Next Article in Journal
Multi-Strain Probiotic Mixture Affects Brain Morphology and Resting State Brain Function in Healthy Subjects: An RCT
Next Article in Special Issue
Characterizing Relevant MicroRNA Editing Sites in Parkinson’s Disease
Previous Article in Journal
Whole-Transcriptome Sequencing Analyses of Nuclear Antixoxidant-1 in Endothelial Cells: Role in Inflammation and Atherosclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Roles of miRNA, lncRNA, circRNA, and Their Cross-Talk in Pituitary Adenoma

1
Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 119 South Forth West Ring, Beijing 100070, China
2
Department of Oncology, First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Hefei 230032, China
*
Authors to whom correspondence should be addressed.
Cells 2022, 11(18), 2920; https://doi.org/10.3390/cells11182920
Submission received: 1 August 2022 / Revised: 14 September 2022 / Accepted: 16 September 2022 / Published: 19 September 2022
(This article belongs to the Special Issue Non-coding RNAs and Neurological Diseases 2022)

Abstract

:
Pituitary adenoma (PA) is a common intracranial tumor without specific biomarkers for diagnosis and treatment. Non-coding RNAs (ncRNAs), including microRNAs (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA), regulate a variety of cellular processes, such as cell proliferation, differentiation, and apoptosis. Increasing studies have shown that the dysregulation of ncRNAs, especially the cross-talk between lncRNA/circRNA and miRNA, is related to the pathogenesis, diagnosis, and prognosis of PA. Therefore, ncRNAs can be considered as promising biomarkers for PA. In this review, we summarize the roles of ncRNAs from different specimens (i.e., tissues, biofluids, cells, and exosomes) in multiple subtypes of PA and highlight important advances in understanding the contribution of the cross-talk between ncRNAs (e.g., competing endogenous RNAs) to PA disease.

1. Introduction

Pituitary adenoma (PA) is the third most common intracranial tumor, accounting for 10 to 15% of all intracranial tumors [1]. The prevalence of PA is 1:10,000 [2]. In addition, it has been shown that the average prevalence of PA is 14.4% in autopsies and 22.5% in radiological series [2]. PAs are usually benign lesions, but some patients have aggressive characteristics, including local invasiveness, rapid growth, and poor responses to conventional treatments [3]. Consequently, they are divided into functional PAs (FPAs) and non-functional PAs (NFPAs). FPAs include growth hormone (GH)-, prolactin (PRL)-, adrenocorticotropic hormone (ACTH)-, thyroid-stimulating hormone (TSH)-, and follicle-stimulating hormone/luteinizing hormone (FSH/LH)-secreting adenomas [4,5]. Some proteins are involved in the pathogenesis of PA subtypes. Patients with either familial or sporadic PA often have aryl hydrocarbon receptor-interacting protein mutations, and guanine nucleotide-binding protein G (s) subunit alpha and G-protein-coupled receptor 101 mutations contribute to excess growth hormone [6]. In addition, abnormal expression levels of the deubiquitinases BRAF, USP8, and USP48 are related to the production of adrenocorticotropic hormone [6]. However, many patients with PA lack early biomarkers due to endocrine diseases and nerve compression [1]. In addition, patients with PA are difficult to identify in the existing medical diagnosis, and many adverse reactions occur during the treatment of patients with PA [7]. Therefore, it is of great clinical significance to further investigate the pathogenesis of PA and find novel molecular targets for the diagnosis and treatment of PA. In recent years, epigenetics, especially non-coding RNAs, has been demonstrated to play an important role in the pathogenesis of PA.
About 75% of the human genome can be transcribed, but only 1.5% of the transcribed sequences code for proteins; i.e., over 98% of non-coding sequences do not have the biological function of coding for proteins (named non-coding RNAs, ncRNAs) [8,9,10,11]. Although ncRNAs cannot encode proteins, they regulate the protein expression processes, including transcription and translation. Furthermore, increasing studies have shown that ncRNAs are involved in the multilevel regulation of gene expression and have a key role in the central dogma of molecular biology [12,13]. Thus, ncRNAs regulate cell differentiation, development, and cellular fate [13]. The most common ncRNAs include microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA), which are all actively involved in the regulation of tumor-suppressor genes and oncogenes [14,15,16]. Numerous studies have suggested that the abnormal expression of ncRNAs is related to the occurrence and progression of PA, and exosome-derived ncRNAs have enriched our understanding of the pathogenesis of PA. Consequently, ncRNAs, especially exosome-derived ncRNAs, may have a key role in the pathogenesis of PA and may become novel diagnostic and therapeutic biomarkers.
In this article, we summarize the biological significance of ncRNAs, exosome-derived ncRNAs, and their target genes in different PA subtypes (e.g., NFPA, IPA, GHPA, etc.). In addition, we also discuss the potentiality of ncRNAs as noninvasive biomarkers for the diagnosis and prognosis of PA.

2. miRNA and PA

The miRNA, a small, endogenous, single-stranded, non-coding RNA with a length of 19 to 25 nucleotides that inhibits post-transcriptional protein synthesis via binding to the 3′-untranslated region (UTR) of target messenger RNAs (mRNAs), plays an important role in a variety of essential and biophysiological processes including cell proliferation, differentiation, and apoptosis. Accumulating studies have shown the dysfunction of miRNAs in human biological fluids and in cell-free environments, suggesting that these miRNAs can function as oncogenes and tumor suppressors. These important findings may contribute to provide novel diagnostic and prognostic biomarkers for PA. Table 1 summarizes miRNAs differentially expressed (DE) in PA and its subtypes.
Table 1. Dysregulation of miRNA in different PA types.
Table 1. Dysregulation of miRNA in different PA types.
Author (Ref.)Type of PASamplemiRNAExpressionTarget GeneBiological Function
Mossakowska [17]CDTissues, AtT-20/D16v-F2 cellshsa-miR-124-3p and hsa-miR-135-5pDownNR3C1, NR3C2Regulation of glucocorticoid receptor
Zhang [18]NFPA SerummiRNA-26b, miRNA-138, miRNA-206, and let-7e DownNATumor-suppressor gene
Chen [19]PATissues, GH3 cellsmiR-424-3pDownJAG1Cell proliferation and EMT
Zhao [20]IPAExosomes, tissues, and GH4C1, MMP, and GH3 cellsmiR-149-5p and miR-99a-3pDownNOVA1, DTL, RAB27BThe expression of EMT- and ECM-related markers and tumor-related genes
Wei [21]PATissues, GH3 cellsmiR-137DownEGFRTumor suppressor, cell proliferation
Wu [22]IPATissuesmiR-193a-3pDown Higher risk of postoperative residual and recurrence
Wang [23]PAGT1.1 and AtT-20 cellsmiR-219a-2-3pDownMDM2Cell proliferation and apoptosis
Németh [24]GHPA, FSH/LH+ PA, hormone-immunonegative PAPlasmamiR-143-3pDownNADistinguishing preoperative plasma samples from normal controls
He [25]NFPA, GHPA, PRLPATissuesmiR-34c-3p, miR-34b-5p, miR-378, miR-338-5p, miR-493-5p, miR-181b-5p, miR-184, and miR-124-3pPRLPA: miR-34c-3p, miR-34b-5p, miR-378, and miR-338-5p were downregulated; NFPA: miR-493-5p and miR-124-3p were downregulated and miR-181b-5p was upregulated; GHPA: miR-184 was upregulated and miR-124-3p was downregulatedNANovel biomarkers
Cai [26]NFPATissuesmiR-370 DownHMGA2miR-370 was decreased by CXCL12 treatment and miR-370 inhibited tumor growth and invasiveness
Song [27]NFPATissues, GH3 cellsmiR-137 DownWIF1Cell proliferation and invasion
Leone [28]GHPA, NFPA miR-23b and miR-130b DownHMGA2, CCNA2Cell proliferation arresting the cells in the G1 and G2 phase of the cell cycle
Gentilin [29]ACTH-secreting PATissues, AtT-20 cellsmiR-26aUpPRKCDDelayed the cell cycle in G1 phase
Trivellin [30]GHPA, NFPATissues, GH3 cellsmiR-107UpAIPCell proliferation
Butz [31]NFPATissuesmiR-135a, miR-140-5p, miR-582-3p, miR-582-5p, and miR-938UpSmad3Tumor size
Butz [32]NFPA, GHPATissuesmiR-128a, miR-155, and miR-516a-3pUpWee1Cell proliferation
Qian [33]PATissueslet-7DownHMGA2Tumor invasion
Bottoni [34]GHPA, PRLPATissuesmiR-15a and miR-16-1Down Tumor diameter
Lyu [35]NFPAExosomehsa-miR-486-5pDownNATumor progression
He [36]PATissues, MMQ and HP75 cellsmiR-448DownBCL2Cell proliferation and migration
Xiong [37]GHPAExosome, tissueshsa-miR-21-5pUpPDCD4Bone formation and trabecula number
Yang [38]PAGH4C1 cellsmiR-34aDownSox7Cell proliferation and apoptosis
Fan [39]GHPATissues, GH3 cellsmiR-185UpSSTR2Cell proliferation and apoptosis
Liao [40]PAGH3 and MMQ cellsmiR-200cUpPTENPituitary tumor formation
Ref., reference; NA, not available; EMT, epithelial–mesenchymal transition; ECM, extracellular matrix.

2.1. Circulating miRNA and PA

In 2008, circulating miRNA (c-miRNA) was detected in peripheral blood [41,42,43]. Compared to that of cellular RNA, the expression of c-miRNA is very stable in the RNase-rich environments of human biofluids due to its binding to specific proteins (e.g., the Argonaute (Ago 2) protein) [44,45]. In addition, c-miRNAs have been shown to be resistant to harmful conditions, such as high temperature, acidic or alkaline environments, and repeated freeze–thaw cycles [41,46]. Therefore, c-miRNAs are ideal candidates for novel noninvasive blood biomarkers for different types of diseases, and c-miRNAs contribute to diagnosis, prognosis, and postoperative monitoring for PA with aggressive behavior and NFPA.
The RNA sequencing (RNA-seq) of plasma samples showed that there were several DE miRNAs in preoperative and postoperative patients with PA, including 3 DE miRNAs in a GH group, 7 in an FSH/LH group, and 66 in a hormone-immunonegative (HN) group [24]. Subsequently, some DE miRNAs (i.e., miR-143-3p in FSH/LH, and miR-26b-5p, miR-126-5p, and miR-148b-3p in HN) were used for validation by real-time polymerase chain reaction (qRT-PCR) [24]. Circulating miR-143-3p was significantly upregulated in patients with preoperative FSH/LH compared to patients with postoperative FSH/LH+ [24].
Furthermore, the tumor size in patients with postoperative FSH/LH decreased significantly, which may result in a decrease in circulating miR-143-3p [24]. Circulating miR-143-3p exhibited a strong differentiation power, with an area under curve (AUC) value of 0.79 between preoperative and postoperative patients with FSH/LH PAs [24]. The plasma miR-143-3p decreases in patients with FSH/LH+ adenoma, but its application in the evaluation of tumor recurrence needs further investigation [24]. These findings suggest that plasma miR-143-3p decreases may be a potential biomarker for patients with FSH/LH+ adenoma after transsphenoidal surgery. In addition, some studies have also found that miR-143 was significantly downregulated in PA tissues [47,48], and inhibited tumor proliferation by targeting the K-Ras gene [47].
Belaya et al. evaluated whether miRNAs were DE in plasma samples from patients with adrenocorticotropic hormone (ACTH)-dependent Cushing’s syndrome (CS) caused by either ectopic ACTH secretion (EAS) or Cushing’s disease (CD) [49]. In their study, 21 miRNAs were measured [49]; the circulating levels of miR-16-5p, miR-145-5p, and miR-7g-5p were altered; and miR-16-5p had the most distinguished power, with an AUC value of 0.879. Therefore, these results indicate that c-miRNAs are promising biomarkers for distinguishing between CD and EAS.
In addition to biomarkers for diagnosis and prognosis, c-miRNAs were also associated with the survival time for PA. Compared to that in healthy controls, the serum expression of miR-16 was decreased in patients with PA, whereas higher levels of miR-16 were accompanied by longer overall survival (OS) times and disease-free survival (DFS) times [50]. In vitro, miR-16 inhibited the proliferation and angiogenesis of PA via regulating the VEGFR2/p38/NF-κB pathway [50]. Consequently, miR-16 may be a potential target for the treatment of PA.
In conclusion, c-miRNAs have an important role for the diagnosis and prognosis of PA, but also for the treatment of PA. In the future, the research directions of c-miRNAs may lead to the translation of potential candidates with important roles in the development of the disease to their implementation as biomarkers for the diagnosis and prognosis of PA in clinical practice.

2.2. miRNA in PA Tissue

Some studies have shown that the abnormal expression of miRNAs in tumor tissues was significantly associated with the levels of miRNAs released into the peripheral circulation [1,7,51], suggesting that the change in miRNAs in PA tissues is implicated in the occurrence and progression of the disease.
In 2005, several DE miRNAs between normal pituitary tissues and PAs were identified [34]. Subsequent studies have shown that miRNAs may be involved in tumorigenesis, invasion, and aggressiveness as oncogenes and as tumor suppressors. Using the HiSeq 2000 sequencing system, several distinctive miRNA expression patterns were identified in three different PAs (i.e., NFPAs, GHPAs, and PRLPAs) [25]. Compared to normal pituitary tissues, significant downregulation of miR-34c-3p, miR-34B-5p, miR-378, and miR-338-5p in PRLPA; downregulation of miR-493-5p and upregulation of miR-181b-5p in NFPA; and significant upregulation of miR-184 in GHPA were observed. In addition, downregulation of miR-124-3p in both NFPA and GHPA was observed [25]. These miRNA signatures may be promising therapeutic biomarkers for different types of PA. A few studies investigated aggressiveness-associated miRNAs in aggressive pituitary tumors; several DE miRNAs between PRLPA and aggressive PA were identified using the GSE46294 miRNA expression profile [52,53]. They suggested that most of the hub genes were modulated by hsa-miR-489 and hsa-miR-520b via the construction of a miRNA–hub gene network; thus, these two miRNAs may provide new targets for the diagnosis and treatment of PRLPA.
In addition to being downregulated in the PAs’ plasma, miR-143 was downregulated in PA tissues, especially in ACTH-secreting pituitary tumors [24,47,48]. Although the expression of miR-143 was not associated with the tumor size and postoperative remission rate [48], miR-143 inhibited cell proliferation and promoted apoptosis by targeting K-Ras [47]. Vicchio et al. found that the downregulation of miR-26b-5p and miR-30a-5p was negatively correlated with ki-67, ‘atypical’ morphological characteristics, and cavernous sinus invasion [54]. These miRNAs can be used as predictors of PA invasion.
In conclusion, exploring the underlying mechanisms between miRNAs and pituitary tumorigenesis may contribute to identifying new potential biomarkers that may be used as an innovative treatment for PA.

2.3. miRNA in PA-Associated Cell Lines

PA-associated cell lines including mouse (e.g., AtT-20 and GT1.1 cells) and rat (e.g., GH3 and MMQ cells) lines were used for exploring the underlying mechanisms of PA. The expression of miR-143 was downregulated in the GH3 and MMQ cell lines except in the circulation and tissue [24,47,48]. Subsequent experiments revealed that miR-143 inhibited the two cells’ proliferation and promoted apoptosis by regulating the oncogene K-Ras [47]. Furthermore, a recent study found that the underexpression of miR-146b-5p was related to the tumor size, the overall survival rate, a poor disease-free survival rate, a poor Knosp grade, and a poor Hardy grade [55]. In addition, miRNA-146b-5p negatively regulated GH3 cell proliferation, invasion, and migration, and induced apoptosis by inhibiting the ephrin receptor A7 (EPHA7) gene via regulating the IRAK4/TRAF6/NF-κB signaling pathway [55].
In murine GT1.1 and AtT-20 cells, the expression of miR-219a-2-3p was significantly downregulated [23]. Moreover, the overexpression of miR-219a-2-3p inhibited cell proliferation and promoted apoptosis as well as reducing MDM2 expression by binding to the 3’-UTR of the MDM2 mRNA and promoting p53 expression [23]. Therefore, miR-219a-2-3p modulated cell proliferation and apoptosis by targeting MDM2/p53 in PA, suggesting that miR-219a-2-3p may be a novel therapeutic marker for PA.
In conclusion, in vitro studies provide greater insight into the pathogenesis of the disease, and the combination of in vitro, in vivo, and clinical specimens (including blood and tissue) will greatly promote the research progress.

2.4. Exosome-Derived miRNA and PA

In human biological fluids, miRNAs accumulate within extracellular vesicles (EVs, including apoptotic bodies, microvesicles, and exosomes) and bind to macromolecules, such as the AGO2 protein or lipoproteins [56]. Exosomes play a key role in the cells’ cross-talk and in the pathogenesis of human diseases.
Recently, our group explored the function of miRNAs in IPAs and the therapeutic strategy of exosome-derived miRNAs for the disease [20]. Twenty DE miRNAs were identified; the two lowest miRNAs, miR-99a-3p and miR-149-5p, were used for the subsequent studies. We found that exosome-derived miR-99a-3p and miR-149-5p inhibited cell viability, migration, and tube formation [20]. Therefore, this study suggested that the upregulation of miR-99a-3p and miR-149-5p can inhibit the progression of IPA by the exosome, and exosome-derived miRNAs represent good potential candidates for future therapies for IPA.
Acromegaly, an endocrine and metabolic disease caused by GHPAs, is partially attributable to an excessive function of the GH and insulin-like growth factor-1 (IGF1) hormones [57,58,59]. In addition, GHPA-derived exosomes contain miRNAs and proteins that regulate cell proliferation and differentiation in distal extremities. Xiong et al. found that GHPA-derived exosomes may be involved in bone formation and osteoblast proliferation via promoting cell viability and DNA replication [37]. Furthermore, they found that exosome-derived miR-21-5p stimulated osteoblast information in the GH/IGF1 pathway. Taken together, exosome-derived miR-21-5p may be a candidate biomarker for the treatment of acromegaly.
RNA-seq was used for identifying exosome-derived miRNAs in six somatotroph adenomas and six healthy pituitary samples [60]. In this study, a total of 169 DE exosomal miRNAs were identified, and miR-423-5p was significantly downregulated in the somatotroph adenoma, whereas pituitary tumor transforming gene (PTTG1), a target of miR-423-5p, was upregulated in patients and contributed to the promotion of proliferation and migration of somatotropic adenoma cells. Thus, their findings indicate that exosome-derived miRNAs, especially miR-423-5p, may be valuable biomarkers for the development of new therapeutic strategies. Next-generation sequencing showed that miR-26b-5p, miR-126-5p, miR-148b-3p, and miR-150-5p were detected in patients with FSH/LH+ adenoma plasma samples, and also in the exosomes of the patients, but others (i.e., miR-6514-3p, miR-6850-5p, and miR-6867-5p) were not detectable in plasma and exosomes [24]. Interestingly, the authors said that the decrease in miR-143-3p in the plasma may be a potential biomarker for patients with FSH/LH+ adenoma after transsphenoidal surgery, but the expression of exosome-derived miR-143-3p in FSH/LH+ samples was not significant, indicating that miR-143-3p is mainly expressed in protein-associated plasma rather than in exosomes.
In conclusion, exosomes are effective markers and promising new therapeutic targets for PA and its complications. However, the role of exosomes, especially intercellular communication, in the pathogenesis of PA needs further investigation.

3. lncRNA and PA

Long non-coding RNAs (lncRNAs) are non-coding RNAs with over 200 nucleotides that cannot encode peptides [61,62]. A variety of functions of lncRNAs have been discovered, such as regulating gene activation and silencing [63,64], post-translational regulation [65,66,67], alternative splicing [68], and X chromosome modification [69]. LncRNAs perform these functions through different mechanisms, including acting as competing endogenous RNAs (ceRNAs) that sponge miRNAs or proteins [70], promoting or inhibiting long-range chromatin interactions [71,72], acting as molecular scaffolds for guiding chromatin-modifying enzymes [73,74,75], and even functioning through the behavior of transcription itself [64,76]. Given the important roles of lncRNAs in the regulation of epigenetic processes, the dysregulation of lncRNAs is implicated in many human diseases [9,77,78,79]. In addition, increasing studies have shown that lncRNA is involved in the progression, proliferation, apoptosis, autophagy, and metastasis of PA. In this section, available data on the role of dysregulated lncRNAs in the pathogenesis of PA will be discussed. Table 2 summarizes DE lncRNAs in PA and its subtypes.
It is well known that miRNAs are processed from primary miRNA transcripts, and some lncRNAs can serve as host transcripts of miRNAs [77,80,81,82]. MIR205HG is a lncRNA that harbors the gene of miR-205. Du et al. showed that MIR205HG regulated growth hormone levels in the anterior pituitary gland independent of miR-205 and regulated the expression of growth hormone and prolactin by interacting with the transcription factor Pit1 [80]. Emerging technologies of high-throughput sequencing and microarray analysis have contributed to systematically identifying abundant lncRNAs. In 2017, a genome-wide study found that there were 839 DE lncRNAs between PA and normal tissues, and some of them might be related to the mTOR signaling pathway [83]. In a microarray analysis, a total of 113 DE lncRNAs were identified in NFPA [84]. The most significantly DE lncRNA, n334366, is a unique satellite in the co-expression network, where it is related to the neuroactive ligand–receptor interaction pathways [84]. Subsequently, a total of 246 DE lncRNAs were identified between IPA and NIPA using a lncRNA microarray analysis [85]. The LOC105371531, LOC105375785, and FAM182B expression levels were significantly decreased in IPA compared to NIPA according to qRT-PCR [85]. ROC curve analysis showed that the expression of LOC105375785 and FAM182B could distinguish IPA from NIPA, suggesting that LOC105375785 and FAM182B might be implicated in the invasiveness of IPAs and might be novel biomarkers for the diagnosis of IPAs. Subsequently, the transcriptional expression of 684 DE lncRNAs was compared between bone invasive pituitary adenoma (BIPA) and NIPA [86]. The inflammatory factor TNF-α, regulated by the lncRNA SNHG24, plays an important role in BIPA [86], suggesting that they might be therapeutic targets.
Moreover, some common lncRNAs have also been investigated in patients with PA. H19 is the first encoding lncRNA discovered and plays opposite roles in different tumors [87,88,89]. The expression of H19 was significantly upregulated in invasive GHPA compared to that in noninvasive GHPA, suggesting that H19 might be a diagnostic and therapeutic target for GHPA [90]. Since H19 knockdown contributes to inhibiting the activation of the NF-κB pathway, it has been speculated that H19 may increase the invasiveness of PA by affecting the transcription of target genes via the NF-κB pathway [91]. Additionally, H19 expression was downregulated in human PA tissues and was negatively correlated with disease progression, while the upregulation of H19 inhibited tumor growth and cell proliferation [92]. Mechanistically, H19 could block mTORC1-mediated 4E-BP1 phosphorylation. These findings indicate that H19 may be a potential target for PA. Notably, exosome-derived H19 was also significantly downregulated in PA tissues and GH3 cells [93]. Exosome-derived H19 expression was negatively correlated with the progression of PA and used as a prognostic biomarker for patients treated with prolactinomas. Mechanistic studies found that H19 inhibited the growth of distal pituitary tumors by regulating 4E-BP1 phosphorylation [93]. Therefore, plasma exosome-derived H19 may be a significant target for predicting prolactinomas’ responses.
In conclusion, lncRNAs are promising therapeutic targets, and the development of these therapies requires the identification of some cell- or tissue-specific lncRNAs, but it remains challenging. In addition, exosome-derived lncRNAs and the cross-talk between lncRNAs and miRNAs are also promising research directions.
Table 2. Dysregulation of lncRNA in different PA types.
Table 2. Dysregulation of lncRNA in different PA types.
Author (Ref.)Type of PASamplelncRNAExpressionTarget GeneBiological Function
Peng [85]IPA TissuesFAM182B, LOC105371531, LOC105375785DownNAFAM182B and LOC105375785 can distinguish IPA from NIPA.
Cheng [94]NFPATissuesLOC101927765, RP11-23N2.4, RP4-533D7.4NANAHigh prediction accuracy for NFPA recurrence.
Lu [95]PATissues, HP75 cell linesIFNG-AS1UpESRP2An oncogene that promoted tumor progression.
Lu [90]GHPATissuesH19, MALAT-1UpNANA
Fu [96]PATissues, HP75 cellsCCTA2UpPTTG1Poor prognosis, cell proliferation, migration, and invasion.
Li [97]NFPATissuesMEG, HOTAIRMEG3: Down; HOTAIR: UpPCNATumor development and invasion.
Cheng [98]NFPATissuesCOA6-AS1, RP11-23N2.4UpNATumor regrowth with a high predictive accuracy.
Zhang [99]PATissues, GH3 and HP75 cellsPVT1UpNACell migration, proliferation, and EMT.
Zhang [93]GHPA, NFPA, PRLPAExosome, GH3 cellsH19Down4E-BP1The prognosis or drug response.
Xing [84]NFPATissuesn334366, n335657, n409198, MEG3, n337303, n340496, n334406, n332607, n333074, n332409Down: n334366, n335657, n409198, MEG3. Up: n337303, n340496, n334406, n332607, n333074, n332409NATumorigenesis.
Ref., reference; NA, not available; EMT, epithelial–mesenchymal transition.

4. circRNA and PA

Most circular RNA (circRNA), a single-stranded, endogenous non-coding RNA, is produced from the back-splicing of exons of precursor mRNAs and is abundant and highly conserved in blood and disease [100,101]. Although circRNA has long been thought to be a transcriptional error, recent advances in RNA-seq and bioinformatics have shown that it plays an important role in human health and diseases [102]. The abnormal expression of circRNA is involved in various physiological and pathological processes [100,103,104], such as epithelial–mesenchymal transition, differentiation, metastasis, and tumorigenesis [105,106]. Several studies revealed that the dysregulation of circRNA is associated with many different tumors, including PA [107,108]. In this section, available data on DE circRNA in PA will be discussed. Table 3 summarizes DE circRNAs in PA and its subtypes.
The circRNA expression profile was identified between NFPA and PA, and the expression of circVPS13C was significantly increased in NFPA tissues and cells but was downregulated in patients’ serum 7 days after transsphenoidal adenoma resection. Mechanistically, the knockout of circVPS13C increased the expression of IFITM1 and activated MAPK/apoptosis-associated downstream genes; further studies showed that circVPS13C inhibited the expression of IFITM1 by competitively interacting with RRBP1. Therefore, circVPS13C is a critical regulator for the proliferation and development of NFPAs through a novel mechanism that regulates mRNA stability by interacting with ribosome-binding proteins [101]. Moreover, the circRNA signature has clinical application value in predicting recurrence and progression in NFPA. Two circRNAs (i.e., hsa_circ_0000066 and hsa_circ_0069707) were related to the progression-free survival in NFPA, and the two circRNAs had a high prediction accuracy for tumor recurrence [109].
In addition to the important role of circRNA in NFPA, it is also involved in GHPA. A circRNA microarray identified the DE circRNA profile in GHPA, of which 1938 circRNAs were upregulated and 1601 were downregulated [110]. Among all the DE circRNAs, hsa_circ_0001368 was significantly upregulated in GHPA and correlated with the invasiveness and serum GH level of GHPA. Further studies found that the knockdown of hsa_circ_0001368 significantly inhibited the cells’ proliferation, invasion, and serum GH level. Moreover, the expression of hsa_circ_0001368 was positively correlated with the pituitary-specific transcription factor Pit-1 [110]. Therefore, hsa_circ_0001368 may represent a novel therapeutic biomarker for GHPA.
In conclusion, although circRNAs have an important role in PA, studies on the association of circRNA with PA are currently limited. Further studies are highly encouraged to explore the role of circRNAs (especially exosome-derived circRNAs) and their downstream target genes or signaling pathways in the pathogenesis of PA.
Table 3. Dysregulation of circRNA in different PA types.
Table 3. Dysregulation of circRNA in different PA types.
Author (Ref.)Type of PASamplecircRNAExpressionTarget GeneBiological Function
Hu [111]NFPATissueshsa_circRNA_102597DownNAAssociated with tumor diameter and Knosp grade, differentiated invasive from noninvasive NFPAs, and predicted tumor progression and recurrence.
Zhang [101]NFPATissues, PDFS cellsCircVPS13CUpRRBP1Promoted proliferation and development.
Du [110]GHPATissueshsa_circ_0001368UpPit-1Associated with the invasiveness and serum GH level; promoted cell proliferation and invasion.
Guo [109]NFPATissueshsa_circ_0000066 and hsa_circ_0069707UpNAAssociated with the PFS, and had a high prediction accuracy for tumor recurrence.
Wang [112]NFPATissueshsa_circ_0054722, hsa_circ_0007362, hsa_circ_0012346, hsa_circ_0062222, hsa_circ_0016403, hsa_circ_0033349Up: hsa_circ_0054722, hsa_circ_0007362, hsa_circ_0012346, Down: hsa_circ_0062222, hsa_circ_0016403, hsa_circ_0033349NAContributed to diagnosis, prognosis, and clinical treatment.
Ref., reference; NA, not available; GH, growth hormone; PFS, progression-free survival.

5. ceRNA and PA

The competing endogenous RNA (ceRNA) hypothesis is that transcripts such as lncRNA and circRNA competitively bind to miRNAs through the miRNA response element (MRE), thus forming a complex regulatory network to realize their respective regulatory functions [113]. In this section, we discuss the available evidence for DE lncRNA and circRNA in PA, respectively. Table 4 summarizes the interactions between lncRNA/circRNA and miRNA in PA and its subtypes.
First, several upregulations of lncRNAs have been implicated in PA [114,115,116]. For example, lncRNA TUG1 was significantly downregulated in PA tissues and PA-associated cell lines [114]. The expression of TUG1 was associated with invasion, Knosp grade, and tumor size, and TUG1 silencing downregulated the expression of NF-κB p65 and κB (IκB)-α and TESC by targeting miR-187-3p [114], suggesting that TUG1 modulates PA progression by regulating the TESC–NF-κB signaling pathway via sponging miR-187-3p. However, lncRNA MEG3 was downregulated in PA tissues and PA-associated cells [117]. The overexpression of MEG3 inhibited the proliferation, invasion, migration, and epithelial–mesenchymal transition (EMT) process and accelerated PA-associated cells’ apoptosis. Further studies found that MEG3 negatively regulated miR-23b-3p expression, while miR-23b-3p negatively regulated FOXO4 expression [117]. Similar to the findings for MEG3 in PA, MEG3 was also decreased in NFPA [118]. Moreover, MEG3 is an enhancer of miR-376B-3P; the overexpression of MEG3 and miR-376B-3p inhibited tumorigenesis and promoted apoptosis; HMGA2, a target gene of miR-376B-3p, is an oncogene in PA and could be negatively regulated by MEG3 via enriching miR-376B-3p [118]. Therefore, the novel regulatory network of the MEG3/MIR-376B-3P/HMGA2 interactions in NFPAs may be helpful for anticancer treatments. In recent years, several studies have shown that some DE lncRNAs were upregulated in IPA tissues and cells; these lncRNAs participated in viability, migration, invasion, and epithelial-mesenchymal transition (EMT) by sponging miRNAs [119,120,121,122]. Therefore, these lncRNAs may be novel valuable therapeutic targets for IPA.
CircRNAs are primarily known to act as miRNA sponges or ceRNAs to regulate transcriptional activity [113]. CircOMA1 (hsa_circRNA_0002316) was significantly decreased but miR-145-5p was upregulated in NFPA samples [123]. The overexpression of miR-145-5p inhibited NFPA cell invasiveness and proliferation as well as promoting cell apoptosis, whereas circOMA1 can reverse these effects by sponging miR-125-5p. Therefore, circOMA1 promoted the tumorigenesis of NFPAs by acting as a sponge of the antioncogene miR-145-5p (123). Moreover, circNFIX (hsa-circ_0005660) was significantly upregulated but miR-34a-5p was downregulated in invasive PA tissues (124). The knockdown of circNFIX or overexpression of miR-34a-5p contributed to inhibiting cell proliferation, migration, and invasion, and circNFIX reversed the promoting effect of miR-34a-5p on the progression of PA by sponging miR-34a-5p (124). Therefore, circNFIX may be a promising target for the treatment of PA.
In conclusion, the interactions between miRNA and lncRNA/circRNA, especially the ceRNA mechanism, play an important role in the pathogenesis of PA and its subtypes.
Table 4. Interaction between lncRNA/circRNA and miRNA in different PA types.
Table 4. Interaction between lncRNA/circRNA and miRNA in different PA types.
Author (Ref.)Type of PASampleLncRNA/ circRNAExpressionTarget miRNAmiRNA TargetBiological Function
Du [123]NFPATissues, pituitary tumor-derived folliculostellate (PDFS) cell linecircOMA1 DownmiR-145-5pTPT1Promoted cell proliferation and invasiveness.
Cheng [124]IPATissues; GT1-1 and GH3 cellscircNFIXUpmiR-34a-5pCCNB1Promoted cell invasion, migration, and proliferation.
Zhu [79]BIPA TissuesMEG8UpmiR-454-3pTNF-αPromoted bone destruction and associated with poor PFS.
Zhao [125]IPATissues; RC-4B/C and GH3 cellsPCAT6UpmiR-139-3pBRD4Regulated the progression of PA.
Wu [78]PRLPATissues, GH3 cellsH19DownmiR-93aATG7H19 had a synergistic effect with dopamine agonist treatment on prolactinomas.
Yue [126]PATissues; GH1, RC-4B/C, GH3 and MMQ cell linesSNHG7UpmiR-449aNAAssociated with poor survival outcomes; increased cell viability, migration, and invasion and decreased apoptosis.
Zhu [86]BIPATissues; GH3 and RAW264.7 cellsNR_033258, SNHG24UpmiR-181c-5p, miR-454-3pNATNFα induced osteoclast differentiation, and NR_033258 and SNHG24 regulated TNFα expression.
Qiu [119]PAHP75 cellsLINC01004UpmiR-323a-3p/miR-136-5pRCN2Promoted disease progression.
Li [120]IPAGH3 and HP75 cellsKCNQ1OT1UpmiR-140-5pRAB11APromoted the EMT, cellular stemness, and proliferation and invasion.
Wu [115]PATissues; RC-4B/C, GH3, and MMQ cellsBBOX1-AS1UpmiR-361-3pE2F1Promoted cell invasion, apoptosis, and proliferation.
Huang [116]PAHP75 cellsLINC01116UpmiR-744-5pHOXB8Promoted cell proliferation, migration, and EMT process.
Li [121]IPATissues; AtT-20 and GT1-1 cellsLINC00473UpmiR-502-3pKMT5APromoted cell proliferation.
Zhang [114]PATissues; HP75 and GH3 cellsTUG1UpmiR-187-3pTESCPromoted cell proliferation, invasion, migration, and EMT, and inhibited apoptosis.
Mao [122]IPATissues, HP75 cellsSNHG6UpmiR-944RAB11APromoted cell viability, migration, invasion, and EMT.
Wang [117]PATissues; GH3 and MMQ cellsMEG3DownmiR-23b-3pFOXO4Promoted cell proliferation, apoptosis, and the EMT process.
Zhu [118]NFPATissues, PDFS cellsMEG3DownmiR-376B-3pHMGA2Tumorigenesis and cell apoptosis
Ref., reference; NA, not available; EMT, epithelial–mesenchymal transition; PFS, progression-free survival.

6. Conclusions and Future Perspective

The abundance of DE ncRNAs in PA has been widely reported. Investigating the roles of ncRNAs is helpful for understanding the occurrence and development of PA, and some ncRNAs can be used as biomarkers for the detection of PA [1,18,43,127], while other ncRNAs can contribute to the diagnosis of PA [52,85,90,112], distinguish the disease subtypes [24,25,34,93], and predict tumor invasion and progression [20,26,35,85,95,110,111,123]; the remaining ncRNAs may be implicated in the pathogenesis of PA [116,117,120,122]. In addition, the regulatory networks of lncRNA/circRNA and miRNA provide promising biomarkers for the noninvasive diagnosis and molecular therapy of PA, and exosome-derived ncRNAs bring a tremendous infusion of hope to the improvement of the disease diagnosis and future potential therapeutic targets for treatments. Therefore, available data on the DE ncRNAs in PA were reviewed and discussed in this study.
Among the abundant DE ncRNAs, miRNAs are the most widely studied. We discuss the role of miRNAs in different PA subtypes in tissues, biofluids, and cell-free environments [17,18,19,20,37]. miRNAs inhibit the expression of target genes at the post-transcriptional level by pairing with the 3′-UTR of mRNA (Figure 1); thus, miRNAs are involved in various pathological and physiological processes by playing a negative role in cell proliferation, differentiation, metabolism, and apoptosis. PA rarely progresses to malignancy but usually presents as aggressive growth, and miRNAs are related to excessive cell proliferation and apoptosis, and tumor size. Therefore, upregulated miRNAs may inhibit the expression of antioncogenes, while downregulated miRNAs cannot inhibit the expression of oncogenes. Moreover, miRNAs play an important role in the diagnosis and prognosis of PA. However, developing biomarkers based on DE miRNAs is inaccurate, as they are also altered in other cancers [128]. To address this issue, here are some research directions for future studies. Firstly, specific miRNAs from specimens should be developed. PA-specific tissue-/serum- and extracellular vesicle (e.g., exosome)-derived miRNAs are highly recommended. Secondly, combined diagnosis should be conducted. Future studies could measure different ncRNAs (e.g., lncRNAs, circRNA, and miRNAs) simultaneously, and perform combined diagnosis to improve the accuracy. In addition, miRNAs can be combined with patients’ medical records, clinical parameters, and radiological examinations to reach an adequate level of accuracy. Importantly, exosome-derived miRNAs participated in cross-talk and transformation between different cells, which raises the possibility of miRNA as circulating biomarkers [129].
Studies on the association of lncRNAs and circRNAs with PA are limited compared to those for miRNAs. On the one hand, lncRNAs and circRNAs participate in PA by regulating the expression of target genes [95,96,101,110]. On the other hand, many studies have shown that the two ncRNAs regulate mRNA by sponging miRNAs [78,79,123,124]. The dysregulation of lncRNAs regulates transcription by controlling the function of transcription factors, resulting in the overexpression of oncogenes or underexpression of antioncogenes. Similar to the role of miRNA in PA, these DE lncRNAs and circRNAs have also been identified in cancers other than PA, suggesting that the development of PA-specific lncRNAs and circRNAs requires more in-depth mechanism studies. Cross-talk between lncRNAs/circRNAs and miRNA, especially ceRNAs, will help us to understand the role of lncRNAs and circRNAs in the pathogenesis of PA [117,121,123,124], and potential regulatory mechanisms of lncRNAs, circRNAs, and miRNAs in PA are shown in Figure 1. In addition, few studies have explored the involvement of exosome-derived lncRNAs and circRNAs in PA, excepting the relationship between exosomal H19 and PA [93]. Therefore, the function of exosome-derived ncRNAs in the pathogenesis of PA remains to be investigated, especially the cross-talk of ncRNAs in different cells mediated by exosomes.
Although increasing studies have been performed to explore the relationship between ncRNAs and PA, the exact mechanism of PA caused by ncRNAs is still unclear. Therefore, some suggestions are highly recommended. Firstly, well-designed experiments should involve a particular signaling pathway to identify specific ncRNAs that are closely associated with PA. Secondly, unknown targets of ncRNAs, including miRNAs targeted by lncRNAs and circRNAs, and mRNAs targeted by the three ncRNAs, should be identified. Thirdly, the pathogenesis of PA and its subtypes should be investigated in combination with population studies and basic experiments as well as combination with genetics and epigenetics. Finally, it is urgent to develop novel drugs effective against ncRNA-associated targets and conduct related clinical trials for the treatment of PA.
With a better understanding of the molecular mechanisms and signaling pathways of cell- and tissue-specific ncRNAs in PA, it is believed that novel clinical diagnostic markers and targeted treatments for PA and its subtypes may be implemented.

Author Contributions

S.G. and X.Z. conceived the idea for the study. W.W. drafted the protocol with input from X.Z., Y.J., Y.X. and L.C. edited and modified the protocol. All authors have read and agreed to the published version of the manuscript.

Funding

S.G. was supported by the Beijing Municipal Science & Technology Commission (Z19110700660000) and Beijing Hospitals Authority Clinical Medicine Development of Special Funding Support (XMLX202108). X.Z. was supported by the National Natural Science Foundation of China (82100628), the Natural Science Foundation of Anhui Province (2108085QH313), the Postdoctoral Science Foundation of China (2021M700183), and the Postdoctoral Science Foundation of Anhui Province (2021B496).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Beylerli, O.; Beeraka, N.M.; Gareev, I.; Pavlov, V.; Yang, G.; Liang, Y.; Aliev, G. MiRNAs as Noninvasive Biomarkers and Therapeutic Agents of Pituitary Adenomas. Int. J. Mol. Sci. 2020, 21, 7287. [Google Scholar] [CrossRef] [PubMed]
  2. Campana, C.; Nista, F.; Castelletti, L.; Caputo, M.; Lavezzi, E.; Marzullo, P.; Ferrero, A.; Gaggero, G.; Canevari, F.R.; Rossi, D.C.; et al. Clinical and radiological presentation of parasellar ectopic pituitary adenomas: Case series and systematic review of the literature. J. Endocrinol. Investig. 2022, 45, 1465–1481. [Google Scholar] [CrossRef] [PubMed]
  3. Giuffrida, G.; D’Argenio, V.; Ferraù, F.; Lasorsa, V.A.; Polito, F.; Aliquò, F.; Ragonese, M.; Cotta, O.R.; Alessi, Y.; Oteri, R.; et al. Methylome Analysis in Nonfunctioning and GH-Secreting Pituitary Adenomas. Front. Endocrinol. 2022, 13, 841118. [Google Scholar] [CrossRef] [PubMed]
  4. Trouillas, J.; Jaffrain-Rea, M.L.; Vasiljevic, A.; Raverot, G.; Roncaroli, F.; Villa, C. How to Classify the Pituitary Neuroendocrine Tumors (PitNET)s in 2020. Cancers 2020, 12, 514. [Google Scholar] [CrossRef]
  5. Spada, A.; Mantovani, G.; Lania, A.G.; Treppiedi, D.; Mangili, F.; Catalano, R.; Carosi, G.; Sala, E.; Peverelli, E. Pituitary Tumors: Genetic and Molecular Factors Underlying Pathogenesis and Clinical Behavior. Neuroendocrinology 2022, 112, 15–33. [Google Scholar] [CrossRef]
  6. Chang, M.; Yang, C.; Bao, X.; Wang, R. Genetic and Epigenetic Causes of Pituitary Adenomas. Front. Endocrinol. 2020, 11, 596554. [Google Scholar] [CrossRef]
  7. Xu, D.; Wang, L. The Involvement of miRNAs in Pituitary Adenomas Pathogenesis and the Clinical Implications. Eur. Neurol. 2022, 85, 171–176. [Google Scholar] [CrossRef] [PubMed]
  8. Zhong, L.; Liu, P.; Fan, J.; Luo, Y. Long non-coding RNA H19: Physiological functions and involvements in central nervous system disorders. Neurochem. Int. 2021, 148, 105072. [Google Scholar] [CrossRef]
  9. DiStefano, J.K.; Gerhard, G.S. Long Noncoding RNAs and Human Liver Disease. Annu. Rev. Pathol. 2022, 17, 1–21. [Google Scholar] [CrossRef]
  10. An integrated encyclopedia of DNA elements in the human genome. Nature 2012, 489, 57–74. [CrossRef] [Green Version]
  11. Kapranov, P.; Cheng, J.; Dike, S.; Nix, D.A.; Duttagupta, R.; Willingham, A.T.; Stadler, P.F.; Hertel, J.; Hackermüller, J.; Hofacker, I.L.; et al. RNA maps reveal new RNA classes and a possible function for pervasive transcription. Science 2007, 316, 1484–1488. [Google Scholar] [CrossRef] [PubMed]
  12. Wery, M.; Kwapisz, M.; Morillon, A. Noncoding RNAs in gene regulation. Wiley Interdiscip. Rev. Syst. Biol. Med. 2011, 3, 728–738. [Google Scholar] [CrossRef] [PubMed]
  13. Bahreini, F.; Jabbari, P.; Gossing, W.; Aziziyan, F.; Frohme, M.; Rezaei, N. The role of noncoding RNAs in pituitary adenoma. Epigenomics 2021, 13, 1421–1437. [Google Scholar] [CrossRef] [PubMed]
  14. Gu, J.; Huang, W.; Wang, X.; Zhang, J.; Tao, T.; Zheng, Y.; Liu, S.; Yang, J.; Chen, Z.S.; Cai, C.Y.; et al. Hsa-miR-3178/RhoB/PI3K/Akt, a novel signaling pathway regulates ABC transporters to reverse gemcitabine resistance in pancreatic cancer. Mol. Cancer 2022, 21, 112. [Google Scholar] [CrossRef]
  15. Zhang, L.X.; Gao, J.; Long, X.; Zhang, P.F.; Yang, X.; Zhu, S.Q.; Pei, X.; Qiu, B.Q.; Chen, S.W.; Lu, F.; et al. The circular RNA circHMGB2 drives immunosuppression and anti-PD-1 resistance in lung adenocarcinomas and squamous cell carcinomas via the miR-181a-5p/CARM1 axis. Mol. Cancer 2022, 21, 110. [Google Scholar] [CrossRef]
  16. Slack, F.J.; Chinnaiyan, A.M. The Role of Non-coding RNAs in Oncology. Cell 2019, 179, 1033–1055. [Google Scholar] [CrossRef] [PubMed]
  17. Mitchell, P.S.; Parkin, R.K.; Kroh, E.M.; Fritz, B.R.; Wyman, S.K.; Pogosova-Agadjanyan, E.L.; Peterson, A.; Noteboom, J.; O’Briant, K.C.; Allen, A.; et al. Circulating microRNAs as stable blood-based markers for cancer detection. Proc. Natl. Acad. Sci. USA 2008, 105, 10513–10518. [Google Scholar] [CrossRef]
  18. Meng, F.; Yu, W.; Chen, C.; Guo, S.; Tian, X.; Miao, Y.; Ma, L.; Zhang, X.; Yu, Y.; Huang, L.; et al. A Versatile Electrochemical Biosensor for the Detection of Circulating MicroRNA toward Non-Small Cell Lung Cancer Diagnosis. Small 2022, 18, e2200784. [Google Scholar] [CrossRef]
  19. Donati, S.; Aurilia, C.; Palmini, G.; Miglietta, F.; Falsetti, I.; Iantomasi, T.; Brandi, M.L. MicroRNAs as Potential Biomarkers in Pituitary Adenomas. Noncoding RNA 2021, 7, 55. [Google Scholar] [CrossRef]
  20. Tabet, F.; Vickers, K.C.; Cuesta Torres, L.F.; Wiese, C.B.; Shoucri, B.M.; Lambert, G.; Catherinet, C.; Prado-Lourenco, L.; Levin, M.G.; Thacker, S.; et al. HDL-transferred microRNA-223 regulates ICAM-1 expression in endothelial cells. Nat. Commun. 2014, 5, 3292. [Google Scholar] [CrossRef] [Green Version]
  21. Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 2011, 13, 423–433. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, X.; Ba, Y.; Ma, L.; Cai, X.; Yin, Y.; Wang, K.; Guo, J.; Zhang, Y.; Chen, J.; Guo, X.; et al. Characterization of microRNAs in serum: A novel class of biomarkers for diagnosis of cancer and other diseases. Cell Res. 2008, 18, 997–1006. [Google Scholar] [CrossRef] [PubMed]
  23. Németh, K.; Darvasi, O.; Likó, I.; Szücs, N.; Czirják, S.; Reiniger, L.; Szabó, B.; Krokker, L.; Pállinger, É.; Igaz, P.; et al. Comprehensive analysis of circulating microRNAs in plasma of patients with pituitary adenomas. J. Clin. Endocrinol. Metab. 2019, 104, 4151–4168. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, J.; Ma, D.; Liu, H.; Wang, J.; Fan, J.; Li, X. MicroRNA-143 shows tumor suppressive effects through inhibition of oncogenic K-Ras in pituitary tumor. Int. J. Clin. Exp. Pathol. 2017, 10, 10969–10978. [Google Scholar]
  25. Amaral, F.C.; Torres, N.; Saggioro, F.; Neder, L.; Machado, H.R.; Silva, W.A., Jr.; Moreira, A.C.; Castro, M. MicroRNAs differentially expressed in ACTH-secreting pituitary tumors. J. Clin. Endocrinol. Metab. 2009, 94, 320–323. [Google Scholar] [CrossRef]
  26. Belaya, Z.; Khandaeva, P.; Nonn, L.; Nikitin, A.; Solodovnikov, A.; Sitkin, I.; Grigoriev, A.; Pikunov, M.; Lapshina, A.; Rozhinskaya, L.; et al. Circulating Plasma microRNA to Differentiate Cushing’s Disease From Ectopic ACTH Syndrome. Front. Endocrinol. 2020, 11, 331. [Google Scholar] [CrossRef]
  27. Lu, B.; Liu, G.L.; Yu, F.; Li, W.J.; Xiang, X.X.; Xiao, H.Z. MicroRNA-16/VEGFR2/p38/NF-κB signaling pathway regulates cell growth of human pituitary neoplasms. Oncol. Rep. 2018, 39, 1235–1244. [Google Scholar] [CrossRef]
  28. Di Ieva, A.; Butz, H.; Niamah, M.; Rotondo, F.; De Rosa, S.; Sav, A.; Yousef, G.M.; Kovacs, K.; Cusimano, M.D. MicroRNAs as biomarkers in pituitary tumors. Neurosurgery 2014, 75, 181–189, discussion 188–189. [Google Scholar] [CrossRef]
  29. Bottoni, A.; Piccin, D.; Tagliati, F.; Luchin, A.; Zatelli, M.C.; degli Uberti, E.C. miR-15a and miR-16-1 down-regulation in pituitary adenomas. J. Cell Physiol. 2005, 204, 280–285. [Google Scholar] [CrossRef]
  30. He, Z.; Chen, L.; Hu, X.; Tang, J.; He, L.; Hu, J.; Fei, F.; Wang, Q. Next-generation sequencing of microRNAs reveals a unique expression pattern in different types of pituitary adenomas. Endocr. J. 2019, 66, 709–722. [Google Scholar] [CrossRef] [PubMed]
  31. Wang, Z.; Gao, L.; Guo, X.; Feng, C.; Deng, K.; Lian, W.; Xing, B. Identification of microRNAs associated with the aggressiveness of prolactin pituitary tumors using bioinformatic analysis. Oncol. Rep. 2019, 42, 533–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Wang, Z.; Gao, L.; Guo, X.; Feng, C.; Deng, K.; Lian, W.; Xing, B. [Corrigendum] Identification of microRNAs associated with the aggressiveness of prolactin pituitary tumors using bioinformatic analysis. Oncol. Rep. 2021, 46, 130. [Google Scholar] [CrossRef] [PubMed]
  33. Vicchio, T.M.; Aliquò, F.; Ruggeri, R.M.; Ragonese, M.; Giuffrida, G.; Cotta, O.R.; Spagnolo, F.; Torre, M.L.; Alibrandi, A.; Asmundo, A.; et al. MicroRNAs expression in pituitary tumors: Differences related to functional status, pathological features, and clinical behavior. J. Endocrinol. Investig. 2020, 43, 947–958. [Google Scholar] [CrossRef]
  34. Lou, X.; Cai, Y.; Zheng, H.; Zhang, Y. MicroRNA-146b-5p/EPHA7 axis regulates cell invasion, metastasis, proliferation, and temozolomide-induced chemoresistance via regulation of IRAK4/TRAF6/NF-κB signaling pathway in aggressive pituitary adenoma. Histol. Histopathol. 2022, 37, 21–33. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, Y.; Zhao, J.; Zhang, C.; Wang, P.; Huang, C.; Peng, H. MiR-219a-2-3p suppresses cell proliferation and promotes apoptosis by targeting MDM2/p53 in pituitary adenomas cells. Biosci. Biotechnol. Biochem. 2020, 84, 911–918. [Google Scholar] [CrossRef] [PubMed]
  36. Russo, F.; Di Bella, S.; Vannini, F.; Berti, G.; Scoyni, F.; Cook, H.V.; Santos, A.; Nigita, G.; Bonnici, V.; Laganà, A.; et al. miRandola 2017: A curated knowledge base of non-invasive biomarkers. Nucleic. Acids Res. 2018, 46, D354-d359. [Google Scholar] [CrossRef] [PubMed]
  37. Zhao, P.; Cheng, J.; Li, B.; Nie, D.; Li, C.; Gui, S.; Wang, H.; Zhang, Y. Up-regulation of the expressions of MiR-149-5p and MiR-99a-3p in exosome inhibits the progress of pituitary adenomas. Cell Biol. Toxicol. 2021, 37, 633–651. [Google Scholar] [CrossRef]
  38. Katznelson, L.; Laws, E.R., Jr.; Melmed, S.; Molitch, M.E.; Murad, M.H.; Utz, A.; Wass, J.A. Acromegaly: An endocrine society clinical practice guideline. J. Clin. Endocrinol. Metab. 2014, 99, 3933–3951. [Google Scholar] [CrossRef]
  39. Cansu, G.B.; Yılmaz, N.; Yanıkoğlu, A.; Özdem, S.; Yıldırım, A.B.; Süleymanlar, G.; Altunbaş, H.A. Assessment of Diastolic Dysfunction, Arterial Stiffness, and Carotid Intima-Media Thickness in Patients with Acromegaly. Endocr. Pract. 2017, 23, 536–545. [Google Scholar] [CrossRef]
  40. Wassenaar, M.J.; Biermasz, N.R.; Bijsterbosch, J.; Pereira, A.M.; Meulenbelt, I.; Smit, J.W.; Roelfsema, F.; Kroon, H.M.; Romijn, J.A.; Kloppenburg, M. Arthropathy in long-term cured acromegaly is characterised by osteophytes without joint space narrowing: A comparison with generalised osteoarthritis. Ann. Rheum. Dis. 2011, 70, 320–325. [Google Scholar] [CrossRef]
  41. Xiong, Y.; Tang, Y.; Fan, F.; Zeng, Y.; Li, C.; Zhou, G.; Hu, Z.; Zhang, L.; Liu, Z. Exosomal hsa-miR-21-5p derived from growth hormone-secreting pituitary adenoma promotes abnormal bone formation in acromegaly. Transl. Res. 2020, 215, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhao, S.; Li, J.; Feng, J.; Li, Z.; Liu, Q.; Lv, P.; Wang, F.; Gao, H.; Zhang, Y. Identification of Serum miRNA-423-5p Expression Signature in Somatotroph Adenomas. Int. J. Endocrinol. 2019, 2019, 8516858. [Google Scholar] [CrossRef] [PubMed]
  43. Mossakowska, B.J.; Kober, P.; Rusetska, N.; Boresowicz, J.; Maksymowicz, M.; Pękul, M.; Zieliński, G.; Styk, A.; Kunicki, J.; Mandat, T.; et al. Difference in miRNA Expression in Functioning and Silent Corticotroph Pituitary Adenomas Indicates the Role of miRNA in the Regulation of Corticosteroid Receptors. Int. J. Mol. Sci. 2022, 23, 2867. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, Q.; Wang, Y.; Zhou, Y.; Zhang, Q.; Xu, C. Potential biomarkers of miRNA in non-functional pituitary adenomas. World J. Surg. Oncol. 2021, 19, 270. [Google Scholar] [CrossRef]
  45. Chen, Y.; Li, B.; Feng, J.; Fang, Q.; Cheng, J.; Xie, W.; Li, C.; Cheng, S.; Zhang, Y.; Gao, H. JAG1, Regulated by microRNA-424-3p, Involved in Tumorigenesis and Epithelial-Mesenchymal Transition of High Proliferative Potential-Pituitary Adenomas. Front. Oncol. 2020, 10, 567021. [Google Scholar] [CrossRef]
  46. Wei, D.; Yu, Z.; Cheng, Y.; Jiawei, H.; Jian, G.; Hua, G.; Guilan, D. Dysregulated miR-137 and its target EGFR contribute to the progression of pituitary adenomas. Mol. Cell. Endocrinol. 2021, 520, 111083. [Google Scholar] [CrossRef]
  47. Su, W.J.; Wang, J.S.; Ye, M.D.; Chen, W.L.; Liao, C.X. Expression and clinical significance of miR-193a-3p in invasive pituitary adenomas. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 7673–7680. [Google Scholar] [CrossRef]
  48. Cai, F.; Dai, C.; Chen, S.; Wu, Q.; Liu, X.; Hong, Y.; Wang, Z.; Li, L.; Yan, W.; Wang, R.; et al. CXCL12-regulated miR-370-3p functions as a tumor suppressor gene by targeting HMGA2 in nonfunctional pituitary adenomas. Mol. Cell. Endocrinol. 2019, 488, 25–35. [Google Scholar] [CrossRef]
  49. Song, W.; Qian, L.; Jing, G.; Jie, F.; Xiaosong, S.; Chunhui, L.; Yangfang, L.; Guilin, L.; Gao, H.; Yazhuo, Z. Aberrant expression of the sFRP and WIF1 genes in invasive non-functioning pituitary adenomas. Mol. Cell. Endocrinol. 2018, 474, 168–175. [Google Scholar] [CrossRef]
  50. Leone, V.; Langella, C.; D’Angelo, D.; Mussnich, P.; Wierinckx, A.; Terracciano, L.; Raverot, G.; Lachuer, J.; Rotondi, S.; Jaffrain-Rea, M.L.; et al. Mir-23b and miR-130b expression is downregulated in pituitary adenomas. Mol. Cell. Endocrinol. 2014, 390, 1–7. [Google Scholar] [CrossRef]
  51. Gentilin, E.; Tagliati, F.; Filieri, C.; Molè, D.; Minoia, M.; Rosaria Ambrosio, M.; Degli Uberti, E.C.; Zatelli, M.C. miR-26a plays an important role in cell cycle regulation in ACTH-secreting pituitary adenomas by modulating protein kinase Cδ. Endocrinology 2013, 154, 1690–1700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Trivellin, G.; Butz, H.; Delhove, J.; Igreja, S.; Chahal, H.S.; Zivkovic, V.; McKay, T.; Patócs, A.; Grossman, A.B.; Korbonits, M. MicroRNA miR-107 is overexpressed in pituitary adenomas and inhibits the expression of aryl hydrocarbon receptor-interacting protein in vitro. Am. J. Physiol. Endocrinol. Metab. 2012, 303, E708–E719. [Google Scholar] [CrossRef] [PubMed]
  53. Butz, H.; Likó, I.; Czirják, S.; Igaz, P.; Korbonits, M.; Rácz, K.; Patócs, A. MicroRNA profile indicates downregulation of the TGFβ pathway in sporadic non-functioning pituitary adenomas. Pituitary 2011, 14, 112–124. [Google Scholar] [CrossRef]
  54. Butz, H.; Likó, I.; Czirják, S.; Igaz, P.; Khan, M.M.; Zivkovic, V.; Bálint, K.; Korbonits, M.; Rácz, K.; Patócs, A. Down-regulation of Wee1 kinase by a specific subset of microRNA in human sporadic pituitary adenomas. J. Clin. Endocrinol. Metab. 2010, 95, E181–E191. [Google Scholar] [CrossRef] [PubMed]
  55. Qian, Z.R.; Asa, S.L.; Siomi, H.; Siomi, M.C.; Yoshimoto, K.; Yamada, S.; Wang, E.L.; Rahman, M.M.; Inoue, H.; Itakura, M.; et al. Overexpression of HMGA2 relates to reduction of the let-7 and its relationship to clinicopathological features in pituitary adenomas. Mod. Pathol. 2009, 22, 431–441. [Google Scholar] [CrossRef] [PubMed]
  56. Lyu, L.; Li, H.; Chen, C.; Yu, Y.; Wang, L.; Yin, S.; Hu, Y.; Jiang, S.; Ye, F.; Zhou, P. Exosomal miRNA Profiling is a Potential Screening Route for Non-Functional Pituitary Adenoma. Front. Cell Dev. Biol. 2021, 9, 771354. [Google Scholar] [CrossRef]
  57. He, C.; Yang, J.; Ding, J.; Li, S.; Wu, H.; Zhou, F.; Teng, L.; Yang, J. MiR-448 targets BLC2 and inhibits the growth of pituitary adenoma cells. Biochem. Cell Biol. 2020, 98, 511–517. [Google Scholar] [CrossRef]
  58. Yang, Z.; Zhang, T.; Wang, Q.; Gao, H. Overexpression of microRNA-34a Attenuates Proliferation and Induces Apoptosis in Pituitary Adenoma Cells via SOX7. Mol. Ther. Oncolytics. 2018, 10, 40–47. [Google Scholar] [CrossRef]
  59. Fan, X.; Mao, Z.; He, D.; Liao, C.; Jiang, X.; Lei, N.; Hu, B.; Wang, X.; Li, Z.; Lin, Y.; et al. Expression of somatostatin receptor subtype 2 in growth hormone-secreting pituitary adenoma and the regulation of miR-185. J. Endocrinol. Investig. 2015, 38, 1117–1128. [Google Scholar] [CrossRef]
  60. Liao, C.; Chen, W.; Fan, X.; Jiang, X.; Qiu, L.; Chen, C.; Zhu, Y.; Wang, H. MicroRNA-200c inhibits apoptosis in pituitary adenoma cells by targeting the PTEN/Akt signaling pathway. Oncol. Res. 2013, 21, 129–136. [Google Scholar] [CrossRef]
  61. Ulitsky, I.; Bartel, D.P. lincRNAs: Genomics, evolution, and mechanisms. Cell 2013, 154, 26–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Kopp, F.; Mendell, J.T. Functional Classification and Experimental Dissection of Long Noncoding RNAs. Cell 2018, 172, 393–407. [Google Scholar] [CrossRef] [PubMed]
  63. Rinn, J.L.; Chang, H.Y. Genome regulation by long noncoding RNAs. Annu. Rev. Biochem. 2012, 81, 145–166. [Google Scholar] [CrossRef] [PubMed]
  64. Engreitz, J.M.; Haines, J.E.; Perez, E.M.; Munson, G.; Chen, J.; Kane, M.; McDonel, P.E.; Guttman, M.; Lander, E.S. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 2016, 539, 452–455. [Google Scholar] [CrossRef]
  65. Willingham, A.T.; Orth, A.P.; Batalov, S.; Peters, E.C.; Wen, B.G.; Aza-Blanc, P.; Hogenesch, J.B.; Schultz, P.G. A strategy for probing the function of noncoding RNAs finds a repressor of NFAT. Science 2005, 309, 1570–1573. [Google Scholar] [CrossRef] [PubMed]
  66. Carrieri, C.; Cimatti, L.; Biagioli, M.; Beugnet, A.; Zucchelli, S.; Fedele, S.; Pesce, E.; Ferrer, I.; Collavin, L.; Santoro, C.; et al. Long non-coding antisense RNA controls Uchl1 translation through an embedded SINEB2 repeat. Nature 2012, 491, 454–457. [Google Scholar] [CrossRef] [PubMed]
  67. Lee, S.; Kopp, F.; Chang, T.C.; Sataluri, A.; Chen, B.; Sivakumar, S.; Yu, H.; Xie, Y.; Mendell, J.T. Noncoding RNA NORAD Regulates Genomic Stability by Sequestering PUMILIO Proteins. Cell 2016, 164, 69–80. [Google Scholar] [CrossRef]
  68. Tripathi, V.; Ellis, J.D.; Shen, Z.; Song, D.Y.; Pan, Q.; Watt, A.T.; Freier, S.M.; Bennett, C.F.; Sharma, A.; Bubulya, P.A.; et al. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol. Cell 2010, 39, 925–938. [Google Scholar] [CrossRef]
  69. Chen, C.K.; Blanco, M.; Jackson, C.; Aznauryan, E.; Ollikainen, N.; Surka, C.; Chow, A.; Cerase, A.; McDonel, P.; Guttman, M. Xist recruits the X chromosome to the nuclear lamina to enable chromosome-wide silencing. Science 2016, 354, 468–472. [Google Scholar] [CrossRef]
  70. Tay, Y.; Rinn, J.; Pandolfi, P.P. The multilayered complexity of ceRNA crosstalk and competition. Nature 2014, 505, 344–352. [Google Scholar] [CrossRef]
  71. Trimarchi, T.; Bilal, E.; Ntziachristos, P.; Fabbri, G.; Dalla-Favera, R.; Tsirigos, A.; Aifantis, I. Genome-wide mapping and characterization of Notch-regulated long noncoding RNAs in acute leukemia. Cell 2014, 158, 593–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Xiang, J.F.; Yin, Q.F.; Chen, T.; Zhang, Y.; Zhang, X.O.; Wu, Z.; Zhang, S.; Wang, H.B.; Ge, J.; Lu, X.; et al. Human colorectal cancer-specific CCAT1-L lncRNA regulates long-range chromatin interactions at the MYC locus. Cell Res. 2014, 24, 513–531. [Google Scholar] [CrossRef] [PubMed]
  73. Gupta, R.A.; Shah, N.; Wang, K.C.; Kim, J.; Horlings, H.M.; Wong, D.J.; Tsai, M.C.; Hung, T.; Argani, P.; Rinn, J.L.; et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature 2010, 464, 1071–1076. [Google Scholar] [CrossRef] [PubMed]
  74. Bond, A.M.; Vangompel, M.J.; Sametsky, E.A.; Clark, M.F.; Savage, J.C.; Disterhoft, J.F.; Kohtz, J.D. Balanced gene regulation by an embryonic brain ncRNA is critical for adult hippocampal GABA circuitry. Nat. Neurosci. 2009, 12, 1020–1027. [Google Scholar] [CrossRef]
  75. Tsai, M.C.; Manor, O.; Wan, Y.; Mosammaparast, N.; Wang, J.K.; Lan, F.; Shi, Y.; Segal, E.; Chang, H.Y. Long noncoding RNA as modular scaffold of histone modification complexes. Science 2010, 329, 689–693. [Google Scholar] [CrossRef]
  76. Li, W.; Notani, D.; Ma, Q.; Tanasa, B.; Nunez, E.; Chen, A.Y.; Merkurjev, D.; Zhang, J.; Ohgi, K.; Song, X.; et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 2013, 498, 516–520. [Google Scholar] [CrossRef]
  77. Zhang, X.; Ji, S.; Cai, G.; Pan, Z.; Han, R.; Yuan, Y.; Xu, S.; Yang, J.; Hu, X.; Chen, M.; et al. H19 Increases IL-17A/IL-23 Releases via Regulating VDR by Interacting with miR675-5p/miR22-5p in Ankylosing Spondylitis. Mol. Ther. Nucleic. Acids 2020, 19, 393–404. [Google Scholar] [CrossRef]
  78. Wu, Z.; Zheng, Y.; Xie, W.; Li, Q.; Zhang, Y.; Ren, B.; Cai, L.; Cheng, Y.; Tang, H.; Su, Z.; et al. The long noncoding RNA-H19/miRNA-93a/ATG7 axis regulates the sensitivity of pituitary adenomas to dopamine agonists. Mol. Cell. Endocrinol. 2020, 518, 111033. [Google Scholar] [CrossRef]
  79. Zhu, H.B.; Li, B.; Guo, J.; Miao, Y.Z.; Shen, Y.T.; Zhang, Y.Z.; Zhao, P.; Li, C.Z. LncRNA MEG8 promotes TNF-α expression by sponging miR-454-3p in bone-invasive pituitary adenomas. Aging 2021, 13, 14342–14354. [Google Scholar] [CrossRef]
  80. Du, Q.; Hoover, A.R.; Dozmorov, I.; Raj, P.; Khan, S.; Molina, E.; Chang, T.C.; de la Morena, M.T.; Cleaver, O.B.; Mendell, J.T.; et al. MIR205HG Is a Long Noncoding RNA that Regulates Growth Hormone and Prolactin Production in the Anterior Pituitary. Dev. Cell 2019, 49, 618–631.e615. [Google Scholar] [CrossRef]
  81. Cesana, M.; Cacchiarelli, D.; Legnini, I.; Santini, T.; Sthandier, O.; Chinappi, M.; Tramontano, A.; Bozzoni, I. A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell 2011, 147, 358–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Dey, B.K.; Pfeifer, K.; Dutta, A. The H19 long noncoding RNA gives rise to microRNAs miR-675-3p and miR-675-5p to promote skeletal muscle differentiation and regeneration. Genes Dev. 2014, 28, 491–501. [Google Scholar] [CrossRef] [PubMed]
  83. Li, J.; Li, C.; Wang, J.; Song, G.; Zhao, Z.; Wang, H.; Wang, W.; Li, H.; Li, Z.; Miao, Y.; et al. Genome-wide analysis of differentially expressed lncRNAs and mRNAs in primary gonadotrophin adenomas by RNA-seq. Oncotarget 2017, 8, 4585–4606. [Google Scholar] [CrossRef] [PubMed]
  84. Xing, W.; Qi, Z.; Huang, C.; Zhang, N.; Zhang, W.; Li, Y.; Qiu, M.; Fang, Q.; Hui, G. Genome-wide identification of lncRNAs and mRNAs differentially expressed in non-functioning pituitary adenoma and construction of an lncRNA-mRNA co-expression network. Biol. Open 2019, 8, bio037127. [Google Scholar] [CrossRef]
  85. Peng, C.; Wang, S.; Yu, J.; Deng, X.; Ye, H.; Chen, Z.; Yao, H.; Cai, H.; Li, Y.; Yuan, Y. lncRNA-mRNA Expression Patterns in Invasive Pituitary Adenomas: A Microarray Analysis. Biomed. Res. Int. 2022, 2022, 1380485. [Google Scholar] [CrossRef]
  86. Zhu, H.; Guo, J.; Shen, Y.; Dong, W.; Gao, H.; Miao, Y.; Li, C.; Zhang, Y. Functions and Mechanisms of Tumor Necrosis Factor-α and Noncoding RNAs in Bone-Invasive Pituitary Adenomas. Clin. Cancer Res. 2018, 24, 5757–5766. [Google Scholar] [CrossRef]
  87. Byun, H.M.; Wong, H.L.; Birnstein, E.A.; Wolff, E.M.; Liang, G.; Yang, A.S. Examination of IGF2 and H19 loss of imprinting in bladder cancer. Cancer Res. 2007, 67, 10753–10758. [Google Scholar] [CrossRef]
  88. Yoshimizu, T.; Miroglio, A.; Ripoche, M.A.; Gabory, A.; Vernucci, M.; Riccio, A.; Colnot, S.; Godard, C.; Terris, B.; Jammes, H.; et al. The H19 locus acts in vivo as a tumor suppressor. Proc. Natl. Acad. Sci. USA 2008, 105, 12417–12422. [Google Scholar] [CrossRef]
  89. Jiang, X.; Yan, Y.; Hu, M.; Chen, X.; Wang, Y.; Dai, Y.; Wu, D.; Wang, Y.; Zhuang, Z.; Xia, H. Increased level of H19 long noncoding RNA promotes invasion, angiogenesis, and stemness of glioblastoma cells. J. Neurosurg. 2016, 124, 129–136. [Google Scholar] [CrossRef]
  90. Lu, T.; Yu, C.; Ni, H.; Liang, W.; Yan, H.; Jin, W. Expression of the long non-coding RNA H19 and MALAT-1 in growth hormone-secreting pituitary adenomas and its relationship to tumor behavior. Int. J. Dev. Neurosci. 2018, 67, 46–50. [Google Scholar] [CrossRef]
  91. Liang, W.; Zou, Y.; Qin, F.; Chen, J.; Xu, J.; Huang, S.; Chen, J.; Dai, S. sTLR4/MD-2 complex inhibits colorectal cancer migration and invasiveness in vitro and in vivo by lncRNA H19 down-regulation. Acta Biochim. Biophys. Sin. 2017, 49, 1035–1041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Wu, Z.R.; Yan, L.; Liu, Y.T.; Cao, L.; Guo, Y.H.; Zhang, Y.; Yao, H.; Cai, L.; Shang, H.B.; Rui, W.W.; et al. Inhibition of mTORC1 by lncRNA H19 via disrupting 4E-BP1/Raptor interaction in pituitary tumours. Nat. Commun. 2018, 9, 4624. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, Y.; Liu, Y.T.; Tang, H.; Xie, W.Q.; Yao, H.; Gu, W.T.; Zheng, Y.Z.; Shang, H.B.; Wang, Y.; Wei, Y.X.; et al. Exosome-Transmitted lncRNA H19 Inhibits the Growth of Pituitary Adenoma. J. Clin. Endocrinol. Metab. 2019, 104, 6345–6356. [Google Scholar] [CrossRef] [PubMed]
  94. Cheng, S.; Guo, J.; Wang, D.; Fang, Q.; Liu, Y.; Xie, W.; Zhang, Y.; Li, C. A Novel Three-LncRNA Signature Predicting Tumor Recurrence in Nonfunctioning Pituitary Adenomas. Front. Genet. 2021, 12, 754503. [Google Scholar] [CrossRef] [PubMed]
  95. Lu, G.; Duan, J.; Zhou, D. Long-noncoding RNA IFNG-AS1 exerts oncogenic properties by interacting with epithelial splicing regulatory protein 2 (ESRP2) in pituitary adenomas. Pathol. Res. Pract. 2018, 214, 2054–2061. [Google Scholar] [CrossRef]
  96. Fu, D.; Zhang, Y.; Cui, H. Long noncoding RNA CCAT2 is activated by E2F1 and exerts oncogenic properties by interacting with PTTG1 in pituitary adenomas. Am. J. Cancer Res. 2018, 8, 245–255. [Google Scholar]
  97. Li, Z.; Li, C.; Liu, C.; Yu, S.; Zhang, Y. Expression of the long non-coding RNAs MEG3, HOTAIR, and MALAT-1 in non-functioning pituitary adenomas and their relationship to tumor behavior. Pituitary 2015, 18, 42–47. [Google Scholar] [CrossRef]
  98. Cheng, S.; Guo, J.; Zhang, Y.; Li, Z.; Li, C. Identification of a multidimensional transcriptome signature predicting tumor regrowth of clinically non-functioning pituitary adenoma. Int. J. Oncol. 2020, 57, 804–812. [Google Scholar] [CrossRef]
  99. Zhang, Y.; Tan, Y.; Wang, H.; Xu, M.; Xu, L. Long Non-Coding RNA Plasmacytoma Variant Translocation 1 (PVT1) Enhances Proliferation, Migration, and Epithelial-Mesenchymal Transition (EMT) of Pituitary Adenoma Cells by Activating β-Catenin, c-Myc, and Cyclin D1 Expression. Med. Sci. Monit. 2019, 25, 7652–7659. [Google Scholar] [CrossRef]
  100. Liu, C.X.; Chen, L.L. Circular RNAs: Characterization, cellular roles, and applications. Cell 2022, 185, 2016–2034. [Google Scholar] [CrossRef]
  101. Zhang, W.; Chen, S.; Du, Q.; Bian, P.; Chen, Y.; Liu, Z.; Zheng, J.; Sai, K.; Mou, Y.; Chen, Z.; et al. CircVPS13C promotes pituitary adenoma growth by decreasing the stability of IFITM1 mRNA via interacting with RRBP1. Oncogene 2022, 41, 1550–1562. [Google Scholar] [CrossRef] [PubMed]
  102. Ali, M.K.; Schimmel, K.; Zhao, L.; Chen, C.K.; Dua, K.; Nicolls, M.R.; Spiekerkoetter, E. The role of circular RNAs in pulmonary hypertension. Eur. Respir. J. 2022, 9, 2200012. [Google Scholar] [CrossRef] [PubMed]
  103. Tang, B.; Yang, Y.; Kang, M.; Wang, Y.; Wang, Y.; Bi, Y.; He, S.; Shimamoto, F. m(6)A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol. Cancer 2020, 19, 3. [Google Scholar] [CrossRef] [PubMed]
  104. Huang, A.; Zheng, H.; Wu, Z.; Chen, M.; Huang, Y. Circular RNA-protein interactions: Functions, mechanisms, and identification. Theranostics 2020, 10, 3503–3517. [Google Scholar] [CrossRef] [PubMed]
  105. Vo, J.N.; Cieslik, M.; Zhang, Y.; Shukla, S.; Xiao, L.; Zhang, Y.; Wu, Y.M.; Dhanasekaran, S.M.; Engelke, C.G.; Cao, X.; et al. The Landscape of Circular RNA in Cancer. Cell 2019, 176, 869–881.e813. [Google Scholar] [CrossRef]
  106. Li, Y.; Zheng, Q.; Bao, C.; Li, S.; Guo, W.; Zhao, J.; Chen, D.; Gu, J.; He, X.; Huang, S. Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 2015, 25, 981–984. [Google Scholar] [CrossRef]
  107. Yu, J.; Xu, Q.G.; Wang, Z.G.; Yang, Y.; Zhang, L.; Ma, J.Z.; Sun, S.H.; Yang, F.; Zhou, W.P. Circular RNA cSMARCA5 inhibits growth and metastasis in hepatocellular carcinoma. J. Hepatol. 2018, 68, 1214–1227. [Google Scholar] [CrossRef]
  108. Jahani, S.; Nazeri, E.; Majidzadeh, A.K.; Jahani, M.; Esmaeili, R. Circular RNA; a new biomarker for breast cancer: A systematic review. J. Cell. Physiol. 2020, 235, 5501–5510. [Google Scholar] [CrossRef]
  109. Guo, J.; Wang, Z.; Miao, Y.; Shen, Y.; Li, M.; Gong, L.; Wang, H.; He, Y.; Gao, H.; Liu, Q.; et al. A two-circRNA signature predicts tumour recurrence in clinical non-functioning pituitary adenoma. Oncol. Rep. 2019, 41, 113–124. [Google Scholar] [CrossRef]
  110. Du, Q.; Zhang, W.; Feng, Q.; Hao, B.; Cheng, C.; Cheng, Y.; Li, Y.; Fan, X.; Chen, Z. Comprehensive circular RNA profiling reveals that hsa_circ_0001368 is involved in growth hormone-secreting pituitary adenoma development. Brain Res. Bull. 2020, 161, 65–77. [Google Scholar] [CrossRef]
  111. Hu, Y.; Zhang, N.; Zhang, S.; Zhou, P.; Lv, L.; Richard, S.A.; Ma, W.; Chen, C.; Wang, X.; Huang, S.; et al. Differential circular RNA expression profiles of invasive and non-invasive non-functioning pituitary adenomas: A microarray analysis. Medicine 2019, 98, e16148. [Google Scholar] [CrossRef] [PubMed]
  112. Wang, J.; Wang, D.; Wan, D.; Ma, Q.; Liu, Q.; Li, J.; Li, Z.; Gao, Y.; Jiang, G.; Ma, L.; et al. Circular RNA In Invasive and Recurrent Clinical Nonfunctioning Pituitary Adenomas: Expression Profiles and Bioinformatic Analysis. World Neurosurg. 2018, 117, e371–e386. [Google Scholar] [CrossRef] [PubMed]
  113. Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, R.; Yang, F.; Fan, H.; Wang, H.; Wang, Q.; Yang, J.; Song, T. Long non-coding RNA TUG1/microRNA-187-3p/TESC axis modulates progression of pituitary adenoma via regulating the NF-κB signaling pathway. Cell Death Dis. 2021, 12, 524. [Google Scholar] [CrossRef] [PubMed]
  115. Wu, H.; Zhou, S.; Zheng, Y.; Pan, Z.; Chen, Y.; Wang, X. LncRNA BBOX1-AS1 promotes pituitary adenoma progression via sponging miR-361-3p/E2F1 axis. Anticancer Drugs 2022, 33, 652–662. [Google Scholar] [CrossRef] [PubMed]
  116. Huang, T.; Cai, M.; Chen, C.; Ling, C.; Zhang, B.; Zheng, W.; Luo, L. LINC01116 boosts the progression of pituitary adenoma via regulating miR-744-5p/HOXB8 pathway. Mol. Cell. Endocrinol. 2021, 536, 111350. [Google Scholar] [CrossRef]
  117. Wang, X.; Li, X.; Wang, Z. lncRNA MEG3 inhibits pituitary tumor development by participating in cell proliferation, apoptosis and EMT processes. Oncol. Rep. 2021, 45, 1–11. [Google Scholar] [CrossRef]
  118. Zhu, D.; Xiao, Z.; Wang, Z.; Hu, B.; Duan, C.; Zhu, Z.; Gao, N.; Zhu, Y.; Wang, H. MEG3/MIR-376B-3P/HMGA2 axis is involved in pituitary tumor invasiveness. J. Neurosurg. 2020, 134, 499–511. [Google Scholar] [CrossRef]
  119. Qiu, P.; Bi, J.; Liu, J.; Lai, C.; Li, X. Long non-coding RNA LINC01004 promotes malignant behaviors of pituitary adenoma via miR-323a-3p/136-5p/RCN2 axis. Pathol. Res. Pract. 2022, 234, 153884. [Google Scholar] [CrossRef]
  120. Li, Z.; Ren, R.; Wang, L.; Wang, Z.; Zong, X.; Sun, P.; Zhu, C.; Guo, M.; Guo, G.; Hu, G.; et al. lncRNA KCNQ1OT1 Promotes EMT, Angiogenesis, and Stemness of Pituitary Adenoma by Upregulation of RAB11A. J. Oncol. 2022, 2022, 4474476. [Google Scholar] [CrossRef]
  121. Li, J.; Qian, Y.; Zhang, C.; Wang, W.; Qiao, Y.; Song, H.; Li, L.; Guo, J.; Lu, D.; Deng, X. LncRNA LINC00473 is involved in the progression of invasive pituitary adenoma by upregulating KMT5A via ceRNA-mediated miR-502-3p evasion. Cell Death Dis. 2021, 12, 580. [Google Scholar] [CrossRef] [PubMed]
  122. Mao, D.; Jie, Y.; Lv, Y. LncRNA SNHG6 Induces Epithelial-Mesenchymal Transition of Pituitary Adenoma Via Suppressing MiR-944. Cancer Biother. Radiopharm. 2022, 37, 246–255. [Google Scholar] [CrossRef] [PubMed]
  123. Du, Q.; Hu, B.; Feng, Y.; Wang, Z.; Wang, X.; Zhu, D.; Zhu, Y.; Jiang, X.; Wang, H. circOMA1-Mediated miR-145-5p Suppresses Tumor Growth of Nonfunctioning Pituitary Adenomas by Targeting TPT1. J. Clin. Endocrinol. Metab. 2019, 104, 2419–2434. [Google Scholar] [CrossRef]
  124. Cheng, J.; Nie, D.; Li, B.; Gui, S.; Li, C.; Zhang, Y.; Zhao, P. CircNFIX promotes progression of pituitary adenoma via CCNB1 by sponging miR-34a -5p. Mol. Cell. Endocrinol. 2021, 525, 111140. [Google Scholar] [CrossRef] [PubMed]
  125. Zhao, P.; Cheng, J.; Li, B.; Nie, D.; Wang, H.; Li, C.; Gui, S.; Zhang, Y. LncRNA PCAT6 regulates the progression of pituitary adenomas by regulating the miR-139-3p/BRD4 axis. Cancer Cell Int. 2021, 21, 14. [Google Scholar] [CrossRef] [PubMed]
  126. Yue, X.; Dong, C.; Ye, Z.; Zhu, L.; Zhang, X.; Wang, X.; Mo, F.; Li, Z.; Pan, B. LncRNA SNHG7 sponges miR-449a to promote pituitary adenomas progression. Metab. Brain Dis. 2021, 36, 123–132. [Google Scholar] [CrossRef] [PubMed]
  127. Yin, H.; Zheng, X.; Tang, X.; Zang, Z.; Li, B.; He, S.; Shen, R.; Yang, H.; Li, S. Potential biomarkers and lncRNA-mRNA regulatory networks in invasive growth hormone-secreting pituitary adenomas. J. Endocrinol. Investig. 2021, 44, 1947–1959. [Google Scholar] [CrossRef]
  128. Kang, Y.; He, W.; Tulley, S.; Gupta, G.P.; Serganova, I.; Chen, C.R.; Manova-Todorova, K.; Blasberg, R.; Gerald, W.L.; Massagué, J. Breast cancer bone metastasis mediated by the Smad tumor suppressor pathway. Proc. Natl. Acad. Sci. USA 2005, 102, 13909–13914. [Google Scholar] [CrossRef]
  129. Inoue, J.; Inazawa, J. Cancer-associated miRNAs and their therapeutic potential. J. Hum. Genet. 2021, 66, 937–945. [Google Scholar] [CrossRef]
Figure 1. The potential mechanisms of non-coding RNAs in pituitary adenoma. (A) miRNA inhibits protein synthesis by binding to the 3’-UTR of mRNA. (BD) miR-142-3p, miR-16, miR-146-5p, and exosome-derived miR-146-5p are involved in the pathogenesis of pituitary adenoma (PA). (E) LncRNA regulates gene activation or suppression by increasing or decreasing transcription via the transcription factor, respectively; antisense lncRNA upregulates the sense mRNA translation process. (FH) MIR205GH, SNHG24, H19, and exosome-derived H19 are involved in the pathogenesis of PA. (IJ) CircVPS13C and hsa_circ_0001368 are involved in the pathogenesis of PA. (K) CircRNA and lncRNA increase the translation process and protein production via sponging miRNA. (L) LncRNA TUG1 modulates PA progression by regulating the TESC–NF-kB pathway via sponging miR-187-3p. (M) CircNFIX promotes PA progression by regulating CCNB1 via sponging miR-34a-5p.
Figure 1. The potential mechanisms of non-coding RNAs in pituitary adenoma. (A) miRNA inhibits protein synthesis by binding to the 3’-UTR of mRNA. (BD) miR-142-3p, miR-16, miR-146-5p, and exosome-derived miR-146-5p are involved in the pathogenesis of pituitary adenoma (PA). (E) LncRNA regulates gene activation or suppression by increasing or decreasing transcription via the transcription factor, respectively; antisense lncRNA upregulates the sense mRNA translation process. (FH) MIR205GH, SNHG24, H19, and exosome-derived H19 are involved in the pathogenesis of PA. (IJ) CircVPS13C and hsa_circ_0001368 are involved in the pathogenesis of PA. (K) CircRNA and lncRNA increase the translation process and protein production via sponging miRNA. (L) LncRNA TUG1 modulates PA progression by regulating the TESC–NF-kB pathway via sponging miR-187-3p. (M) CircNFIX promotes PA progression by regulating CCNB1 via sponging miR-34a-5p.
Cells 11 02920 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, W.; Cao, L.; Jia, Y.; Xiao, Y.; Zhang, X.; Gui, S. Emerging Roles of miRNA, lncRNA, circRNA, and Their Cross-Talk in Pituitary Adenoma. Cells 2022, 11, 2920. https://doi.org/10.3390/cells11182920

AMA Style

Wu W, Cao L, Jia Y, Xiao Y, Zhang X, Gui S. Emerging Roles of miRNA, lncRNA, circRNA, and Their Cross-Talk in Pituitary Adenoma. Cells. 2022; 11(18):2920. https://doi.org/10.3390/cells11182920

Chicago/Turabian Style

Wu, Wentao, Lei Cao, Yanfei Jia, Youchao Xiao, Xu Zhang, and Songbai Gui. 2022. "Emerging Roles of miRNA, lncRNA, circRNA, and Their Cross-Talk in Pituitary Adenoma" Cells 11, no. 18: 2920. https://doi.org/10.3390/cells11182920

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