acad. Sci. USA 2011, 108, 15840–15845. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  237. Zhao, L.W.; Yu, A.J.; Zhang, Y.J.; Wang, X.C.; Han, B.; Wang, X.H. MicroRNA-149 suppresses the malignant phenotypes of ovarian cancer via downregulation of MSI2 and inhibition of PI3K/AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 55–64. [Google Scholar] [CrossRef] [PubMed]
  238. Pan, S.J.; Zhan, S.K.; Pei, B.G.; Sun, Q.F.; Bian, L.G.; Sun, B.M. MicroRNA-149 inhibits proliferation and invasion of glioma cells via blockade of AKT1 signaling. Int. J. Immunopathol. Pharmacol. 2012, 25, 871–881. [Google Scholar] [CrossRef] [PubMed]
  239. Pfeffer, S.R.; Grossmann, K.F.; Cassidy, P.B.; Yang, C.H.; Fan, M.; Kopelovich, L.; Leachman, S.A.; Pfeffer, L.M. Detection of Exosomal miRNAs in the Plasma of Melanoma Patients. J. Clin. Med. 2015, 4, 2012–2027. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  240. Niu, Y.; Zhang, J.; Tong, Y.; Li, J.; Liu, B. miR-145-5p restrained cell growth, invasion, migration and tumorigenesis via modulating RHBDD1 in colorectal cancer via the EGFR-associated signaling pathway. Int. J. Biochem. Cell Biol. 2019, 117, 105641. [Google Scholar] [CrossRef] [PubMed]
  241. Xu, Y.; Qiu, Z.; Chen, J.; Huang, L.; Zhang, J.; Lin, J. LINC00460 promotes neuroblastoma tumorigenesis and cisplatin resistance by targeting miR-149-5p/DLL1 axis and activating Notch pathway in vitro and in vivo. Drug Deliv. Transl. Res. 2024, 14, 2003–2018. [Google Scholar] [CrossRef] [PubMed]
  242. Tian, P.; Yan, L. Inhibition of MicroRNA-149-5p Induces Apoptosis of Acute Myeloid Leukemia Cell Line THP-1 by Targeting Fas Ligand (FASLG). Med. Sci. Monit. 2016, 22, 5116–5123. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  243. Lin, R.J.; Lin, Y.C.; Yu, A.L. miR-149* induces apoptosis by inhibiting Akt1 and E2F1 in human cancer cells. Mol. Carcinog. 2010, 49, 719–727. [Google Scholar] [CrossRef] [PubMed]
  244. Wang, D.T.; Ma, Z.L.; Li, Y.L.; Wang, Y.Q.; Zhao, B.T.; Wei, J.L.; Qi, X.; Zhao, X.T.; Jin, Y.X. miR-150, p53 protein and relevant miRNAs consist of a regulatory network in NSCLC tumorigenesis. Oncol. Rep. 2013, 30, 492–498. [Google Scholar] [CrossRef] [PubMed]
  245. Lei, B.; Huang, Y.; Zhou, Z.; Zhao, Y.; Thapa, A.J.; Li, W.; Cai, W.; Deng, Y. Circular RNA hsa_circ_0076248 promotes oncogenesis of glioma by sponging miR-181a to modulate SIRT1 expression. J. Cell Biochem. 2019, 120, 6698–6708. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  246. Chen, Y.; Liao, W.; Yuan, A.; Xu, H.; Yuan, R.; Cao, J. MiR-181a reduces radiosensitivity of non-small-cell lung cancer via inhibiting PTEN. Panminerva Med. 2022, 64, 374–383. [Google Scholar] [CrossRef] [PubMed]
  247. Lin, F.; Li, Y.; Yan, S.; Liu, S.; Qian, W.; Shen, D.; Lin, Q.; Mao, W. MicroRNA-181a inhibits tumor proliferation, invasiveness, and metastasis and is downregulated in gastric cancer. Oncol. Res. 2015, 22, 75–84. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  248. Yuan, Y.; Long, Z. LncRNA LINC01232 Enhances Proliferation, Angiogenesis, Migration and Invasion of Colon Adenocarcinoma Cells by Downregulating miR-181a-5p. J. Microbiol. Biotechnol. 2023, 33, 398–409. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  249. Cai, G.; Wang, Y.; Houda, T.; Yang, C.; Wang, L.; Gu, M.; Mueck, A.; Croteau, S.; Ruan, X.; Hardy, P. MicroRNA-181a suppresses norethisterone-promoted tumorigenesis of breast epithelial MCF10A cells through the PGRMC1/EGFR-PI3K/Akt/mTOR signaling pathway. Transl. Oncol. 2021, 14, 101068. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  250. Ranjbar, R.; Karimian, A.; Aghaie Fard, A.; Tourani, M.; Majidinia, M.; Jadidi-Niaragh, F.; Yousefi, B. The importance of miRNAs and epigenetics in acute lymphoblastic leukemia prognosis. J. Cell Physiol. 2019, 234, 3216–3230. [Google Scholar] [CrossRef] [PubMed]
  251. Xu, G.; Yang, Y.; Yang, J.; Xiao, L.; Wang, X.; Qin, L.; Gao, J.; Xuan, R.; Wu, X.; Chen, Z.; et al. Screening and identification of miR-181a-5p in oral squamous cell carcinoma and functional verification in vivo and in vitro. BMC Cancer 2023, 23, 162. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  252. Maroof, H.; Irani, S.; Arianna, A.; Vider, J.; Gopalan, V.; Lam, A.K. Interactions of Vascular Endothelial Growth Factor and p53 with miR-195 in Thyroid Carcinoma: Possible Therapeutic Targets in Aggressive Thyroid Cancers. Curr. Cancer Drug Targets 2019, 19, 561–570. [Google Scholar] [CrossRef] [PubMed]
  253. Selvi, R.B.; Swaminathan, A.; Chatterjee, S.; Shanmugam, M.K.; Li, F.; Ramakrishnan, G.B.; Siveen, K.S.; Chinnathambi, A.; Zayed, M.E.; Alharbi, S.A.; et al. Inhibition of p300 lysine acetyltransferase activity by luteolin reduces tumor growth in head and neck squamous cell carcinoma (HNSCC) xenograft mouse model. Oncotarget 2015, 6, 43806–43818. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  254. Wang, Y.; Mu, L.; Huang, M. MicroRNA-195 suppresses rectal cancer growth and metastasis via regulation of the PI3K/AKT signaling pathway. Mol. Med. Rep. 2019, 20, 4449–4458. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  255. Lu, H.; Kang, F. Down-regulating NEAT1 inhibited the viability and vasculogenic mimicry formation of sinonasal squamous cell carcinoma cells via miR-195-5p/VEGFA axis. Biosci. Rep. 2020, 40, BSR20201373. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  256. Duan, X.; Shen, N.; Chen, J.; Wang, J.; Zhu, Q.; Zhai, Z. Circular RNA MYLK serves as an oncogene to promote cancer progression via microRNA-195/cyclin D1 axis in laryngeal squamous cell carcinoma. Biosci. Rep. 2019, 39, BSR20190227. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  257. Boldrin, E.; Gaffo, E.; Niedermayer, A.; Boer, J.M.; Zimmermann, M.; Weichenhan, D.; Claus, R.; Münch, V.; Sun, Q.; Enzenmüller, S.; et al. MicroRNA-497/195 is tumor suppressive and cooperates with CDKN2A/B in pediatric acute lymphoblastic leukemia. Blood 2021, 138, 1953–1965. [Google Scholar] [CrossRef] [PubMed]
  258. Liang, M.; Huang, G.; Liu, Z.; Wang, Q.; Yu, Z.; Liu, Z.; Lin, H.; Li, M.; Zhou, X.; Zheng, Y. Elevated levels of hsa_circ_006100 in gastric cancer promote cell growth and metastasis via miR-195/GPRC5A signalling. Cell Prolif. 2019, 52, e12661. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  259. Jin, Y.; Wang, M.; Hu, H.; Huang, Q.; Chen, Y.; Wang, G. Overcoming stemness and chemoresistance in colorectal cancer through miR-195-5p-modulated inhibition of notch signaling. Int. J. Biol. Macromol. 2018, 117, 445–453. [Google Scholar] [CrossRef] [PubMed]
  260. Bai, E.; Dong, M.; Lin, X.; Sun, D.; Dong, L. Expressional and functional characteristics of checkpoint kinase 1 as a prognostic biomarker in hepatocellular carcinoma. Transl. Cancer Res. 2022, 11, 4272–4288. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  261. Tamura, M.; Sasaki, Y.; Kobashi, K.; Takeda, K.; Nakagaki, T.; Idogawa, M.; Tokino, T. CRKL oncogene is downregulated by p53 through miR-200s. Cancer Sci. 2015, 106, 1033–1040. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  262. Kim, T.; Veronese, A.; Pichiorri, F.; Lee, T.J.; Jeon, Y.J.; Volinia, S.; Pineau, P.; Marchio, A.; Palatini, J.; Suh, S.S.; et al. p53 regulates epithelial-mesenchymal transition through microRNAs targeting ZEB1 and ZEB2. J. Exp. Med. 2011, 208, 875–883. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  263. Lin, H.; Yang, L.; Tian, F.; Nie, S.; Zhou, H.; Liu, J.; Chen, W. Up-regulated LncRNA-ATB regulates the growth and metastasis of cholangiocarcinoma via miR-200c signals. Onco Targets Ther. 2019, 12, 7561–7571. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  264. Qiu, M.; Liang, Z.; Chen, L.; Tan, G.; Liu, L.; Wang, K.; Chen, H.; Liu, J. MicroRNA-200c suppresses cell growth and metastasis by targeting Bmi-1 and E2F3 in renal cancer cells. Exp. Ther. Med. 2017, 13, 1329–1336. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  265. Muñoz-Hidalgo, L.; San-Miguel, T.; Megías, J.; Serna, E.; Calabuig-Fariñas, S.; Monleón, D.; Gil-Benso, R.; Cerdá-Nicolás, M.; López-Ginés, C. The Status of EGFR Modulates the Effect of miRNA-200c on ZEB1 Expression and Cell Migration in Glioblastoma Cells. Int. J. Mol. Sci. 2020, 22, 368. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  266. Liu, P.L.; Liu, W.L.; Chang, J.M.; Chen, Y.H.; Liu, Y.P.; Kuo, H.F.; Hsieh, C.C.; Ding, Y.S.; Chen, W.W.; Chong, I.W. MicroRNA-200c inhibits epithelial-mesenchymal transition, invasion, and migration of lung cancer by targeting HMGB1. PLoS ONE 2017, 12, e0180844. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  267. Boominathan, L. The tumor suppressors p53, p63, and p73 are regulators of microRNA processing complex. PLoS ONE 2010, 5, e10615. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  268. Funamizu, N.; Lacy, C.R.; Kamada, M.; Yanaga, K.; Manome, Y. MicroRNA-203 induces apoptosis by upregulating Puma expression in colon and lung cancer cells. Int. J. Oncol. 2015, 47, 1981–1988. [Google Scholar] [CrossRef] [PubMed]
  269. Pal, D.; Mukhopadhyay, D.; Ramaiah, M.J.; Sarma, P.; Bhadra, U.; Bhadra, M.P. Regulation of Cell Proliferation and Migration by miR-203 via GAS41/miR-10b Axis in Human Glioblastoma Cells. PLoS ONE 2016, 11, e0159092. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  270. Han, N.; Li, H.; Wang, H. MicroRNA-203 inhibits epithelial-mesenchymal transition, migration, and invasion of renal cell carcinoma cells via the inactivation of the PI3K/AKT signaling pathway by inhibiting CAV1. Cell Adh. Migr. 2020, 14, 227–241. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  271. Siu, M.K.; Abou-Kheir, W.; Yin, J.J.; Chang, Y.S.; Barrett, B.; Suau, F.; Casey, O.; Chen, W.Y.; Fang, L.; Hynes, P.; et al. Correction: Loss of EGFR signaling-regulated miR-203 promotes prostate cancer bone metastasis and tyrosine kinase inhibitors resistance. Oncotarget 2018, 9, 32403, Erratum in Oncotarget 2014, 5, 3770–3784. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  272. Diao, Y.; Guo, X.; Jiang, L.; Wang, G.; Zhang, C.; Wan, J.; Jin, Y.; Wu, Z. miR-203, a tumor suppressor frequently down-regulated by promoter hypermethylation in rhabdomyosarcoma. J. Biol. Chem. 2014, 289, 529–539. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  273. Odar, K.; Boštjančič, E.; Gale, N.; Glavač, D.; Zidar, N. Differential expression of microRNAs miR-21, miR-31, miR-203, miR-125a-5p and miR-125b and proteins PTEN and p63 in verrucous carcinoma of the head and neck. Histopathology 2012, 61, 257–265. [Google Scholar] [CrossRef] [PubMed]
  274. Zhang, K.; Dai, L.; Zhang, B.; Xu, X.; Shi, J.; Fu, L.; Chen, X.; Li, J.; Bai, Y. miR-203 is a direct transcriptional target of E2F1 and causes G1 arrest in esophageal cancer cells. J. Cell Physiol. 2015, 230, 903–910. [Google Scholar] [CrossRef] [PubMed]
  275. He, J.; Feng, X.; Hua, J.; Wei, L.; Lu, Z.; Wei, W.; Cai, H.; Wang, B.; Shi, W.; Ding, N.; et al. miR-300 regulates cellular radiosensitivity through targeting p53 and apaf1 in human lung cancer cells. Cell Cycle 2017, 16, 1943–1953. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  276. Xu, X.H.; Li, D.W.; Feng, H.; Chen, H.M.; Song, Y.Q. MiR-300 regulate the malignancy of breast cancer by targeting p53. Int. J. Clin. Exp. Med. 2015, 8, 6957–6966. [Google Scholar] [PubMed] [PubMed Central]
  277. Wang, R.; Yu, Z.; Chen, F.; Xu, H.; Shen, S.; Chen, W.; Chen, L.; Su, Q.; Zhang, L.; Bi, J.; et al. miR-300 regulates the epithelial-mesenchymal transition and invasion of hepatocellular carcinoma by targeting the FAK/PI3K/AKT signaling pathway. Biomed. Pharmacother. 2018, 103, 1632–1642. [Google Scholar] [CrossRef] [PubMed]
  278. Chen, L.; Fang, C.; Yuan, X.; Liu, M.; Wu, P.; Zhong, L.; Chen, Z. Has-miR-300—GADD45B promotes melanoma growth via cell cycle. Aging 2023, 15, 13920–13943. [Google Scholar] [CrossRef]
  279. Guan, X.; Shi, A.; Zou, Y.; Sun, M.; Zhan, Y.; Dong, Y.; Fan, Z. EZH2-Mediated microRNA-375 Upregulation Promotes Progression of Breast Cancer via the Inhibition of FOXO1 and the p53 Signaling Pathway. Front. Genet. 2021, 12, 633756. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  280. Liu, Y.; Xing, R.; Zhang, X.; Dong, W.; Zhang, J.; Yan, Z.; Li, W.; Cui, J.; Lu, Y. miR-375 targets the p53 gene to regulate cellular response to ionizing radiation and etoposide in gastric cancer cells. DNA Repair 2013, 12, 741–750. [Google Scholar] [CrossRef] [PubMed]
  281. Saika, M.; Nakashiro, K.I.; Tokuzen, N.; Shirai, H.; Uchida, D. Possible Role of miR-375-3p in Cervical Lymph Node Metastasis of Oral Squamous Cell Carcinoma. Cancers 2024, 16, 1492. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  282. Hu, C.; Lv, L.; Peng, J.; Liu, D.; Wang, X.; Zhou, Y.; Huo, J. MicroRNA-375 suppresses esophageal cancer cell growth and invasion by repressing metadherin expression. Oncol. Lett. 2017, 13, 4769–4775. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  283. Yang, M.Y.; Lin, P.M.; Liu, Y.C.; Hsiao, H.H.; Yang, W.C.; Hsu, J.F.; Hsu, C.M.; Lin, S.F. Induction of cellular senescence by doxorubicin is associated with upregulated miR-375 and induction of autophagy in K562 cells. PLoS ONE 2012, 7, e37205. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  284. Liu, L.; Shen, W.; Zhu, Z.; Lin, J.; Fang, Q.; Ruan, Y.; Zhao, H. Combined inhibition of EGFR and c-ABL suppresses the growth of fulvestrant-resistant breast cancer cells through miR-375-autophagy axis. Biochem. Biophys. Res. Commun. 2018, 498, 559–565. [Google Scholar] [CrossRef] [PubMed]
  285. Gong, G.; Han, Z.; Wang, W.; Xu, Q.; Zhang, J. Silencing hsa_circRNA_0008035 exerted repressive function on osteosarcoma cell growth and migration by upregulating microRNA-375. Cell Cycle 2020, 19, 2139–2147. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  286. Liu, G.; Huang, K.; Jie, Z.; Wu, Y.; Chen, J.; Chen, Z.; Fang, X.; Shen, S. CircFAT1 sponges miR-375 to promote the expression of Yes-associated protein 1 in osteosarcoma cells. Mol. Cancer 2018, 17, 170. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  287. Li, P.; Lv, H.; Wu, Y.; Xu, K.; Xu, M.; Ma, Y. E2F transcription factor 1 is involved in the phenotypic modulation of esophageal squamous cell carcinoma cells via microRNA-375. Bioengineered 2021, 12, 10047–10062. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  288. Huang, G.; Zhong, X.; Yao, L.; Ma, Q.; Liao, H.; Xu, L.; Zou, J.; Sun, R.; Wang, D.; Guo, X. MicroRNA-449a inhibits cell proliferation and migration by regulating mutant p53 in MDA-MB-468 cells. Exp. Ther. Med. 2021, 22, 1020. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  289. Kumar, P.; Sharad, S.; Petrovics, G.; Mohamed, A.; Dobi, A.; Sreenath, T.L.; Srivastava, S.; Biswas, R. Loss of miR-449a in ERG-associated prostate cancer promotes the invasive phenotype by inducing SIRT1. Oncotarget 2016, 7, 22791–22806. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  290. Bach, D.H.; Kim, D.; Bae, S.Y.; Kim, W.K.; Hong, J.Y.; Lee, H.J.; Rajasekaran, N.; Kwon, S.; Fan, Y.; Luu, T.T.; et al. Targeting Nicotinamide N-Methyltransferase and miR-449a in EGFR-TKI-Resistant Non-Small-Cell Lung Cancer Cells. Mol. Ther. Nucleic Acids 2018, 11, 455–467. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  291. Jiang, T.; Liu, J.; Mu, J. Downregulation of microRNA-449a-5p promotes esophageal squamous cell carcinoma cell proliferation via cyclin D1 regulation. Mol. Med. Rep. 2018, 18, 848–854. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  292. Kawasaki, H.; Takeuchi, T.; Ricciardiello, F.; Lombardi, A.; Biganzoli, E.; Fornili, M.; De Bortoli, D.; Mesolella, M.; Cossu, A.M.; Scrima, M.; et al. Definition of miRNA Signatures of Nodal Metastasis in LCa: miR-449a Targets Notch Genes and Suppresses Cell Migration and Invasion. Mol. Ther. Nucleic Acids 2020, 20, 711–724. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  293. Zhang, Y.; Xu, H. LncRNA FAL1 Upregulates SOX4 by Downregulating miR-449a to Promote the Migration and Invasion of Cervical Squamous Cell Carcinoma (CSCC) Cells. Reprod. Sci. 2020, 27, 935–939. [Google Scholar] [CrossRef] [PubMed]
  294. Lizé, M.; Pilarski, S.; Dobbelstein, M. E2F1-inducible microRNA 449a/b suppresses cell proliferation and promotes apoptosis. Cell Death Differ. 2010, 17, 452–458. [Google Scholar] [CrossRef] [PubMed]
  295. Chang, H.; Zhang, X.; Li, B.; Meng, X. PARP1 Is Targeted by miR-519a-3p and Promotes the Migration, Invasion, and Tube Formation of Ovarian Cancer Cells. Cancer Biother. Radiopharm. 2022, 37, 824–836. [Google Scholar] [CrossRef] [PubMed]
  296. Tu, K.; Liu, Z.; Yao, B.; Han, S.; Yang, W. MicroRNA-519a promotes tumor growth by targeting PTEN/PI3K/AKT signaling in hepatocellular carcinoma. Int. J. Oncol. 2016, 48, 965–974. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  297. Yu, G.; Zhang, T.; Jing, Y.; Bao, Q.; Tang, Q.; Zhang, Y. miR-519 suppresses nasopharyngeal carcinoma cell proliferation by targeting oncogene URG4/URGCP. Life Sci. 2017, 175, 47–51. [Google Scholar] [CrossRef] [PubMed]
  298. Huang, Y.; Guerrero-Preston, R.; Ratovitski, E.A. Phospho-ΔNp63α-dependent regulation of autophagic signaling through transcription and micro-RNA modulation. Cell Cycle 2012, 11, 1247–1259. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  299. Wang, Q.; Selth, L.A.; Callen, D.F. MiR-766 induces p53 accumulation and G2/M arrest by directly targeting MDM4. Oncotarget 2017, 8, 29914–29924. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  300. Ding, Y.; Zhang, M.; Hu, S.; Zhang, C.; Zhou, Y.; Han, M.; Li, J.; Li, F.; Ni, H.; Fang, S.; et al. MiRNA-766-3p inhibits gastric cancer via targeting COL1A1 and regulating PI3K/AKT signaling pathway. J. Cancer 2024, 15, 990–998. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  301. Li, Y.C.; Li, C.F.; Chen, L.B.; Li, D.D.; Yang, L.; Jin, J.P.; Zhang, B. MicroRNA-766 targeting regulation of SOX6 expression promoted cell proliferation of human colorectal cancer. Onco Targets Ther. 2015, 8, 2981–2988. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  302. Yan, H.; Chen, X.; Li, Y.; Fan, L.; Tai, Y.; Zhou, Y.; Chen, Y.; Qi, X.; Huang, R.; Ren, J. MiR-1205 functions as a tumor suppressor by disconnecting the synergy between KRAS and MDM4/E2F1 in non-small cell lung cancer. Am. J. Cancer Res. 2019, 9, 312–329. [Google Scholar] [PubMed] [PubMed Central]
  303. Zhao, X.; Cui, D.; Yan, F.; Yang, L.; Zhang, M.; Huang, B. Circ_0006174 promotes the malignancy of colorectal cancer cell via the miR-1205/CCBE1/Wnt pathway. Mol. Med. Rep. 2022, 26, 251. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  304. Qin, Y.; Zhang, S.; Deng, S.; An, G.; Qin, X.; Li, F.; Xu, Y.; Hao, M.; Yang, Y.; Zhou, W.; et al. Epigenetic silencing of miR-137 induces drug resistance and chromosomal instability by targeting AURKA in multiple myeloma. Leukemia 2017, 31, 1123–1135. [Google Scholar] [CrossRef] [PubMed]
  305. Neault, M.; Mallette, F.A.; Richard, S. miR-137 Modulates a Tumor Suppressor Network-Inducing Senescence in Pancreatic Cancer Cells. Cell Rep. 2016, 14, 1966–1978. [Google Scholar] [CrossRef] [PubMed]
  306. Wang, J.; Wang, Z.; Yuan, J.; Wang, Q.; Shen, X. Upregulation of miR-137 Expression Suppresses Tumor Growth and Progression via Interacting with DNMT3a Through Inhibiting the PTEN/Akt Signaling in HCC. Onco Targets Ther. 2021, 14, 165–176. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  307. Zeng, X.; Xu, Z.; Gu, J.; Huang, H.; Gao, G.; Zhang, X.; Li, J.; Jin, H.; Jiang, G.; Sun, H.; et al. Induction of miR-137 by Isorhapontigenin (ISO) Directly Targets Sp1 Protein Translation and Mediates Its Anticancer Activity Both In Vitro and In Vivo. Mol. Cancer Ther. 2016, 15, 512–522. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  308. Luo, Y.; Li, X.; Dong, J.; Sun, W. microRNA-137 is downregulated in thyroid cancer and inhibits proliferation and invasion by targeting EGFR. Tumour Biol. 2016, 37, 7749–7755. [Google Scholar] [CrossRef] [PubMed]
  309. Wang, X.; Zhang, G.; Cheng, Z.; Dai, L.; Jia, L.; Jing, X.; Wang, H.; Zhang, R.; Liu, M.; Jiang, T.; et al. Knockdown of LncRNA-XIST Suppresses Proliferation and TGF-β1-Induced EMT in NSCLC Through the Notch-1 Pathway by Regulation of miR-137. Genet. Test. Mol. Biomark. 2018, 22, 333–342. [Google Scholar] [CrossRef] [PubMed]
  310. Yao, F.; Zhang, C.; Du, W.; Liu, C.; Xu, Y. Identification of Gene-Expression Signatures and Protein Markers for Breast Cancer Grading and Staging. PLoS ONE 2015, 10, e0138213. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  311. Huang, Y.; Gu, M.; Tang, Y.; Sun, Z.; Luo, J.; Li, Z. Systematic review and meta-analysis of prognostic microRNA biomarkers for survival outcome in laryngeal squamous cell cancer. Cancer Cell Int. 2021, 21, 316. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  312. Zou, W.; Hu, X.; Wang, D.; Jiang, L. Prognostic Value of MiRNAs in Patients with Laryngeal Cancer: A Systematic Review and Meta-Analysis. Curr. Cancer Drug Targets 2020, 20, 802–810. [Google Scholar] [CrossRef] [PubMed]
  313. Hegazy, M.; Elkady, M.A.; Yehia, A.M.; Elsakka, E.G.E.; Abulsoud, A.I.; Abdelmaksoud, N.M.; Elshafei, A.; Abdelghany, T.M.; Elkhawaga, S.Y.; Ismail, A.; et al. The role of miRNAs in laryngeal cancer pathogenesis and therapeutic resistance—A focus on signaling pathways interplay. Pathol. Res. Pract. 2023, 246, 154510. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representation of miRs with prognostic value in laryngeal carcinoma and their association with major oncogenes/pathways (Euler diagram). Asterix displays available data for association with FAT1 gene.
Figure 1. Representation of miRs with prognostic value in laryngeal carcinoma and their association with major oncogenes/pathways (Euler diagram). Asterix displays available data for association with FAT1 gene.
Ijms 25 13468 g001
Table 1. Search strategy summary.
Table 1. Search strategy summary.
The Search Strategy Summary
ItemsSpecification
Databases and other sources searchedPubMed/MEDLINE
Search terms used“laryngeal carcinoma” OR “laryngeal cancer” AND “microRNA” OR “miRNA” AND “prognostic marker” OR “prognosis”
Timeframe2002–2024
Inclusion and exclusion criteriaInclusion criteria: without predefined restriction as to the study type
Exclusion criteria: restricted to articles published in English
Selection processK.K. and T.P. independently screened data sources. Data analysis was conducted by K.K. and T.P.
Table 2. Studies reporting microRNAs with prognostic value associated with laryngeal carcinoma along with methodology and patient characteristics.
Table 2. Studies reporting microRNAs with prognostic value associated with laryngeal carcinoma along with methodology and patient characteristics.
Ref.MicroRNAStageMethodSample MaterialPoor PrognosisEndpoint
Measurement
[7]miR-9T1–T4qRT-PCRFresh frozenUpregulatedOS
[8]miR-19aT1–T4 qPCRFresh frozenUpregulatedOS
[9]miR-20b-5pT1–T4RT-qPCRFresh frozenUpregulatedOS; DFS: RFS
[10,11,12]miR-21T1–T4Microarray
qRT-PCR
Fresh frozen
Blood sample
UpregulatedOS
[13]miR-23aT1–T4qRT-PCRFresh frozenUpregulatedOS
[14,15]miR-26a/bT1–T4
I–IV
RT-qPCRFresh frozen
Blood sample
UpregulatedOS
OS; DFS
[16]miR-31I–IVqRT-PCRFresh frozenUpregulatedOS
[17,18,19,20,21]miR-34T1–T4
I–IV
T3–T4
T3–T4
T1–T4
RT-qPCR
Bisulfite pyrosequencing
Fresh frozen
FFPE
Upregulated
Downregulated
Hypermethylation
OS; DFS
[22]miR-93-5pT3–T4Microarray
RT-qPCR
Fresh frozenUpregulatedRFS
[21]miR-146a-5pT1–T4RT-qPCRFresh frozenUpregulatedOS
[23,24]miR-196bT1–T4
T2–T4
qRT-PCRFresh frozenUpregulatedOS
[22]miR-210-3p T3–T4Microarray
RT-qPCR
Fresh frozenUpregulatedRFS
[25]miR-301a-3pT1–T2 (6 patients)
T1–T4
qRT-PCRFresh frozen
FFPE
UpregulatedOS
[21,26]miR-378c-5pT1–T4
I–IV
RT-qPCRFresh frozen
Blood sample
UpregulatedOS;
RFS
[27]miR-1246T1–T4qRT-PCRFresh frozen
FFPE
Blood sample
UpregulatedOS
[28]miR-296-5pT1–T2TLDA
qRT- PCR
FFPEUpregulatedDFS
[29]miR-155T2–T4qRT-PCRTissue samples (not specified)UpregulatedOS
[30]miR-632I–IV RT-PCR
qRT-LSCCR assay
Serum samplesUpregulatedOS; DFS
[31]miR-101T1–T4qRT-PCRFresh frozen
FFPE
DownregulatedOS
[32]miR-125b-5pNot specifiedqRT-PCRFresh frozenDownregulatedOS
[33,34]miR-143-3pI–IVMicroarray
RT-qPCR
Fresh frozenDownregulatedOS
[22,35]miR-144-3pT3–T4Microarray
RT-qPCR
Fresh frozen
FFPE
Downregulated
Upregulated
OS; RFS
[36]miR-145-5pI–IVqRT-PCRFresh frozenDownregulated
Hypermethylated
OS
[37]miR-149I–IVqRT-PCRFresh frozenDownregulatedOS
[38,39]miR-195T1–T4
I–IV
qRT-PCRFresh frozenDownregulatedOS
[40]miR-203T1–T4qRT-PCRFresh frozenDownregulatedOS
[41]miR-300T1–T2qRT-PCRFresh frozenDownregulatedOS
[42]miR-375T1–T4qRT-PCRFresh frozen DownregulatedOS
[21]miR-449a-5pT1–T4RT-qPCRFresh frozenDownregulatedOS
[43]miR-519a I–IV qRT-PCRFresh frozenDownregulatedDFS
[44]miR-29c-3pT1–T4qRT-PCRFFPEDownregulatedOS
[45]miR-200a/cT1–T4qRT-PCRFFPEDownregulatedRFS
[46]miR-137T1–T4 Methylation-specific PCR
qRT-PCR
FFPEHypermethylatedOS
[47]miR-181aT2–T4qRT-PCRTissue samples (not specified)DownregulatedOS
[48]miR-766-5pI–IV qRT-PCRTissue samples (not specified)DownregulatedOS
[49]miR-1205 T1–T4qRT-PCRTissue samples (not specified)DownregulatedOS; DFS
[50]miR-126 T1–T4qRT-PCRTissue samples (not specified)
Blood samples
DownregulatedOS
[12]miR-10a-5pT1–T4Microarray
qRT-PCR
Blood sampleDownregulatedOS
OS—Overall survival. DFS—disease-free survival. RFS—recurrence free survival. Numbers in the table represent references.
Table 3. Studies reporting molecular association between the most prominently affected oncogenes and pathways in laryngeal carcinoma and miRs with prognostic value. Numbers in the table represent references. Light green–existent data for regulatory association; grey–lack of regulatory association.
Table 3. Studies reporting molecular association between the most prominently affected oncogenes and pathways in laryngeal carcinoma and miRs with prognostic value. Numbers in the table represent references. Light green–existent data for regulatory association; grey–lack of regulatory association.
MicroRNAp53PTEN/PI3KCCND1CDKN2A
/p16
EGFRNOTCHP63FAT1FADDTRAF3E2F1
miR-9[59][60,61][62][63][64][65] [66]
miR-19a[67][68] [69] [70]
miR-20b-5p [71] [72] [73]
miR-21[74,75][76,77][78][79][80][81] [82]
miR-23a[83,84,85][86,87][88][89][90]
miR-26a[91,92,93][94,95] [96][97] [98] [99]
miR-26b[100,101][102,103] [104] [98] [105]
miR-31[106,107,108][109,110][111][112][113][114] [115]
miR-34[116,117][118,119][120] [121][122] [123]
miR-93-5p[124][125,126][127] [128] [127]
miR-146a-5p[129,130][131,132][130][133][134][135] [136][137]
miR-155[138,139,140][141,142][143][144][145][146][147] [148]
miR-196b [149,150]
miR-210-3p [151,152][153,154][155] [156]
miR-296-5p[157][158][159] [160]
miR-301a-3p [161,162][163] [164] [165]
miR-378c-5p[166,167][168][166] [169]
miR-632 [170]
miR-1246[171,172][173][174] [175][171] [174]
miR-10a-5p[176,177][178,179][180]
miR-29c-3p[181,182][183,184][185][186][187] [188]
miR-101[189,190][191,192][26][193][194][195][196] [197]
miR-125b-5p[198,199][200,201][202][203][204][205][206]
miR-126 [207][208,209][210][211][212]
miR-143-3p[213,214][215,216][217] [218][219]
miR-144-3p[220][221,222][223] [224][225][226]
miR-145-5p[227,228][229,230][231] [232][233] [234]
miR-149[235,236][237][238][239][240][241] [242] [243]
miR-181a[244,245][246][247][248][249][250] [251]
miR-195[252,253][254,255][256][257][258][259] [260]
miR-200a[261,262] [263][264][265][266][267]
miR-203[268,269][270] [271][272][273] [274]
miR-300[275,276][277] [278]
miR-375[279,280][281][282][283][284][285] [286] [287]
miR-449a-5p[288,289][290][291] [292][267][293] [294]
miR-519a [295][296][297] [298]
miR-766-5p[299][300][301]
miR-1205 [302] [303] [50]
miR-137[304,305][306][307][28][308][309]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Komitova, K.S.; Dimitrov, L.D.; Stancheva, G.S.; Kyurkchiyan, S.G.; Petkova, V.; Dimitrov, S.I.; Skelina, S.P.; Kaneva, R.P.; Popov, T.M. A Critical Review on microRNAs as Prognostic Biomarkers in Laryngeal Carcinoma. Int. J. Mol. Sci. 2024, 25, 13468. https://doi.org/10.3390/ijms252413468

AMA Style

Komitova KS, Dimitrov LD, Stancheva GS, Kyurkchiyan SG, Petkova V, Dimitrov SI, Skelina SP, Kaneva RP, Popov TM. A Critical Review on microRNAs as Prognostic Biomarkers in Laryngeal Carcinoma. International Journal of Molecular Sciences. 2024; 25(24):13468. https://doi.org/10.3390/ijms252413468

Chicago/Turabian Style

Komitova, Kristina S., Lyuben D. Dimitrov, Gergana S. Stancheva, Silva G. Kyurkchiyan, Veronika Petkova, Stoyan I. Dimitrov, Silviya P. Skelina, Radka P. Kaneva, and Todor M. Popov. 2024. "A Critical Review on microRNAs as Prognostic Biomarkers in Laryngeal Carcinoma" International Journal of Molecular Sciences 25, no. 24: 13468. https://doi.org/10.3390/ijms252413468

APA Style

Komitova, K. S., Dimitrov, L. D., Stancheva, G. S., Kyurkchiyan, S. G., Petkova, V., Dimitrov, S. I., Skelina, S. P., Kaneva, R. P., & Popov, T. M. (2024). A Critical Review on microRNAs as Prognostic Biomarkers in Laryngeal Carcinoma. International Journal of Molecular Sciences, 25(24), 13468. https://doi.org/10.3390/ijms252413468

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

Article Metrics

Back to TopTop