miRNA expression profiling regulates necroptotic cell death in hepatocellular carcinoma

  • Authors:
    • Maria Visalli
    • Marcello Bartolotta
    • Francesca Polito
    • Rosaria Oteri
    • Adalberto Barbera
    • Roberto Arrigo
    • Rosa Maria Di Giorgio
    • Giuseppe Navarra
    • M'hammed Aguennouz
  • View Affiliations

  • Published online on: May 17, 2018     https://doi.org/10.3892/ijo.2018.4410
  • Pages: 771-780
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) is one of the most aggressive types of cancer and is among the leading causes of cancer-related mortality worldwide. Although the dysregulation of microRNAs (miRNAs or miRs) has often been reported in HCC, the precise molecular mechanisms by which miRNAs modulate the process of tumorigenesis and the behavior of cancer cells are not yet clearly understood. In this study, we identified a novel three‑miRNA signature, including miR‑371-5p, miR‑373 and miR‑543, that appears to orchestrate programmed cell necrosis in HCC by directly targeting the caspase‑8 gene (Casp‑8). Our results demonstrated that miR‑371-5p, miR‑373 and miR‑543 were overexpressed in HCC tissues compared with paired adjacent normal tissues. The upregulation of these miRNAs specifically and markedly downregulated the expression of Casp‑8, as well as significantly enhanced the Z-VAD/TNF‑α-induced necroptosis of HCC cells. By contrast, the selective knockdown of miRNA expression led to a significant increase in Casp‑8 levels and a marked reduction in programmed cell necrosis. Intriguingly, the sustained overexpression of Casp‑8 reversed the pro‑necroptotic effects exerted by miRNA mimics. Finally, a strong inverse association between the level of miR‑223 and the expression levels of nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing-3 inflammasome was observed in our HCC specimens. On the whole, the present study revealed a molecular link between the three‑miRNA signature, comprising miR‑371-5p, miR‑373 and miR‑543, and the negative necroptotic regulator Casp‑8, and presents evidence for its employment as a novel potential diagnostic, prognostic and therapeutic target in HCC.

Introduction

Hepatocellular carcinoma (HCC) is one of the most aggressive malignant tumors with respect to the disease course and mortality rate (1) Although numerous therapeutic strategies have been introduced, the prognosis of patients with HCC remains poor due to the frequent metastasis and recurrence (2,3). Therefore, it is crucial to identify specific molecular targets and provide the basis for early diagnosis of HCC and targeted therapy, in order to achieve optimal outcomes and improve the quality of life and life expectancy of HCC patients.

MicroRNAs (miRNAs or miRs) are small, non-coding, single-stranded RNAs that regulate key cell processes, such as proliferation, differentiation and apoptosis (49), and exert major functional effects on various disorders, including numerous, if not all, cancers (1013). Abnormalities in miRNA expression profiles may be caused by different mechanisms, such as amplifications, deletions, or sequence changes in the miRNA loci, epigenetic silencing and the dysregulation of transcription factors targeting specific miRNAs (14). Over the past decades, miRNAs have provided a powerful tool for health care intervention in a variety of tumor types (1013). Increasing interest in the development of miRNAs as novel therapeutic biomarkers stems from the ability to interfere with miRNA levels through selective miRNA mimics or anti-miRs (15).

Recently, a number of miRNAs have been implicated as modulators of cell necroptosis (16,17), a newly discovered pathway of regulated cell necrosis governed by receptor-interacting protein (RIP)1, RIP3 and mixed lineage kinase domain-like (MLKL) protein, which constitutes a critical cell-killing mechanism in response to severe stress, chemotherapy or inflammatory factors (18). Previous studies have demonstrated that necroptosis is associated with cancer initiation and progression, playing a dual and opposing role. Accordingly, while enhanced necroptosis concurs with cancer cell death, extensive cell death also increases the risk of proliferation and the metastasis of surviving cells by promoting the formation of reactive oxygen species, triggering the inflammatory process and inhibiting the immune response (19). In this regard, emphasis has been placed on the crucial role of certain miRNAs in the transcriptional regulation of the nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing−3 (NRLP3) inflammasome, an intracellular multiprotein complex that contributes to necroptosis upon the production of pro-inflammatory cytokines (20), with activity involved in the pathogenesis and progression of several cancers, including HCC (21,22). In this context, recent investigations revealed that the caspase-8 gene (Casp-8) is a key controller of NLRP3 activation (2326); however, the mechanisms underlying this process have not yet been fully elucidated.

The aim of the present study was to identify potential interactions between a panel of miRNAs predicted by computational scoring as necroptotic pathway regulators and Casp-8 in HCC specimens and cell lines. We identified three miRNAs, namely miR-371-5p, miR-373 and miR-543, which were upregulated in HCC tissues compared with adjacent non-cancerous tissues. These miRNAs have the ability to bind to their target Casp-8 mRNA transcript in order to facilitate the necroptosis of HCC cells. Additionally, the data presented herein confirm the well-known association between miR-223 and NLRP3, as already reported in different disease contexts (2731).

The findings of this study identified a key piece in the complex molecular puzzle of HCC, thereby providing new insight for a better understanding of the pathogenic mechanisms of this type of tumor. The assessment of the necroptotic pathway regulation by miRNAs depicts the starting point for a novel miRNA-based diagnostic, prognostic and therapeutic approach in HCC.

Materials and methods

Patient selection

This a retrospective study of tumor tissues from 38 patients with HCC treated by surgical resection at the University Hospital of Messina between 2011 and 2015. This study was approved by the local Institutional Review Board (IRB) of the University Hospital of Messina, Messina, Italy. The study protocol conforms to the ethical guidelines of the 1975 Declaration of Helsinki. Written informed consent was obtained from all subjects at the moment of the surgical procedures. For research purposes, no consent for retrospective de-identified data analysis was required as deemed by the IRB.

No patients received neoadjuvant therapy (radiotherapy and/or chemotherapy). The diagnosis of HCC was based on explant evaluation according to the revised WHO classification tumors (32). The clinical characteristics of the patients are presented in Table I.

Table I

Clinical characteristics of the patients with HCC.

Table I

Clinical characteristics of the patients with HCC.

Sex (male/female)27/11
Mean age (range, years)68±11
HBsAg/Anti-HCV/NBNC8/13/17
Child-Pugh grading (A/B/C)21/17/0
Stage (I/II/IIIA/IIIB/IIIC/IV)
0/26/12/0/0/0
Total bilirubin (mg/dl)44±10
Albumin (g/dl)3.5± 0.4
PT (INR)4±0.8
Ascites (−/+)20/18

[i] HBsAg, hepatitis B surface antigen; anti-HCV, antibodies against hepatitis C; NBNC, non-B non-C hepatitis.

Tissue samples

HCC tissues and their normal liver counterparts were frozen immediately after surgery and stored at −80°C for biomolecular analyses.

Cells and cell culture

The Hep3B-hCG cells were purchased from Sigma-Aldrich Italy (Milan, Italy) and maintained in Dulbecco's modified Eagle's medium (DMEM) with high glucose (HyClone, Logan, UT, USA) supplemented with 10% FBS. The hNHeps™ (human normal hepatocytes) cells were purchased from Lonza (Lonza, Walkersville, MD, USA) and maintained in hepatocyte culture medium (Lonza).

RNA extraction, cDNA synthesis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was extracted from the HCC tissues and cell cultures using TRIzol reagent (Invitrogen/Thermo Fisher Scientific, Inc., Waltham, MA USA), according to the manufacturer's instructions. Total RNA concentration and integrity were examined using an Agilent Bioanalyzer (Agilent Technologies, Inc., Santa Clara, CA, USA). Subsequently, 300 ng of total mRNA per sample was reverse transcribed into cDNA using the High Capacity cDNA Reverse Transcription kit (Applied Biosystems/Thermo Fisher Scientific, Inc.).

RT-qPCR for Casp-8 and NLRP3 expression was performed using a standard TaqMan PCR kit procedure on an AB-7300 RT-PCR system (Applied Biosystems/Thermo Fisher Scientific, Inc.) The reaction was performed with one cycle of 95°C for 5 min followed by 40 cycles at 95°C for 15 sec, 52°C for 20 sec and 72°C for 30 sec. The mRNA levels of Casp-8 and NLRP3 were normalized to endogenous β-actin (Applied Biosystems). Primers and probes were as previously reported (33,34).

The control was represented by hNHeps™ cells (described above) whose expression was considered as equal to 1

Relative fold expression and changes were calculated using the 2−∆∆Cq method (35).

miRNA isolation, cDNA synthesis and qPCR analysis

miRNAs were extracted from the HCC tissues and cells using the miRVana Isolation kit (Ambion/Thermo Fisher Scientific, Inc.), following the manufacturer's instructions. The enriched miRNA fraction was converted in cDNA using the TaqMan MicroRNA Reverse Transcriptase kit (Applied Biosystems/Thermo Fisher Scientific, Inc.). hsa-miR-137, hsa-miR-223, hsa-miR-324, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373, hsa-miR-455, hsa-miR-543, hsa-miR-595, and hsa-miR-661 were predicted by computational scoring as necroptotic pathway regulators (36,37).

For miRNA quantification, 2 µl of cDNA was used for each specific miRNA TaqMan Small RNA assay (Applied Biosystems/Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. All reverse transcription reactions, including no-template controls and reverse transcription controls, were performed in triplicate. RNU6 small nuclear RNA was used to normalize miRNA expression levels owing to its claimed expression stability and its wide use as loading control in several published miRNA expression studies (38,39). Relative fold expression and changes were calculated using the 2−ΔΔCq method (35).

Target prediction tools

miRNA/Casp-8 mRNA interactions were identified by means of bioinformatics algorithms that predict miRNA target sites (40). Specifically, three online databases, microRNA.org (www.microrna.org), miRDB (http://mirdb.org/miRDB/), and TargetScan (www.targetscan.org), were used.

Oligonucleotide transfection

hsa-miR-371-5p, hsa-miR-373 and hsa-miR-543 mimics/inhibitors (Qiagen, Milan, Italy) were transfected into the Hep3B-hCG cells using HiPerFect according to the manufacturer's instructions (Qiagen). The cells were transfected twice with 100 pmol of oligonucleotide per well (0.5×106 cells) at 24-h intervals. The transfected cells were assayed 48 h after the second transfection.

Plasmid constructs and transient transfection

The plasmid containing the full-length Casp-8 cDNA was obtained as previously described (33). The 3′-UTR of Casp-8 and the mutation sequences were amplified by PCR using the primers with a BglII restriction site on each 5′ or 3′ strand. The PCR products were inserted into the BglII sites of the pGL3-control vector (Promega, Madison, WI, USA) and identified by DNA sequencing.

The wild-type plasmids were created containing the 3′UTR of Casp-8 with the complementary sequence of miR-371-5p (pGL3-Casp-8-3′UTR wild 1), miR-373 (pGL3-Casp-8-3′UTR wild 2) and miR-543 (pGL3-Casp-8-3′UTR wild 3). Three mutant plasmids were generated without the complementary sequence of miR-371-5p (pGL3-Casp-8-3′UTR mut 1), miR-373 (pGL3-Casp-8-3′UTR mut 2) and miR-543 (pGL3-Casp-8-3′UTR mut 3).

For the luciferase reporter assay, the cells were seeded on 24-well plates and co-transfected using Lipofectamine 2000 (Invitrogen/Thermo Fisher Scientific, Inc.) with 100 ng per well of the resulting luciferase UTR-report vector, 2 ng per well of pRLCMV vector (internal control, Promega), and 20 ng per well of miR-371-5p, miR-373 and miR-543 mimics or inhibitors following the manufacturer's instructions (Qiagen). After 24 h, the cells were lysed, and the relative luciferase activity was assessed with the Dual-Luciferase Assay Reporter System (Promega).

Necroptosis assay

The Hep3B-hCG cells (2×103 cells/well) were pre-treated with the pan-caspase inhibitor, Z-VAD (10 µM), for 1 h in order to induce RIPK3-dependent necroptosis, in the presence or absence of miRNA mimics and inhibitors. Tumor necrosis factor (TNF)-α (50 ng/ml) was then added and the cells were incubated for an additional 72 h. Cell viability was assessed by MTT assay. In brief, 20 µl of 5 mg/ml stock solution of 3-(4,5-dimetylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (Sigma-Aldrich) was added to each well, and the cells were incubated for 1 h at 37°C in a humidified atmosphere of 5% CO2. The supernatants were carefully removed, and 100 µl of DMSO (vehicle) was added to each well. The plates were subsequently shaken for 20 min at room temperature, and the absorbance of the solution was measured at 570 nm. The absorbance of the untreated cells was set as 100% and cell survival was expressed as a percentage of this value.

Statistical analysis

Statistical analysis was performed using INSTAT, version 3.0 and PRISM version 4.0 software (GraphPad, San Diego, CA, USA). One-way analysis of variance (ANOVA), followed by a pair-wise multiple comparison test (Bonferroni correction), was performed to identify the differences among the groups. The association between Casp-8 mRNA expression levels and clinicopathological characteristics was evaluated using Pearson's Chi-square test. Pearson's correlation analysis was used to estimate the correlation between the Casp-8 mRNA levels and the miR-371-5p, miR-373 and miR-543 expression profiles, as well as between NLRP3 abundance and miR-223 expression. A value of P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of Casp-8 in HCC samples

Casp-8 is a cysteine protease that is a key regulator of cell apoptosis; however, its functional significance as a negative regulator of programmed cell necrosis has attracted growing attention over the years (4143). Since different programmed cell death pathways, including necroptosis, have been recently implicated in hepatocarcinogenesis (44), in this study, we addressed the question of whether Casp-8 plays a role in the programmed necrotic death of HCC cells. The values for Casp-8 mRNA levels were first determined in the normal liver (NL) samples in order to determine the cut-off point for Casp-8 expression levels in the HCC samples. Casp-8 expression levels (as defined by the ratio between the values measured in NL samples over those of hNHeps™ cells) were between 0.95 and 1.6 (mean, 1.29±0.16). Values ≤0.81 (determined as the mean minus 3 s.d.) were considered to represent the underexpression of Casp-8. As shown in Fig. 1A, a marked decrease in the Casp-8 mRNA level was found in 34/38 (89.5%) HCC samples. Box plots showing the individual means and scores of Casp-8 mRNA expression in the NL and HCC specimens are presented in Fig. 2B. Likewise, a marked decrease in Casp-8 expression levels was observed in the HCC cell line, Hep3B-hCG, compared with the normal hepatocyte cell line, hNHeps™ (Fig. 1C). In addition, in order to gain insight into the role of Casp-8 in HCC, we analyzed the association between Casp-8 expression and various patient characteristics, including gender, age, Child-Pugh grade and stage (Table II). No significant association was observed between Casp-8 expression and the clinical variables considered (P>0.05).

Table II

Association between Casp-8 expression and clinicopathological characteristics.

Table II

Association between Casp-8 expression and clinicopathological characteristics.

VariablesCasp-8 mRNA expression
P-value
Low (n-34)High (n=4)
Sex0.854
 Male243
 Female101
Age (years)0.911
 ≤60162
 ≥60182
Child-Pugh grading0.401
 A183
 B161
Stage0.765
 II233
 IIIA111
Prediction of miRNAs regulating necroptotic cell death that target Casp-8

To identify the potential upstream executioners of Casp-8 signaling in necrotic cascades, we first explored the expression profiles of a panel of miRNAs predicted by computational scoring as necroptotic pathway regulators (36,37). Significantly higher levels of hsa-miR-137, hsa-miR-324, hsa-miR-371-5p, hsa-miR-372, hsa-miR-373, hsa-miR-455, hsa-miR-543, hsa-miR-595 and hsa-miR-661, and lower levels of miR-223, were observed in the HCC tissues compared with the NL specimens (Fig. 2A). A similar expression pattern of this miRNA panel was noted in the HCC cell line, Hep3B-hCG, compared with the normal hepatocyte cell line, hNHeps™ (Fig. 2B). Subsequently, among these miRNAs, we searched for those that may have seed regions able to bind to the 3′UTR of Casp-8 by using predictive bioinformatics programs (microRNA.org, miRDB and TargetScan). We identified miR-371-5p, miR-373 and miR-543 as potential putative Casp-8-targeting miRNAs. Of note, Pearson's correlation analysis revealed a strong negative correlation between the miR-371-5p, miR-373 and miR-543 expression profiles and Casp-8 mRNA levels in the paired HCC tumor tissues (r=−0.689 for miR-371-5p; r=−0.611 for miR-373; and r=−0.606 for miR-543).

miR-371-5p, miR-373 and miR-543 participate in the regulation of Casp-8 expression

To validate the direct interaction of miR-371-5p, miR-373 and miR-543 with Casp-8 mRNA (Fig. 3A), wild-type plasmids containing the 3′UTR of Casp-8 with complementary sequence of miR-371-5p (pGL3-Casp-8-3′UTR wild 1), miR-373 (pGL3-Casp-8-3′UTR wild 2) and miR-543 (pGL3-Casp-8-3′UTR wild 3) and the mutant plasmids without complementary sequence of miR-371-5p (pGL3-Casp-8-3′UTR mut 1), miR-373 (pGL3-Casp-8-3′UTR mut 2) and miR-543 (pGL3-Casp-8-3′UTR mut 3) were co-transfected into the Hep3B-hCG cells together with specific mimics or inhibitors. We found that the enforced expression of miR-371-5p, miR-373 and miR-543 induced a marked decrease in Casp-8 expression (Fig. 3B), accompanied by a significant decrease in the luciferase activity of pGL3-Casp-8-3′UTR wild-type plasmids (Fig. 3C; P<0.001), whereas the transcription of the pGL3-Casp-8-3′UTR mut vectors was not affected (Fig. 3C). Upon blocking the expression of the three miRNAs with specific inhibitors, luciferase intensity markedly increased in cells transfected with the pGL3-Casp-8-3′UTR wild-type vectors (P<0.001). No change was observed in the cells transfected with the pGL3-Casp-8-3′UTR mut plasmids (Fig. 3D). These results provide evidence of Casp-8 as a target gene of miR-371-5p, miR-373 and miR-543 in our setting.

miR-371-5p, miR-373 and miR-543 regulated necroptosis of HCC cells by targeting Casp-8

Only a limited number of studies to date have reported the mechanisms through which miRNAs regulate necroptotic cell death (16,17). Of note, miR-874 has been shown to enhance necroptosis by targeting Casp-8 (45). Hence, given the functional interaction of miR-371-5p, miR-373 and miR-543 with Casp-8, in this study, we attempted to elucidate whether these miRNAs play a role in the necrotic cascade orchestrated by Casp-8. Hep3B-hCG cells were first pre-treated with the pan-caspase inhibitor, Z-VAD (10 µM), in the presence or absence of miRNA mimics and inhibitors, and then exposed to TNF-α. The enforced expression of miR-371-5p, miR-373 and miR-543 markedly increased Z-VAD/TNF-α-induced cell necroptosis, whereas the knockdown of miR-371-5p, miR-373 and miR-543 led to a marked reduction of programmed cell necrosis. Of note, the pro-necroptotic effects exerted by the miRNA mimics were abolished when Casp-8 was ectopically overexpressed, thus indicating that this three-miRNA signature was able to regulate the necroptosis of Hep3B-hCG cells via targeting Casp-8 (Fig. 4).

Inverse association between miR-223 and NLRP3 in HCC samples

The miR-223-dependent post-transcriptional regulation of the NLRP3 inflammasome has been well validated in a variety of cell types (27,30); however, this functional link has not been established yet in HCC. Therefore, in this study, we proceeded to investigate whether an inverse correlation between miR-223 abundance and NLRP3 mRNA expression also exists in HCC. A significant decrease in the miR-223 level was accompanied by a corresponding increase in NLRP3 in the HCC samples compared with the NL specimens (r=−0.3455, P<0.01), supporting the hypothesis that miR-223 likely also targets NLRP3 in HCC (Fig. 5).

Discussion

Necroptosis has recently attracted increasing attention as a newly identified type of programmed cell death that occurs when cells are subjected to severe stress or are exposed to chemotherapeutic agents or inflammatory factors (4649). This type of regulated necrosis has been implicated in the pathogenesis of a variety of human diseases, such as neurodegeneration, ischemic reperfusion injury, Gaucher's disease, progressive atherosclerotic lesions, TNF-mediated hypothermia and systemic inflammation (50). However, the role of necroptosis in cancer remains controversial, as some authors consider this phenomenon a double-edged sword (19). Indeed, although a number of studies have reported that necroptosis prevents the onset and progression of cancer and promotes its therapy by proving a plethora of cancer cell lines highly sensitive and responsive to necroptosis inducers (18), emerging data have revealed that necroptosis may in fact contribute to cancer development (5153). The rapid and extensive release of damage-associated molecules [e.g., interleukin (IL)-1 family cytokines] from necroptotic cancer cells may act as a potent inducer of inflammation (54), which in turn leads to tumorigenesis through the stimulation of angiogenesis and metastasis, as well as to the mitigation of the adaptive immune responses (55,56). miRNAs were recently identified as new participants in regulated necrosis (57); however, the underlying mechanisms have not yet been fully elucidated. The findings of the present study indicated that a novel three-miRNA signature, including miR-371-5p, miR-373 and miR-543, is critically involved in programmed necroptotic death of HCC cells. This assertion was strongly supported by functional experiments in which enforced expression of these miRNAs markedly increased Z-VAD/TNF-α-induced necroptosis in Hep3B-hCG cells, whereas their specific knockdown was able to significantly attenuate programmed necrosis (Fig. 4).

Indeed, among all the miRNAs initially predicted by computational scoring as necroptotic pathway regulators, we restricted our investigations to those targeting the necroptosis inhibitor Casp-8 gene, i.e., miR-371-5p, miR-373 and miR-543. Consequently, since miR-137, miR-324, miR-371-5p, miR-372, miR-373, miR-455, miR-543, miR-595 and miR-661 did not present regions able to bind to the 3′UTR of Casp-8 according to the target prediction results, they were not included in our subsequent experimental analyses. However, these miRNAs will be the subject of our further (in-progress) studies aimed at defining other metabolic pathways involved in necroptosis.

From the results obtained by the luciferase assay experiments, we found that miR-371-5p, miR-373 and miR-543 suppressed the expression of Casp-8 through direct interaction with the 3′UTR of the target (Fig. 3). Of note, emerging evidence indicates that Casp-8 is an important negative regulator of necroptosis (43). Treatment with Z-VAD, a pan-caspase inhibitor, was found to promote necroptosis, since Casp-8, when inactivated, cannot cleave the two key inducers of necroptosis, namely RIP1 and RIP3 (43,58,59). Moreover, the pharmacological inhibition of Casp-8 induces the autocrine production of TNF, which triggers necroptosis (60). The results reported herein may shed new light into the mechanisms regulating programmed necrosis in HCC, revealing that the three miRNAs of interest induce necroptosis by suppressing Casp-8. Accordingly, we observed that the pro-necroptotic effects exerted by miRNA mimics were abolished when Casp-8 was ectopically overexpressed (Fig. 4). Higher expression levels of miR-371-5p, miR-373 and miR-543 were significantly associated with lower amounts of Casp-8 mRNA in HCC tissues compared with paired normal tissues (Figs. 1 and 2), thereby supporting the key role of these miRNAs in controlling Casp-8 expression.

Of note, miR-373 has been previously implicated in several types of cancers as an oncomiR (61), with its down-regulation resulting in the growth arrest and the apoptosis of tumor cells (62). Moreover, the increased expression levels of miR-371-5p have been associated with a shorter survival and a poor prognosis of patients with pancreatic cancer (63). Similarly, miR-543 has been found to be closely associated with tumor size, TNM stage and metastasis, as well as with a poor overall and disease-free survival (64). In this context, our study may provide new insight into the necroptotic function of this three-miRNA signature, further providing valuable insights into its role in HCC and its value as a potential predictor of poor prognosis. Indeed, the present findings are in line with those of a recent study reporting that pro-necroptotic pathways promote inflammatory cytokine production, thereby sustaining tumor growth (51).

Another meaningful result of this study was the significant inverse correlation between the expression pattern of miR-223-3p and that of the NLRP3 inflammasome found in the resected tissue specimens from HCC patients (Fig. 5). Specifically, we demonstrated that the downregulation of miR-223 was closely associated with increased NLRP3 mRNA levels. This finding effectively supports the hypothesis that miR-223 acts as a negative regulator of NLRP3 inflammasome activity, as it was already observed in other pathological conditions (27,29). It is well recognized that NLRP3 is an important mediator involved in cell necroptosis (30), and it has been suggested that its activation may be modulated by Casp-8 (23,26). Of note, a recent study demonstrated that miR-223-3p prevents the ischemia/reperfusion-induced cardiac necroptosis via targeting NLRP3 inflammasome signaling (17). Therefore, the data presented herein may uncover a novel role for miR-223-3p and its target gene, NLRP3, in the context of HCC, strengthening the inextricable association between these molecular effectors involved in programmed cell necrosis. Defining the role of miR-223-3p-mediated NLRP3 signaling in these and other potential necroptotic models is mandatory for the better understanding the complex crosstalk regulating programmed cell death and for testing new disease-specific medical treatments.

In conclusion, the results from the present study may provide new insight into the complex miRNA-governed network of regulated necrosis, and suggest that the newly identified signature of necroptosis-regulating miRNAs may represent a promising diagnostic and prognostic biomarker as well an effective therapeutic target for HCC.

However, further studies are required to verify the interplay between these miRNAs, their target genes and HCC progression, and to validate our findings for applicability in the clinical setting.

Acknowledgments

Not applicable.

Funding

No funding was received.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

MV, FP, RMDG, MB and MA contributed to the conception and design of the study; MB, AB and GN were involved in the selection of the biopsy tissues. FP, RO, RA and MA acquired the data; MV, FP and MA analyzed and interpreted the data; RMDG, MB and GN participated in the revision of the article. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

This study was approved by the local Institutional Review Board (IRB) of the University Hospital of Messina, Messina, Italy. The study protocol conforms to the ethical guidelines of the 1975 Declaration of Helsinki. Written informed consent was obtained from all subjects at the moment of the surgical procedures. For research purposes, no consent for retrospective de-identified data analysis was required as deemed by the IRB.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Mlynarsky L, Menachem Y and Shibolet O: Treatment of hepatocellular carcinoma: Steps forward but still a long way to go. World J Hepatol. 7:566–574. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Tang ZY: Hepatocellular carcinoma - cause, treatment and metastasis. World J Gastroenterol. 7:445–454. 2001. View Article : Google Scholar

4 

Herranz H and Cohen SM: MicroRNAs and gene regulatory networks: Managing the impact of noise in biological systems. Genes Dev. 24:1339–1344. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Arner P and Kulyté A: MicroRNA regulatory networks in human adipose tissue and obesity. Nat Rev Endocrinol. 11:276–288. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Fang S, Deng Y, Gu P and Fan X: MicroRNAs regulate bone development and regeneration. Int J Mol Sci. 16:8227–8253. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Kohlhapp FJ, Mitra AK, Lengyel E and Peter ME: MicroRNAs as mediators and communicators between cancer cells and the tumor microenvironment. Oncogene. 34:5857–5868. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Su Y, Wu H, Pavlosky A, Zou LL, Deng X, Zhang ZX and Jevnikar AM: Regulatory non-coding RNA: New instruments in the orchestration of cell death. Cell Death Dis. 7:e23332016. View Article : Google Scholar : PubMed/NCBI

9 

Tran DDH, Kessler C, Niehus SE, Mahnkopf M, Koch A and Tamura T: Myc target gene, long intergenic noncoding RNA, Linc00176 in hepatocellular carcinoma regulates cell cycle and cell survival by titrating tumor suppressor microRNAs. Oncogene. 37:75–85. 2018. View Article : Google Scholar

10 

Allegra A, Alonci A, Campo S, Penna G, Petrungaro A, Gerace D and Musolino C: Circulating microRNAs: New biomarkers in diagnosis, prognosis and treatment of cancer (Review). Int J Oncol. 41:1897–1912. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Kunej T, Godnic I, Ferdin J, Horvat S, Dovc P and Calin GA: Epigenetic regulation of microRNAs in cancer: An integrated review of literature. Mutat Res. 717:77–84. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Zhang B, Pan X, Cobb GP and Anderson TA: microRNAs as oncogenes and tumor suppressors. Dev Biol. 302:1–12. 2007. View Article : Google Scholar

13 

Mizuguchi Y, Takizawa T, Yoshida H and Uchida E: Dysregulated miRNA in progression of hepatocellular carcinoma: A systematic review. Hepatol Res. 46:391–406. 2016. View Article : Google Scholar

14 

Croce CM: Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 10:704–714. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Krützfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M and Stoffel M: Silencing of microRNAs in vivo with 'antagomirs'. Nature. 438:685–689. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Afonso MB, Rodrigues PM, Simão AL, Gaspar MM, Carvalho T, Borralho P, Bañales JM, Castro RE and Rodrigues CMP: miRNA-21 ablation protects against liver injury and necroptosis in cholestasis. Cell Death Differ. Dec 11–2017.Epub ahead of print. PubMed/NCBI

17 

Qin D, Wang X, Li Y, Yang L, Wang R, Peng J, Essandoh K, Mu X, Peng T, Han Q, et al: MicroRNA-223-5p and -3p cooperatively suppress necroptosis in ischemic/reperfused hearts. J Biol Chem. 291:20247–20259. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Chen D, Yu J and Zhang L: Necroptosis: An alternative cell death program defending against cancer. Biochim Biophys Acta. 1865:228–236. 2016.PubMed/NCBI

19 

Wang T, Jin Y, Yang W, Zhang L, Jin X, Liu X, He Y and Li X: Necroptosis in cancer: An angel or a demon? Tumour Biol. 39:1010428317711539. 2017. View Article : Google Scholar

20 

Sutterwala FS, Haasken S and Cassel SL: Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci. 1319:82–95. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Davis BK, Wen H and Ting JP: The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu Rev Immunol. 29:707–735. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Wei Q, Mu K, Li T, Zhang Y, Yang Z, Jia X, Zhao W, Huai W, Guo P and Han L: Deregulation of the NLRP3 inflammasome in hepatic parenchymal cells during liver cancer progression. Lab Invest. 94:52–62. 2014. View Article : Google Scholar

23 

Gurung P, Anand PK, Malireddi RK, Vande Walle L, Van Opdenbosch N, Dillon CP, Weinlich R, Green DR, Lamkanfi M and Kanneganti TD: FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes. J Immunol. 192:1835–1846. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Weng D, Marty-Roix R, Ganesan S, Proulx MK, Vladimer GI, Kaiser WJ, Mocarski ES, Pouliot K, Chan FK, Kelliher MA, et al: Caspase-8 and RIP kinases regulate bacteria-induced innate immune responses and cell death. Proc Natl Acad Sci USA. 111:7391–7396. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Gurung P and Kanneganti TD: Novel roles for caspase-8 in IL-1β and inflammasome regulation. Am J Pathol. 185:17–25. 2015. View Article : Google Scholar :

26 

Kang TB, Yang SH, Toth B, Kovalenko A and Wallach D: Caspase-8 blocks kinase RIPK3-mediated activation of the NLRP3 inflammasome. Immunity. 38:27–40. 2013. View Article : Google Scholar

27 

Xie XJ, Ma LG, Xi K, Fan DM, Li JG, Zhang Q and Zhang W: Effects of microRNA-223 on morphine analgesic tolerance by targeting NLRP3 in a rat model of neuropathic pain. Mol Pain. 13:1744806917706582. 2017. View Article : Google Scholar

28 

Neudecker V, Haneklaus M, Jensen O, Khailova L, Masterson JC, Tye H, Biette K, Jedlicka P, Brodsky KS, Gerich ME, et al: Myeloid-derived miR-223 regulates intestinal inflammation via repression of the NLRP3 inflammasome. J Exp Med. 214:1737–1752. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Yang Z, Zhong L, Xian R and Yuan B: MicroRNA-223 regulates inflammation and brain injury via feedback to NLRP3 inflammasome after intracerebral hemorrhage. Mol Immunol. 65:267–276. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Bauernfeind F, Rieger A, Schildberg FA, Knolle PA, Schmid-Burgk JL and Hornung V: NLRP3 inflammasome activity is negatively controlled by miR-223. J Immunol. 189:4175–4181. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Haneklaus M, Gerlic M, Kurowska-Stolarska M, Rainey AA, Pich D, McInnes IB, Hammerschmidt W, O'Neill LA and Masters SL: Cutting edge: miR-223 and EBV miR-BART15 regulate the NLRP3 inflammasome and IL-1β production. J Immunol. 189:3795–3799. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Venza M, Visalli M, Biondo C, Oteri R, Agliano F, Morabito S, Teti D and Venza I: Epigenetic marks responsible for cadmiuminduced melanoma cell overgrowth. Toxicol In Vitro. 29:242–250. 2015. View Article : Google Scholar

34 

Vilaysane A, Chun J, Seamone ME, Wang W, Chin R, Hirota S, Li Y, Clark SA, Tschopp J, Trpkov K, et al: The NLRP3 inflammasome promotes renal inflammation and contributes to CKD. J Am Soc Nephrol. 21:1732–1744. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−∆ ∆ C(T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar

36 

Vlachos IS, Kostoulas N, Vergoulis T, Georgakilas G, Reczko M, Maragkakis M, Paraskevopoulou MD, Prionidis K, Dalamagas T and Hatzigeorgiou AG: DIANA miRPath v.2.0: Investigating the combinatorial effect of microRNAs in pathways. Nucleic Acids Res. 40W:W498–W504. 2012. View Article : Google Scholar

37 

Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, Franz M, Grouios C, Kazi F, Lopes CT, et al: The GeneMANIA prediction server: Biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 38(Suppl 2): W214–W220. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Peltier HJ and Latham GJ: Normalization of microRNA expression levels in quantitative RT-PCR assays: Identification of suitable reference RNA targets in normal and cancerous human solid tissues. RNA. 14:844–852. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Song J, Bai Z, Han W, Zhang J, Meng H, Bi J, Ma X, Han S and Zhang Z: Identification of suitable reference genes for qPCR analysis of serum microRNA in gastric cancer patients. Dig Dis Sci. 57:897–904. 2012. View Article : Google Scholar

40 

Krek A, Grün D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, MacMenamin P, da Piedade I, Gunsalus KC, Stoffel M, et al: Combinatorial microRNA target predictions. Nat Genet. 37:495–500. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Feng S, Yang Y, Mei Y, Ma L, Zhu DE, Hoti N, Castanares M and Wu M: Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain. Cell Signal. 19:2056–2067. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Günther C, Martini E, Wittkopf N, Amann K, Weigmann B, Neumann H, Waldner MJ, Hedrick SM, Tenzer S, Neurath MF, et al: Caspase-8 regulates TNF-α-induced epithelial necroptosis and terminal ileitis. Nature. 477:335–339. 2011. View Article : Google Scholar

43 

Oberst A, Dillon CP, Weinlich R, McCormick LL, Fitzgerald P, Pop C, Hakem R, Salvesen GS and Green DR: Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature. 471:363–367. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Vucur M, Reisinger F, Gautheron J, Janssen J, Roderburg C, Cardenas DV, Kreggenwinkel K, Koppe C, Hammerich L, Hakem R, et al: RIP3 inhibits inflammatory hepatocarcinogenesis but promotes cholestasis by controlling caspase-8- and JNK-dependent compensatory cell proliferation. Cell Reports. 4:776–790. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Wang K, Liu F, Zhou LY, Ding SL, Long B, Liu CY, Sun T, Fan YY, Sun L and Li PF: miR-874 regulates myocardial necrosis by targeting caspase-8. Cell Death Dis. 4:e7092013. View Article : Google Scholar : PubMed/NCBI

46 

Chan FK, Shisler J, Bixby JG, Felices M, Zheng L, Appel M, Orenstein J, Moss B and Lenardo MJ: A role for tumor necrosis factor receptor-2 and receptor-interacting protein in programmed necrosis and antiviral responses. J Biol Chem. 278:51613–51621. 2003. View Article : Google Scholar : PubMed/NCBI

47 

Han W, Li L, Qiu S, Lu Q, Pan Q, Gu Y, Luo J and Hu X: Shikonin circumvents cancer drug resistance by induction of a necroptotic death. Mol Cancer Ther. 6:1641–1649. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Newton K and Manning G: Necroptosis and Inflammation. Annu Rev Biochem. 85:743–763. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Long JS and Ryan KM: New frontiers in promoting tumour cell death: Targeting apoptosis, necroptosis and autophagy. Oncogene. 31:5045–5060. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Zhou W and Yuan J: Necroptosis in health and diseases. Semin Cell Dev Biol. 35:14–23. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Liu X, Zhou M, Mei L, Ruan J, Hu Q, Peng J, Su H, Liao H, Liu S, Liu W, et al: Key roles of necroptotic factors in promoting tumor growth. Oncotarget. 7:22219–22233. 2016.PubMed/NCBI

52 

Seifert L, Werba G, Tiwari S, Giao Ly NN, Alothman S, Alqunaibit D, Avanzi A, Barilla R, Daley D, Greco SH, et al: The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature. 532:245–249. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Strilic B, Yang L, Albarrán-Juárez J, Wachsmuth L, Han K, Müller UC, Pasparakis M and Offermanns S: Tumour-cell-induced endothelial cell necroptosis via death receptor 6 promotes metastasis. Nature. 536:215–218. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Pasparakis M and Vandenabeele P: Necroptosis and its role in inflammation. Nature. 517:311–320. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature. 454:436–444. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Korniluk A, Koper O, Kemona H and Dymicka-Piekarska V: From inflammation to cancer. Ir J Med Sci. 186:57–62. 2017. View Article : Google Scholar :

57 

Su Z, Yang Z, Xu Y, Chen Y and Yu Q: MicroRNAs in apoptosis, autophagy and necroptosis. Oncotarget. 6:8474–8490. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Geserick P, Hupe M, Moulin M, Wong WW, Feoktistova M, Kellert B, Gollnick H, Silke J and Leverkus M: Cellular IAPs inhibit a cryptic CD95-induced cell death by limiting RIP1 kinase recruitment. J Cell Biol. 187:1037–1054. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Welz PS, Wullaert A, Vlantis K, Kondylis V, Fernández-Majada V, Ermolaeva M, Kirsch P, Sterner-Kock A, van Loo G and Pasparakis M: FADD prevents RIP3-mediated epithelial cell necrosis and chronic intestinal inflammation. Nature. 477:330–334. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Hitomi J, Christofferson DE, Ng A, Yao J, Degterev A, Xavier RJ and Yuan J: Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell. 135:1311–1323. 2008. View Article : Google Scholar : PubMed/NCBI

61 

Huang Q, Gumireddy K, Schrier M, le Sage C, Nagel R, Nair S, Egan DA, Li A, Huang G, Klein-Szanto AJ, et al: The microRNAs miR-373 and miR-520c promote tumour invasion and metastasis. Nat Cell Biol. 10:202–210. 2008. View Article : Google Scholar : PubMed/NCBI

62 

Stelzer Y, Sagi I and Benvenisty N: Involvement of parental imprinting in the antisense regulation of onco-miR-372-373. Nat Commun. 4:27242013. View Article : Google Scholar : PubMed/NCBI

63 

He D, Miao H, Xu Y, Xiong L, Wang Y, Xiang H, Zhang H and Zhang Z: MiR-371-5p facilitates pancreatic cancer cell proliferation and decreases patient survival. PLoS One. 9:e1129302014. View Article : Google Scholar : PubMed/NCBI

64 

Zhai F, Cao C, Zhang L and Zhang J: miR-543 promotes colorectal cancer proliferation and metastasis by targeting KLF4. Oncotarget. 8:59246–59256. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 53 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Visalli M, Bartolotta M, Polito F, Oteri R, Barbera A, Arrigo R, Di Giorgio RM, Navarra G and Aguennouz M: miRNA expression profiling regulates necroptotic cell death in hepatocellular carcinoma. Int J Oncol 53: 771-780, 2018
APA
Visalli, M., Bartolotta, M., Polito, F., Oteri, R., Barbera, A., Arrigo, R. ... Aguennouz, M. (2018). miRNA expression profiling regulates necroptotic cell death in hepatocellular carcinoma. International Journal of Oncology, 53, 771-780. https://doi.org/10.3892/ijo.2018.4410
MLA
Visalli, M., Bartolotta, M., Polito, F., Oteri, R., Barbera, A., Arrigo, R., Di Giorgio, R. M., Navarra, G., Aguennouz, M."miRNA expression profiling regulates necroptotic cell death in hepatocellular carcinoma". International Journal of Oncology 53.2 (2018): 771-780.
Chicago
Visalli, M., Bartolotta, M., Polito, F., Oteri, R., Barbera, A., Arrigo, R., Di Giorgio, R. M., Navarra, G., Aguennouz, M."miRNA expression profiling regulates necroptotic cell death in hepatocellular carcinoma". International Journal of Oncology 53, no. 2 (2018): 771-780. https://doi.org/10.3892/ijo.2018.4410