Abstract

Context

Silent corticotroph adenoma (SCA) exhibits more tumor aggressiveness features than functioning adenomas (FCAs).

Objective

We aimed to investigate proprotein convertase subtilisin/kexin type 1 inhibitor (PCSK1N) expression in CA and examine if endoplasmic reticulum (ER) stress-induced responses affect cell survival in a corticotroph tumor cell model.

Methods

Clinical and imaging characteristics were recorded in 33 patients with FCA (20 women, 11 macroadenomas) and 18 SCAs (8 women, all macroadenomas). Gene expression of pro-opiomelanocortin (POMC), T-box transcription factor 19(TBX19)/TPIT, proprotein convertase subtilisin/kexin type 1 (PCSK1)/PC1/3, and its inhibitor PCSK1N, was measured by reverse transcription–quantitative polymerase chain reaction in adenoma tissue. Mouse pituitary corticotroph tumor (AtT-20) cells were treated with tanespimycin (17-AAG), an HSP90 chaperone inhibitor, to induce ER stress, followed by gene and protein analyses.

Results

POMC, TPIT, and PCSK1 expression were higher, whereas PCSK1N was lower in FCA compared to SCA. PCSK1N correlated with POMC (rs = −0.514; P < .001), TPIT (rs = −0.386; P = .005), PCSK1 (rs = −0.3691; P = .008), and tumor largest diameter (rs = 0.645; P < .001), in all CA. Induction of ER stress by 17-AAG in AtT-20 cells led to a decrease of Pomc and an increase of Pcsk1n gene expression at 24 hours. Moreover, a downregulation of cell cycle, apoptosis, and senescence pathways, and alterations in cell adhesion and cytoskeleton, were observed at the protein level.

Conclusion

PCSK1N is higher in SCA compared with FCA, and associated with corticotroph cell markers and tumor size. PCSK1N is likely to be part of the adaptive response to ER stress, potentially conferring a survival advantage to the corticotroph tumor cell in conjunction with other proteins.

Corticotroph pituitary adenomas (CAs) develop from corticotroph cells and are classified by their immunohistochemistry (IHC) staining for adrenocorticotropic hormone (ACTH) and/or the corresponding transcription factor T-box transcription factor 19 (TBX19)/TPIT (1). In a clinical setting, CA present with different degrees of functionality, based on the level of ACTH secretion, ranging from silent CA (SCA) to whispering, and ultimately to functioning CA (FCA) (2, 3). Due to the clinical symptoms caused by ACTH-stimulated cortisol secretion leading to Cushing disease, FCAs are generally detected at an earlier stage as compared to SCAs (4). SCAs are usually detected by the symptoms caused by the tumor volume, as they are mostly macroadenomas with extrasellar expansion, whereas FCAs most often are intrasellar microadenomas at presentation (3, 5). SCAs represent up to 15% of all nonfunctioning pituitary adenomas (6), and about 3% of all pituitary neuroendocrine tumors, while FCAs comprise up to 2% to 6% (7).

Dysfunctional processing of pro-opiomelanocortin (POMC) into ACTH has been hypothesized to contribute to the silence of SCA. Indeed, a previous study showed that SCA secrete more inactive ACTH molecules (8). The main prohormone-converting enzyme in the anterior lobe of the pituitary gland is proprotein convertase subtilisin/kexin type 1 (PCSK1) or PC1/3, sequentially cleaving POMC to ACTH (9). Previous studies have shown that SCAs have lower expression of PCSK1 as compared to their functional counterpart, potentially causing the diminished ACTH production (9, 10). In addition, it has been shown that PCSK1 expression is increased in certain cases of SCA that later develop into FCA (3, 11). In the early 2000, studies of the AtT-20 mouse pituitary corticotroph tumor cell line revealed that overproduction of a small protein called proprotein convertase subtilisin/kexin type 1 inhibitor (PCSK1N) or proSAAS, encoded by the PCSK1N gene, markedly reduced the processing of POMC, and this protein was shown to specifically inhibit PCSK1 (12, 13). We have previously shown that SCAs express higher protein levels of PCSK1N (14).

SCAs are recognized to have an aggressive clinical behavior due to rapid and invasive growth, tumor heterogeneity, and higher recurrence rates (5, 15). In recent years, several differences in the molecular profiles between FCA and SCA have been described. For example, SCA exhibited several features of epithelial-mesenchymal transition (EMT), with increased expression of mesenchymal genes (16), dysregulation of several lipid metabolic pathways (17), a distinct expression of extracellular matrix genes, and a reduced endoplasmic reticulum (ER) protein-folding ability (14). Impaired protein processing in the ER potentially leads to ER stress and the activation of several compensatory pathways, among them, the unfolded protein response (UPR) signaling pathway, to reestablish protein homeostasis (18). Recently, PCSK1N has been described as a stress-responsive protein, with an antiaggregation function, and produced as part of a physiological defense mounted by cells to reduce cell damage and maintain cell viability (19).

Several molecules are known to induce ER stress in experimental settings, some with the potential to be further developed into anticancer drugs (20). One of these molecules is 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) or tanespimycin, an inhibitor of the 90-kDa heat shock protein (Hsp90), which has been used in cancer treatment clinical trials (21). Hsp90 plays important roles in protein-folding processes in the ER, and 17-AAG has been known to activate one branch of UPR pathways called IRE1 in myeloma cancer cells (22). The inhibition of Hsp90 by 17-AAG also induces compensatory adaptation for cell survival (23). However, how the Hsp90 inhibitor interacts with PCSK1 and PCSK1N is not clearly defined.

We hypothesized that PCSK1N plays important roles involving cell growth and survival processes, facilitating CA growth. PCSK1N is known to function as a PCSK1 inhibitor (12), and is an ER stress-responsive protein (19). Based on our previous data showing that PCSK1N protein is higher expressed in SCA (14), we here aimed to assess the expression of PCSK1N, in addition to corticotroph cell markers (POMC, TPIT, and PCSK1), in a large cohort of FCA and SCA, and correlate its expression to clinical features such as tumor diameter. Furthermore, we investigated the protein repertoire initiated by ER stress response in AtT-20 cells to decipher the mechanisms behind the growth potential/cell survival in SCA.

Material and Methods

Study Design and Sample Selection

Patients operated on for pituitary adenomas at our single center were included in a retrospective study between 1998 and 2009 (n = 240) and a prospective study between 2014 and 2022 (n = 216). For the present study, we included 51 patients (29 from the retrospective and 22 from the prospective study) diagnosed and operated on for Cushing disease (n = 33) and clinically nonfunctioning pituitary adenomas showing positive staining for TPIT and/or ACTH (SCAs) (n = 18) with available tumor tissue (38 tumors were snap-frozen samples and 13 were optimum cutting temperature embedded). All SCAs were clinically classified as nonfunctioning at the time of surgery, whereas the patients with FCAs presented with signs and symptoms of hypercortisolism. Twenty-four FCA and 5 SCA patients were previously included in published cohorts (14, 24, 25). The IHC subclassification of SCA was performed in a research setting as described (26, 27). Tumor diameter as described in the preoperative pituitary magnetic resonance imaging investigation was recorded as the largest diameter. Tumors with a diameter of less than 10 mm were defined as microadenomas, and macroadenomas were 10 mm or larger. Two SCA tumors had a diameter greater than 40 mm (57 and 62 mm). Morning preoperative plasma ACTH (pmol/L) and cortisol (nmol/L) concentrations were measured in the routine clinical setting. ACTH was available in 28 FCAs and 7 SCAs, whereas cortisol was available in 29 FCAs and 10 SCAs.

Written informed consent was obtained from all patients. This study was approved by the regional ethics committee (REK No. 2014/1680, and REK No. 2020/22301) and the hospital authority.

Sample Preparation

Tumor tissue was homogenized in Trizol (Life Technologies [RRID:SCR_008817], catalog No. 15596018). RNA was extracted by QIAGEN miRNeasy Mini Kit (QIAGEN [RRID:SCR_008539], catalog No. 74106), including RNase-Free DNase (QIAGEN [RRID:SCR_008539], catalog No. 79256) treatment, according to the manufacturer's protocol. RNA integrity was determined using the Agilent 2100 Bioanalyzer (Agilent 2100 Bioanalyzer Instrument [RRID:SCR_018043]), and concentrations were measured by optical density (OD) readings on a Nanodrop ND-1000 Spectrophotometer (Nanodrop Technologies [RRID:SCR_016517]) as described previously (28). RNA integrity numbers were above 7 in 41 patients, and between 5.6 and 6.9 in 10 patients.

Reverse Transcription–Quantitative Polymerase Chain Reaction

Reverse transcription (RT) was performed using a high-capacity complementary DNA (cDNA) reverse transcription kit (Applied Biosystems [RRID:SCR_005039], catalog No. 4368814) by a Labnet MultiGene Gradient Thermal Cycler (Labnet MultiGene Gradient PCR Thermal Cycler [RRID:SCR_025065], Labnet International Inc) according to the manufacturer's protocol, using a total of 1 µg RNA. After the reaction, the cDNA was diluted to a ratio of 1:10. RT-qPCR was performed in an ABI 7900 (Applied Biosystems [RRID:SCR_005039]) using Power SYBR Green Master Mix (Applied Biosystems [RRID:SCR_005039], catalog No. 4367659). Samples were dispensed in the corresponding wells by an automated pipetting system (Eppendorf epMotion 5070 CB [RRID:SCR_025064]). Samples were run in duplicates with 10 μL per reaction, including 2 μL of the cDNA at 1:10 dilution. Primer concentration per reaction was 375 nM. Primer pairs were found either in the literature or in the primer bank (29). All primers were tested for specificity using BLAST analysis (NCBI BLAST [RRID:SCR_004870]). All the polymerase chain reaction (PCR) reactions had an efficiency between 90% and 100% (−3.6 ≥ slope ≥ −3.3). The amplification efficiency and correlation coefficients for these primers (Sigma-Aldrich [RRID:SCR_008988]) were obtained from the slope of the standard curves. To avoid false-positive amplification due to DNA contamination, the genomic DNA sequence (downloaded from Ensemble Data Base, Ensembl [RRID:SCR_002344]) was used to check exon-intron borders by matching the primers to their location. Target gene cycle threshold (Ct) value was adjusted by using the ΔCt method for the geometrical Ct mean of 2 reference genes (GAPDH and ALAS1), as previously validated for pituitary tumor samples (28), and to Actb for AtT-20 samples. Human primers are presented in Supplementary Table S1 (30). Gene expression data for ER protein-processing genes (CALR, GRP78, GRP94, and UGGT2) were previously presented (14). In our CA tumor cohort the gene expression Ct values range were as follows: POMC (12.5-27.8), TPIT (18.2-29.7), PCSK1 (17.5-26.8), PCSK1N (18.1-29.9), GAPDH (17.2-20.5), and ALAS1 (21.7-26.3) cycles.

Cell Culture and 90-kDa Heat Shock Protein Inhibition

AtT-20 mouse pituitary corticotroph tumor cells (ATCC catalog No. CRL-1795, RRID:CVCL_4109) were cultured in Dulbecco's Modified Eagle's medium-high glucose (Sigma-Aldrich [RRID:SCR_008988], catalog No. D6429) supplemented with 10% fetal bovine serum (FBS, South America, Premium, Biowest [RRID:SCR_025063], catalog No. S181B) and 1% penicillin/streptomycin (Gibco Life Technologies [RRID:SCR_008817], catalog No. 15140122) at 37 °C in a 5% CO2 incubator.

The cells were seeded in triplicate in 12-well plates containing 1.5 × 105 cells/well and cultured for 72 hours before stimulation for 6 and 24 hours by 17-AAG (50 μM, MedChemExpress [RRID:SCR_025062], catalog No. HY-10211). Dimethyl sulfoxide (DMSO) was used as control. Mouse primers are presented in Supplementary Table S2 (30). For proteomic analysis, the media was removed, and cells were washed with phosphate-buffered saline and lysed using radioimmunoprecipitation assay lysis buffer (Merck Millipore, Merck [RRID:SCR_001287], catalog No. 20-188) and halt protease and phosphatase inhibitor cocktail (Thermo Fisher Scientific [RRID:SCR_008452], catalog No. 78441), according to the manufacturer's protocol.

Quantitative Label-Free Mass Spectrometry–Based Proteomic Analysis

The protein lysates from cells treated for 24 hours with 17-AAG (4 biological replicates) and controls (DMSO, 7 biological replicates) were subjected to quantitative label-free mass spectrometry (MS)–based proteomic analysis. Protein concentration was first estimated by bicinchoninic acid assay (Pierce BCA Protein Assay Kits, Thermo Fisher Scientific [RRID:SCR_008452], catalog No. 23225), and for each replicate an equal amount (10 μg) of protein was precipitated on amine beads (31). The precipitated proteins were dissolved in 50-mM ammonium bicarbonate, reduced, alkylated, and digested with trypsin (1:50 enzyme:protein ratio; Promega) at 37 °C overnight. Digested peptides were acidified, and the peptides were loaded into Evosep C18 tips.

Liquid chromatography (LC)-MS/MS analysis was carried out using an Evosep LC system (Evosep [RRID:SCR_024590]) coupled to a timsTOF fleX mass spectrometer (Bruker Daltonics [RRID:SCR_023608]), using a CaptiveSpray nanoelectrospray ion source (Bruker Daltonics [RRID:SCR_023608]). A total of 200 ng of peptide digest was loaded on a capillary C18 Evosep column (15 cm length, 150 μm inner diameter, 1.5 μm particle size, 120; Evosep [RRID:SCR_024590]). Peptides were separated at 50 °C using a 44-minute gradient. The timsTOF fleX was operated in PASEF mode. Mass spectra for MS and MS/MS scans were recorded between m/z 100 and 1700. Ion mobility resolution was set to 0.60 to 1.60 V s/cm over a ramp time of 100 ms. Data-dependent acquisition was performed using 10 PASEF MS/MS scans per cycle with a near 100% duty cycle. A polygon filter was applied in the m/z and ion mobility space to exclude low m/z, singly charged ions from PASEF precursor selection. An active exclusion time of 0.4 minutes was applied to precursors that reached 20 000 intensity units. Collisional energy was ramped stepwise as a function of ion mobility.

Raw files from LC-MS/MS analyses were submitted to MaxQuant 2.0.3.0 software for protein identification and label-free quantification (MaxQuant [RRID:SCR_014485]). Parameters were set as follows: Carbamidomethyl (C) was set as a fixed modification and protein N-acetylation and methionine oxidation as variable modifications. First search error window of 20 ppm and mains search error of 6 ppm. Trypsin without proline restriction enzyme option was used, with 2 allowed miscleavages. Minimal unique peptides were set to one, and the false discovery rate (FDR) allowed was 0.01 (1%) for peptide and protein identification. The Uniprot mouse database was used (UniProt [RRID:SCR_002380]). Generation of reversed sequences was selected to assign FDR rate. Known contaminants as provided by MaxQuant and identified in the samples were excluded from further analysis.

Statistics

Statistical analyses were performed using IBM SPSS version 27.0.0.0 (IBM SPSS Statistics [RRID:SCR_016479]). Data are presented as mean ± 1 SD or as median (minimum-maximum) or interquartile range depending on distribution for continuous measures. Correlation analyses were performed using 2-tailed Spearman correlation tests, and correlation coefficient (rs) and statistical significance level (P) presented. The Mann-Whitney U test was used to test the differences between the groups. Perseus version 1.6.15.0 (Perseus [RRID:SCR_015753]) was used for further analysis of MaxQuant data: label-free quantitation intensity values were lg10 transformed, a minimum of 50% valid values in at least one group was required, and missing values were imputed from the low end of normal distribution. The t test between the groups (permutation-based FDR < 0.05) was performed to find the significant differentially expressed proteins (DEPs), and q (P value adjusted) less than .05 was considered statistically significant. The heat map was generated in R version 4.2.2 using the gplots2::heatmap.2 function (parameter: scale = “row”).

Results

Patient Clinical Characteristics

Clinical characteristics of the patients are presented in Table 1. The groups were similar regarding sex, whereas patients with FCAs were significantly younger and had smaller tumors. All SCAs and half of the FCAs were macroadenomas. Morning plasma ACTH and cortisol levels, measured preoperatively, were significantly higher in FCA compared with SCA patients.

Table 1.

Patient characteristics

VariablesFCA
n = 33
SCA
n = 18
 
Clinical
 Women/Men20/138/10
 Age at surgery, y47.3 (12.6)56.8 (11.1)P = .015
 Tumor largest diameter, mm8 (1-36); n = 2926 (10-62); n = 17P < .001
 Macro/Micro/Missing11/22/118/0/0
Biochemical
 ACTH, pmol/L11.5 (3.7-63.7); n = 285.6 (2.4-46.4); n = 7P = .01
 Cortisol, nmol/L659 (253-1487); n = 29361 (249-574); n = 10P < .001
Relative gene expressiona
POMC166.10 (0.04-562.10)9.60 (0.01-100.90)P < .001
TPIT2.23 (0.01-6.79)0.88 (0.05-5.06)P = .004
PCSK10.84 (0.02-7.85)0.27 (0.01-1.02)P < .001
PCSK1N0.229 (0.003-2.658)2.265 (0.042-7.206)P < .001
VariablesFCA
n = 33
SCA
n = 18
 
Clinical
 Women/Men20/138/10
 Age at surgery, y47.3 (12.6)56.8 (11.1)P = .015
 Tumor largest diameter, mm8 (1-36); n = 2926 (10-62); n = 17P < .001
 Macro/Micro/Missing11/22/118/0/0
Biochemical
 ACTH, pmol/L11.5 (3.7-63.7); n = 285.6 (2.4-46.4); n = 7P = .01
 Cortisol, nmol/L659 (253-1487); n = 29361 (249-574); n = 10P < .001
Relative gene expressiona
POMC166.10 (0.04-562.10)9.60 (0.01-100.90)P < .001
TPIT2.23 (0.01-6.79)0.88 (0.05-5.06)P = .004
PCSK10.84 (0.02-7.85)0.27 (0.01-1.02)P < .001
PCSK1N0.229 (0.003-2.658)2.265 (0.042-7.206)P < .001

Data are given as mean (1 SD) or median (minimum-maximum) depending on distribution for continuous measures, and total (n) for categorical measures.

Abbreviations: ACTH, adrenocorticotropic hormone; FCA, functioning corticotroph adenomas; n; number of patients; PCSK1, proprotein convertase subtilisin/kexin type 1; PCSK1N, proprotein convertase subtilisin/kexin type 1 inhibitor; POMC, pro-opiomelanocortin; SCA, silent corticotroph adenoma.

aData are presented as ΔCt-adjusted values (ie, 2^−(Ct target – Ct geometric mean (GAPDH;ALAS1)).

Table 1.

Patient characteristics

VariablesFCA
n = 33
SCA
n = 18
 
Clinical
 Women/Men20/138/10
 Age at surgery, y47.3 (12.6)56.8 (11.1)P = .015
 Tumor largest diameter, mm8 (1-36); n = 2926 (10-62); n = 17P < .001
 Macro/Micro/Missing11/22/118/0/0
Biochemical
 ACTH, pmol/L11.5 (3.7-63.7); n = 285.6 (2.4-46.4); n = 7P = .01
 Cortisol, nmol/L659 (253-1487); n = 29361 (249-574); n = 10P < .001
Relative gene expressiona
POMC166.10 (0.04-562.10)9.60 (0.01-100.90)P < .001
TPIT2.23 (0.01-6.79)0.88 (0.05-5.06)P = .004
PCSK10.84 (0.02-7.85)0.27 (0.01-1.02)P < .001
PCSK1N0.229 (0.003-2.658)2.265 (0.042-7.206)P < .001
VariablesFCA
n = 33
SCA
n = 18
 
Clinical
 Women/Men20/138/10
 Age at surgery, y47.3 (12.6)56.8 (11.1)P = .015
 Tumor largest diameter, mm8 (1-36); n = 2926 (10-62); n = 17P < .001
 Macro/Micro/Missing11/22/118/0/0
Biochemical
 ACTH, pmol/L11.5 (3.7-63.7); n = 285.6 (2.4-46.4); n = 7P = .01
 Cortisol, nmol/L659 (253-1487); n = 29361 (249-574); n = 10P < .001
Relative gene expressiona
POMC166.10 (0.04-562.10)9.60 (0.01-100.90)P < .001
TPIT2.23 (0.01-6.79)0.88 (0.05-5.06)P = .004
PCSK10.84 (0.02-7.85)0.27 (0.01-1.02)P < .001
PCSK1N0.229 (0.003-2.658)2.265 (0.042-7.206)P < .001

Data are given as mean (1 SD) or median (minimum-maximum) depending on distribution for continuous measures, and total (n) for categorical measures.

Abbreviations: ACTH, adrenocorticotropic hormone; FCA, functioning corticotroph adenomas; n; number of patients; PCSK1, proprotein convertase subtilisin/kexin type 1; PCSK1N, proprotein convertase subtilisin/kexin type 1 inhibitor; POMC, pro-opiomelanocortin; SCA, silent corticotroph adenoma.

aData are presented as ΔCt-adjusted values (ie, 2^−(Ct target – Ct geometric mean (GAPDH;ALAS1)).

Gene Expression of Corticotroph Cell Markers

Fig. 1 shows that gene expression of POMC (Fig. 1A), TPIT (Fig. 1B), and PCSK1 (Fig. 1C) were significantly higher, whereas PCSK1N (Fig. 1D) was lower, in FCAs as compared to SCAs. As a median, the POMC, TPIT, and PCSK1 gene expression fold change between FCA and SCA was 16.6, 2.4, and 3.2 times higher. Conversely, PCSK1N was a median of 9.9 times higher in SCA as compared to FCA (see Table 1). All the corticotroph cell markers and the housekeeping genes were highly expressed in our CA tumor cohort (Ct values < 30 cycles).

Gene expression of corticotroph cell markers and PCSK1N. FCAs express higher levels of POMC (A), TPIT (B), and PCSK1 (C) and lower levels of PCSK1N (D) as compared to SCAs. Data are presented as scatter dot plots with median and interquartile range, and individual dots represent each patient. Relative gene expression is measured by ΔCt method (ie, 2^−(Ct target gene – Ct geometric mean (GAPDH;ALAS1)). FCA, functioning corticotroph adenoma; POMC, pro-opiomelanocortin; TPIT, T box transcription factor; PCSK1, proprotein convertase subtilisin/kexin type 1; PCSK1N, proprotein convertase subtilisin/kexin type 1 inhibitor; SCA, silent corticotroph adenoma.
Figure 1.

Gene expression of corticotroph cell markers and PCSK1N. FCAs express higher levels of POMC (A), TPIT (B), and PCSK1 (C) and lower levels of PCSK1N (D) as compared to SCAs. Data are presented as scatter dot plots with median and interquartile range, and individual dots represent each patient. Relative gene expression is measured by ΔCt method (ie, 2^−(Ct target gene – Ct geometric mean (GAPDH;ALAS1)). FCA, functioning corticotroph adenoma; POMC, pro-opiomelanocortin; TPIT, T box transcription factor; PCSK1, proprotein convertase subtilisin/kexin type 1; PCSK1N, proprotein convertase subtilisin/kexin type 1 inhibitor; SCA, silent corticotroph adenoma.

Correlation Analyses in the Entire Cohort of Corticotroph Tumors

POMC gene expression correlated positively with TPIT (rs = 0.720; P < .001; n = 51) and PCSK1 (rs = 0.569; P < .001; n = 51), and negatively with tumor size (rs = −0.491; P < .001; n = 46).

PCSK1N correlated negatively with POMC (rs = −0.514; P < .001; n = 51) (Fig. 2A), TPIT (rs = −0.386; P = .005; n = 51), and cortisol (rs = −0.356; P = .026; n = 39). However, a positive correlation was shown with tumor size (rs = 0.645; P < .001; n = 46) (Fig. 2B). The correlation between PCSK1N and tumor size was also statistically significant in the FCA group alone (rs = 0.655; P < .001) but not in the SCA group alone (rs = 0.264; P > .05). PCSK1N correlated negatively with genes involved in ER processing (CALR, GRP78, GRP94, and UGGT2) (−0.57 < rs < −0.35; P < .05; n = 44). No correlation of PCSK1N with plasma ACTH levels was found (rs = −0.208; P = not significant; n = 35).

Correlation plots between gene expression of PCSK1N and POMC (A), and tumor size (B). Spearman correlation plots show negative correlation between relative gene expression (lg10) of PCSK1N and POMC (n = 51) (A), and positive correlation between PCSK1N and tumor size (n = 46) (B) in the entire cohort of corticotroph adenomas. Blue dots represent FCA, orange squares represent SCA.
Figure 2.

Correlation plots between gene expression of PCSK1N and POMC (A), and tumor size (B). Spearman correlation plots show negative correlation between relative gene expression (lg10) of PCSK1N and POMC (n = 51) (A), and positive correlation between PCSK1N and tumor size (n = 46) (B) in the entire cohort of corticotroph adenomas. Blue dots represent FCA, orange squares represent SCA.

Abbreviations: PCSK1N, proprotein convertase subtilisin/kexin type 1 inhibitor; POMC, pro-opiomelanocortin; FCA, functioning corticotroph adenoma; SCA, silent corticotroph adenoma.

Induction of Endoplasmic Reticulum Stress Response in AtT-20 Cells

Treatment of the AtT-20 mouse corticotroph tumor cell line with 17-AAG (50 µM), an HSP90 chaperone inhibitor, to induce ER stress, led to a decrease of Pomc gene expression at 24 hours (Fig. 3A) and an increase of Pcsk1n expression at 6 and 24 hours (Fig. 3B). Gene expression of several ER chaperones, including Grp94, Calr, and Grp78, increased at 24 hours, as expected, as a consequence of induced ER stress (Fig. 3C-3E). Furthermore, Uggt1, a molecule that provides quality control by selectively reglycosylating unfolded glycoproteins, showed increased expression at both time points (Fig. 3F).

ER stress response in AtT-20 mouse corticotroph tumor cell line. AtT-20 mouse corticotroph tumor cell line was stimulated with 17-AAG (50 µM) for 6 and 24 hours. Bar graphs show gene expression adjusted to β-actin of Pomc (A), Pcsk1n (B), and genes involved in ER protein folding/stress (C-K). DMSO is used as control. Data are presented as mean ± SD of 3 biological and 3 technical replicates. ****P less than .0001; ***P less than or equal to .002; **P less than or equal to .005; *P less than .05.
Figure 3.

ER stress response in AtT-20 mouse corticotroph tumor cell line. AtT-20 mouse corticotroph tumor cell line was stimulated with 17-AAG (50 µM) for 6 and 24 hours. Bar graphs show gene expression adjusted to β-actin of Pomc (A), Pcsk1n (B), and genes involved in ER protein folding/stress (C-K). DMSO is used as control. Data are presented as mean ± SD of 3 biological and 3 technical replicates. ****P less than .0001; ***P less than or equal to .002; **P less than or equal to .005; *P less than .05.

Abbreviations: 17-AAG, 17-N-allylamino-17-demethoxygeldanamycin; DMSO, dimethyl sulfoxide; ER, endoplasmic reticulum; PCSK1N, proprotein convertase subtilisin/kexin type 1 inhibitor; POMC, pro-opiomelanocortin.

As the induction of ER stress response leads to the activation of the UPR pathway, genes involved in UPR pathway were also measured. Ire1, showed a trend toward increased expression at 6 hours, becoming significant just at 24 hours, after 17-AAG treatment (Fig. 3G). Furthermore, a significant decrease of the s/uXbp1 ratio, one of the main readouts of the IRE1 pathway, was observed (Fig. 3L), suggesting a compensatory response of the cells already at 6 hours, becoming even more prominent at 24 hours. Atf4 and Atf6 showed similar patterns of regulation as for Ire1, but reached statistical significance only for Atf4 at 6 hours (Fig. 3H and 3I).

The ratio of Bax/Bcl-XL, molecules involving cell death and survival as an end readout of the entire UPR response pathway, showed increased expression both at 6 and 24 hours (Fig. 3J). In contrast, Bax/Bcl-2 ratio was not significantly regulated (Fig. 3K).

Proteomic Analyses of AtT-20 Cell Lysates

To assess the ER stress response at the protein level, we performed proteomic analysis and identified 1118 DEPs regulated by 17-AAG treatment at 24 hours (Supplementary Excel file) (30). Six Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways were selected based on the expected response to ER protein-folding inhibition and their relevance to the study. All the DEPs within these pathways are presented as a heat map (Fig. 4). Furthermore, based on literature review and each protein's fold change, we present the most relevant proteins within these 6 pathways in Table 2.

Differentially expressed proteins regulated by 17-AAG in ATt-20, involved in 6 selected KEGG signaling pathways. The heat map shows cell lysate protein expression profiles of AtT-20 mouse corticotroph tumor cell line treated with 17-AAG (4 biological replicates) compared with controls (DMSO, 7 biological replicates) for 24 hours. Functional annotation is shown based on KEGG pathways. Protein names are shown on the right side. Data were normalized by Z score before clustering. Each row indicates a protein, each column represents a sample. For the color scale, red indicates increase expression, white represents no differences, and blue indicates decrease expression after 17-AAG treatment.
Figure 4.

Differentially expressed proteins regulated by 17-AAG in ATt-20, involved in 6 selected KEGG signaling pathways. The heat map shows cell lysate protein expression profiles of AtT-20 mouse corticotroph tumor cell line treated with 17-AAG (4 biological replicates) compared with controls (DMSO, 7 biological replicates) for 24 hours. Functional annotation is shown based on KEGG pathways. Protein names are shown on the right side. Data were normalized by Z score before clustering. Each row indicates a protein, each column represents a sample. For the color scale, red indicates increase expression, white represents no differences, and blue indicates decrease expression after 17-AAG treatment.

Abbreviations: 17-AAG, 17-N-allylamino-17-demethoxygeldanamycin; Con, control; DMSO, dimethyl sulfoxide; KEGG, Kyoto Encyclopedia of Genes and Genomes.

Table 2.

Selected proteins belonging to the 6 signaling pathways differentially regulated in AtT-20 mouse corticotroph tumor cell line after treatment with 17-N-allylamino-17-demethoxygeldanamycin for 24 hours

KEGG pathwayProtein short nameProtein descriptionProteomics fold change
Protein processing in endoplasmic reticulum
ko04141
Hspa5/BiP/Grp78Heat shock protein family A (Hsp70) member 53.5
Hsph1Heat shock protein family H (Hsp110) member 13.3
Pdia6Protein disulfide isomerase family A member 62.2
Syvn1/Hrd1Synoviolin 12.1
Hsp90b1/Grp94Heat shock protein 90 β family member 12.0
Os9OS9 ER lectin1.9
Erlec1ER lectin 11.7
Uggt1UDP-glucose glycoprotein glucosyltransferase 11.7
Sel1LSEL1L adaptor subunit of ERAD E3 ubiquitin ligase1.7
PrkcshGlucosidase 2 subunit β/Protein kinase C substrate 80K-H1.5
CanxCalnexin1.5
VcpValosin-containing protein1.4
Edem3ER degradation enhancing α-mannosidase like protein 31.3
Nploc4NPL4 homologue, ubiquitin recognition factor1.2
Dnajc10DnaJ heat shock protein family (Hsp40) member C101.2
Ptpn1/Ptp1bProtein tyrosine phosphatase non-receptor type 1−1.1
Apoptosis
ko04210
CtsdCathepsin D1.7
Capn1Calpain 1−1.2
Eif2s1Eukaryotic translation initiation factor 2 subunit α−1.4
TraddTNFRSF1A associated via death domain−2.1
EndogEndonuclease G−2.4
Itpr3Inositol 14,5-trisphosphate receptor type 3−2.6
AtmATM serine/threonine kinase−6.5
Cell cycle
ko04110
Rad21RAD21 cohesin complex component1.3
Smc3Structural maintenance of chromosomes 31.3
Mad1l1Mitotic arrest deficient 1 like 1−2.3
Cdk1Cyclin-dependent kinase 1−10.6
Adhesion
ko04510
ko04514
Col1a1Collagen type I α 1 chain1.9
AlcamActivated leukocyte cell adhesion molecule1.5
Ctnnb1Catenin β 11.4
Itgb1Integrin subunit β 11.3
Ncam1Neural cell adhesion molecule 1−1.3
Arhgap35Rho GTPase activating protein 35−1.7
Gsk3bGlycogen synthase kinase 3 β−2.4
Cldn3Claudin-3−2.9
Cytoskeleton
ko04810
VclVinculin1.7
Actn4Actinin α 41.3
Itga6Integrin subunit α 61.3
MsnMoesin1.2
Iqgap1IQ motif containing GTPase activating protein 1−1.1
Pik3r1Phosphoinositide-3-kinase regulatory subunit 1−1.5
Cellular senescence
ko04218
Sqstm1/p62Sequestosome-14.6
Ppp1cbProtein phosphatase 1 catalytic subunit β−1.4
Map2k1Mitogen-activated protein kinase kinase 1−1.4
Smad2SMAD family member 2−1.5
MtorMechanistic target of rapamycin kinase−1.5
Mapk3Mitogen-activated protein kinase 3−1.7
Raf1Raf-1 proto-oncogene, serine/threonine kinase−1.8
RelaRELA proto-oncogene, NF-κB subunit−1.8
Cdk2Cyclin-dependent kinase 2−2.3
Foxo3Forkhead box O3−2.7
Map2k3Mitogen-activated protein kinase kinase 3−3.5
Cdk4Cyclin-dependent kinase 4−5.4
KEGG pathwayProtein short nameProtein descriptionProteomics fold change
Protein processing in endoplasmic reticulum
ko04141
Hspa5/BiP/Grp78Heat shock protein family A (Hsp70) member 53.5
Hsph1Heat shock protein family H (Hsp110) member 13.3
Pdia6Protein disulfide isomerase family A member 62.2
Syvn1/Hrd1Synoviolin 12.1
Hsp90b1/Grp94Heat shock protein 90 β family member 12.0
Os9OS9 ER lectin1.9
Erlec1ER lectin 11.7
Uggt1UDP-glucose glycoprotein glucosyltransferase 11.7
Sel1LSEL1L adaptor subunit of ERAD E3 ubiquitin ligase1.7
PrkcshGlucosidase 2 subunit β/Protein kinase C substrate 80K-H1.5
CanxCalnexin1.5
VcpValosin-containing protein1.4
Edem3ER degradation enhancing α-mannosidase like protein 31.3
Nploc4NPL4 homologue, ubiquitin recognition factor1.2
Dnajc10DnaJ heat shock protein family (Hsp40) member C101.2
Ptpn1/Ptp1bProtein tyrosine phosphatase non-receptor type 1−1.1
Apoptosis
ko04210
CtsdCathepsin D1.7
Capn1Calpain 1−1.2
Eif2s1Eukaryotic translation initiation factor 2 subunit α−1.4
TraddTNFRSF1A associated via death domain−2.1
EndogEndonuclease G−2.4
Itpr3Inositol 14,5-trisphosphate receptor type 3−2.6
AtmATM serine/threonine kinase−6.5
Cell cycle
ko04110
Rad21RAD21 cohesin complex component1.3
Smc3Structural maintenance of chromosomes 31.3
Mad1l1Mitotic arrest deficient 1 like 1−2.3
Cdk1Cyclin-dependent kinase 1−10.6
Adhesion
ko04510
ko04514
Col1a1Collagen type I α 1 chain1.9
AlcamActivated leukocyte cell adhesion molecule1.5
Ctnnb1Catenin β 11.4
Itgb1Integrin subunit β 11.3
Ncam1Neural cell adhesion molecule 1−1.3
Arhgap35Rho GTPase activating protein 35−1.7
Gsk3bGlycogen synthase kinase 3 β−2.4
Cldn3Claudin-3−2.9
Cytoskeleton
ko04810
VclVinculin1.7
Actn4Actinin α 41.3
Itga6Integrin subunit α 61.3
MsnMoesin1.2
Iqgap1IQ motif containing GTPase activating protein 1−1.1
Pik3r1Phosphoinositide-3-kinase regulatory subunit 1−1.5
Cellular senescence
ko04218
Sqstm1/p62Sequestosome-14.6
Ppp1cbProtein phosphatase 1 catalytic subunit β−1.4
Map2k1Mitogen-activated protein kinase kinase 1−1.4
Smad2SMAD family member 2−1.5
MtorMechanistic target of rapamycin kinase−1.5
Mapk3Mitogen-activated protein kinase 3−1.7
Raf1Raf-1 proto-oncogene, serine/threonine kinase−1.8
RelaRELA proto-oncogene, NF-κB subunit−1.8
Cdk2Cyclin-dependent kinase 2−2.3
Foxo3Forkhead box O3−2.7
Map2k3Mitogen-activated protein kinase kinase 3−3.5
Cdk4Cyclin-dependent kinase 4−5.4

The table shows KEGG pathways with protein name, protein description, and proteomic fold change. Proteins upregulated in 17-AAG–treated cells are presented with positive fold change values.

Abbreviations: 17-AAG, 17-N-allylamino-17-demethoxygeldanamycin; ER, endoplasmic reticulum; KEGG, Kyoto Encyclopedia of Genes and Genomes; NF-κB; nuclear factor κB.

Table 2.

Selected proteins belonging to the 6 signaling pathways differentially regulated in AtT-20 mouse corticotroph tumor cell line after treatment with 17-N-allylamino-17-demethoxygeldanamycin for 24 hours

KEGG pathwayProtein short nameProtein descriptionProteomics fold change
Protein processing in endoplasmic reticulum
ko04141
Hspa5/BiP/Grp78Heat shock protein family A (Hsp70) member 53.5
Hsph1Heat shock protein family H (Hsp110) member 13.3
Pdia6Protein disulfide isomerase family A member 62.2
Syvn1/Hrd1Synoviolin 12.1
Hsp90b1/Grp94Heat shock protein 90 β family member 12.0
Os9OS9 ER lectin1.9
Erlec1ER lectin 11.7
Uggt1UDP-glucose glycoprotein glucosyltransferase 11.7
Sel1LSEL1L adaptor subunit of ERAD E3 ubiquitin ligase1.7
PrkcshGlucosidase 2 subunit β/Protein kinase C substrate 80K-H1.5
CanxCalnexin1.5
VcpValosin-containing protein1.4
Edem3ER degradation enhancing α-mannosidase like protein 31.3
Nploc4NPL4 homologue, ubiquitin recognition factor1.2
Dnajc10DnaJ heat shock protein family (Hsp40) member C101.2
Ptpn1/Ptp1bProtein tyrosine phosphatase non-receptor type 1−1.1
Apoptosis
ko04210
CtsdCathepsin D1.7
Capn1Calpain 1−1.2
Eif2s1Eukaryotic translation initiation factor 2 subunit α−1.4
TraddTNFRSF1A associated via death domain−2.1
EndogEndonuclease G−2.4
Itpr3Inositol 14,5-trisphosphate receptor type 3−2.6
AtmATM serine/threonine kinase−6.5
Cell cycle
ko04110
Rad21RAD21 cohesin complex component1.3
Smc3Structural maintenance of chromosomes 31.3
Mad1l1Mitotic arrest deficient 1 like 1−2.3
Cdk1Cyclin-dependent kinase 1−10.6
Adhesion
ko04510
ko04514
Col1a1Collagen type I α 1 chain1.9
AlcamActivated leukocyte cell adhesion molecule1.5
Ctnnb1Catenin β 11.4
Itgb1Integrin subunit β 11.3
Ncam1Neural cell adhesion molecule 1−1.3
Arhgap35Rho GTPase activating protein 35−1.7
Gsk3bGlycogen synthase kinase 3 β−2.4
Cldn3Claudin-3−2.9
Cytoskeleton
ko04810
VclVinculin1.7
Actn4Actinin α 41.3
Itga6Integrin subunit α 61.3
MsnMoesin1.2
Iqgap1IQ motif containing GTPase activating protein 1−1.1
Pik3r1Phosphoinositide-3-kinase regulatory subunit 1−1.5
Cellular senescence
ko04218
Sqstm1/p62Sequestosome-14.6
Ppp1cbProtein phosphatase 1 catalytic subunit β−1.4
Map2k1Mitogen-activated protein kinase kinase 1−1.4
Smad2SMAD family member 2−1.5
MtorMechanistic target of rapamycin kinase−1.5
Mapk3Mitogen-activated protein kinase 3−1.7
Raf1Raf-1 proto-oncogene, serine/threonine kinase−1.8
RelaRELA proto-oncogene, NF-κB subunit−1.8
Cdk2Cyclin-dependent kinase 2−2.3
Foxo3Forkhead box O3−2.7
Map2k3Mitogen-activated protein kinase kinase 3−3.5
Cdk4Cyclin-dependent kinase 4−5.4
KEGG pathwayProtein short nameProtein descriptionProteomics fold change
Protein processing in endoplasmic reticulum
ko04141
Hspa5/BiP/Grp78Heat shock protein family A (Hsp70) member 53.5
Hsph1Heat shock protein family H (Hsp110) member 13.3
Pdia6Protein disulfide isomerase family A member 62.2
Syvn1/Hrd1Synoviolin 12.1
Hsp90b1/Grp94Heat shock protein 90 β family member 12.0
Os9OS9 ER lectin1.9
Erlec1ER lectin 11.7
Uggt1UDP-glucose glycoprotein glucosyltransferase 11.7
Sel1LSEL1L adaptor subunit of ERAD E3 ubiquitin ligase1.7
PrkcshGlucosidase 2 subunit β/Protein kinase C substrate 80K-H1.5
CanxCalnexin1.5
VcpValosin-containing protein1.4
Edem3ER degradation enhancing α-mannosidase like protein 31.3
Nploc4NPL4 homologue, ubiquitin recognition factor1.2
Dnajc10DnaJ heat shock protein family (Hsp40) member C101.2
Ptpn1/Ptp1bProtein tyrosine phosphatase non-receptor type 1−1.1
Apoptosis
ko04210
CtsdCathepsin D1.7
Capn1Calpain 1−1.2
Eif2s1Eukaryotic translation initiation factor 2 subunit α−1.4
TraddTNFRSF1A associated via death domain−2.1
EndogEndonuclease G−2.4
Itpr3Inositol 14,5-trisphosphate receptor type 3−2.6
AtmATM serine/threonine kinase−6.5
Cell cycle
ko04110
Rad21RAD21 cohesin complex component1.3
Smc3Structural maintenance of chromosomes 31.3
Mad1l1Mitotic arrest deficient 1 like 1−2.3
Cdk1Cyclin-dependent kinase 1−10.6
Adhesion
ko04510
ko04514
Col1a1Collagen type I α 1 chain1.9
AlcamActivated leukocyte cell adhesion molecule1.5
Ctnnb1Catenin β 11.4
Itgb1Integrin subunit β 11.3
Ncam1Neural cell adhesion molecule 1−1.3
Arhgap35Rho GTPase activating protein 35−1.7
Gsk3bGlycogen synthase kinase 3 β−2.4
Cldn3Claudin-3−2.9
Cytoskeleton
ko04810
VclVinculin1.7
Actn4Actinin α 41.3
Itga6Integrin subunit α 61.3
MsnMoesin1.2
Iqgap1IQ motif containing GTPase activating protein 1−1.1
Pik3r1Phosphoinositide-3-kinase regulatory subunit 1−1.5
Cellular senescence
ko04218
Sqstm1/p62Sequestosome-14.6
Ppp1cbProtein phosphatase 1 catalytic subunit β−1.4
Map2k1Mitogen-activated protein kinase kinase 1−1.4
Smad2SMAD family member 2−1.5
MtorMechanistic target of rapamycin kinase−1.5
Mapk3Mitogen-activated protein kinase 3−1.7
Raf1Raf-1 proto-oncogene, serine/threonine kinase−1.8
RelaRELA proto-oncogene, NF-κB subunit−1.8
Cdk2Cyclin-dependent kinase 2−2.3
Foxo3Forkhead box O3−2.7
Map2k3Mitogen-activated protein kinase kinase 3−3.5
Cdk4Cyclin-dependent kinase 4−5.4

The table shows KEGG pathways with protein name, protein description, and proteomic fold change. Proteins upregulated in 17-AAG–treated cells are presented with positive fold change values.

Abbreviations: 17-AAG, 17-N-allylamino-17-demethoxygeldanamycin; ER, endoplasmic reticulum; KEGG, Kyoto Encyclopedia of Genes and Genomes; NF-κB; nuclear factor κB.

As expected due to the compensatory mechanisms of the ER inhibition, there were many upregulated proteins involved in protein processing in ER (see Fig. 4). Among these, several ER chaperones (GRP78, GRP94, PDIA6, CANX) and ER-associated degradation (ERAD) proteins (HRD1/SYVN1, OS9, ERLEC1, SEL1L, VCP, EDEM3, and DNAJC10) were observed (see Table 2). Furthermore, several proteins that are involved in the IRE1 pathway were regulated, such as increased PDIA6 and PRKCSH and decreased PTPN1 (see Table 2). No clear evidence for the activation at the protein level of the UPR-activated signaling pathways ATF4 and ATF6 was found.

Overall, the 17-AAG treatment resulted in a downregulation of apoptosis, cell cycle, and senescence pathways in addition to changes in cell adhesion and cytoskeleton protein composition indicating less proliferative and less-cohesive cells (see Fig. 4 and Table 2).

Discussion

CAs represent a continuum of tumors from SCA, to whispering and FCA, having in common the expression of ACTH and/or TPIT. Here, we have shown that PCSK1N expression is closely associated with tumor size in both subtypes. Furthermore, it may be a part of the ER stress-activated response facilitating corticotroph tumor cell survival, and potentially contributing to tumor growth.

PCSK1N has been described as an endogenous inhibitor of PCSK1 in AtT-20 cells. Higher expression in SCA might reduce proteolytic cleavage of POMC, and consecutively lower ACTH production and secretion. Interestingly, our data showed a negative association between PCSK1N gene expression and plasma cortisol, but not ACTH levels, suggesting an effect on POMC cleavage and functional ACTH formation in human CAs. Furthermore, in concordance with our data, PCSK1 has been shown to be higher expressed in FCA than in SCA in several studies (10, 11, 32), suggesting an impairment of the POMC processing ability in SCA. In addition, the present results show an overlap in the expression of corticotroph cell markers between FCA and SCA, supporting that they are not entirely separate entities, but represent a continuum of the same type of tumors.

Previous studies have indicated that PCSK1N might have other functions besides inhibiting PCSK1, as it has been found to be increased during disease progression in neurodegenerative diseases (33). Moreover, expression of PCSK1N and PCSK1 were not coregulated in the adult brain of PCSK1N-knockout mice and in rat islet cells (34). Recently, PCSK1N has been described as a potent antiaggregation chaperone, preventing β-amyloid aggregation into fibrils in a dose-dependent manner (35, 36). Following ER stress, Pcsk1n levels increase, rescuing the cells from toxicity and providing a survival advantage (19). Since PCSK1N has an antiaggregation function, the increase of PCSK1N cellular levels may represent an adaptation to prevent more protein aggregation that will increase the severity of ER stress. In addition to the increase of Pcsk1n, our data show that ER stress activation leads to a downregulation of apoptosis, cell cycle, and senescence signaling pathways, which could confer a survival advantage to the corticotroph tumor cell, thereby contributing to tumor growth in the longer perspective. As such, this mechanism could explain the strong association between PCSK1N expression and tumor size.

The UPR pathway is activated by 17-AAG treatment, as shown by the upregulation of the ER stress-response molecules Grp78, Grp94, and Uggt1 in our data, to maintain homeostasis. GRP78 has been found to be overexpressed in several tumor types, including lung cancer (37) and showed to be induced by oxidative stress to protect the cell from damage and death (38). Therefore, GRP78 overexpression is an important factor for tumor resistance to various stressors and tumor development. One of the mechanisms by which GRP78 inhibits apoptosis is stabilizing mitochondrial permeability and reducing cytochrome C release through the Raf-1 proto-oncogene, serine/threonine kinase (Raf-1) expression (37). Raf-1 protein expression was shown to be decreased by 17-AAG treatment in our data set. However, Raf-1 itself has been known to involve different pathways including cell cycle and nuclear factor κB activation, so the downregulation in our data might reflect other biological roles (39). Besides antiapoptotic activity, GRP78 has also been shown to be involved in tumor invasion. Upregulation of GRP78 can induce EMT and contribute to metastasis, whereas downregulation strongly inhibits in vitro tumor cell invasion and metastasis (40, 41). GRP94 is another protein involved in tumor proliferation, invasion, and EMT (42, 43). The knockdown of GRP94 in a lung cancer cell line was shown to inhibit tumor cell proliferation and increase apoptosis through caspase-7 (44). Based on our data, we suggest that the increase of both GRP78 and GRP94 by ER stress promotes cell survival by counteracting the cell death signal initiated by the proapoptotic BAX.

XBP1, as a member of the IRE1 pathway, is essential for cell survival during tumor growth and hypoxic conditions (45). During the ER stress response, the targets of the spliced form (sXBP1) include ER chaperones (GRP78, GRP94), autophagic response, and ERAD components. The unspliced form (uXBP1) is a negative feedback regulator of sXBP1, and the overexpression of uXBP1 is involved in sXBP1 degradation during the recovery phase of ER stress (46). Thus, the decreased s/uXbp ratio in our data suggests that the cells were in the recovery phase after ER stress induction, and this seems to occur very early within the first 6 hours after 17-AAG treatment. However, despite the early decrease in sXbp1 expression, the expression of Grp78 and Grp94 still continued to increase at 24 hours, potentially suggesting a compensatory mechanism.

Among the proteins involved in ERAD, SYVN1 plays a role to antagonize ER stress-induced apoptotic cell death by promoting IRE1 ubiquitination and degradation (47). Besides involving the fate of cell death or survival, IRE1 has also been known to potentiate tumor growth. Several proteins involved in IRE1 pathways were regulated in our study. Among these, PDIA6 and PRKCSH enhance IRE1 activation, so the upregulation, shown in our proteomics data, contributes most probably to the resistance to ER stress-induced cell death (48, 49). On the contrary, the absence of PTPN1 or PTP-1B can impair ER stress-induced apoptosis (50). Being downregulated by 17-AAG in our proteomics results, this may antagonize apoptosis induced by ER stress, again contributing to cell survival.

The observed decrease in NCAM1, a neuron-neuron adhesion and neural development protein, might lead to decreased cell adhesion, and influence cell attachment and migration properties. Downregulation of NCAM1 has been shown to be correlated with poor prognosis in colorectal cancer, increased migration of pancreatic cancer cells, and tumor metastasis in thyroid tumor (51, 52).

Lastly, we found several molecules involved in autophagy, among them CALR, GRP78, and SQSTM1/p62, to be upregulated as a response to 17-AAG–induced ER stress. During ER stress that is not severe enough to cause cell death, autophagy is always activated (53) and contributes to the protection against apoptotic cell death. Autophagy is a conserved process, which involves recycling of molecules within the lysosomes (54). This process is important for cell survival during stress conditions since UPR needs 2 degradation systems, ERAD and selective autophagy, to reduce the load of unfolded proteins (55).

To summarize, ER stress induction by 17-AAG treatment in AtT-20 cells resulted in the increase of genes and proteins involved in ER stress response, including ER chaperones, activation of UPR response (mainly IRE1/XBP1 pathway), and induction of ERAD. The final outcome is cell protection against apoptotic cell death. Decreasing the apoptosis may in the long term result in increased growth potential that contributes to the increase of tumor size observed in SCA.

Study Limitations

Assessment of ACTH or cortisol overproduction was not performed systematically in the SCA, as they were diagnosed by tumor IHC analysis after surgery. Second, the lack of standardized magnetic resonance imaging–based measurement of tumor volume may weaken the correlations using size as an aggressiveness marker. However, almost all the SCAs were diagnosed at a later stage of tumor development and most of them showed extrasellar growth, making tumor volume of little use as a predictor for aggressiveness. Lastly, ER stress induction in vitro shows data limited to 24 hours’ exposure, which serves only as a model for the mechanisms that may occur in vivo in tumors with longstanding ER stress. The prolonged ER stress occurring in the corticotroph tumor probably involves several other mechanisms that play a role in increased cell proliferation and escaping cell death.

Conclusion

PCSK1N is negatively associated with corticotroph pituitary cell markers, and positively with tumor size, being likely a part of the adaptive response initiated by ER stress, and together with other proteins, may contribute to a survival advantage of the corticotroph tumor cell.

Funding

M.A. received scholarships provided by the South-Eastern Norway Regional Health Authority (Open Project No. 2020081/2020). The study did not receive any external funding. Mass spectrometry–based proteomic analyses were performed by the Proteomics Core Facility, Department of Immunology, University of Oslo and Oslo University Hospital, which is supported by the Core Facilities program of the South-Eastern Norway Regional Health Authority. This core facility is also a member of the National Network of Advanced Proteomics Infrastructure (NAPI), funded by the Research Council of Norway INFRASTRUKTUR-program (project No. 295910).

Disclosures

There is no conflict of interest that could be perceived as prejudicing the impartiality of this study.

Data Availability

Some or all data sets generated during and/or analyzed during the present study are not publicly available but are available from the corresponding author on reasonable request.

References

1

Asa
SL
,
Mete
O
,
Perry
A
,
Osamura
RY
.
Overview of the 2022 WHO classification of pituitary tumors
.
Endocr Pathol
.
2022
;
33
(
1
):
6
26
.

2

Drummond
J
,
Roncaroli
F
,
Grossman
AB
,
Korbonits
M
.
Clinical and pathological aspects of silent pituitary adenomas
.
J Clin Endocrinol Metab
.
2019
;
104
(
7
):
2473
2489
.

3

Ben-Shlomo
A
,
Cooper
O
.
Silent corticotroph adenomas
.
Pituitary
.
2018
;
21
(
2
):
183
193
.

4

Tampourlou
M
,
Ntali
G
,
Ahmed
S
, et al.
Outcome of nonfunctioning pituitary adenomas that regrow after primary treatment: a study from two large UK centers
.
J Clin Endocrinol Metab
.
2017
;
102
(
6
):
1889
1897
.

5

Raverot
G
,
Ilie
MD
,
Lasolle
H
, et al.
Aggressive pituitary tumours and pituitary carcinomas
.
Nat Rev Endocrinol
.
2021
;
17
(
11
):
671
684
.

6

Kolnes
AJ
,
Øystese
KAB
,
Sjöstedt
E
, et al.
TGFBR3L is associated with gonadotropin production in non-functioning gonadotroph pituitary neuroendocrine tumours
.
Pituitary
.
2023
;
26
(
2
):
227
236
.

7

Karavitaki
N
.
Prevalence and incidence of pituitary adenomas
.
Ann Endocrinol (Paris)
.
2012
;
73
(
2
):
79
80
.

8

Ueyama
T
,
Tamaki
N
,
Kondoh
T
,
Kurata
H
.
Large and invasive silent corticotroph-cell adenoma with elevated serum ACTH: a case report
.
Surg Neurol
.
1998
;
50
(
1
):
30
31
;
discussion 32
.

9

Harno
E
,
Gali Ramamoorthy
T
,
Coll
AP
,
White
A
.
POMC: the physiological power of hormone processing
.
Physiol Rev
.
2018
;
98
(
4
):
2381
2430
.

10

Ohta
S
,
Nishizawa
S
,
Oki
Y
,
Yokoyama
T
,
Namba
H
.
Significance of absent prohormone convertase 1/3 in inducing clinically silent corticotroph pituitary adenoma of subtype I–immunohistochemical study
.
Pituitary
.
2002
;
5
(
4
):
221
223
.

11

Righi
A
,
Faustini-Fustini
M
,
Morandi
L
, et al.
The changing faces of corticotroph cell adenomas: the role of prohormone convertase 1/3
.
Endocrine
.
2017
;
56
(
2
):
286
297
.

12

Qian
Y
,
Devi
LA
,
Mzhavia
N
,
Munzer
S
,
Seidah
NG
,
Fricker
LD
.
The C-terminal region of proSAAS is a potent inhibitor of prohormone convertase 1
.
J Biol Chem
.
2000
;
275
(
31
):
23596
23601
.

13

Basak
A
,
Koch
P
,
Dupelle
M
, et al.
Inhibitory specificity and potency of proSAAS-derived peptides toward proprotein convertase 1
.
J Biol Chem
.
2001
;
276
(
35
):
32720
32728
.

14

Eieland
AK
,
Normann
KR
,
Sundaram
AYM
, et al.
Distinct pattern of endoplasmic reticulum protein processing and extracellular matrix proteins in functioning and silent corticotroph pituitary adenomas
.
Cancers (Basel)
.
2020
;
12
(
10
):
2980
.

15

Melmed
S
,
Kaiser
UB
,
Lopes
MB
, et al.
Clinical biology of the pituitary adenoma
.
Endocr Rev
.
2022
;
43
(
6
):
1003
1037
.

16

Zhang
D
,
Hugo
W
,
Bergsneider
M
, et al.
Single-cell RNA sequencing in silent corticotroph tumors confirms impaired POMC processing and provides new insights into their invasive behavior
.
Eur J Endocrinol
.
2022
;
187
(
1
):
49
64
.

17

Zhang
Q
,
Yao
B
,
Long
X
, et al.
Single-cell sequencing identifies differentiation-related markers for molecular classification and recurrence prediction of PitNET
.
Cell Rep Med
.
2023
;
4
(
2
):
100934
.

18

Wiseman
RL
,
Mesgarzadeh
JS
,
Hendershot
LM
.
Reshaping endoplasmic reticulum quality control through the unfolded protein response
.
Mol Cell
.
2022
;
82
(
8
):
1477
1491
.

19

Shakya
M
,
Yildirim
T
,
Lindberg
I
.
Increased expression and retention of the secretory chaperone proSAAS following cell stress
.
Cell Stress Chaperones
.
2020
;
25
(
6
):
929
941
.

20

Jaskulska
A
,
Janecka
AE
,
Gach-Janczak
K
.
Thapsigargin-from traditional medicine to anticancer drug
.
Int J Mol Sci
.
2020
;
22
(
1
):
4
.

21

Neckers
L
,
Workman
P
.
Hsp90 molecular chaperone inhibitors: are we there yet?
Clin Cancer Res
.
2012
;
18
(
1
):
64
76
.

22

Davenport
EL
,
Moore
HE
,
Dunlop
AS
, et al.
Heat shock protein inhibition is associated with activation of the unfolded protein response pathway in myeloma plasma cells
.
Blood
.
2007
;
110
(
7
):
2641
2649
.

23

Taiyab
A
,
Sreedhar
AS
,
Rao Ch
M
.
Hsp90 inhibitors, GA and 17AAG, lead to ER stress-induced apoptosis in rat histiocytoma
.
Biochem Pharmacol
.
2009
;
78
(
2
):
142
152
.

24

Evang
JA
,
Bollerslev
J
,
Casar-Borota
O
,
Lekva
T
,
Ramm-Pettersen
J
,
Berg
JP
.
Different levels of various glucocorticoid-regulated genes in corticotroph adenomas
.
Endocrine
.
2013
;
44
(
1
):
220
227
.

25

Øystese
KAB
,
Casar-Borota
O
,
Berg-Johnsen
J
,
Berg
JP
,
Bollerslev
J
.
Distribution of E- and N-cadherin in subgroups of non-functioning pituitary neuroendocrine tumours
.
Endocrine
.
2022
;
77
(
1
):
151
159
.

26

Sjöstedt
E
,
Bollerslev
J
,
Mulder
J
,
Lindskog
C
,
Pontén
F
,
Casar-Borota
O
.
A specific antibody to detect transcription factor T-Pit: a reliable marker of corticotroph cell differentiation and a tool to improve the classification of pituitary neuroendocrine tumours
.
Acta Neuropathol
.
2017
;
134
(
4
):
675
677
.

27

Øystese
KA
,
Casar-Borota
O
,
Normann
KR
,
Zucknick
M
,
Berg
JP
,
Bollerslev
J
.
Estrogen receptor α, a sex-dependent predictor of aggressiveness in nonfunctioning pituitary adenomas: SSTR and sex hormone receptor distribution in NFPA
.
J Clin Endocrinol Metab
.
2017
;
102
(
9
):
3581
3590
.

28

Normann
KR
,
Oystese
KA
,
Berg
JP
, et al.
Selection and validation of reliable reference genes for RT-qPCR analysis in a large cohort of pituitary adenomas
.
Mol Cell Endocrinol
.
2016
;
437
:
183
189
.

29

Spandidos
A
,
Wang
X
,
Wang
H
,
Seed
B
.
PrimerBank: a resource of human and mouse PCR primer pairs for gene expression detection and quantification
.
Nucleic Acids Res
.
2010
;
38
(
suppl_1
):
D792
D799
.

30

Abusdal
M
,
Nyman
T
,
Olarescu
N.
JCEM Supplementary files—Primers and DEPs. Dryad Digital Repository. doi:. Accessed 5 April 2024.

31

Batth
TS
,
Tollenaere
MX
,
Rüther
P
, et al.
Protein aggregation capture on microparticles enables multipurpose proteomics sample preparation
.
Mol Cell Proteomics
.
2019
;
18
(
5
):
1027
1035
.

32

Tateno
T
,
Izumiyama
H
,
Doi
M
,
Akashi
T
,
Ohno
K
,
Hirata
Y
.
Defective expression of prohormone convertase 1/3 in silent corticotroph adenoma
.
Endocr J
.
2007
;
54
(
5
):
777
782
.

33

Lindberg
I
,
Shu
Z
,
Lam
H
, et al.
The proSAAS chaperone provides neuroprotection and attenuates transsynaptic α-synuclein spread in rodent models of Parkinson's disease
.
Journal of Parkinson's Disease
.
2022
;
12
(
5
):
1463
1478
.

34

Alarcon
C
,
Verchere
CB
,
Rhodes
CJ
.
Translational control of glucose-induced islet amyloid polypeptide production in pancreatic islets
.
Endocrinology
.
2012
;
153
(
5
):
2082
2087
.

35

Jarvela
TS
,
Lam
HA
,
Helwig
M
, et al.
The neural chaperone proSAAS blocks α-synuclein fibrillation and neurotoxicity
.
Proceedings of the National Academy of Sciences
.
2016
;
113
(
32
):
E4708
E4715
.

36

Hoshino
A
,
Helwig
M
,
Rezaei
S
,
Berridge
C
,
Eriksen
JL
,
Lindberg
I
.
A novel function for proSAAS as an amyloid anti-aggregant in Alzheimer's disease
.
J Neurochem
.
2014
;
128
(
3
):
419
430
.

37

Xia
S
,
Duan
W
,
Liu
W
,
Zhang
X
,
Wang
Q
.
GRP78 in lung cancer
.
J Transl Med
.
2021
;
19
(
1
):
118
.

38

Dauer
P
,
Sharma
NS
,
Gupta
VK
, et al.
ER stress sensor, glucose regulatory protein 78 (GRP78) regulates redox status in pancreatic cancer thereby maintaining “stemness”
.
Cell Death Dis
.
2019
;
10
(
2
):
132
.

39

McCubrey
JA
,
Steelman
LS
,
Chappell
WH
, et al.
Roles of the Raf/MEK/ERK pathway in cell growth, malignant transformation and drug resistance
.
Biochim Biophys Acta
.
2007
;
1773
(
8
):
1263
1284
.

40

Chiu
CC
,
Lin
CY
,
Lee
LY
, et al.
Glucose-regulated protein 78 regulates multiple malignant phenotypes in head and neck cancer and may serve as a molecular target of therapeutic intervention
.
Mol Cancer Ther
.
2008
;
7
(
9
):
2788
2797
.

41

Zhang
L
,
Li
Z
,
Fan
Y
,
Li
H
,
Li
Z
,
Li
Y
.
Overexpressed GRP78 affects EMT and cell-matrix adhesion via autocrine TGF-β/Smad2/3 signaling
.
Int J Biochem Cell Biol
.
2015
;
64
:
202
211
.

42

Wei
PL
,
Huang
CY
,
Tai
CJ
, et al.
Glucose-regulated protein 94 mediates metastasis by CCT8 and the JNK pathway in hepatocellular carcinoma
.
Tumour Biol
.
2016
;
37
(
6
):
8219
8227
.

43

Duan
X
,
Iwanowycz
S
,
Ngoi
S
,
Hill
M
,
Zhao
Q
,
Liu
B
.
Molecular chaperone GRP94/GP96 in cancers: oncogenesis and therapeutic target
.
Front Oncol
.
2021
;
11
:
629846
.

44

Duan
XF
,
Xin
YW
.
Overexpression of molecule GRP94 favors tumor progression in lung adenocarcinoma by interaction with regulatory T cells
.
Thorac Cancer
.
2020
;
11
(
3
):
704
712
.

45

Romero-Ramirez
L
,
Cao
H
,
Nelson
D
, et al.
XBP1 is essential for survival under hypoxic conditions and is required for tumor growth
.
Cancer Res
.
2004
;
64
(
17
):
5943
5947
.

46

Oh-Hashi
K
,
Hirata
Y
,
Kiuchi
K
.
Transcriptional regulation of mouse mesencephalic astrocyte-derived neurotrophic factor in Neuro2a cells
.
Cell Mol Biol Lett
.
2013
;
18
(
3
):
398
415
.

47

Gao
B
,
Lee
S-M
,
Chen
A
, et al.
Synoviolin promotes IRE1 ubiquitination and degradation in synovial fibroblasts from mice with collagen-induced arthritis
.
EMBO Rep
.
2008
;
9
(
5
):
480
485
.

48

Eletto
D
,
Eletto
D
,
Dersh
D
,
Gidalevitz
T
,
Argon
Y
.
Protein disulfide isomerase A6 controls the decay of IRE1α signaling via disulfide-dependent association
.
Mol Cell
.
2014
;
53
(
4
):
562
576
.

49

Li
H
,
Korennykh
AV
,
Behrman
SL
,
Walter
P
.
Mammalian endoplasmic reticulum stress sensor IRE1 signals by dynamic clustering
.
Proc Natl Acad Sci
.
2010
;
107
(
37
):
16113
16118
.

50

Gu
F
,
Nguyên
DT
,
Stuible
M
,
Dubé
N
,
Tremblay
ML
,
Chevet
E
.
Protein-tyrosine phosphatase 1B potentiates IRE1 signaling during endoplasmic reticulum stress*
.
J Biol Chem
.
2004
;
279
(
48
):
49689
49693
.

51

Sowparani
S
,
Mahalakshmi
P
,
Sweety
JP
,
Francis
AP
,
Dhanalekshmi
UM
,
Selvasudha
N
.
Ubiquitous neural cell adhesion molecule (NCAM): potential mechanism and valorisation in cancer pathophysiology, drug targeting and molecular transductions
.
Mol Neurobiol
.
2022
;
59
(
9
):
5902
5924
.

52

Fernández-Briera
A
,
García-Parceiro
I
,
Cuevas
E
,
Gil-Martín
E
.
Effect of human colorectal carcinogenesis on the neural cell adhesion molecule expression and polysialylation
.
Oncology
.
2010
;
78
(
3-4
):
196
204
.

53

Holczer
M
,
Márton
M
,
Kurucz
A
,
Bánhegyi
G
,
Kapuy
O
.
A comprehensive systems biological study of autophagy-apoptosis crosstalk during endoplasmic reticulum stress
.
Biomed Res Int
.
2015
;
2015
:
319589
.

54

Kim
KH
,
Lee
M-S
.
Autophagy—a key player in cellular and body metabolism
.
Nat Rev Endocrinol
.
2014
;
10
(
6
):
322
337
.

55

Park
JS
,
Oh
SY
,
Lee
DH
, et al.
P62/SQSTM1 is required for the protection against endoplasmic reticulum stress-induced apoptotic cell death
.
Free Radic Res
.
2016
;
50
(
12
):
1408
1421
.

Abbreviations

     
  • 17-AAG

    17-N-allylamino-17-demethoxygeldanamycin

  •  
  • ACTH

    adrenocorticotropic hormone

  •  
  • CAs

    corticotroph pituitary adenomas

  •  
  • cDNA

    complementary DNA

  •  
  • Ct

    cycle threshold

  •  
  • DEPs

    differentially expressed proteins

  •  
  • DMSO

    dimethyl sulfoxide

  •  
  • EMT

    epithelial-mesenchymal transition

  •  
  • ER

    endoplasmic reticulum

  •  
  • FCA

    functioning corticotroph adenoma

  •  
  • FDR

    false discovery rate

  •  
  • Hsp90

    90-kDa heat shock protein

  •  
  • IHC

    immunohistochemistry

  •  
  • MS

    mass spectrometry

  •  
  • PCSK1

    proprotein convertase subtilisin/kexin type 1

  •  
  • PCSK1N

    proprotein convertase subtilisin/kexin type 1 inhibitor

  •  
  • POMC

    pro-opiomelanocortin

  •  
  • RT-qPCR

    reverse transcription–quantitative polymerase chain reaction

  •  
  • SCA

    silent corticotroph adenoma

  •  
  • TBX19

    T-box transcription factor 19

  •  
  • UPR

    unfolded protein response

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs licence (https://creativecommons.org/licenses/by-nc-nd/4.0/), which permits non-commercial reproduction and distribution of the work, in any medium, provided the original work is not altered or transformed in any way, and that the work is properly cited. For commercial re-use, please contact [email protected] for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact [email protected]. See the journal About page for additional terms.