Next Article in Journal
Platelet-Rich Plasma Injections in Chronic Lateral Ankle Instability: A Case Series
Previous Article in Journal
The Essence of Lipoproteins in Cardiovascular Health and Diseases Treated by Photodynamic Therapy
Previous Article in Special Issue
Gastrointestinal Disease: New Diagnostic and Therapeutic Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Expression of Selected miRNAs in Undifferentiated Carcinoma with Osteoclast-like Giant Cells (UCOGC) of the Pancreas: Comparison with Poorly Differentiated Pancreatic Ductal Adenocarcinoma

1
Department of Pathology, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic
2
Department of Surgery, 3rd Faculty of Medicine, Charles University, University Hospital Královské Vinohrady, 100 00 Prague, Czech Republic
3
Department of Medical Biophysics and Medical Informatics, 3rd Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic
4
Institute of Pathology, 1st Faculty of Medicine, Charles University, General University Hospital, 100 00 Prague, Czech Republic
5
Clinical and Transplant Pathology Centre, Institute for Clinical and Experimental Medicine, 140 00 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(5), 962; https://doi.org/10.3390/biomedicines12050962
Submission received: 23 February 2024 / Revised: 18 April 2024 / Accepted: 23 April 2024 / Published: 26 April 2024

Abstract

:
Undifferentiated carcinoma with osteoclast-like giant cells (UCOGC) of the pancreas represents a rare subtype of pancreatic ductal adenocarcinoma (PDAC). Despite a distinct morphology and specific clinical behavior, UCOGCs exhibit unexpected similarities in regard to DNA mutational profiles with conventional PDAC. Treating pancreatic ductal adenocarcinoma is particularly challenging, with limited prospects for cure. As with many other malignant neoplasms, the exploration of microRNAs (miRNAs, miRs) in regulating the biological characteristics of pancreatic cancer is undergoing extensive investigation to enhance tumor diagnostics and unveil the therapeutic possibilities. Herein, we evaluated the expression of miR-21, -96, -148a, -155, -196a, -210, and -217 in UCOGCs and poorly differentiated (grade 3, G3) PDACs. The expression of miR-21, miR-155, and miR-210 in both UCOGCs and G3 PDACs was significantly upregulated compared to the levels in normal tissue, while the levels of miR-148a and miR-217 were downregulated. We did not find any significant differences between cancerous and normal tissues for the expression of miR-96 and miR-196a in G3 PDACs, whereas miR-196a was slightly, but significantly, downregulated in UCOGCs. On the other hand, we have not observed significant differences in the expression of the majority of miRNAs between UCOGC and G3 PDAC, with the exception of miR-155. UCOGC samples demonstrated lower mean levels of miR-155 in comparison with those in G3 PDACs.

1. Introduction

In the latest WHO classification, undifferentiated carcinoma with osteoclast-like giant cells (UCOGC) of the pancreas is considered a rare subtype of pancreatic ductal adenocarcinoma (PDAC) [1], accounting for approximately 0.4% of pancreatic carcinomas [2]. The initial description of UCOGC was reported in 1954 [3]. Fourteen years later, Rosai labeled the lesion “carcinoma simulating giant cell tumor of bone” [4].
UCOGC exhibits a distinct morphology and typically carries a grim prognosis. The tumor comprises three main cell types: (1) neoplastic cytokeratin-positive mononuclear cells exhibiting cytonuclear atypia, (2) non-neoplastic mononuclear macrophages, and (3) non-neoplastic osteoclast-like multinucleated giant cells, which vary in quantity and are often accompanied by hemorrhage and necrosis. Despite their morphological differences, the DNA mutational profiles of UCOGCs surprisingly mirror those of conventional PDAC, frequently featuring KRAS activation and the inactivation of SMAD4, TP53, and CDKN2A [5,6,7,8]. Many neoplastic pathways in pancreatic cancer are regulated by microRNAs. However, little is known about miRNA expression profiles in UCOGCs and eventual differences in comparison with PDACs.
MicroRNAs (miRNAs) represent a group of single-stranded RNA molecules ranging from 15 to 27 nucleotides long. These compact, non-coding RNAs play pivotal roles in regulating various cellular processes within both healthy and cancerous cells. Functionally, miRNAs can bind to messenger RNAs (mRNAs), thereby modulating the translation process. Consequently, this interaction can lead to the downregulation of specific protein synthesis, serving as a crucial regulatory step in gene expression [9,10,11]. This post-transcriptional mechanism ensures precise control over essential cellular processes, including proliferation, migration, and apoptosis, which are fundamental to cancer growth, dissemination, and resistance to therapy. MiRNAs play a crucial role in the intricate interplay between cancer cells and other components within the tumor microenvironment. Given the challenging prognosis of PDAC, despite available treatments, there is considerable interest in studying the regulatory functions orchestrated by miRNAs in this malignancy. Such investigations aim to identify miRNAs as potential targets for innovative therapeutic strategies [12,13,14,15,16,17,18,19]. MiRNAs have been proposed to function as both oncogenes and tumor suppressors, and alterations in miRNA expression have been observed in neoplastic precursor lesions [20,21]. The dysregulation of miRNA expression profiles is a common feature in various cancer types, including PDAC [12,13,22]. Zhang et al. showed substantial variations in the relative expression levels of miRNAs across individual cases of pancreatic ductal adenocarcinoma, with changes spanning six orders of magnitude, ranging from 0.01 to 10,000 [23]. Aberrantly expressed microRNAs, documented across numerous studies, have been suggested as potential predictive factors for disease progression, chemotherapy response, and patient survival [24,25,26,27].
In this study, the expression of seven selected miRNAs (miR-21, miR-96 miR-148a, miR-155, miR-196a, miR-210, and miR-217), described to be deregulated in PDACs [28,29,30], was analyzed in UCOGC and poorly differentiated (G3) PDAC. In greater detail, these seven miRNAs were chosen due to their substantial involvement in pancreatic ductal adenocarcinoma, impacting tumor morphology, progression, and clinical prognosis. Notably, among the selected miRNAs, miR-21, miR-155, and miR-217 have been specifically associated with differential expression patterns correlated with tumor advancement [25,27,28]. MiR-21, miR-155, miR-196a, and miR-210 were chosen because they have been suggested as potential diagnostic and prognostic markers in previous studies [24,28,31]. Additionally, miR-96 and miR-148a were included in the selection due to their established roles as tumor suppressors in PDAC [32,33]. The aim of this work was to characterize the expression of these miRNAs in UCOGC and to identify possible differences in miRNAs expression between UCOGC and G3 PDAC.

2. Materials and Methods

2.1. Patients and Samples

A cohort of 10 patients (8 male, 2 female, aged 50–76 years) who underwent surgical treatment for pancreatic UCOGC and were histologically diagnosed in three participating institutions from 2003 to 2019 were included in the study. From those, nine were primary tumor resection specimens, and of those, four patients displayed regional lymph node metastasis at time of surgery. One case was a specimen of lymph node metastasis obtained from explorative laparotomy, without pancreas resection. There were two cases of UCOGC associated with conventional PDAC and one case associated with intraductal papillary mucinous neoplasm (IPMN); the remaining seven patients exhibited pure UCOGCs. The tumor size at surgery varied from 5mm to 140mm; two cases were pT1, two cases were pT2, five cases were pT3, and one case was pTX stage, respectively. Due to the rarity of UCOGC, we utilized cases from our previously published studies [8,34].
The second comparative group consisted of 12 patients (8 male, 4 female, age 53–83 years) with surgically resected, poorly differentiated (G3) PDAC diagnosed between 2008–2015. From these 12 cancers, 11 were pT3, and 1 was pT4 stage, respectively. Ten patients with G3 PDAC displayed regional lymph node metastases at surgery. Comparative group PDAC cases were selected randomly among patients with high grade and advanced disease.
Representative tissue blocks of formalin-fixed and paraffin embedded (FFPE) tumor tissue were selected for miRNA extraction from the archives of the departments of pathology in all three involved institutions.

2.2. MicroRNA Isolation and Reverse Transcription

For miRNA extraction, one to three 6 µm thick unstained paraffin-embedded tissue sections were obtained and processed using the miRNeasy FFPE kit from Qiagen (Venlo, The Netherlands). Following extraction, two microliters of the isolated RNA were utilized for assessing RNA quantity and purity. This analysis involved measuring the optical density at 260 and 280 nm using the multi-detection microplate reader Synergy HT from BioTek (Winooski, VT, USA), equipped with the Take3 micro-volume plate. RNA integrity was evaluated through denaturing agarose gel electrophoresis, and GeneTools 3.08 software from SynGene (Bengaluru, India) was employed for analysis. Subsequently, reverse transcription was conducted using RevertAid Reverse Transcriptase from Thermo Fisher Scientific (Waltham, MA, USA), following the manufacturer’s protocol. The reaction mixture, containing 1 μg of DNA-free RNA and a mixture of eight stem–loop primers (20 pmol of each stem–loop primer), was utilized for miRNA reverse transcription. These primers were designed using MiRNA Primer Designer software (version 1.0), generously provided by Dr. Fuliang Xie from East Carolina University, USA. The sequences of the stem–loop primers for the pancreatic miRNAs under investigation and the internal control are detailed in Table 1. Additionally, artificial spike RNA (miR-39 from C. elegans, 5 × 108 copies) was included in the reaction as an internal control.

2.3. Real-Time qPCR

cDNA samples were amplified in duplicate, using the Applied Biosystems 7500 Fast real-time PCR system and Hot FirePol EvaGreen qPCR Mix Plus (Solis BioDyne, Tartu, Estonia), as described in our previous work [17], primers listed in Table 2. The ΔΔCt method was applied to measure the miRNA expression values of interest [35].

2.4. Statistical Analysis

All statistical analyses were conducted using S.A.S. release 9.4 (SAS Inc., Cary, NC, USA) and SPSS 25 (IBM Corporation, Armonk, NY, USA), unless otherwise specified. The expression levels of miRNAs in neoplastic and normal cells were compared using a Mann–Whitney test. All hypotheses were tested as two-sided. A significance level of alpha = 0.05 was chosen; thus, p-values less than 0.05 were deemed statistically significant.

3. Results

Abnormal miRNA Expression in Pancreatic Cancers
We investigated the expression of seven miRNAs isolated from FFPE samples of pancreatic adenocarcinomas from 12 patients, as well as 10 samples of UCOGC. The following microRNAs were selected: miR-21, which promotes cell proliferation and may accelerate tumorigenesis [14,36,37]; miR-155, which interacts with tumor protein p53 inducible nuclear protein 1 (TP53INP1) and transforming growth factor β (TGF-β) [31,38,39]; miR-96 and miR-217, which may act as a tumor suppressors, inhibiting the KRAS-signaling pathway [40,41]; as well as miR-148a, miR-196a, and miR-210, which are frequently included in experimental panels for pancreatic carcinoma diagnosis [23,28,42,43,44].
In comparison with normal pancreatic tissue, miR-21 demonstrated increased expression levels, with great variations, from 7-fold up to 44.79-fold in UCOGC samples, and from 3.76-fold up to 53.51-fold in PDAC (Table 3, Table 4 and Table 5; p < 0.05). The miR-155 was also overexpressed in UCOGC (from 1.42-fold to 16.63-fold; p < 0.05). This miRNA varied more significantly in PDACs, from 0.86-fold to 232.36-fold (Table 3, Table 4 and Table 5; p < 0.05). Expression of miR-210 was also significantly upregulated in both types of tumors: up to 90.7-fold in UCOGC, as well as 65.34-fold in PDAC (Table 3, Table 4 and Table 5; p < 0.05). However, we did not detect significant differences between tumor and normal tissue for the expression of miR-96 and miR-196a in G3 PDACs (Table 5, p > 0.05) and miR-96 in UCOGC, while miR-196a was slightly but significantly downregulated in the latter (p < 0.05, Table 5). The expression of miR-148a in the tumor samples was downregulated in both UCOGC and PDAC (Table 3, Table 4 and Table 5; p < 0.05). Specifically, miR-217 displayed a substantial decrease, up to one hundred times in both types of tumors (p < 0.05, Table 3, Table 4 and Table 5).
We have not observed significant differences in the expression of the majority of miRNAs between UCOGC and G3 PDAC (p > 0.05. Table 5), with the exception of miR-155. UCOGC demonstrated lower mean levels of miR-155 in comparison with PDACs (p < 0.05. Table 5). On the other hand, miR-196a was slightly, but significantly, downregulated in UCOGCs (p < 0.05. Table 5), whereas significant differences between PDACs and normal tissue samples were not detected (p > 0.05. Table 5, Figure 1).

4. Discussion

UCOGC is a well-defined WHO subtype of pancreatic cancer that may accompany conventional PDAC or mucinous pancreatic tumors, such as IPMN or mucinous cystic neoplasm (MCN) [2]. UCOGC has been thought to have a poor prognosis [45,46,47], although better than that of conventional PDAC and undifferentiated carcinoma without giant cells [2]. However, there have been descriptions of individual UCOGC cases with favorable clinical courses, including a small subset of “unexpected long survivors” [2,48,49]. To date, programmed death ligand 1 (PD-L1), frequently expressed in UCOGC and rarely in PDAC [50], is the only robust prognostic marker in UCOGC, whereas the PD-L1-negative UCOGC is characterized by long survival and favorable outcome [5,34]. There are two successful clinical trials using anti-PD-L1 treatment in UCOGC described in the literature [51,52].
Of note, there is evidence pointing towards a worse prognosis of UCOGC associated with PDAC compared to pure UCOGC, which has been explained by a higher degree of the epithelial–mesenchymal transitional phenotype [53].
However, little is known about the genomic background and biological rationale for the peculiar morphology and immunohistochemical phenotype of UCOGC. A recent large genomic landscape study analyzing 2568 PDAC cases revealed the canonical mutations of KRAS (85.1%), TP53 (69.1%), CDKN2A (35.4%), and SMAD4 (19.4%) to be the most common. Additionally, mutations in CDKN2B, ARID1A, STK11, MYC, KDM6A, and DNMT3A were among top ten DNA abnormalities [54]. A review by Bazzichetto presented a similar distribution of driver mutations in UCOGC for KRAS (70–100%), TP53 (50–100%), SMAD4 (10–50%), and CDKN2A (25%) as non-canonical possibly oncogenic mutations. SERPINA3, MAGEB4, GLI3, MEGF8, TTN, and BRCA2 have been described in a small number of UCOGCs [6]. Our group recently reported 10 KRAS, 9 TP53, 4 CDKN2A, and 1 SMAD4 mutations when analyzing 13 UCOGC cases [9]. Bergmann et al. identified druggable genomic targets (L1CAM, cyclooxygenase 2, epidermal growth factor receptor) in a high proportion of undifferentiated pancreatic carcinomas, including UCOGC [55]. To the best of our knowledge, except for a single case analysis [56], there are no other studies comparing the spectrum of RNA expression in UCOGC compared to PDAC.
Many cancer pathways in PDACs are regulated by microRNAs. However, little is known about miRNA expression profiles in UCOGC. Aberrant expression levels of miRNAs have been extensively documented in various malignant tumors, playing a crucial role in the formation, development, and prognosis of pancreatic cancers. These miRNAs can generally be categorized into two major groups: oncomirs and tumor suppressors. In pancreatic cancer, a significant number of oncomirs are found to be overexpressed, contributing to disease progression and aggressiveness. Nakata et al. and Eun et al. reported that miR-21, miR-155, and miR-210 were aberrantly expressed in PDAC [22,57]. These miRNAs stimulate PDAC cell proliferation, invasion, and chemoresistance and prevent apoptosis, as well as promote tumor angiogenesis and survival in hypoxic conditions [36,39,58,59,60,61,62] (Table 6).
MiR-21 is among the earliest identified oncomirs and is known for promoting cancer development. It targets nearly 30 genes, including tumor suppressors such as CDK2AP1, Pdcd4, and BCL2 [63]. MiR-21 stimulates pancreatic ductal adenocarcinoma (PDAC) cell proliferation, invasion, and chemoresistance, while also preventing apoptosis [14,15,36,58,68]. MiR-21 has been discovered to activate the epidermal growth factor (EGF) pathway by binding to sprouty RTK signaling antagonist 2 (Spry2). This interaction leads to heightened cell proliferation and triggers downstream signaling pathways such as MAPK/ERK and PI3K/Akt [69,70]. Nakamura et al. investigated exosomal miRNAs in pancreatic juice, finding that miR-21 and miR-155 can be used to differentiate PDAC patients from CP patients [65]. Wang et al. established that elevated serum levels of miR-21 might serve as a predictor for chemoresistance in PDAC [71]. Karasek et al. described the association of poor patient overall survival (OS) and elevated plasma levels of miR-21 [72]. On the other hand, Khan et al. observed worse progression-free survival (PFS) in patients with inoperable PDAC and elevated plasma miR-21, but no association between circulating miR-21 levels and OS was detected [73]. The levels of miR-21 expression show an increase in pancreatic ductal adenocarcinoma (PDAC) and exhibit considerable variation across different research studies [28,29,36,42]. Bloomston et al. observed a median 2.2-fold rise in miR-21 expression levels in formalin-fixed paraffin-embedded (FFPE) tumor samples [28]. In contrast, Zhang et al. identified significantly elevated levels of miR-21 expression, showing an increase of up to 6888-fold in various tumor samples [23]. In this study, the expression level of miR-21 was significantly increased in PDACs, as well as in UCOGCs in comparison with that in normal tissue (p < 0.05, Table 5).
MiR-155 acts as an inhibitor of tumor protein 53-induced nuclear protein 1 (TP53INP1) and FOXO3a expression, consequently promoting cell proliferation and malignant transformation [39,59]. Moreover, miR-155 is linked to the JAK/STAT pathway, exerting a negative regulatory effect on SOCS1. This correlation contributes to the enhanced migration and invasion capabilities of PDAC cells [64]. The expression levels of six miRNAs, among them miR-155, in tumor biopsies collected from human pancreatic ductal adenocarcinoma (PDAC) patients were discovered to be significantly associated with a higher risk of lymph node metastasis. This finding not only enhances the accuracy of diagnosing lymph node metastasis, but also, when combined with serum CA19-9 levels [74], MiR-155 has also been associated with advanced tumor stage and poor survival outcomes [26]. Mikamori et al. demonstrated that both OS and disease-free survival (DFS) were significantly shorter in the group with high miR-155 expression in microdissected formalin-fixed paraffin-embedded (FFPE) pancreatic cancer samples [75]. Similarly to miR-21, the expression of miR-155, miR-196a, and miR-10b has been correlated with increased invasiveness and poor OS in pancreatic cancer patients [76]. Pang et al. illustrated that PDAC cells have the ability to induce the differentiation of cancer-associated fibroblasts (CAFs) or CAF-like cells from normal fibroblasts by releasing exosomes containing miR-155. These miR-155-containing exosomes can directly interact with TP53INP1, facilitating this differentiation process [69,77]. Recently published pooled results on the role of increased levels of miR-155 in cancer diagnosis and prognosis showed a remarkable diagnostic value of miR-155 in regards to several cancers, including PDAC [78]. Kim et al. recommended several serum miRNAs, including miR-21 and miR-155, for accurate PDAC diagnosis [79]. In various studies, the overexpression of miR-155 in paraffin-embedded pancreatic ductal adenocarcinoma samples and pancreatic cancer cell lines has been observed, with the fold change ranging from 1.8 to 2.9 [23,28,80]. On the other hand, Zhang et al. reported an up to 52-fold increase in individual cases [23]. In our group of patients, we detected a mean 4.91-fold and 45.8-fold increase in miR-155 expression in UCOGCs and G3 PDACs, respectively (p < 0.05, Table 5).
MiR-210 is necessary for tumor angiogenesis, cell cycle regulation, and cancer survival in hypoxic conditions [60,61,62,67]. Research has revealed that miR-210 imposes a negative regulatory influence on EFNA3 expression and actively engages in the PI3K/AKT/VEGFA or Wnt/β-catenin/RHOA pathways. This participation facilitates tumor angiogenesis and enhances cellular permeability. In vivo studies have corroborated that exosomal miR-210 plays a significant role in driving the progression of pancreatic ductal adenocarcinoma [81,82]. Yu et al. analyzed plasma levels of miR-196a and miR-210 with RT-qPCR in a cohort of 31 PDAC patients. High miR-196a expression was associated with poor OS, whereas high miR-210 expression was significantly associated with improved survival [83]. An increase in miR-210 levels has been consistently reported across numerous studies [31,43]. In their study, Greither et al. identified a significant increase of up to 39.9-fold in miR-210 levels in snap-frozen surgical resection specimens [31]. Wang et al. reported a substantial elevation ranging from 2 to 28-fold in miR-210 plasma levels among patients diagnosed with pancreatic PDAC [30]. In this study, the expression level of miR-210 was significantly increased in G3 PDACs, as well as in UCOGCs, in comparison with the level in normal tissue (p < 0.05, Table 5).
The miR-196a is an oncomir reported to be frequently dysregulated in PDAC [28,31]. Serum levels of miR-196a could potentially serve as a valuable marker for distinguishing between resectable and unresectable PDAC [84]. Nuclear factor-kappa-B-inhibitor alpha (NFKBIA) is a direct target for miR-196a. This miRNA promotes cancer cell proliferation, migration, and invasion. Bloomston et al. have associated high expression levels of miR-196a in formalin-fixed paraffin-embedded (FFPE) samples obtained from PDAC patients, as measured by microarrays, with shorter OS [28]. Kong et al. similarly reported a correlation between elevated levels of miR-196a measured in the blood sera of pancreatic ductal adenocarcinoma patients and poorer survival outcomes, as well as advanced disease stage [26]. Abnormalities in miR-196a expression have been documented, not only in pancreatic cancer, but also in various other malignancies. [23,30,85,86]. Zhang et al. observed variations in tumor miR-196 expression ranging from 0.35 to 1557-fold [23]. Meanwhile, Wang et al. demonstrated a significant 16.05-fold increase in miR-196 expression levels in plasma samples [30]. We did not find significant differences in miR-196a expression between G3 PDACs and normal tissues (p > 0.05, Table 5). On the other hand, this miRNA was slightly but significantly downregulated in UCOGCs samples (p < 0.05, Table 5).
The microRNAs miR-96, miR-148a, and miR-217 are considered to be tumor suppressors, and they are inhibited in PDACs. These miRNAs may inhibit cell proliferation and prevent cancer cells chemoresistance, migration, and invasions; additionally, they induce cellular senescence and apoptosis [32,33,40,41], Table 7.
MiR-96 may act as a tumor suppressor, inhibiting the KRAS-signaling pathway [40]. Indeed, the expression of miR-96 in PDAC appears to be inconsistent across various studies, with some reporting downregulation [29,40,43], while others suggest upregulation [28]. On the other hand, we did not find any significant differences in miR-96 expression levels between UCOGCs, G3 PDACs, and normal tissues (p > 0.05, Table 5).
MiR-148a targets may affect the cell cycle and apoptosis [32]. The expression of microRNA miR-148a is reported to be downregulated in pancreatic ductal adenocarcinoma as a result of promoter hypermethylation. This phenomenon represents an early event in the process of pancreatic carcinogenesis [91]. Bloomston et al. and Jamieson et al. both measured a significant average decrease in miR-148a expression, with Bloomston et al. observing a 5.5-fold decrease, and Jamieson et al. reporting a 7.14-fold decrease [25,28]. a decrease in miR-148a levels was identified in UCOGC, as well as in G3 PDAC tissue samples (p < 0.05, Table 5).
MiR-217 has been shown to inhibit in vitro tumor cell growth, and it acts as a potential tumor suppressor by influencing the Akt/KRAS signaling pathway. Consequently, miR-217 is often found to be downregulated in PDAC [41]. Conversely, miR-217 is typically expressed in normal pancreatic tissue [92], and downregulated in pancreatic dysplasia [93]. In the study by Szafranszka et al., microRNA miR-217 was found to be downregulated by 10-fold [43]. However, Greither et al. determined only a mean 2-fold decrease [31]. Hong et al. discovered that the expression of miR-217 was downregulated by up to 62.5-fold in PDACs [29]. Both UCOGC and G3 PDAC samples demonstrated a significant downregulation of miR-217, with an up to-100-fold decrease (p < 0.05, Table 5).
In the current study, no significant differences in the expression of the majority of miRNAs were observed between UCOGC and G3 PDAC (p > 0.05, Table 5), with the exception of miR-155. UCOGC demonstrated distinctly lower mean levels of miR-155 in comparison with those of the PDACs (p < 0.05, Table 5). MiR-155 is an oncomir overexpressed in early pancreatic adenocarcinoma precursors and invasive PDAC [38]. High expression of this miRNA was associated with lymph node metastasis in PDAC patients [94,95]. The microRNA miR-155 was found to induce epithelial–mesenchymal transitions (EMT) in breast, liver, and kidney cancer [96,97,98,99]. MiR-155 promotes (EMT), as well as regulates cancer cell invasion and migration, by modulating the STAT3 signaling pathway in pancreatic cancer cells [64]. Mattiolo et al. reported that EMT is more frequently activated in undifferentiated carcinomas (UC) (10/10 cases) than in UCOGC (8/16 cases). Furthermore, in UCOGC, EMT was activated with a higher frequency in cases with an associated PDAC component. Snai2 was the most frequently and strongly expressed marker of EMT in both tumor types (10/10 UC, 8/16 UCOGC), and its expression was higher in UC than in UCOGC [53]. Therefore, our results indicating that poorly differentiated G3 PDACs demonstrated higher levels of miR-155 in comparison with UCOGC are in concordance with the findings mentioned above. On the other hand, our work is of a preliminary nature. We believe that a cohort of 10 UCOGC patients is not enough for conducting statistical analysis on the correlation between miRNA fold change and patient outcomes. Consequently, future investigations involving a larger number of patients are warranted. Additionally, our study only examined seven miRNAs. Therefore, analyzing the entire miRNome, using RNA microarray or sequencing techniques, may be instrumental in identifying miRNAs that are highly beneficial for PDAC and UCOGC diagnostics, therapy, or predicting patient outcomes.

5. Conclusions

We have demonstrated that UCOGC, as well as PDAC, display similar pattern of changes in miRNAs expression in comparison with those of normal tissues. Oncomirs, including miR-21, miR-155, and miR-210, were significantly upregulated. Tumor suppressors, such as miR-148a and miR-217, were downregulated in both UCOGC and PDAC. We did not find any significant differences between cancerous and normal tissues for the expression levels of miR-96 and miR-196a in PDAC, whereas miR-196a was downregulated in UCOGC. We have not observed significant differences in the expression of the majority of miRNAs between UCOGC and PDAC, with the exception of miR-155.

Author Contributions

Conceptualization, J.H. and V.M.; methodology, A.P. and J.V.; formal analysis, J.H., A.S. and A.P.; investigation, A.P., J.H., A.S., J.V., V.C. and J.M.; resources, M.O. and Z.Š.; writing—original draft preparation, A.P., J.H., V.M., J.V. and A.S.; writing—review and editing, J.H., A.P., M.O. and V.M.; supervision, J.H. and V.M.; funding acquisition, J.H. and V.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the Charles University project Cooperatio Medical Diagnostics and Basic Medical Sciences.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Ethics Committee of the University Hospital Královské Vinohrady (protocol code EK_VP/2/20l1, 29 June 2011).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All data supporting reported results can be found in the authors’ institutions or in public repositories.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hruban, R.H.; Adsay, N.V.; Esposito, I.; Fukushima, N.; Furukawa, T.; Klöppel, G.; Maitra, A.; Notohara, K.; Offerhaus, G.J.A.; Ohike, N.; et al. Pancreatic ductal adenocarcinoma. In WHO Classification of Tumours Editorial Board. Digestive System Tumours; International Agency for Research on Cancer: Lyon, France, 2019; pp. 329–330. ISBN 978-92-832-4499-8. [Google Scholar]
  2. Muraki, T.; Reid, M.D.; Basturk, O.; Jang, K.T.; Bedolla, G.; Bagci, P.; Mittal, P.; Memis, B.; Katabi, N.; Bandyopadhyay, S.; et al. Undifferentiated Carcinoma with Osteoclastic Giant Cells of the Pancreas: Clinicopathologic Analysis of 38 Cases Highlights a More Protracted Clinical Course than Currently Appreciated. Am. J. Surg. Pathol. 2016, 40, 1203–1216. [Google Scholar] [CrossRef]
  3. Sommers, S.C.; Meissner, W.A. Unusual Carcinomas of the Pancreas. AMA Arch. Pathol. 1954, 58, 101–111. [Google Scholar]
  4. Rosai, J. Carcinoma of Pancreas Simulating Giant Cell Tumor of Bone. Electron-Microscopic Evidence of Its Acinar Cell Origin. Cancer 1968, 22, 333–344. [Google Scholar] [CrossRef]
  5. Luchini, C.; Cros, J.; Pea, A.; Pilati, C.; Veronese, N.; Rusev, B.; Capelli, P.; Mafficini, A.; Nottegar, A.; Brosens, L.A.A.; et al. PD-1, PD-L1, and CD163 in Pancreatic Undifferentiated Carcinoma with Osteoclast-like Giant Cells: Expression Patterns and Clinical Implications. Hum. Pathol. 2018, 81, 157–165. [Google Scholar] [CrossRef]
  6. Bazzichetto, C.; Luchini, C.; Conciatori, F.; Vaccaro, V.; Di Cello, I.; Mattiolo, P.; Falcone, I.; Ferretti, G.; Scarpa, A.; Cognetti, F.; et al. Morphologic and Molecular Landscape of Pancreatic Cancer Variants as the Basis of New Therapeutic Strategies for Precision Oncology. Int. J. Mol. Sci. 2020, 21, 8841. [Google Scholar] [CrossRef]
  7. Mugaanyi, J.; Lu, C.; Huang, J.; Lu, C. Undifferentiated Pancreatic Carcinomas, Clinical Features and Therapeutic Options: What We Know. Cancers 2022, 14, 6102. [Google Scholar] [CrossRef]
  8. Hrudka, J.; Kalinová, M.; Ciprová, V.; Moravcová, J.; Dvořák, R.; Matěj, R. Undifferentiated Carcinoma with Osteoclast-like Giant Cells of the Pancreas: Molecular Genetic Analysis of 13 Cases. Int. J. Mol. Sci. 2024, 25, 3285. [Google Scholar] [CrossRef]
  9. Ali Syeda, Z.; Langden, S.S.S.; Munkhzul, C.; Lee, M.; Song, S.J. Regulatory Mechanism of MicroRNA Expression in Cancer. Int. J. Mol. Sci. 2020, 21, 1723. [Google Scholar] [CrossRef]
  10. Shang, R.; Lee, S.; Senavirathne, G.; Lai, E.C. microRNAs in Action: Biogenesis, Function and Regulation. Nat. Rev. Genet. 2023, 24, 816–833. [Google Scholar] [CrossRef] [PubMed]
  11. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  12. Sassen, S.; Miska, E.A.; Caldas, C. MicroRNA—Implications for Cancer. Virchows Archiv. 2008, 452, 1–10. [Google Scholar] [CrossRef] [PubMed]
  13. Iorio, M.V.; Croce, C.M. MicroRNA Dysregulation in Cancer: Diagnostics, Monitoring and Therapeutics. A Comprehensive Review. EMBO Mol. Med. 2012, 4, 143–159. [Google Scholar] [CrossRef]
  14. Moriyama, T.; Ohuchida, K.; Mizumoto, K.; Yu, J.; Sato, N.; Nabae, T.; Takahata, S.; Toma, H.; Nagai, E.; Tanaka, M. MicroRNA-21 Modulates Biological Functions of Pancreatic Cancer Cells Including Their Proliferation, Invasion, and Chemoresistance. Mol. Cancer Ther. 2009, 8, 1067–1074. [Google Scholar] [CrossRef] [PubMed]
  15. Hu, G.; Tao, F.; Wang, W.; Ji, K. Prognostic Value of microRNA-21 in Pancreatic Ductal Adenocarcinoma: A Meta-Analysis. World J. Surg. Oncol. 2016, 14, 82. [Google Scholar] [CrossRef] [PubMed]
  16. Daoud, A.Z.; Mulholland, E.J.; Cole, G.; McCarthy, H.O. MicroRNAs in Pancreatic Cancer: Biomarkers, Prognostic, and Therapeutic Modulators. BMC Cancer 2019, 19, 1130. [Google Scholar] [CrossRef] [PubMed]
  17. Szabo, A.; Gurlich, R.; Liberko, M.; Soumarova, R.; Vernerova, Z.; Mandys, V.; Popov, A. Expression of Selected microRNAs in Pancreatic Ductal Adenocarcinoma: Is There a Relation to Tumor Morphology, Progression and Patient’s Outcome? Neoplasma 2020, 67, 1170–1181. [Google Scholar] [CrossRef] [PubMed]
  18. Popov, A.; Mandys, V. Senescence-Associated miRNAs and Their Role in Pancreatic Cancer. Pathol. Oncol. Res. 2022, 28, 1610156. [Google Scholar] [CrossRef] [PubMed]
  19. Prinz, C.; Fehring, L.; Frese, R. MicroRNAs as Indicators of Malignancy in Pancreatic Ductal Adenocarcinoma (PDAC) and Cystic Pancreatic Lesions. Cells 2022, 11, 2374. [Google Scholar] [CrossRef] [PubMed]
  20. Shenouda, S.K.; Alahari, S.K. MicroRNA Function in Cancer: Oncogene or a Tumor Suppressor? Cancer Metastasis Rev. 2009, 28, 369–378. [Google Scholar] [CrossRef]
  21. Zhang, B.; Pan, X.; Cobb, G.; Anderson, T. microRNAs as Oncogenes and Tumor Suppressors. Dev. Biol. 2007, 302, 1–12. [Google Scholar] [CrossRef]
  22. Eun, J.L.; Gusev, Y.; Jiang, J.; Nuovo, G.J.; Lerner, M.R.; Frankel, W.L.; Morgan, D.L.; Postier, R.G.; Brackett, D.J.; Schmittgen, T.D. Expression Profiling Identifies microRNA Signature in Pancreatic Cancer. Int. J. Cancer 2007, 120, 1046–1054. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, Y.; Li, M.; Wang, H.; Fisher, W.E.; Lin, P.H.; Yao, Q.; Chen, C. Profiling of 95 MicroRNAs in Pancreatic Cancer Cell Lines and Surgical Specimens by Real-Time PCR Analysis. World J. Surg. 2009, 33, 698–709. [Google Scholar] [CrossRef] [PubMed]
  24. Negoi, I.; Hostiuc, S.; Sartelli, M.; Negoi, R.I.; Beuran, M. MicroRNA-21 as a Prognostic Biomarker in Patients with Pancreatic Cancer—A Systematic Review and Meta-Analysis. Am. J. Surg. 2017, 214, 515–524. [Google Scholar] [CrossRef]
  25. Jamieson, N.B.; Morran, D.C.; Morton, J.P.; Ali, A.; Dickson, E.J.; Carter, C.R.; Sansom, O.J.; Evans, T.R.J.; McKay, C.J.; Oien, K.A. MicroRNA Molecular Profiles Associated with Diagnosis, Clinicopathologic Criteria, and Overall Survival in Patients with Resectable Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. 2012, 18, 534–545. [Google Scholar] [CrossRef] [PubMed]
  26. Kong, X.; Du, Y.; Wang, G.; Gao, J.; Gong, Y.; Li, L.; Zhang, Z.; Zhu, J.; Jing, Q.; Qin, Y.; et al. Detection of Differentially Expressed microRNAs in Serum of Pancreatic Ductal Adenocarcinoma Patients: miR-196a Could Be a Potential Marker for Poor Prognosis. Dig. Dis. Sci. 2011, 56, 602–609. [Google Scholar] [CrossRef]
  27. Papaconstantinou, I.G.; Manta, A.; Gazouli, M.; Lyberopoulou, A.; Lykoudis, P.M.; Polymeneas, G.; Voros, D. Expression of microRNAs in Patients with Pancreatic Cancer and Its Prognostic Significance. Pancreas 2013, 42, 67–71. [Google Scholar] [CrossRef] [PubMed]
  28. Bloomston, M.; Frankel, W.L.; Petrocca, F.; Volinia, S.; Alder, H.; Hagan, J.P.; Liu, C.G.; Bhatt, D.; Taccioli, C.; Croce, C.M. MicroRNA Expression Patterns to Differentiate Pancreatic Adenocarcinoma from Normal Pancreas and Chronic Pancreatitis. JAMA 2007, 297, 1901–1908. [Google Scholar] [CrossRef] [PubMed]
  29. Hong, T.H.; Park, I.Y. MicroRNA Expression Profiling of Diagnostic Needle Aspirates from Surgical Pancreatic Cancer Specimens. Ann. Surg. Treat. Res. 2014, 87, 290. [Google Scholar] [CrossRef] [PubMed]
  30. Wang, J.; Chen, J.; Chang, P.; LeBlanc, A.; Li, D.; Abbruzzesse, J.L.; Frazier, M.L.; Killary, A.M.; Sen, S. MicroRNAs in Plasma of Pancreatic Ductal Adenocarcinoma Patients as Novel Blood-Based Biomarkers of Disease. Cancer Prev. Res. 2009, 2, 807–813. [Google Scholar] [CrossRef]
  31. Greither, T.; Grochola, L.F.; Udelnow, A.; Lautenschläger, C.; Würl, P.; Taubert, H. Elevated Expression of microRNAs 155, 203, 210 and 222 in Pancreatic Tumors Is Associated with Poorer Survival. Int. J. Cancer 2010, 126, 73–80. [Google Scholar] [CrossRef]
  32. Delpu, Y.; Lulka, H.; Sicard, F.; Saint-Laurent, N.; Lopez, F.; Hanoun, N.; Buscail, L.; Cordelier, P.; Torrisani, J. The Rescue of miR-148a Expression in Pancreatic Cancer: An Inappropriate Therapeutic Tool. PLoS ONE 2013, 8, e55513. [Google Scholar] [CrossRef] [PubMed]
  33. Feng, J.; Yu, J.; Pan, X.; Li, Z.; Chen, Z.; Zhang, W.; Wang, B.; Yang, L.; Xu, H.; Zhang, G.; et al. HERG1 Functions as an Oncogene in Pancreatic Cancer and Is Downregulated by miR-96. Oncotarget 2014, 5, 5832–5844. [Google Scholar] [CrossRef]
  34. Hrudka, J.; Lawrie, K.; Waldauf, P.; Ciprová, V.; Moravcová, J.; Matěj, R. Negative Prognostic Impact of PD-L1 Expression in Tumor Cells of Undifferentiated (Anaplastic) Carcinoma with Osteoclast-like Giant Cells of the Pancreas: Study of 13 Cases Comparing Ductal Pancreatic Carcinoma and Review of the Literature. Virchows Arch. 2020, 477, 687–696. [Google Scholar] [CrossRef] [PubMed]
  35. Pfaffl, M.W. A New Mathematical Model for Relative Quantification in Real-Time RT-PCR. Nucleic Acids Res. 2001, 29, e45. [Google Scholar] [CrossRef]
  36. Du Rieu, M.C.; Torrisani, J.; Selves, J.; Al Saati, T.; Souque, A.; Dufresne, M.; Tsongalis, G.J.; Suriawinata, A.A.; Carrère, N.; Buscail, L.; et al. MicroRNA-21 Is Induced Early in Pancreatic Ductal Adenocarcinoma Precursor Lesions. Clin. Chem. 2010, 56, 603–612. [Google Scholar] [CrossRef]
  37. Qi, L.; Bart, J.; Tan, L.P.; Platteel, I.; van der Sluis, T.; Huitema, S.; Harms, G.; Fu, L.; Hollema, H.; van den Berg, A. Expression of miR-21 and Its Targets (PTEN, PDCD4, TM1) in Flat Epithelial Atypia of the Breast in Relation to Ductal Carcinoma in Situ and Invasive Carcinoma. BMC Cancer 2009, 9, 163. [Google Scholar] [CrossRef]
  38. Habbe, N.; Koorstra, J.B.M.; Mendell, J.T.; Offerhaus, G.J.; Ji, K.R.; Feldmann, G.; Mullendore, M.E.; Goggins, M.G.; Hong, S.M.; Maitra, A. MicroRNA miR-155 Is a Biomarker of Early Pancreatic Neoplasia. Cancer Biol. Ther. 2009, 8, 340–346. [Google Scholar] [CrossRef] [PubMed]
  39. Gironella, M.; Seux, M.; Xie, M.-J.; Cano, C.; Tomasini, R.; Gommeaux, J.; Garcia, S.; Nowak, J.; Yeung, M.L.; Jeang, K.-T.; et al. Tumor Protein 53-Induced Nuclear Protein 1 Expression Is Repressed by miR-155, and Its Restoration Inhibits Pancreatic Tumor Development. Proc. Natl. Acad. Sci. USA 2007, 104, 16170–16175. [Google Scholar] [CrossRef]
  40. Yu, S.; Lu, Z.; Liu, C.; Meng, Y.; Ma, Y.; Zhao, W.; Liu, J.; Yu, J.; Chen, J. miRNA-96 Suppresses KRAS and Functions as a Tumor Suppressor Gene in Pancreatic Cancer. Cancer Res. 2010, 70, 6015–6025. [Google Scholar] [CrossRef] [PubMed]
  41. Zhao, W.-G.; Yu, S.-N.; Lu, Z.-H.; Ma, Y.-H.; Gu, Y.-M.; Chen, J. The miR-217 microRNA Functions as a Potential Tumor Suppressor in Pancreatic Ductal Adenocarcinoma by Targeting KRAS. Carcinogenesis 2010, 31, 1726–1733. [Google Scholar] [CrossRef]
  42. Szafranska, A.E.; Davison, T.S.; John, J.; Cannon, T.; Sipos, B.; Maghnouj, A.; Labourier, E.; Hahn, S.A. MicroRNA Expression Alterations Are Linked to Tumorigenesis and Non-Neoplastic Processes in Pancreatic Ductal Adenocarcinoma. Oncogene 2007, 26, 4442–4452. [Google Scholar] [CrossRef]
  43. Szafranska, A.E.; Doleshal, M.; Edmunds, H.S.; Gordon, S.; Luttges, J.; Munding, J.B.; Barth, R.J.; Gutmann, E.J.; Suriawinata, A.A.; Pipas, J.M.; et al. Analysis of microRNAs in Pancreatic Fine-Needle Aspirates Can Classify Benign and Malignant Tissues. Clin. Chem. 2008, 54, 1716–1724. [Google Scholar] [CrossRef]
  44. Link, A.; Becker, V.; Goel, A.; Wex, T.; Malfertheiner, P. Feasibility of Fecal microRNAs as Novel Biomarkers for Pancreatic Cancer. PLoS ONE 2012, 7, e42933. [Google Scholar] [CrossRef]
  45. Molberg, K.H.; Heffess, C.; Delgado, R.; Albores-Saavedra, J. Undifferentiated Carcinoma with Osteoclast-like Giant Cells of the Pancreas and Periampullary Region. Cancer 1998, 82, 1279–1287. [Google Scholar] [CrossRef]
  46. Paal, E.; Thompson, L.D.; Frommelt, R.A.; Przygodzki, R.M.; Heffess, C.S. A Clinicopathologic and Immunohistochemical Study of 35 Anaplastic Carcinomas of the Pancreas with a Review of the Literature. Ann. Diagn. Pathol. 2001, 5, 129–140. [Google Scholar] [CrossRef]
  47. Rosai and Ackerman’s Surgical Pathology—2 Volume Set—11th Edition. Available online: https://www.elsevier.com/books/rosai-and-ackermans-surgical-pathology-2-volume-set/goldblum/978-0-323-26339-9 (accessed on 23 May 2023).
  48. Dworak, O.; Wittekind, C.; Koerfgen, H.P.; Gall, F.P. Osteoclastic Giant Cell Tumor of the Pancreas. An Immunohistological Study and Review of the Literature. Pathol. Res. Pract. 1993, 189, 228–234. [Google Scholar] [CrossRef]
  49. Reid, M.D.; Muraki, T.; HooKim, K.; Memis, B.; Graham, R.P.; Allende, D.; Shi, J.; Schaeffer, D.F.; Singh, R.; Basturk, O.; et al. Cytologic Features and Clinical Implications of Undifferentiated Carcinoma with Osteoclastic Giant Cells of the Pancreas: An Analysis of 15 Cases. Cancer Cytopathol. 2017, 125, 563–575. [Google Scholar] [CrossRef]
  50. Lehrke, H.D.; Graham, R.P.; McWilliams, R.R.; Lam-Himlin, D.M.; Smyrk, T.C.; Jenkins, S.; Dong, H.; Zhang, L. Undifferentiated Pancreatic Carcinomas Display Enrichment for Frequency and Extent of PD-L1 Expression by Tumor Cells. Am. J. Clin. Pathol. 2017, 148, 441–449. [Google Scholar] [CrossRef]
  51. Besaw, R.J.; Terra, A.R.; Malvar, G.L.; Chapman, T.R.; Hertan, L.M.; Schlechter, B.L. Durable Response to PD-1 Blockade in a Patient with Metastatic Pancreatic Undifferentiated Carcinoma with Osteoclast-Like Giant Cells. J. Natl. Compr. Canc. Netw. 2021, 19, 247–252. [Google Scholar] [CrossRef]
  52. Obayashi, M.; Shibasaki, Y.; Koakutsu, T.; Hayashi, Y.; Shoji, T.; Hirayama, K.; Yamazaki, M.; Takayanagi, Y.; Shibata, H.; Nakamura, M.; et al. Pancreatic Undifferentiated Carcinoma with Osteoclast-like Giant Cells Curatively Resected after Pembrolizumab Therapy for Lung Metastases: A Case Report. BMC Gastroenterol. 2020, 20, 220. [Google Scholar] [CrossRef]
  53. Mattiolo, P.; Fiadone, G.; Paolino, G.; Chatterjee, D.; Bernasconi, R.; Piccoli, P.; Parolini, C.; El Aidi, M.; Sperandio, N.; Malleo, G.; et al. Epithelial-Mesenchymal Transition in Undifferentiated Carcinoma of the Pancreas with and without Osteoclast-like Giant Cells. Virchows Arch. 2021, 478, 319–326. [Google Scholar] [CrossRef]
  54. Sakakida, T.; Ishikawa, T.; Doi, T.; Morita, R.; Kataoka, S.; Miyake, H.; Yamaguchi, K.; Moriguchi, M.; Sogame, Y.; Yasuda, H.; et al. Genomic Landscape and Clinical Features of Rare Subtypes of Pancreatic Cancer: Analysis with the National Database of Japan. J. Gastroenterol. 2023, 58, 575–585. [Google Scholar] [CrossRef]
  55. Bergmann, F.; Moldenhauer, G.; Herpel, E.; Gaida, M.M.; Strobel, O.; Werner, J.; Esposito, I.; Müerköster, S.S.; Schirmacher, P.; Kern, M.A. Expression of L1CAM, COX-2, EGFR, c-KIT and Her2/Neu in Anaplastic Pancreatic Cancer: Putative Therapeutic Targets? Histopathology 2010, 56, 440–448. [Google Scholar] [CrossRef]
  56. Wang, X.; Miao, J.; Wang, S.; Shen, R.; Zhang, S.; Tian, Y.; Li, M.; Zhu, D.; Yao, A.; Bao, W.; et al. Single-Cell RNA-Seq Reveals the Genesis and Heterogeneity of Tumor Microenvironment in Pancreatic Undifferentiated Carcinoma with Osteoclast-like Giant-Cells. Mol. Cancer 2022, 21, 133. [Google Scholar] [CrossRef]
  57. Nakata, K.; Ohuchida, K.; Mizumoto, K.; Kayashima, T.; Ikenaga, N.; Sakai, H.; Lin, C.; Fujita, H.; Otsuka, T.; Aishima, S.; et al. MicroRNA-10b Is Overexpressed in Pancreatic Cancer, Promotes Its Invasiveness, and Correlates with a Poor Prognosis. Surgery 2011, 150, 916–922. [Google Scholar] [CrossRef]
  58. Park, J.-K.; Lee, E.J.; Esau, C.; Schmittgen, T.D. Antisense Inhibition of microRNA-21 or -221 Arrests Cell Cycle, Induces Apoptosis, and Sensitizes the Effects of Gemcitabine in Pancreatic Adenocarcinoma. Pancreas 2009, 38, e190. [Google Scholar] [CrossRef]
  59. Wang, P.; Zhu, C.; Ma, M.; Chen, G.; Song, M.; Zeng, Z.; Lu, W.; Yang, J.; Wen, S.; Chiao, P.J.; et al. Micro-RNA-155 Is Induced by K-Ras Oncogenic Signal and Promotes ROS Stress in Pancreatic Cancer. Oncotarget 2015, 6, 21148–21158. [Google Scholar] [CrossRef]
  60. Fasanaro, P.; D’Alessandra, Y.; Di Stefano, V.; Melchionna, R.; Romani, S.; Pompilio, G.; Capogrossi, M.C.; Martelli, F. MicroRNA-210 Modulates Endothelial Cell Response to Hypoxia and Inhibits the Receptor Tyrosine Kinase Ligand Ephrin-A3. J. Biol. Chem. 2008, 283, 15878–15883. [Google Scholar] [CrossRef]
  61. Zhang, Z.; Sun, H.; Dai, H.; Walsh, R.M.; Imakura, M.; Schelter, J.; Burchard, J.; Dai, X.; Chang, A.N.; Diaz, R.L.; et al. MicroRNA miR-210 Modulates Cellular Response to Hypoxia through the MYC Antagonist MNT. Cell Cycle 2009, 8, 2756–2768. [Google Scholar] [CrossRef]
  62. Huang, X.; Le, Q.-T.; Giaccia, A.J. MiR-210–Micromanager of the Hypoxia Pathway. Trends Mol. Med. 2010, 16, 230–237. [Google Scholar] [CrossRef]
  63. Buscaglia, L.E.B.; Li, Y. Apoptosis and the Target Genes of microRNA-21. Chin. J. Cancer 2011, 30, 371–380. [Google Scholar] [CrossRef]
  64. Huang, C.; Li, H.; Wu, W.; Jiang, T.; Qiu, Z. Regulation of miR-155 Affects Pancreatic Cancer Cell Invasiveness and Migration by Modulating the STAT3 Signaling Pathway through SOCS1. Oncol. Rep. 2013, 30, 1223–1230. [Google Scholar] [CrossRef]
  65. Nakamura, S.; Sadakari, Y.; Ohtsuka, T.; Okayama, T.; Nakashima, Y.; Gotoh, Y.; Saeki, K.; Mori, Y.; Nakata, K.; Miyasaka, Y.; et al. Pancreatic Juice Exosomal MicroRNAs as Biomarkers for Detection of Pancreatic Ductal Adenocarcinoma. Ann. Surg. Oncol. 2019, 26, 2104–2111. [Google Scholar] [CrossRef]
  66. Huang, F.; Tang, J.; Zhuang, X.; Zhuang, Y.; Cheng, W.; Chen, W.; Yao, H.; Zhang, S. MiR-196a Promotes Pancreatic Cancer Progression by Targeting Nuclear Factor Kappa-B-Inhibitor Alpha. PLoS ONE 2014, 9, e87897. [Google Scholar] [CrossRef]
  67. Giovannetti, E.; Funel, N.; Peters, G.J.; Del Chiaro, M.; Erozenci, L.A.; Vasile, E.; Leon, L.G.; Pollina, L.E.; Groen, A.; Falcone, A.; et al. MicroRNA-21 in Pancreatic Cancer: Correlation with Clinical Outcome and Pharmacologic Aspects Underlying Its Role in the Modulation of Gemcitabine Activity. Cancer Res. 2010, 70, 4528–4538. [Google Scholar] [CrossRef]
  68. Huang, X.; Zuo, J. Emerging Roles of miR-210 and Other Non-Coding RNAs in the Hypoxic Response. Acta Biochim. Et Biophys. Sin. 2014, 46, 220–232. [Google Scholar] [CrossRef]
  69. Zhao, Q.; Chen, S.; Zhu, Z.; Yu, L.; Ren, Y.; Jiang, M.; Weng, J.; Li, B. miR-21 Promotes EGF-Induced Pancreatic Cancer Cell Proliferation by Targeting Spry2. Cell Death Dis. 2018, 9, 1157. [Google Scholar] [CrossRef]
  70. Mok, E.T.Y.; Chitty, J.L.; Cox, T.R. miRNAs in Pancreatic Cancer Progression and Metastasis. Clin. Exp. Metastasis 2024. [Google Scholar] [CrossRef]
  71. Wang, P.; Zhuang, L.; Zhang, J.; Fan, J.; Luo, J.; Chen, H.; Wang, K.; Liu, L.; Chen, Z.; Meng, Z. The Serum miR-21 Level Serves as a Predictor for the Chemosensitivity of Advanced Pancreatic Cancer, and miR-21 Expression Confers Chemoresistance by Targeting FasL. Mol. Oncol. 2013, 7, 334. [Google Scholar] [CrossRef]
  72. Karásek, P.; Gablo, N.; Hlavsa, J.; Kiss, I.; Vychytilová-Faltejsková, P.; Hermanová, M.; Kala, Z.; Slabý, O.; Procházka, V. Pre-Operative Plasma miR-21-5p Is a Sensitive Biomarker and Independent Prognostic Factor in Patients with Pancreatic Ductal Adenocarcinoma Undergoing Surgical Resection. Cancer Genom. Proteom. 2018, 15, 321–327. [Google Scholar] [CrossRef]
  73. Khan, K.; Cunningham, D.; Peckitt, C.; Barton, S.; Tait, D.; Hawkins, M.; Watkins, D.; Starling, N.; Rao, S.; Begum, R.; et al. miR-21 Expression and Clinical Outcome in Locally Advanced Pancreatic Cancer: Exploratory Analysis of the Pancreatic Cancer Erbitux, Radiotherapy and UFT (PERU) Trial. Oncotarget 2016, 7, 12672–12681. [Google Scholar] [CrossRef] [PubMed]
  74. Nishiwada, S.; Sho, M.; Banwait, J.K.; Yamamura, K.; Akahori, T.; Nakamura, K.; Baba, H.; Goel, A. A MicroRNA Signature Identifies Pancreatic Ductal Adenocarcinoma Patients at Risk for Lymph Node Metastases. Gastroenterology 2020, 159, 562–574. [Google Scholar] [CrossRef] [PubMed]
  75. Mikamori, M.; Yamada, D.; Eguchi, H.; Hasegawa, S.; Kishimoto, T.; Tomimaru, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K.; et al. MicroRNA-155 Controls Exosome Synthesis and Promotes Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma. Sci. Rep. 2017, 7, 42339. [Google Scholar] [CrossRef] [PubMed]
  76. Khan, M.A.; Zubair, H.; Srivastava, S.K.; Singh, S.; Singh, A.P. Insights into the Role of microRNAs in Pancreatic Cancer Pathogenesis: Potential for Diagnosis, Prognosis, and Therapy. Adv. Exp. Med. Biol. 2015, 889, 71–87. [Google Scholar] [CrossRef] [PubMed]
  77. Pang, W.; Su, J.; Wang, Y.; Feng, H.; Dai, X.; Yuan, Y.; Chen, X.; Yao, W. Pancreatic Cancer-Secreted miR-155 Implicates in the Conversion from Normal Fibroblasts to Cancer-Associated Fibroblasts. Cancer Sci. 2015, 106, 1362–1369. [Google Scholar] [CrossRef] [PubMed]
  78. Wu, Y.; Hong, Q.; Lu, F.; Zhang, Z.; Li, J.; Nie, Z.; He, B. The Diagnostic and Prognostic Value of miR-155 in Cancers: An Updated Meta-Analysis. Mol. Diagn. Ther. 2023, 27, 283–301. [Google Scholar] [CrossRef]
  79. Kim, M.W.; Koh, H.; Kim, J.Y.; Lee, S.; Lee, H.; Kim, Y.; Hwang, H.K.; Kim, S.I. Tumor-Specific miRNA Signatures in Combination with CA19-9 for Liquid Biopsy-Based Detection of PDAC. Int. J. Mol. Sci. 2021, 22, 13621. [Google Scholar] [CrossRef]
  80. Wang, J.; Raimondo, M.; Guha, S.; Chen, J.; Diao, L.; Dong, X.; Wallace, M.B.; Killary, A.M.; Frazier, M.L.; Woodward, T.A.; et al. Circulating microRNAs in Pancreatic Juice as Candidate Biomarkers of Pancreatic Cancer. J. Cancer 2014, 5, 696–705. [Google Scholar] [CrossRef] [PubMed]
  81. Lian, M.; Mortoglou, M.; Uysal-Onganer, P. Impact of Hypoxia-Induced miR-210 on Pancreatic Cancer. Curr. Issues Mol. Biol. 2023, 45, 9778–9792. [Google Scholar] [CrossRef]
  82. Wu, G.; Ding, X.; Quan, G.; Xiong, J.; Li, Q.; Li, Z.; Wang, Y. Hypoxia-Induced miR-210 Promotes Endothelial Cell Permeability and Angiogenesis via Exosomes in Pancreatic Ductal Adenocarcinoma. Biochem. Res. Int. 2022, 2022, 7752277. [Google Scholar] [CrossRef]
  83. Yu, Q.; Xu, C.; Yuan, W.; Wang, C.; Zhao, P.; Chen, L.; Ma, J. Evaluation of Plasma MicroRNAs as Diagnostic and Prognostic Biomarkers in Pancreatic Adenocarcinoma: miR-196a and miR-210 Could Be Negative and Positive Prognostic Markers, Respectively. BioMed Res. Int. 2017, 2017, 6495867. [Google Scholar] [CrossRef] [PubMed]
  84. Wnuk, J.; Strzelczyk, J.K.; Gisterek, I. Clinical Value of Circulating miRNA in Diagnosis, Prognosis, Screening and Monitoring Therapy of Pancreatic Ductal Adenocarcinoma–A Review of the Literature. Int. J. Mol. Sci. 2023, 24, 5113. [Google Scholar] [CrossRef] [PubMed]
  85. Luthra, R.; Singh, R.R.; Luthra, M.G.; Li, Y.X.; Hannah, C.; Romans, A.M.; Barkoh, B.A.; Chen, S.S.; Ensor, J.; Maru, D.M.; et al. MicroRNA-196a Targets Annexin A1: A microRNA-Mediated Mechanism of Annexin A1 Downregulation in Cancers. Oncogene 2008, 27, 6667–6678. [Google Scholar] [CrossRef]
  86. Lee, Y.S.; Kim, H.; Kim, H.W.; Lee, J.-C.; Paik, K.-H.; Kang, J.; Kim, J.; Yoon, Y.-S.; Han, H.-S.; Sohn, I.; et al. High Expression of MicroRNA-196a Indicates Poor Prognosis in Resected Pancreatic Neuroendocrine Tumor. Medicine 2015, 94, e2224. [Google Scholar] [CrossRef] [PubMed]
  87. Balzeau, J.; Menezes, M.R.; Cao, S.; Hagan, J.P. The LIN28/Let-7 Pathway in Cancer. Front. Genet. 2017, 8, 31. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, H.; Chirshev, E.; Hojo, N.; Suzuki, T.; Bertucci, A.; Pierce, M.; Perry, C.; Wang, R.; Zink, J.; Glackin, C.A.; et al. The Epithelial–Mesenchymal Transcription Factor SNAI1 Represses Transcription of the Tumor Suppressor miRNA Let-7 in Cancer. Cancers 2021, 13, 1469. [Google Scholar] [CrossRef] [PubMed]
  89. Bhutia, Y.D.; Hung, S.W.; Krentz, M.; Patel, D.; Lovin, D.; Manoharan, R.; Thomson, J.M.; Govindarajan, R. Differential Processing of Let-7a Precursors Influences RRM2 Expression and Chemosensitivity in Pancreatic Cancer: Role of LIN-28 and SET Oncoprotein. PLoS ONE 2013, 8, e53436. [Google Scholar] [CrossRef] [PubMed]
  90. Idichi, T.; Seki, N.; Kurahara, H.; Fukuhisa, H.; Toda, H.; Shimonosono, M.; Okato, A.; Arai, T.; Kita, Y.; Mataki, Y.; et al. Molecular Pathogenesis of Pancreatic Ductal Adenocarcinoma: Impact of Passenger Strand of Pre-miR-148a on Gene Regulation. Cancer Sci. 2018, 109, 2013–2026. [Google Scholar] [CrossRef]
  91. Hanoun, N.; Delpu, Y.; Suriawinata, A.A.; Bournet, B.; Bureau, C.; Selves, J.; Tsongalis, G.J.; Dufresne, M.; Buscail, L.; Cordelier, P.; et al. The Silencing of MicroRNA 148a Production by DNA Hypermethylation Is an Early Event in Pancreatic Carcinogenesis. Clin. Chem. 2010, 56, 1107–1118. [Google Scholar] [CrossRef]
  92. Deng, S.; Zhu, S.; Wang, B.; Li, X.; Liu, Y.; Qin, Q.; Gong, Q.; Niu, Y.; Xiang, C.; Chen, J.; et al. Chronic Pancreatitis and Pancreatic Cancer Demonstrate Active Epithelial-Mesenchymal Transition Profile, Regulated by miR-217-SIRT1 Pathway. Cancer Lett. 2014, 355, 184–191. [Google Scholar] [CrossRef]
  93. Chang, X.; Yu, C.; Li, J.; Yu, S.; Chen, J. Hsa-miR-96 and Hsa-miR-217 Expression Down-Regulates with Increasing Dysplasia in Pancreatic Intraepithelial Neoplasias and Intraductal Papillary Mucinous Neoplasms. Int. J. Med. Sci. 2017, 14, 412–418. [Google Scholar] [CrossRef] [PubMed]
  94. Lemberger, M.; Loewenstein, S.; Lubezky, N.; Nizri, E.; Pasmanik-Chor, M.; Barazovsky, E.; Klausner, J.M.; Lahat, G. MicroRNA Profiling of Pancreatic Ductal Adenocarcinoma (PDAC) Reveals Signature Expression Related to Lymph Node Metastasis. Oncotarget 2019, 10, 2644–2656. [Google Scholar] [CrossRef] [PubMed]
  95. Kent, O.A.; Mullendore, M.; Wentzel, E.A.; López-Romero, P.; Tan, A.C.; Alvarez, H.; West, K.; Ochs, M.F.; Hidalgo, M.; Arking, D.E.; et al. A Resource for Analysis of microRNA Expression and Function in Pancreatic Ductal Adenocarcinoma Cells. Cancer Biol. Ther. 2009, 8, 2013–2024. [Google Scholar] [CrossRef] [PubMed]
  96. Liu, X.; Li, Y.; Li, Z.; Hou, T. miR-155 Promotes Proliferation and Epithelial-Mesenchymal Transition of MCF-7 Cells. Exp. Ther. Med. 2021, 21, 218. [Google Scholar] [CrossRef] [PubMed]
  97. Kong, W.; Yang, H.; He, L.; Zhao, J.; Coppola, D.; Dalton, W.S.; Cheng, J.Q. MicroRNA-155 Is Regulated by the Transforming Growth Factor Beta/Smad Pathway and Contributes to Epithelial Cell Plasticity by Targeting RhoA. Mol. Cell. Biol. 2008, 28, 6773–6784. [Google Scholar] [CrossRef] [PubMed]
  98. Kong, X.; Liu, F.; Gao, J. MiR-155 Promotes Epithelial-Mesenchymal Transition in Hepatocellular Carcinoma Cells through the Activation of PI3K/SGK3/β-Catenin Signaling Pathways. Oncotarget 2016, 7, 66051–66060. [Google Scholar] [CrossRef]
  99. Kulkarni, P.; Dasgupta, P.; Hashimoto, Y.; Shiina, M.; Shahryari, V.; Tabatabai, Z.L.; Yamamura, S.; Tanaka, Y.; Saini, S.; Dahiya, R.; et al. A lncRNA TCL6-miR-155 Interaction Regulates the Src-Akt-EMT Network to Mediate Kidney Cancer Progression and Metastasis. Cancer Res. 2021, 81, 1500–1512. [Google Scholar] [CrossRef]
Figure 1. Comparison of miRNA expression fold change in UCOGC and G3 PDAC, shown graphically.
Figure 1. Comparison of miRNA expression fold change in UCOGC and G3 PDAC, shown graphically.
Biomedicines 12 00962 g001
Table 1. Stem–loop primers for the miRNAs.
Table 1. Stem–loop primers for the miRNAs.
miRNA Name:Stem–Loop Primer Sequence:
miR-39
C. elegans
GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACT-
GGATACGACTATTAC
mir-21GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCAC-
TGGATACGACTCAACA
miR-96GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATA-
CGACAGCAAAAATGTG
miR-148aGTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTG-
GATACGACAGTCGGAG
miR-155GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATA-
CGACACCCCTATCACG
miR-196aGTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATA-
CGACCCCAACAACATG
miR-210GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATAC-
GACTCAGCCGCTGTC
miR-217GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATA-
CGACTCCAATCAGTTC
Table 2. Real-time qPCR primers.
Table 2. Real-time qPCR primers.
Primer Name:Primer Sequence:
Universal primerATCCAGTGCAGGGTCCGAGG
mir-39 C. elegansGCGGCGGAGCTGATTTCGTCTTG
mir-21GCGGCGGTAGCTTATCAGACTG
miR-96GCGGCGGTTTGGCACTAGCAC
miR-148aGCGGCGGAAAGTTCTGAGACACTCC
miR-155GCGGCGGTTAATGCTAATCGTG
miR-196aGCGGCGGTAGGTAGTTTCATGTTG
miR-210GCGGCGGCTGTGCGTGTGACAG
Table 3. MicroRNAs expression fold change and clinical data in UCOGC.
Table 3. MicroRNAs expression fold change and clinical data in UCOGC.
IDClinical DatamiRNA
DiagnosisSexAgeStageSurvival (Months)StatusmiR-21miR-96miR-148amiR-155miR-196amiR-210miR-217
1UCOGCM73pTXN14Dead13.250.420.486.341.4845.880.06
2UCOGCF76pT3N00.1Dead12.420.260.613.730.9614.840.03
3UCOGCM61pT3N09Dead10.920.560.393.350.914.490.026
4UCOGCM53pT3N158Alive16.680.271.062.180.437.920.02
5UCOGCM63pT1cN057Alive44.790.380.765.610.8939.980.08
6UCOGCM67pT3N11Dead71.990.791.420.964.060.17
7UCOGC + PDACM63pT2N16Dead15.790.920.1716.630.4490.70.07
8UCOGC + IPMNF50pT1bN086Alive34.392.210.463.190.4515.880.06
9UCOGCM59pT2N0168Dead11.626.222.121.691.144.390.05
10UCOGC + PDACM75pT3N16Dead19.390.140.374.920.753.990.01
Min and max values are shown in red.
Table 4. MicroRNAs expression fold change and clinical data in G3 PDAC.
Table 4. MicroRNAs expression fold change and clinical data in G3 PDAC.
IDClinical DatamiRNA
DiagnosisSexAgeStageSurvival (Months)StatusmiR-21miR-96miR-148amiR-155miR-196amiR-210miR-217
1PDACM69pT3N123Dead7.830.4560.683.461.37211.170.025
2PDACM68pT3N118Dead13.160.8241.416.620.79728.070.105
3PDACM83pT3N129Dead3.760.3960.8472.270.553.90.083
4PDACF71pT3N17Dead11.110.6631.2710.581.0697.960.031
5PDACM63pT3N145Dead53.511.450.13780.671.5126.880.001
6PDACM59pT3N115Dead8.080.550.1780.861.043.610.006
7PDACM60pT3N19Dead4.611.912.167.741.2059.440.075
8PDACF66pT3N123Dead12.2810.4939.181.2813.810.125
9PDACM64pT3N112Dead5.90.410.11654.260.99641.980.002
10PDACF53pT4N111Dead14.960.760.232232.361.095.90.031
11PDACM67pT3N027Dead28.431.4970.519121.051.4221.350.236
12PDACF77pT3N09Dead35.40.7920.05310.561.465.340.103
Min and max values are shown in red.
Table 5. Comparison of miRNAs expression fold change in UCOGC and G3 PDAC. Data are presented as means ± standard deviation (SD). Statistically significant differences between cancer and normal tissue samples are shown in bold. Significant differences between studied types of pancreatic cancers are highlighted in red. The Mann–Whitney test was used for the calculations. p-values of p < 0.05 were considered statistically significant.
Table 5. Comparison of miRNAs expression fold change in UCOGC and G3 PDAC. Data are presented as means ± standard deviation (SD). Statistically significant differences between cancer and normal tissue samples are shown in bold. Significant differences between studied types of pancreatic cancers are highlighted in red. The Mann–Whitney test was used for the calculations. p-values of p < 0.05 were considered statistically significant.
Cancer TypemiRNAs Fold Change
miR-21miR-96miR-148amiR-155miR-196amiR-210miR-217
UCOGC18.62± 8.5391.34 ± 1.2810.72± 0.3694.91± 2.7750.84± 0.25923.21±21.3840.05± 0.031
PDAC16.58± 11.2630.89 ± 0.3810.67± 0.49845.8± 50.8561.14 ± 0.2219.95± 13.9770.06± 0.052
Table 6. Target genes for the oncomirs in pancreatic tumors.
Table 6. Target genes for the oncomirs in pancreatic tumors.
Oncomirs
(Upregulated)
miRNA TargetsmiRNA
Enhances
References
miR-21CDK2AP1, Pdcd, BCL2, PTEN, and almost 30 genesproliferation, invasion, chemoresistance, tumor survival[14,15,63,64]
miR-155TP53INP1, FOXO3a, and SOCS1proliferation, transformation, migration, invasion[38,59,65]
miR-196aNFKBIAproliferation, migration, invasion[66]
miR-210Ephrin-A3, MNTproliferation, angiogenesis, tumor growth, survival[26,67]
Table 7. Tumor-suppressing miRNAs in pancreatic cancer.
Table 7. Tumor-suppressing miRNAs in pancreatic cancer.
Tumor Suppressors (Downregulated)miRNA TargetsmiRNA
Inhibits
References
miR-96KRASproliferation, migration[87,88,89]
miR-148aPHLDA2, LPCAT2, and AP1S3proliferation, migration, invasion[32,90]
miR-217KRAS, SIRT1proliferation, migration[37,65]
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

Popov, A.; Hrudka, J.; Szabó, A.; Oliverius, M.; Šubrt, Z.; Vránová, J.; Ciprová, V.; Moravcová, J.; Mandys, V. Expression of Selected miRNAs in Undifferentiated Carcinoma with Osteoclast-like Giant Cells (UCOGC) of the Pancreas: Comparison with Poorly Differentiated Pancreatic Ductal Adenocarcinoma. Biomedicines 2024, 12, 962. https://doi.org/10.3390/biomedicines12050962

AMA Style

Popov A, Hrudka J, Szabó A, Oliverius M, Šubrt Z, Vránová J, Ciprová V, Moravcová J, Mandys V. Expression of Selected miRNAs in Undifferentiated Carcinoma with Osteoclast-like Giant Cells (UCOGC) of the Pancreas: Comparison with Poorly Differentiated Pancreatic Ductal Adenocarcinoma. Biomedicines. 2024; 12(5):962. https://doi.org/10.3390/biomedicines12050962

Chicago/Turabian Style

Popov, Alexey, Jan Hrudka, Arpád Szabó, Martin Oliverius, Zdeněk Šubrt, Jana Vránová, Vanda Ciprová, Jana Moravcová, and Václav Mandys. 2024. "Expression of Selected miRNAs in Undifferentiated Carcinoma with Osteoclast-like Giant Cells (UCOGC) of the Pancreas: Comparison with Poorly Differentiated Pancreatic Ductal Adenocarcinoma" Biomedicines 12, no. 5: 962. https://doi.org/10.3390/biomedicines12050962

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