Open Access

Altered expression of the IGF2‑H19 locus and mitochondrial respiratory complexes in adrenocortical carcinoma

  • Authors:
    • Patrick Scicluna
    • Stefano Caramuta
    • Hanna Kjellin
    • Cheng Xu
    • Robin Fröbom
    • Monira Akhtar
    • Jiwei Gao
    • Hao Shi
    • Magnus Kjellman
    • Malin Almgren
    • Anders Höög
    • Jan Zedenius
    • Tomas J. Ekström
    • Robert Bränström
    • Weng-Onn Lui
    • Catharina Larsson
  • View Affiliations

  • Published online on: September 27, 2022     https://doi.org/10.3892/ijo.2022.5430
  • Article Number: 140
  • Copyright: © Scicluna et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Abnormalities of the insulin‑like growth factor 2 (IGF2)‑H19 locus with the overexpression of IGF2 are frequent findings in adrenocortical carcinoma (ACC). The present study assessed the expression of RNAs and microRNAs (miRNAs/miRs) from the IGF2‑H19 locus using PCR‑based methods in ACC and adrenocortical adenoma (ACA). The results were associated with proteomics data. IGF2 was overexpressed in ACC, and its expression correlated with that of miR‑483‑3p and miR‑483‑5p hosted by IGF2. The downregulated expression of H19 in ACC compared to ACA correlated with miR‑675 expression hosted by H19. Several proteins exhibited an inverse correlation in expression and were predicted as targets of miR‑483‑3p, miR‑483‑5p or miR‑675. Subsets of these proteins were differentially expressed between ACC and ACA. These included several proteins involved in mitochondrial metabolism. Among the mitochondrial respiratory complexes, complex I and IV were significantly decreased in ACC compared to ACA. The protein expression of NADH:ubiquinone oxidoreductase subunit C1 (NDUFC1), a subunit of mitochondrial respiratory complex I, was further validated as being lower in ACC compared to ACA and normal adrenals. The silencing of miR‑483‑5p increased NDUFC1 protein expression and reduced both oxygen consumption and glycolysis rates. On the whole, the findings of the present study reveal the dysregulation of the IGF2‑H19 locus and mitochondrial respiration in ACC. These findings may provide a basis for the further understanding of the pathogenesis of ACC and may have potential values for diagnostics and treatment.

Introduction

Adrenocortical carcinoma (ACC) is a rare, yet highly aggressive tumor type (1). In addition to gross genomic alterations, mutational events in several key genes involved in the β-catenin pathway (e.g., CTNNB1 and ZNRF3), the p53/Rb pathway (e.g., TP53, CDKN2A and RB1) and chromatin remodeling (e.g., MEN1, TERT and DAXX) are associated with ACC tumorigenesis (1-3). The overexpression of insulin-like growth factor (IGF) II and the IGF2 gene is observed in the majority of ACC; however, this does not occur in adrenocortical adenoma (ACA) or normal adrenal glands (4-6).

The IGF2 gene is located within 90 kb from the H19 gene in chromosomal region 11p15.5 (7). H19 produces an untranslated, yet spliced and polyadenylated transcript with possible tumor suppressor function (8), and IGF2 encodes the growth factor, IGFII. The IGF2-H19 locus is subjected to parental imprinting, which is frequently lost in cancer by the loss of imprinting, leading to the overexpression of IGF2/IGFII (9-11). Furthermore, IGFII has been reported to promote malignant transformation in the mammary gland in vivo (12). However, it has not been found to exert an effect on the tumor phenotype in vitro (13), and only mildly contributes to the development of ACC in vivo (14,15). Given that IGFII acts on the IGF1 receptor (IGF1R), the application of an IGF1R blocker has been attempted in anticancer therapy to counteract IGFII signaling in ACC (16-21). In ACC, H19 expression has been reported to be decreased as compared to that in ACA (6), while IGF2/IGFII expression is known to be markedly increased in the vast majority of ACC cases (4,6,22-24).

MicroRNAs (miRNAs/miRs) are short non-coding RNAs that generally suppress the expression of target genes by mRNA degradation or translational repression (25). Three miRNAs are known to be transcribed from the IGF2-H19 locus, i.e., miR-483-3p and miR-483-5p from IGF2 intron 7, and miR-675 from H19. miR-483-3p is overexpressed in different tumor types, including ACC (13,26-28). Furthermore, the p53 upregulated modulator of apoptosis (PUMA) is a target of miR-483-3p, and the inhibition of miR-483-3p inhibits proliferation and increases apoptosis in vitro, and tumorigenicity in vivo (13,27). miR-483-5p is also overexpressed in ACC (26-29) and their inhibition in vitro reduces cell growth, although it does not affect the apoptosis of ACC cells (27). On the other hand, miR-483-5p has been shown to regulate the N-Myc downstream-regulated gene 2 and promote the invasion of ACC cells (30). Notably, miR-483-5p is detectable in serum samples of patients with ACC, and its high expression level is associated with a degree of malignancy and a poor survival (31-33). The overexpression of miR-675 has been reported in various types of cancer, e.g. gastric, colorectal, esophageal and breast cancer (34-37); however, the decreased expression has also been observed in non-small cell lung cancer (38). In adrenocortical tumors, miR-675 has only been analyzed in a small cohort of 13 patients (4 patients with ACC and 9 patients with ACA), while the significance of its deregulation in ACC remains to be investigated (39).

The present study aimed to comprehensively characterize the expression of RNAs and miRNAs generated from the IGF2-H19 locus in adrenocortical tissues and associate their expression levels with global protein expression profiles in ACA and ACC. The analysis revealed the expression of several proteins involved in mitochondrial respiratory complexes that inversely correlated with the miR-483-5p level. Functionally, miR-483-5p was demonstrated to regulate the expression of the NADH:ubiquinone oxidoreductase subunit C1 (NDUFC1) and the mitochondrial oxygen consumption rate.

Materials and methods

Tissue material

Fresh-frozen tumor tissues were obtained from the Karolinska University Hospital Biobank for patients treated surgically for ACC (n=35) or ACA (n=43) between 1986 and 2010. Tumors were classified according to the WHO criteria (40). In addition, normal adrenal gland samples were obtained from patients undergoing nephrectomy for other reasons, or from patients with adrenocortical tumors, and histologically verified as non-malignant. The samples were obtained with informed consent and the study of the tissue material was approved by the Ethical Committee of the Karolinska University Hospital.

The clinical information for all cases is presented in Table SI. The screening series included 6 cases with ACA and 8 cases with ACC used for proteomics analysis in a previous study by the authors (41). An extended series of samples from nine normal adrenal glands, 43 ACA and 29 ACC (30 samples) was used for the reverse transcription-quantitative PCR (RT-qPCR) analysis of RNAs and miRNAs. The results from the quantification analyses of miR-483-3p and miR-483-5p have been previously published for most cases (27). Additionally, samples from 13 normal adrenal glands, 25 ACA and 25 ACC (29 samples) were used for western blot analyses. The ACA and ACC cases included in the different analyses are presented in Table SI.

Data analysis from TCGA and genotype-tissue expression (GTEx)

Comparisons of IGF2 or H19 RNA expression levels between ACCs and normal adrenal glands, and principal component analysis (PCA) were performed in Gene Expression Profiling Interactive Analysis [GEPIA; (42)]. The RNA expression levels were based on the RNA sequencing expression data of 77 ACC cases from The Cancer Genome Atlas (TCGA) and 128 normal adrenal glands from GTEx. PCA was performed using the 46 genes whose proteins inversely correlated with miR-483-5p and significantly differentially expressed between ACA and ACC (P<0.05, Table SII). Correlations between the expression of IGF2 and H19, IGF2 and miR-483-3p or miR-483-5p, H19 and miR-675 were assessed in the StarBase Pan-Cancer Analysis Platform (43). This platform includes RNA and miRNA sequencing data of 79 ACC samples from TCGA.

Cell line and cell culture

All in vitro cell assays were performed using the NCI-H295R ACC cell line purchased from the American Type Culture Collection (cat. no. CRL-2128; ATCC). The cells were maintained in DMEM:F12 medium (cat. no. 31330038; Gibco; Thermo Fisher Scientific, Inc.) supplemented with 2.5% NuSerum Growth medium (Corning 355100; Thermo Fisher Scientific, Inc.) and 1% insulin-transferrin-selenium basal medium supplement (cat. no. 41400045; Thermo Fisher Scientific, Inc.). The cells were incubated at 37°C in a humidified CO2 incubator. The authenticity of the cell line was re-verified prior to the experiments by genotyping of short tandem repeats (STRs) performed by the National Genomics Infrastructure-Uppsala (SciLifeLab, Uppsala University, Uppsala, Sweden). The NCI-H295R cell line is the only human ACC cell line that is capable of secreting major adrenocortical steroids (44), representing the cellular origin of ACC cells, and it is the most extensively used cell line in in vitro models of ACC.

RT-qPCR analysis

The expression levels of mature miRNAs and RNAs were evaluated using RT-qPCR with an Applied Biosystems 7900HT Fast Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc.). cDNA for miRNA analysis was synthesized from 25 ng total RNA using the TaqMan MicroRNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). miRNA assays for miR-483-3p (ID 002339), miR-483-5p (ID 002338) and miR-675 (ID 002005) were purchased from Applied Biosystems (Thermo Fisher Scientific, Inc.) and normalization was performed using RNU6B (ID 001093). The high Capacity cDNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was adopted to generate cDNA from 100 ng total RNA for RNA expression analysis and specific TaqMan probes targeting IGF2 (ID 00277496_s1) and H19 (ID 00399293_g1) were used for RT-qPCR. Normalization was performed using 18S (ID 99999901). All TaqMan probes were labeled with FAM-MGB (Applied Biosystems); the reverse transcription and qPCR conditions followed the manufacturer's instructions. All reactions were performed in triplicate and the relative expression levels were determined using the ΔΔCq method and reported as 2−ΔΔCq (45).

Proteomics data analysis

Proteomics profiling for 6 cases of ACA and 8 cases of ACC in the screening series has been described in a previous study by the authors (41). The protein expression levels of the screening series were correlated with their corresponding IGF2, H19, miR-483-3p, miR-483-5p and miR-675 transcripts using Pearson's correlation analysis. The mRNA targets of miR-483-3p, miR-483-5p and miR-675 were predicted using TargetScan 7.1 (https://www.targetscan.org/vert_71/). Ingenuity pathway analysis of complex 'omics' data (IPA; IPA version 14400082; Ingenuity® Systems, www. ingenuity.com) was employed to assess the theoretical interactions and cellular networks of the proteins that correlated with IGF2.

Gene ontology (GO) enrichment analysis

The gene signature corresponding to the 46 proteins that inversely correlated with miR-483-5p, and significantly differentially expressed between ACA and ACC in the proteomics cohort (Table SII) was annotated based on biological processes using GO enrichment analysis (http://geneontology.org/). The analysis was based on the PANTHER overrepresentation test (released on August 3, 2019; http://www.pantherdb.org/) and the GO database (released on February 2, 2019). P-values <0.05 following the Bonferroni correction were considered to indicate statistically significant differences.

Transfections with miRNA inhibitors

Transfections were performed by suspending 3×106 NCI-H295R cells in 100 µl Ingenio solution (Mirus Bio) together with 100 pmol mirVana miR-483-5p inhibitor, miR-483-3p inhibitor or Negative Control #1 (MH12629, MH12478 and 4464076, respectively; Thermo Fisher Scientific, Inc.) in Ingenio cuvettes (MIR50121; Mirus Bio). The cells were electroporated using the program T-20 of the Amaxa Nucleofector device (Lonza Group, Ltd.) and then transferred into a culture plate containing pre-warmed complete DMEM:F12 medium. The plate was incubated at 37°C in a humidified CO2 incubator. The cell culture medium was replaced at 24 h post-transfection and harvested for metabolic profiling or western blot analysis following 48 h of transfection. At least three biological replicates were performed for each transfection. The efficiency of miR-483-5p silencing was assessed using RT-qPCR.

Real-time metabolic profiling

Mitochondrial and glycolytic functions were measured using the Mito Stress Test and Glycolysis Stress Test, respectively (Agilent Technologies, Inc.) using the Seahorse XFe24 Analyser (Agilent Technologies, Inc.). The transfected cells were seeded at 1.5×105/well in 24-well plates (Seahorse XF24 V7 PS Culture Microplates, Agilent Technologies, Inc.). The culture media were replaced after 24 h and incubated for a further 24 h at 37°C. Culture media were changed to the Seahorse XF Base Medium (Agilent Technologies, Inc.) supplemented with 5.5 mM glucose, 1 mM L-glutamine and 1 mM sodium pyruvate at pH 7.4 and incubated at 37°C with CO2 for 1 h. For the evaluation of mitochondrial respiration, the oxygen consumption rate (OCR) was measured at the basal level and following sequential loading with 1 µM oligomycin (ATP synthase inhibitor), 0.5 µM carbonyl cyanide p-trifluoromethoxy-phenylhydrazone (FCCP, an uncoupler of oxidative phosphorylation), 1 µM rotenone and antimycin A (complex I and III inhibitor, respectively). For the assessment of glycolysis, the extracellular acidification rate (ECAR) was analyzed at basal conditions and after sequential loading with 1 µM oligomycin (inhibiting ATP synthase in the mitochondria leading to enhanced glycolysis dependency), and 50 mM 2-deoxyglucose (2-DG, a competitive inhibitor of glucose). Both OCR and ECAR were normalized to the total protein content in each well as measured using the Bradford assay (Bio-Rad Laboratories, Inc.).

Western blot analysis

Cell pellets or tissue samples were suspended in RIPA lysis and extraction buffer (cat. no. 89901; Thermo Fisher Scientific, Inc.) supplemented with 1 mM phenylmethanesulfonyl fluoride (PMSF; Sigma-Aldrich; Merck KGaA), 1% cOmplete™ proteinase inhibitor cocktail (Sigma-Aldrich; Merck KGaA) and 1% phosphatase inhibitor cocktail 2 and 3 (Sigma-Aldrich; Merck KGaA). The lysates were vortexed repeatedly and left on ice for 60 min or until homogenization. The protein concentration was quantified using the Bradford assay (Bio-Rad Laboratories). Denatured lysates (40 µg) were separated in 10% NuPAGE™ Bis-Tris denaturing pre-cast gels (Invitrogen; Thermo Fisher Scientific, Inc.) and transferred onto nitrocellulose membranes (cat. no. 88013, Thermo Fisher Scientific, Inc.) at 4°C. The membranes were blocked in 5% BSA or 1% non-fat dried milk (for total OXPHOS WB) in TBS containing 0.1% Tween-20 (Sigma-Aldrich; Merck KGaA). The membranes were then incubated overnight at 4°C with the primary antibodies, anti-NDUFC1 (cat. no. PA5-68240; Thermo Fisher Scientific, Inc.) at a 1:500 dilution and total OXPHOS WB Antibody Cocktail (ab110413, Abcam) at a 1:250 dilution. Anti-β-actin (A1978, Sigma-Aldrich; Merck KGaA) diluted to 1:2,500 was used for normalization purposes. IRDye 800CW goat anti-mouse IgG (LI-COR Biosciences) at a 1:10,000 dilution or HRP-conjugated goat anti-rabbit IgG (cat. no. 31466, Invitrogen; Thermo Fisher Scientific, Inc.) at a 1:2,000 dilution or goat anti-mouse IgG (cat. no. 62-6520 Invitrogen; Thermo Fisher Scientific, Inc.) at a 1:10,000 dilution were used as secondary antibodies. Novex Sharp pre-stained protein standard (LC5800, Thermo Fisher Scientific, Inc.) and Precision Plus Protein All Blue protein standards were used as molecular weight markers. Chemiluminescent signals were detected using the SuperSignal™ West Femto Maximum Sensitivity Substrate (cat. no. 34096, Thermo Fisher Scientific, Inc.) and the Odyssey Fc imaging system (LI-COR Biosciences) and quantified using Image Studio Lite 5.2 software (LI-COR Biosciences).

Statistical analyses

Statistica 10.0 (StatSoft, Inc., Tulsa, OK) or IBM SPSS Statistics version 24.0 (IBM Corp., Armonk, NY) was used for all statistical analyses, unless otherwise specified. Differences in expression levels between two sample groups were calculated using the Mann-Whitney U-test or the Student's t-test. One-way ANOVA with post hoc Tukey's test was used to compare the three transfection conditions for the metabolic assays. Correlations between IGF2 and H19 and miRNA (miR-483-3p, miR-483-5p and miR-675) expression levels were assessed using Spearman's rank order correlation analysis. Difference in NDUFC1 levels in transfection experiments were evaluated using the Student's t-test. All analyses were two-tailed and P-values <0.05 were considered to indicate statistically significant differences.

Results

Expression of RNAs and miRNAs from the IGF2-H19 locus

The RNA and miRNA transcripts generated from IGF2 and H19 (Fig. 1A) were quantified using RT-qPCR in an extended series of normal adrenal glands, ACAs and ACCs. The results are illustrated in Fig. 1B-D. Elevated levels of IGF2 and decreased levels of H19 were observed in the majority of ACC tissues as compared to ACA tissues and normal adrenals. Concordant with the findings in the present cohort, the ACC samples (n=77) in TCGA dataset also exhibited higher levels of IGF2 RNA and lower levels of H19, as compared to normal adrenal glands from the GTEx (n=128) datasets. miR-483-3p and miR-483-5p generated from IGF2 were both upregulated in ACC vs. ACA and normal adrenals, as previously published for a subset of the cases (27). miR-675 transcribed from H19 was underexpressed in ACC as compared to ACA and normal adrenals.

A comparison of the RNAs and miRNAs analyzed in the extended series of normal adrenal glands, ACA and ACC revealed a moderately inverse correlation between IGF2 and H19 (Fig. 2A). Strong positive correlations were found between miR-483-3p and miR-483-5p and their host IGF2, as well as between miR-675 and its host H19 (Fig. 2A). Similar findings were subsequently revealed in TCGA dataset with 79 ACC cases (Fig. 2B). A weak inverse correlation, although not statistically significant, was observed between H19 and IGF2. Strong positive correlations were observed between IGF2 and miR-483-3p and miR-483-5p, as well as between H19 and miR-675.

Comparative analysis of transcripts with the proteome

Protein expression profiles determined using mass spectrometry for the 6 cases of ACA and 8 cases of ACC in the screening series (41) were compared with the expression levels of the RNAs (IGF2 and H19) and miRNAs (miR-483-3p, miR-483-5p and miR-675). The numbers of proteins overlapping with the various transcripts are illustrated in Venn diagrams in Fig. 3A and B. In total, six proteins were associated with both IGF2 and miR-483-3p, 48 proteins were associated with IGF2 and miR-483-5p, and 120 proteins were associated with IGF2 and both miR-483-3p and miR-483-5p. Only four proteins were associated with H19 and miR-675.

A comparison of the proteomics data with the expression of IGF2 or H19 identified 222 proteins that correlated with IGF2 (Table SIII) and 71 that correlated with H19 (Table SIV). The proteomics data were also investigated to identify proteins exhibiting an inverse expression to the three miRNAs, since such proteins would represent potential targets. This identified seven proteins whose expression inversely correlated with that of miR-483-3p, including one predicted target using TargetScan 7.1 (Table SV). For miR-483-5p, 101 inversely correlated proteins were revealed including nine predicted targets (Table SII). For miR-675, 11 inversely expressed proteins were identified, including one predicted target (Table SVI).

Among the 101 proteins that were inversely correlated with miR-483-5p, 46 of them were differentially expressed between ACA and ACC based on the proteomics data (Table SII). Based on these 46 protein expression patterns, all ACC cases (apart from ACC case no. 10) were clustered together and separated from the ACA cluster (Fig. 3C). Using TCGA and GTEx datasets, it was also observed that this 46-gene signature could distinguish ACC from normal adrenal glands (Fig. 3D). GO analysis of these 46 genes revealed a significant enrichment of biological processes related to mitochondrial metabolism (Fig. 3E). Notably, six proteins (NDUFV1, NDUFC1, NDUFC2, NDUFB9, NDUFB3 and NDUFB7) are subunits of the mitochondrial respiratory chain complex I. Among these six subunits, only NDUFC1 is a predicted target of miR-483-5p according to TargetScan 7.1.

Protein expression of mitochondrial respiratory complexes in adrenal cortical tissues

To further assess the association between mitochondrial respiratory complexes and ACC, the expression levels of mitochondrial respiratory complex I-V were examined in nine adrenal glands, 10 ACA tissues and 10 ACC tissues using western blot analysis, and the total OXPHOS antibody cocktail representing each of the complexes (Fig. 4A). Of note, complex I and IV were lower in ACC compared to ACA (P=0.009 and P=0.002, respectively; Fig. 4B). On the other hand, complex II and III were higher in ACC compared to adrenal glands (P=0.013 and P=0.008, respectively), whereas they did not differ significantly compared to ACA (Fig. 4B).

Figure 4

Expression levels of mitochondrial respiratory complexes in normal adrenals and adrenocortical tumors, and the effect of miR-483-5p on NDUFC1. (A) Mitochondrial respiratory complexes I-V were evaluated in nine adrenal gland (N), 10 ACA and 10 ACC cases using western blot analysis. β-actin used on the same western blot was used for normalization, whereby the lower band corresponding to the expected ~42 kDa for β-actin was used for quantification and the upper band was the leftover signal from the Complex III band of the total OXPHOS antibody cocktail. (B) Graphs representing quantification of each mitochondrial complexes in each sample group and the difference was evaluated using the Mann-Whitney U-test. (C) Western blot analysis of NDUFC1 in 13 adrenal gland, 25 ACA and 29 ACC samples (from 25 patients with ACC). β-actin used on the same western blot was used for normalization. The three blots shown in the lower panel were from the same blots used in panel A and their β-actin blots are the same as those in panel A. (D) Quantification of NDUFC1 in each group from the blots shown in panel C. P-values were obtained using the Mann-Whitney U-test. (E) Reverse transcription-quantitative PCR of miR-483-5p expression in the NCI-H295R ACC cells treated with anti-miR-CTR or anti-miR-483-5p. Error bars represent the SEM of the mean from three independent experiments. P-values were obtained using the Student's t-test. (F) Upper panel, representative western blot image illustrating the effects of anti-miR-483-5p and anti-miR-CTR on NDUFC1 expression. β-actin was used as a loading control. Lower panel, quantification of NDUFC1 levels in miR-483-5p inhibition. Error bars represent the SEM of the mean from nine independent experiments. P-values were calculated using the Student's t-test. NDUFC1, NADH:ubiquinone oxidoreductase subunit C1; ACC, adrenocortical carcinoma; ACA, adrenocortical adenoma.

To validate the involvement of complex I in ACCs, NDUFC1 protein expression was also evaluated in an extended series of clinical samples, consisting of 13 normal adrenal glands, 25 ACA and 29 ACC samples (from 25 patients with ACC), using western blot analysis (Fig. 4C). Concordantly, NDUFC1 expression was lower in ACC compared to ACA and normal adrenals (P=0.043 and P=0.046, respectively; Fig. 4D). The inhibition of miR-483-5p, using anti-miR-483-5p, in the NCI-H295R ACC cell line increased NDUFC1 protein expression (Fig. 4E and F), suggesting a putative role of miR-483-5p in mitochondrial respiratory activities.

Role of miR-483 in the cellular metabolism of ACC cells

To further validate the effects of miR-483-5p on metabolic activities, the mitochondrial OCR and ECAR were measured using a Seahorse system. The OCR profile exhibited a significant decrease in the basal and maximum respiration upon the inhibition of miR-483-5p, but not that of miR-483-3p, using anti-miR-483-5p and anti-miR-483-3p, respectively (Fig. 5A and B). Additionally, ATP production generated from the mitochondrial respiration was also lower in the cells transfected with anti-miR-483-5p, compared to those transfected with anti-miR-483-3p or negative control-transfected cells (Fig. 5B).

The measurement of the ECAR, an indicator of glycolysis, revealed a decrease in basal glycolysis, glycolytic capacity and reserve in the anti-miR-483-5p-transfected cells. Similar effects were also observed upon the inhibition of miR-483-3p (Fig. 5A and B). To obtain an overview of the bioenergetics profile, the OCR vs. ECAR was plotted at basal conditions. As illustrated in Fig. 5C, the cells in which miR-483-5p was inhibited were least energetic, as demonstrated by lower respiration and glycolysis rates, whereas the cells in which miR-483-3p was inhibited were less glycolytic, but were similarly oxidative as the anti-miR-CTR-transfected cells.

Discussion

Several studies have demonstrated that the overexpression of IGF2 is a very frequent alteration in ACC, which suggests a role for this growth factor in ACC tumor development (4-6). The present study further analyzed this alteration by measuring RNA and miRNA transcripts from the IGF2 and H19 loci and associated these results with global protein levels determined using proteomics. The present study confirmed previous findings (4,5) that IGF2 expression is elevated in ACC compared to normal adrenal and to ACA. The higher transcription of IGF2 was, as expected, paralleled by an increased expression of miR-483-3p and miR-483-5p (hosted by IGF2) in ACC. It was also demonstrated that H19 expression was generally higher in ACA compared to normal adrenal, and lower in ACC compared to ACA and normal adrenals. This observation corroborates previous findings of low H19 levels in some hormonally active ACC (24). Furthermore, the expression of miR-675 (hosted by H19) correlated with H19 levels in the present cohort and in TCGA dataset.

In the ACC group, the concerted upregulation of IGF2 together with miR-483-3p and miR-483-5p, as well as the downregulation of H19 together with miR-675 was observed. Each of these molecules could potentially contribute to ACC development alone or cooperatively. The present study then investigated protein expression profiles [obtained using mass spectrometry (41)] of proteins from ACC and ACA, in order to identify proteins that correlate with the transcription from the H19-IGF2 locus. The correlation analysis of IGF2 and H19 transcripts with the proteome took into account both up- and downregulated proteins. Mass spectrometry revealed a correlation between IGF2 mRNA and IGFII protein levels, as well as an increased IGFII protein content in ACC (Table SIII), demonstrating that translation of increased IGF2 mRNAs indeed occur in ACC. Among the 222 proteins overlapping with IGF2 mRNA expression, 15 proteins (NDUFV1, ANTXR1, RAB20, PTGR1, ECE1, TNS1, MGST1, UGGT1, H6PD, AOC3, TM9SF2, MGST2, SCAMP2, AUP1 and DNAJC16) were significantly downregulated in ACC compared to ACA (Table SIII). One of these, NDUFV1, is associated with mitochondrial oxidative phosphorylation. This indicates that energy metabolism is an important marker for the ACC phenotype.

The present study also applied IPA to the IGF2 mRNA correlated proteins, to elucidate their associations and ontology. The top two molecular and cellular functions were energy metabolism and lipid metabolism. Further IPA identified hepatocyte nuclear factor 4A and TP53 as the top two upstream regulators, both of which are central to cancer (Table SVII). In addition, IPA suggested seven of the molecules (COX17, NDUFV1, OGDH, PRDX3, TXN2, TXNRD2 and UQCRB) to be associated with mitochondrial dysfunction (Table SVII). Notably, one of the Weiss criteria for the classification of ACC is the presence of <25% clear cells (46). Such tumors instead have a majority of oxyphilic cells, which are characterized by increased amounts of mitochondria (giving the positive eosin staining). It is not known whether these mitochondria are functional or whether they represent a compensatory increase due to a defective mitochondrial function. The present study also found 71 proteins that correlated with H19 expression, and almost all exhibited a lower prevalence in ACC vs. ACA (Table SIV). Of the 71 identified proteins, 23 displayed significant differences between ACC and ACA. Of note, the protein with the greatest difference between ACC and ACA in association with H19 transcription is encoded by the potassium channel gene KCNQ1, which is imprinted and associated with for example Beckwith-Wiedemann syndrome (47). Other notable proteins are two mitochondrial NDUF (NADH dehydrogenase sub-complex) proteins, supporting the importance of energy metabolism.

As it is assumed that the role of miRNAs is the negative regulation of target genes, the present study analyzed the inverse correlation of these with the proteome. In total, 11 proteins correlated inversely with miR-675, one of which was predicted as a target. This apoptosis-related protein, tumor necrosis factor alpha-induced protein 2 (TNFAIP2), is induced by tumor necrosis factor-α and associated with various cancer types (48-50). miR-483-5p exhibited an inverse correlation with 101 proteins. By contrast, its companion miR-483-3p only negatively correlated with seven proteins. Of note, all except two of these were found in the list of proteins inversely correlating with miR-483-5p. However, these two proteins (H6PD and UBE2G2) were not predicted as targets using TargetScan 7.1. The PUMA, a potential target of miR-483-3p in ACC (27), was not detected using mass spectrometry. Among the proteins inversely correlating with miR-483-5p, a group of mitochondrial proteins/enzymes may be noted, such as members of the NADH dehydrogenase complex, as well as other mitochondrial molecules. In total, 46 of the proteins with an inverse correlation with miR-483-5p were differentially expressed between ACC and ACA. This signature was also validated as a classifier for ACC in independent cohorts using TCGA and GTEx datasets. GO analysis revealed the enrichment of biological processes related to mitochondrial respiration. In particular, six subunits of mitochondrial respiratory complex I were downregulated in ACC. The decreased expression of mitochondrial complex I was also validated in an extended series of clinical samples using western blot analysis, suggesting the deficiency of complex I in ACC. In line with these findings, Kimmel et al (51) previously demonstrated a poor oxygen uptake by the tumor mitochondria of the rat ACC 494 model using the tricarboxylic acid cycle substrates (α-ketoglutarate, malate and isocitrate), which led them to propose that the tumor mitochondria could be deficiency in the flavoprotein dehydrogeneases for NADH and NADPH oxidation. Notably, the effect of several anti-ACC drugs (mitotane, niclosamide and ATR-101) is also tightly linked to the disruption of mitochondrial function (52-54). In addition, a deficiency in mitochondrial respiratory complex I has also been demonstrated to promote tumor progression in other tumor types, such as breast cancer and hepatoma (55,56).

One of the mitochondrial respiratory complex I subunits, NDUFC1, is also a predicted target of miR-483-5p. The expression of this target was upregulated upon miR-483-5p inhibition. The involvement of miR-483-5p in mitochondrial respiration was also supported by the Mito Stress Test using the Seahorse technology. Additionally, the inhibition of miR-483-5p also reduced glycolysis, which led to a low metabolic state resembling cellular quiescence. These results are in line with the observations that the inhibition of miR-483-5p had no effect on cell proliferation or apoptosis in ACC (27,30). Although the target(s) of miR-483-5p involved in glycolysis remains unknown, the present study noted a common protein, hexose-6-phosphate-dehydrogenase (H6PD), which inversely correlated with both miR-483-3p and miR-483-5p. H6PD is an enzyme that produces NAPDH in the endoplasmic reticulum for glucose metabolism and glycolysis (57,58). Even though this gene was not predicted as a target of miR-483-3p and miR-483-5p by TargetScan 7.1, the possibility of H6PD as a direct or indirect target of these miRNAs has not yet been excluded.

The present data may be utilized to identify differentially expressed molecules that may be developed into diagnostic markers for the identification of ACC cases in the absence of metastasis at the time of diagnosis. Furthermore, it may potentially aid in the identification of those patients who could benefit from adjuvant mitotane therapy (59,60). The main findings of the present study were that correlations exist between specific miRNA(s) and RNA(s) in the IGF2-H19 locus and the proteomes of ACC and ACA. The patterns of RNA transcription from this locus form specific networks of protein expression that appear to be associated with ACC. The present study also reveals a link of the IGF2-H19 locus to energy metabolism and deficiency of mitochondrial respiratory complex I in ACC, suggesting the importance of mitochondrial dysfunction and IGF2-H19 regulatory network in ACC development.

Supplementary Data

Availability of data and materials

The analyzed datasets presented in the present study are available in the Gene Expression Profiling Interactive Analysis (http://gepia.cancer-pku.cn/), StarBase Pan-Cancer Analysis Platform (https://starbase.sysu.edu.cn/panCancer.php) and ProteomeXchange Consortium (http://proteomecentral.proteomexchange.org; PXD000604).

Authors' contributions

PS, TJE, RB, WOL and CL conceived and designed the study. PS, SC, HK, CX, RF, MoA, JG, HS and MaA performed the experiments and analyzed the data. MK, AH, JZ and RB contributed to the clinical materials and data. WOL and CL supervised the study, and reviewed and approved the authenticity of all the raw data. All authors critically reviewed, and have read and approved the final version of the manuscript.

Ethics approval and consent to participate

The use of the tumor samples (Dnr 01-136; Dnr 2020-04226) and normal adrenals (Dnr 01-353; Dnr 2020-04226) in the present study were approved by the Ethics Committee of Karolinska Institutet (Dnr 01-136; Dnr 01-353) and by the Swedish Ethical Review Authority (Dnr 2020-04226). Tissue samples were collected with informed consent and was obtained from the patients prior to surgery.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Ms. Lisa Ånfalk (Karolinska University Hospital) for assisting with the collection of all tissue samples, and the Strategic Research Programme Diabetes Facility for the Seahorse system.

Funding

The present study was supported by grants from the Swedish Research Council (2021-03006), the Swedish Cancer Society (20 0843 and 20 0859), the Cancer Society in Stockholm (201223), the Gustav V Jubilee Foundation (204103), the Stockholm County Council (RS 2019-1054) and Funds from Karolinska Institutet (2020-01851 and 2020-01768).

References

1 

Assié G, Letouzé E, Fassnacht M, Jouinot A, Luscap W, Barreau O, Omeiri H, Rodriguez S, Perlemoine K, René-Corail F, et al: Integrated genomic characterization of adrenocortical carcinoma. Nat Genet. 46:607–612. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Juhlin CC, Goh G, Healy JM, Fonseca AL, Scholl UI, Stenman A, Kunstman JW, Brown TC, Overton JD, Mane SM, et al: Whole-exome sequencing characterizes the landscape of somatic mutations and copy number alterations in adrenocortical carcinoma. J Clin Endocrinol Metab. 100:E493–E502. 2015. View Article : Google Scholar

3 

Zheng S, Cherniack AD, Dewal N, Moffitt RA, Danilova L, Murray BA, Lerario AM, Else T, Knijnenburg TA, Ciriello G, et al: Comprehensive pan-genomic characterization of adrenocortical carcinoma. Cancer Cell. 29:723–736. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Boulle N, Logié A, Gicquel C, Perin L and Le Bouc Y: Increased levels of insulin-like growth factor II (IGF-II) and IGF-binding protein-2 are associated with malignancy in sporadic adrenocortical tumors. J Clin Endocrinol Metab. 83:1713–1720. 1998.PubMed/NCBI

5 

Gicquel C, Bertagna X, Schneid H, Francillard-Leblond M, Luton JP, Girard F and Le Bouc Y: Rearrangements at the 11p15 locus and overexpression of insulin-like growth factor-II gene in sporadic adrenocortical tumors. J Clin Endocrinol Metab. 78:1444–1453. 1994.PubMed/NCBI

6 

Gicquel C, Raffin-Sanson ML, Gaston V, Bertagna X, Plouin PF, Schlumberger M, Louvel A, Luton JP and Le Bouc Y: Structural and functional abnormalities at 11p15 are associated with the malignant phenotype in sporadic adrenocortical tumors: Study on a series of 82 tumors. J Clin Endocrinol Metab. 82:2559–2565. 1997.PubMed/NCBI

7 

Larsson C: Epigenetic aspects on therapy development for gastroenteropancreatic neuroendocrine tumors. Neuroendocrinology. 97:19–25. 2013. View Article : Google Scholar

8 

Brannan CI, Dees EC, Ingram RS and Tilghman SM: The product of the H19 gene may function as an RNA. Mol Cell Biol. 10:28–36. 1990.PubMed/NCBI

9 

Ogawa O, Eccles MR, Szeto J, McNoe LA, Yun K, Maw MA, Smith PJ and Reeve AE: Relaxation of insulin-like growth factor II gene imprinting implicated in Wilms' tumour. Nature. 362:749–751. 1993. View Article : Google Scholar : PubMed/NCBI

10 

Rainier S, Johnson LA, Dobry CJ, Ping AJ, Grundy PE and Feinberg AP: Relaxation of imprinted genes in human cancer. Nature. 362:747–749. 1993. View Article : Google Scholar : PubMed/NCBI

11 

Weksberg R, Shen DR, Fei YL, Song QL and Squire J: Disruption of insulin-like growth factor 2 imprinting in Beckwith-Wiedemann syndrome. Nat Genet. 5:143–150. 1993. View Article : Google Scholar : PubMed/NCBI

12 

Pravtcheva DD and Wise TL: Metastasizing mammary carcinomas in H19 enhancers-Igf2 transgenic mice. J Exp Zool. 281:43–57. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Veronese A, Lupini L, Consiglio J, Visone R, Ferracin M, Fornari F, Zanesi N, Alder H, D'Elia G, Gramantieri L, et al: Oncogenic role of miR-483-3p at the IGF2/483 locus. Cancer Res. 70:3140–3149. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Drelon C, Berthon A, Ragazzon B, Tissier F, Bandiera R, Sahut-Barnola I, de Joussineau C, Batisse-Lignier M, Lefrancois-Martinez AM, Bertherat J, et al: Analysis of the role of Igf2 in adrenal tumour development in transgenic mouse models. PLoS One. 7:e441712012. View Article : Google Scholar : PubMed/NCBI

15 

Heaton JH, Wood MA, Kim AC, Lima LO, Barlaskar FM, Almeida MQ, Fragoso MC, Kuick R, Lerario AM, Simon DP, et al: Progression to adrenocortical tumorigenesis in mice and humans through insulin-like growth factor 2 and β-catenin. Am J Pathol. 181:1017–1033. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Barlaskar FM, Spalding AC, Heaton JH, Kuick R, Kim AC, Thomas DG, Giordano TJ, Ben-Josef E and Hammer GD: Preclinical targeting of the type I insulin-like growth factor receptor in adrenocortical carcinoma. J Clin Endocrinol Metab. 94:204–212. 2009. View Article : Google Scholar :

17 

Fassnacht M, Berruti A, Baudin E, Demeure MJ, Gilbert J, Haak H, Kroiss M, Quinn DI, Hesseltine E, Ronchi CL, et al: Linsitinib (OSI-906) versus placebo for patients with locally advanced or metastatic adrenocortical carcinoma: A double-blind, randomised, phase 3 study. Lancet Oncol. 16:426–435. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Haluska P, Worden F, Olmos D, Yin D, Schteingart D, Batzel GN, Paccagnella ML, de Bono JS, Gualberto A and Hammer GD: Safety, tolerability, and pharmacokinetics of the anti-IGF-1R monoclonal antibody figitumumab in patients with refractory adrenocortical carcinoma. Cancer Chemother Pharmacol. 65:765–773. 2010. View Article : Google Scholar :

19 

Jones RL, Kim ES, Nava-Parada P, Alam S, Johnson FM, Stephens AW, Simantov R, Poondru S, Gedrich R, Lippman SM, et al: Phase I study of intermittent oral dosing of the insulin-like growth factor-1 and insulin receptors inhibitor OSI-906 in patients with advanced solid tumors. Clin Cancer Res. 21:693–700. 2015. View Article : Google Scholar

20 

Lerario AM, Worden FP, Ramm CA, Hesseltine EA, Stadler WM, Else T, Shah MH, Agamah E, Rao K and Hammer GD: The combination of insulin-like growth factor receptor 1 (IGF1R) antibody cixutumumab and mitotane as a first-line therapy for patients with recurrent/metastatic adrenocortical carcinoma: A multi-institutional NCI-sponsored trial. Horm Cancer. 5:232–239. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Naing A, Kurzrock R, Burger A, Gupta S, Lei X, Busaidy N, Hong D, Chen HX, Doyle LA, Heilbrun LK, et al: Phase I trial of cixutumumab combined with temsirolimus in patients with advanced cancer. Clin Cancer Res. 17:6052–6060. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Ilvesmäki V, Kahri AI, Miettinen PJ and Voutilainen R: Insulin-like growth factors (IGFs) and their receptors in adrenal tumors: High IGF-II expression in functional adrenocortical carcinomas. J Clin Endocrinol Metab. 77:852–858. 1993.PubMed/NCBI

23 

Laurell C, Velázquez-Fernández D, Lindsten K, Juhlin C, Enberg U, Geli J, Höög A, Kjellman M, Lundeberg J, Hamberger B, et al: Transcriptional profiling enables molecular classification of adrenocortical tumours. Eur J Endocrinol. 161:141–152. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Liu J, Kahri AI, Heikkilä P, Ilvesmäki V and Voutilainen R: H19 and insulin-like growth factor-II gene expression in adrenal tumors and cultured adrenal cells. J Clin Endocrinol Metab. 80:492–496. 1995.PubMed/NCBI

25 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Doghman M, El Wakil A, Cardinaud B, Thomas E, Wang J, Zhao W, Peralta-Del Valle MH, Figueiredo BC, Zambetti GP and Lalli E: Regulation of insulin-like growth factor-mammalian target of rapamycin signaling by microRNA in childhood adrenocortical tumors. Cancer Res. 70:4666–4675. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Özata DM, Caramuta S, Velázquez-Fernández D, Akçakaya P, Xie H, Höög A, Zedenius J, Bäckdahl M, Larsson C and Lui WO: The role of microRNA deregulation in the pathogenesis of adrenocortical carcinoma. Endocr Relat Cancer. 18:643–655. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Soon PS, Tacon LJ, Gill AJ, Bambach CP, Sywak MS, Campbell PR, Yeh MW, Wong SG, Clifton-Bligh RJ, Robinson BG and Sidhu SB: miR-195 and miR-483-5p identified as predictors of poor prognosis in adrenocortical cancer. Clin Cancer Res. 15:7684–7692. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Patterson EE, Holloway AK, Weng J, Fojo T and Kebebew E: MicroRNA profiling of adrenocortical tumors reveals miR-483 as a marker of malignancy. Cancer. 117:1630–1639. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Agosta C, Laugier J, Guyon L, Denis J, Bertherat J, Libé R, Boisson B, Sturm N, Feige JJ, Chabre O and Cherradi N: MiR-483-5p and miR-139-5p promote aggressiveness by targeting N-myc downstream-regulated gene family members in adrenocortical cancer. Int J Cancer. 143:944–957. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Chabre O, Libé R, Assie G, Barreau O, Bertherat J, Bertagna X, Feige JJ and Cherradi N: Serum miR-483-5p and miR-195 are predictive of recurrence risk in adrenocortical cancer patients. Endocr Relat Cancer. 20:579–594. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Decmann A, Bancos I, Khanna A, Thomas MA, Turai P, Perge P, Pintér JZ, Tóth M, Patócs A and Igaz P: Comparison of plasma and urinary microRNA-483-5p for the diagnosis of adrenocortical malignancy. J Biotechnol. 297:49–53. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Patel D, Boufraqech M, Jain M, Zhang L, He M, Gesuwan K, Gulati N, Nilubol N, Fojo T and Kebebew E: MiR-34a and miR-483-5p are candidate serum biomarkers for adrenocortical tumors. Surgery. 154:1224–1229. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Li H, Yu B, Li J, Su L, Yan M, Zhu Z and Liu B: Overexpression of lncRNA H19 enhances carcinogenesis and metastasis of gastric cancer. Oncotarget. 5:2318–2329. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Tsang WP, Ng EK, Ng SS, Jin H, Yu J, Sung JJ and Kwok TT: Oncofetal H19-derived miR-675 regulates tumor suppressor RB in human colorectal cancer. Carcinogenesis. 31:350–358. 2010. View Article : Google Scholar

36 

Zhai LL, Wang P, Zhou LY, Yin JY, Tang Q, Zhang TJ, Wang YX, Yang DQ, Lin J and Deng ZQ: Over-expression of miR-675 in formalin-fixed paraffin-embedded (FFPE) tissues of breast cancer patients. Int J Clin Exp Med. 8:11195–11201. 2015.PubMed/NCBI

37 

Zhou YW, Zhang H, Duan CJ, Gao Y, Cheng YD, He D, Li R and Zhang CF: miR-675-5p enhances tumorigenesis and metastasis of esophageal squamous cell carcinoma by targeting REPS2. Oncotarget. 7:30730–30747. 2016. View Article : Google Scholar : PubMed/NCBI

38 

He D, Wang J, Zhang C, Shan B, Deng X, Li B, Zhou Y, Chen W, Hong J, Gao Y, et al: Down-regulation of miR-675-5p contributes to tumor progression and development by targeting pro-tumorigenic GPR55 in non-small cell lung cancer. Mol Cancer. 14:732015. View Article : Google Scholar : PubMed/NCBI

39 

Schmitz KJ, Helwig J, Bertram S, Sheu SY, Suttorp AC, Seggewiss J, Willscher E, Walz MK, Worm K and Schmid KW: Differential expression of microRNA-675, microRNA-139-3p and microRNA-335 in benign and malignant adrenocortical tumours. J Clin Pathol. 64:529–535. 2011. View Article : Google Scholar : PubMed/NCBI

40 

DeLellis RA, Lloyd RV, Heitz PU and Eng C: Pathology and genetics of tumours of endocrine organs. World Health Organization Classification of Tumours. 3rd edition. 8. IARC Press; Lyon, France: 2004

41 

Kjellin H, Johansson H, Höög A, Lehtiö J, Jakobsson PJ and Kjellman M: Differentially expressed proteins in malignant and benign adrenocortical tumors. PLoS One. 9:e879512014. View Article : Google Scholar : PubMed/NCBI

42 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45(W1): W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Li JH, Liu S, Zhou H, Qu LH and Yang JH: starBase v2.0: Decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 42(Database Issue): D92–D97. 2014. View Article : Google Scholar

44 

Gazdar AF, Oie HK, Shackleton CH, Chen TR, Triche TJ, Myers CE, Chrousos GP, Brennan MF, Stein CA and La Rocca RV: Establishment and characterization of a human adrenocortical carcinoma cell line that expresses multiple pathways of steroid biosynthesis. Cancer Res. 50:5488–5496. 1990.PubMed/NCBI

45 

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

46 

Weiss LM: Comparative histologic study of 43 metastasizing and nonmetastasizing adrenocortical tumors. Am J Surg Pathol. 8:163–169. 1984. View Article : Google Scholar : PubMed/NCBI

47 

Valente FM, Sparago A, Freschi A, Hill-Harfe K, Maas SM, Frints SGM, Alders M, Pignata L, Franzese M, Angelini C, et al: Transcription alterations of KCNQ1 associated with imprinted methylation defects in the Beckwith-Wiedemann locus. Genet Med. 21:1808–1820. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Corrêa GT, Bandeira GA, Cavalcanti BG, de Carvalho Fraga CA, dos Santos EP, Silva TF, Gomez RS, Guimarães AL and De Paula AM: Association of-308 TNF-α promoter polymorphism with clinical aggressiveness in patients with head and neck squamous cell carcinoma. Oral Oncol. 47:888–894. 2011. View Article : Google Scholar

49 

Schteingart DE, Giordano TJ, Benitez RS, Burdick MD, Starkman MN, Arenberg DA and Strieter RM: Overexpression of CXC chemokines by an adrenocortical carcinoma: A novel clinical syndrome. J Clin Endocrinol Metab. 86:3968–3974. 2001. View Article : Google Scholar : PubMed/NCBI

50 

Shih CM, Lee YL, Chiou HL, Chen W, Chang GC, Chou MC and Lin LY: Association of TNF-alpha polymorphism with susceptibility to and severity of non-small cell lung cancer. Lung Cancer. 52:15–20. 2006. View Article : Google Scholar : PubMed/NCBI

51 

Kimmel GL, Péron FG, Haksar A, Bedigian E, Robidoux WF Jr and Lin MT: Ultrastructure, steroidogenic potential, and energy metabolism of the Snell adrenocortical carcinoma 494. A comparison with normal adrenocortical tissue. J Cell Biol. 62:152–163. 1974. View Article : Google Scholar : PubMed/NCBI

52 

Cheng Y, Kerppola RE and Kerppola TK: ATR-101 disrupts mitochondrial functions in adrenocortical carcinoma cells and in vivo. Endocr Relat Cancer. 23:1–19. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Poli G, Guasti D, Rapizzi E, Fucci R, Canu L, Bandini A, Cini N, Bani D, Mannelli M and Luconi M: Morphofunctional effects of mitotane on mitochondria in human adrenocortical cancer cells. Endocr Relat Cancer. 20:537–550. 2013. View Article : Google Scholar : PubMed/NCBI

54 

Satoh K, Zhang L, Zhang Y, Chelluri R, Boufraqech M, Nilubol N, Patel D, Shen M and Kebebew E: Identification of niclosamide as a novel anticancer agent for adrenocortical carcinoma. Clin Cancer Res. 22:3458–3466. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Lee JH, Lee YK, Lim JJ, Byun HO, Park I, Kim GH, Xu WG, Wang HJ and Yoon G: Mitochondrial respiratory dysfunction induces claudin-1 expression via reactive oxygen species-mediated heat shock factor 1 activation, leading to hepatoma cell invasiveness. J Biol Chem. 290:21421–21431. 2015. View Article : Google Scholar : PubMed/NCBI

56 

Santidrian AF, Matsuno-Yagi A, Ritland M, Seo BB, LeBoeuf SE, Gay LJ, Yagi T and Felding-Habermann B: Mitochondrial complex I activity and NAD+/NADH balance regulate breast cancer progression. J Clin Invest. 123:1068–1081. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Marbet P, Klusonova P, Birk J, Kratschmar DV and Odermatt A: Absence of hexose-6-phosphate dehydrogenase results in reduced overall glucose consumption but does not prevent 11β-hydroxysteroid dehydrogenase-1-dependent glucocorticoid activation. FEBS J. 285:3993–4004. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Marini C, Ravera S, Buschiazzo A, Bianchi G, Orengo AM, Bruno S, Bottoni G, Emionite L, Pastorino F, Monteverde E, et al: Discovery of a novel glucose metabolism in cancer: The role of endoplasmic reticulum beyond glycolysis and pentose phosphate shunt. Sci Rep. 6:250922016. View Article : Google Scholar : PubMed/NCBI

59 

Berruti A, Grisanti S, Pulzer A, Claps M, Daffara F, Loli P, Mannelli M, Boscaro M, Arvat E, Tiberio G, et al: Long-term outcomes of adjuvant mitotane therapy in patients with radically resected adrenocortical carcinoma. J Clin Endocrinol Metab. 102:1358–1365. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Calabrese A, Basile V, Puglisi S, Perotti P, Pia A, Saba L, Berchialla P, Porpiglia F, Veltri A, Volante M, et al: Adjuvant mitotane therapy is beneficial in non-metastatic adrenocortical carcinoma at high risk of recurrence. Eur J Endocrinol. 180:387–396. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2022
Volume 61 Issue 5

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
Scicluna P, Caramuta S, Kjellin H, Xu C, Fröbom R, Akhtar M, Gao J, Shi H, Kjellman M, Almgren M, Almgren M, et al: Altered expression of the <em>IGF2‑H19</em> locus and mitochondrial respiratory complexes in adrenocortical carcinoma. Int J Oncol 61: 140, 2022
APA
Scicluna, P., Caramuta, S., Kjellin, H., Xu, C., Fröbom, R., Akhtar, M. ... Larsson, C. (2022). Altered expression of the <em>IGF2‑H19</em> locus and mitochondrial respiratory complexes in adrenocortical carcinoma. International Journal of Oncology, 61, 140. https://doi.org/10.3892/ijo.2022.5430
MLA
Scicluna, P., Caramuta, S., Kjellin, H., Xu, C., Fröbom, R., Akhtar, M., Gao, J., Shi, H., Kjellman, M., Almgren, M., Höög, A., Zedenius, J., Ekström, T. J., Bränström, R., Lui, W., Larsson, C."Altered expression of the <em>IGF2‑H19</em> locus and mitochondrial respiratory complexes in adrenocortical carcinoma". International Journal of Oncology 61.5 (2022): 140.
Chicago
Scicluna, P., Caramuta, S., Kjellin, H., Xu, C., Fröbom, R., Akhtar, M., Gao, J., Shi, H., Kjellman, M., Almgren, M., Höög, A., Zedenius, J., Ekström, T. J., Bränström, R., Lui, W., Larsson, C."Altered expression of the <em>IGF2‑H19</em> locus and mitochondrial respiratory complexes in adrenocortical carcinoma". International Journal of Oncology 61, no. 5 (2022): 140. https://doi.org/10.3892/ijo.2022.5430