Abstract Prolactinoma is the most prevalent pituitary neuroendocrine tumor and dopamine agonists (DAs) targeting dopamine D2 receptor (DRD2) are recommended as the first-line treatment. However, varying degrees of DA resistance limit patient benefit. Our study used transcriptome sequencing of surgical tumor samples and found abnormal cholesterol metabolism in prolactinoma, especially in DA-resistant tumors. We found that cholesterol significantly enhanced the resistance of prolactinoma MMQ cell lines to cabergoline in vitro and in vivo xenografts. Further, cholesterol did not affect the total protein level of DRD2, but changed the distribution of DRD2 with downregulation of its membrane abundance and upregulation of cytoplasmic localization. Mechanistically, immunoprecipitation combined with mass spectrometry revealed cholesterol increased binding affinity between DRD2 and stress granules (SGs) core proteins, such as G3BP1. Western blot experiment of G3BP1 and fluorescent probe were used to confirm the formation of SGs after cholesterol treatment in MMQ cells and tumor xenografts, as well as in surgical tumor samples. Interfering the formation of SGs by overexpressing of USP10 and using the small molecule ISRIB reversed cholesterol’s effect on DRD2 cellular distribution and DA resistance in MMQ cells. Finally, a non-specificity inhibitor of SGs, anisomycin identified by drug repositioning analysis, could attenuate cholesterol-induced cabergoline resistance in vitro. Taken together, our findings suggest that abnormal cholesterol metabolism reduces DRD2 membrane localization via stress granules formation, which may be an important reason for the DA resistance of prolactinoma patients. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-025-01986-1. Keywords: Prolactinoma, Drug resistance, Cholesterol, Stress granules Highlights Abnormal cholesterol metabolism has been identified in prolactinoma, especially in dopamine agonist-resistant tumors. Cholesterol promoted prolactinoma dopamine agonist resistance by downregulating the membrane localization of DRD2. Targeting of stress granules can attenuate cholesterol-regulated reduction of DRD2 membrane localization. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-025-01986-1. Introduction Pituitary neuroendocrine tumors (PitNETs) include nonfunctional and functional (hormone-secreting) types, with prolactinomas accounting for 50% of all pituitary adenomas in both males and females according to the latest consensus [[40]1]. Prolactinomas can cause hyperprolactinemia and hypogonadism, as well as a greater risk of metabolic syndrome and obesity [[41]1]. The dopamine agonist cabergoline is the primary medical therapy for patients with microprolactinomas and well-encased macroprolactinomas [[42]1]. Nevertheless, approximately 10% of prolactinoma patients are resistant to cabergoline at the recommended doses, and the resistance rate among patients with macroprolactinomas is 20% [[43]1–[44]4]. Primary or secondary resistance to dopamine agonists in prolactinoma patients may involve multiple molecular mechanisms that are not yet fully understood; furthermore, the expression of dopamine receptor D2 (DRD2) is a prerequisite for normal dopamine agonist function [[45]5]. Previous studies have reported that molecules such as filamin A (FLNA) and β-arrestin2 are necessary for signal transduction of DRD2 activated by dopamine agonists [[46]5]. FLNA is required for DRD2 cell surface expression and prevents its lysosomal degradation, and drug-resistant prolactinomas exhibit lower FLNA expression [[47]6]. β-arrestin2 supports DRD2 in reducing the phosphorylation of AKT, and its silencing weakens the ability of dopamine agonists to inhibit lactotroph proliferation [[48]7]. Currently, the posttranslational transport process of DRD2 remains unclear. Rab4 and Rab11 participate in the constitutive and ligand-dependent endosomal recycling of membrane DRD2, respectively [[49]8]. Furthermore, the regulatory network of DRD2 cellular localization in prolactinomas requires further research. Previous work by our team revealed that patients with PitNETs were more susceptible to hypercholesterolemia [[50]9]. A recent study showed that the overexpression of patched domain-containing 1 (PTCHD1), which is a cholesterol transporter protein, could directly induce the formation of stress granules in the absence of other stressors [[51]10]. SGs are membraneless aggregates that are formed under stress conditions and include a variety of biomolecules, such as G3BP1, USP10, and Caprin-1 [[52]11]. Caprin-1 promotes the formation of SGs, whereas USP10 inhibits their formation [[53]11]. A high-cholesterol diet can induce the accumulation of SGs in macrophages, the liver and vascular smooth muscle cells in mice [[54]12, [55]13]. Moreover, hydroxycholesterol can induce the formation of SGs in human neuroblastoma, and saturated fatty acids can increase the content of G3BP1 in [56]P18000 pellets (containing SGs) during cell lysis [[57]14–[58]16]. SGs are reportedly involved in the drug resistance of various tumors, and the inhibition of SGs can overcome resistance to chemotherapy agents (such as 5-fluorouracil) and targeted therapy (including sorafenib) [[59]17, [60]18]. However, the roles of cholesterol metabolism and SGs in the resistance of prolactinomas to dopamine agonists require further research. In this study, we performed transcriptome sequencing on peritumoral gland tissue and sensitive and resistant prolactinoma samples. The data revealed the presence of abnormal cholesterol metabolism in prolactinomas and its correlation with dopamine agonist resistance. Further cell and animal experiments revealed that cholesterol was able to reduce the membrane localization of DRD2 in MMQ cells, thus underscoring the importance of continued management of the hyperlipidemia that commonly accompanies prolactinomas [[61]19]. The interference of the formation of SGs can attenuate the effect of cholesterol on the membrane localization of DRD2. We recommend special consideration of the roles of abnormal cholesterol metabolism and SGs in dopamine agonist-resistant prolactinomas in the future. Materials and methods Cell culture and lentiviral transfection Rat prolactinoma cell lines MMQ (CRL-10609) and GH3 (CCL-82.1) were purchased from the American Type Culture Collection. Cells were cultured in F-12 K medium (PYG0036, Boster Biological Technology) with 15% donor horse serum (04-124-1 A, Biological Industries), 2.5% certified fetal bovine serum (04-001-1 A, Biological Industries) and 1% penicillin-streptomycin solution (03-031-1B, Biological Industries). Cells were cultured in an incubator containing 5% CO2 and 95% air at 37 °C. USP10 and control overexpression lentiviral (Genechem, Shanghai, China) were transfected into MMQ cells according to the manufacturer’s instructions. Specifically, we selected 10^^5 well-grown MMQ cells in a 6-well plate, added lentivirus and pro-transfection agent polybrene (Genechem, Shanghai, China) with a Multiplicity of Infection (MOI) of 100, and replaced fresh media containing puromycin after 72 h. Stable clones were obtained by using puromycin (2 µg/ml) to for 72 h. Transfection efficiency was determined by western blot experiments. Patients and surgical samples Patients presented to the Multidisciplinary Center for Pituitary Adenomas in Xinqiao Hospital from January 2020 to December 2023 were considered. Responsiveness to dopamine agonists was determined by two experienced clinicians according to previously reported criteria [[62]20]. Resistance of prolactinomas was defined, according to the latest consensus, as the failure to normalize prolactin serum levels or achieve a significant reduction in tumor size (a decrease of ≥ 30% in the maximum diameter) when treated with standard doses of dopamine agonists (7.5–10 mg/day of bromocriptine or 2.0 mg/week of cabergoline) for a minimum duration of 6 months [[63]1]. The types of dopamine agonists utilized, doses and duration of treatment for 29 patients were provided in supplementary Table [64]e1. And the reasons for surgery in sensitive patients were provided in supplementary Table [65]2. To explore the correlation between serum lipid and drug responsiveness, patients formerly consumed lipid-lowering medications were excluded, and finally included 29 cases. The patient’s blood cholesterol and triglyceride level were reported by preoperative biochemical detection. The samples used for sequencing were obtained through transsphenoidal surgery, and 5 peritumoral gland tissue, 5sensitive and 4 resistant prolactinoma tissue were selected. RNA sequencing The operation of RNA sequencing was carried out as we described before [[66]21]. In brief, total RNA of human prolactinoma tissue and peritumoral gland tissue was extracted by TRIzol (Invitrogen) according to manufacturer’s instructions. The mRNA after quality control was enriched by magnetic beads and then fragmented, and cDNA was obtained by reverse transcription for end repair. After amplification and quantification, the library was sequenced with Illumina Hiseq xten (2 × 150 bp read length). SeqPrep ([67]https://github.com/jstjohn/SeqPrep) and Sickle ([68]https://github.com/najoshi/sickle) were used for quality control of raw reads data. Then using HiSat2([69]http://ccb.jhu.edu/software/hisat2/index.shtml) for reads mapping [[70]22]. WGCNA analysis and identification of differentially expressed genes The “WGCNA’” R package was used to found key genes related to prolactinoma [[71]23]. After construction of sample clustering and co-expression network, the soft-threshold was set to further detect the module. Finally, the modules significantly related to traits were selected for analysis. Differentially expressed genes (DEGs) were analyzed by “edgeR” R package and defined as|log(Fold Change)| > 1.5 and p-value < 0.05 [[72]24]. Then Toppgene website ([73]https://toppgene.cchmc.org/) was used for the gene ontology and pathway enrichment analyses of DGEs. Z-score of GO term and pathway term was automatically calculated by “GOplot” R package [[74]25]. The Z-score referred to here is not the conventional statistical measure but rather a metric calculated based on the fold change and the number of differentially expressed genes as described on ‘[75]https://wencke.github.io/’. This metric serves to indicate the likelihood of a biological process, molecular function, or cellular component being upregulated (positive value) or downregulated (negative value) [[76]25]. Sequencing data of pituitary gland from female wild mice and pituitary hyperplasia (anterior pituitary were already enlarged) from female prolactin receptor deficient mice are publicly available under accession number [77]GSE100334 [[78]26]. Cell viability assay MMQ cells were seeded in 96-well plates at a density of 5000 cells per well and cultured with palmitic acid (HY-N0830, MedChemExpress), cabergoline (HY-15296, MedChemExpress) and cholesterol/Mβ-CD (C4951, Sigma Aldrich) for different times. The solution of palmitic acid was developed as described previously [[79]27]. DMSO (HY-Y0320, MedChemExpress) and Mβ-CD (C4555, Sigma Aldrich) were used as vehicle for cabergoline and cholesterol, respectively. After adding CCK-8 solution (HY-K0301, MedChemExpress), the optical density at 450 nm (OD450) was measured after 1 h of culture in an incubator containing 5% CO2. Lipid depleted fetal bovine serum (C3840, Vivacell) was utilized instead of standard fetal bovine serum and donor horse serum to formulate the medium, thereby minimizing the impact of endogenous lipids. The kill effect of cabergoline was assessed by inhibition rate defined as (vehicle[OD450]-cabergoline[OD450])/vehicle[OD450] × 100%. Apoptosis analysis An annexin V-FITC apoptosis detection kit (C1062S, Beyotime) was used to measure the apoptosis level of MMQ cells. In brief, MMQ cells were collected and mixed with 195ul binding buffer, 5ul Annexin-V-FITC and 10 µl propidium iodide. After incubated at room temperature for 20 min, the MMQ cells were immediately analyzed using flow cytometry (Guava easyCyte, Cytek). Xenograft experiments Three weeks old female BALB/cA-nu mice were purchased from Beijing Vital River Laboratory Animal Technology Co., Ltd. and fed of high-cholesterol diet (XT109C, Xietong Pharmaceutical Bio-engineering) or control diet (XT102C, Xietong Pharmaceutical Bio-engineering) for two weeks in SPF conditions. Then nude mice were subcutaneously inoculated with 2 × 10^6 MMQ cells and divided into four groups: high-fat diet with cabergoline administration, high-fat diet with vehicle administration, control diet with cabergoline administration, and control diet with vehicle administration. Cabergoline was administered at a dose of 0.5 mg/kg, once every two days, starting 8 days after cell inoculation. Tumor volumes were calculated using the following formula: V (mm3) = [ab^2]/2 (a was the tumor maximum length and b was the minimum length). The tumors were resected and the mice heart blood was preserved for the measurement of triglycerides and cholesterol three weeks later. All animal experimental procedures were conducted under review and approval by the Internal Animal Care and Use Committee. Reverse transcription and qPCR experiment Total RNA was extracted using RNA extraction kit (AG21022, Accurate Biotechnology). PrimeScript RT Reagent Kit (PR047A, TaKaRa) was used to obtain cDNA from 1 µg RNA. Then qPCR experiment was performed by using TB Green Premix Ex Taq II (RR820A, TaKaRa) and CFX96 Real-time System (Bio-Rad). The primers were described as follows, forward (Drd2): AGTTTCCCAGTGAACAGGCG, reverse (Drd2): CGTGCCCATTCTTCTCTGGT, forward (Actb): GAGGGAAATCGTGCGTGAC, reverse (Actb): GCATCGGAACCGCTCATT. Western blot Total protein was obtained by using RIPA lysis buffer (WB001, Invent Biotechnologies) with protease inhibitor cocktail (GRF101, Epizyme Biotech). Plasma membrane and cytoplasmic proteins were isolated by using Minute™ plasma membrane protein isolation and cell fractionation kit (SM-005, Invent Biotechnologies). Protein was separated by TGX Stain-Free™ FastCast™ acrylamide kits (1610183, Bio-Rad) and transferred to PVDF membranes (Millipore, ISEQ00010). After blocking with 5% skim milk mixed in TBST solution, the membranes were incubated with primary antibodies listed as follows, DRD2 (55084-1-AP, Proteintech, 1:1000), G3BP1 (66486-1-Ig, Proteintech, 1:1000), USP10 (19374-1-AP, Proteintech, 1:1000), Beta-Actin (81115-1-RR, Proteintech, 1:4000). Then membranes were incubated with HRP-conjugated secondary antibodies (SA00001-2/SA00001-1, Proteintech, 1:2000) and detected with ECL substrate (BG0001, Bioground Biotech). Immunofluorescence Before immunostaining, GH3 cells were cultured in 24-well plate coated with poly-L-lysine (ST509, Beyotime) and MMQ cells were cultured in 24-well plate with Shi-fix™ coverslips (SB-Shifix-25, SHIKHAR BIOTECH). The treated cells were fixed with 4% paraformaldehyde and permeabilized with Saponin (P0095, Beyotime). Then the cells underwent sequential steps of blocking, primary antibody incubation, Alexa Fluor™ 594-conjugated secondary antibody (A-11012, Invitrogen) incubation, and finally mounting with an antifade mounting medium containing DAPI (P0131, Beyotime). Immunohistochemistry Tumors implanted subcutaneously in nude mice were fixed in 4% paraformaldehyde and embedded with paraffin, and then sliced into 4 μm sections. The subsequent immunohistochemical staining was performed following the previously reported procedures [[80]9]. The intensity of immunostaining was measured by the IHC toolbox plug-in provided by Fiji [[81]28]. Stress granules detection Protein isolation of stress granules was consistent with previously reported procedures [[82]15]. The lysis buffer was composed of 100mmol/L potassium acetate, 50mmol/L Tris-HCl with pH 7.4, 0.5mmol/L dithiothreitol, 2mmol/L magnesium acetate, 0.5% NP-40 solution, 50 µg/ml heparin and protease inhibitor cocktail. Collected cells were lysed in lysis buffer, followed by centrifugation at 1000 g for 5 min, and the supernatant was collected as whole cell lysate (Wcl). After determining the concentration by using BCA protein assay kit (P0010, Beyotime), equal amounts of protein were further centrifuged at 18,000 g for 20 min and the pellet was rich in stress granules ([83]P18000). The previously reported small molecule fluorescent probe (TASG) was used for fluorescence detection of stress granules [[84]29]. The TASG was quantified by Multi-point tool provided by Fiji to count the granules in each cell [[85]28]. Immunoprecipitation (IP) According to the manufacturer ‘s instructions, Magnetic Protein A/G IP/Co-IP Kit (YJ201, Epizyme Biotech) was used to explore the substrates binding with DRD2. In brief, MMQ cells were collected and washed with PBS, then incubated with lysis buffer provided in IP kit. The protein lysis after centrifugation was incubated with primary antibody against DRD2 overnight, then mixed with magnetic beads for 4 h at 4℃. Then the proteins were eluted from the magnetic beads for mass spectrometry sequencing. Protein–protein interaction (PPI) network construction STRING database ([86]https://cn.string-db.org/) was used for the protein-protein interaction network construction and using Cytoscape (v3.9) for visualization. The importance of proteins in the network was ranked by the algorithm provided by cytoHubba plug-in of Cytoscape [[87]30]. GTEx database analysis The GEPIA2 web server ([88]http://gepia2.cancer-pku.cn/) was used to analysis the molecules expression correlation in normal pituitary tissue in GTEx database [[89]31]. Drug repositioning analysis The repositioning analysis of potential drugs for DA resistance prolactinoma was performed by R package “cogena” basing on the CMap database [[90]32]. Statistical analysis Data in this study was presented as the mean ± standard deviation (SD). The chi-squared test (one side) was used to determine the correlation between hypercholesterolemia and DA resistance. Significant differences between the two groups were ascertained by employing an unpaired t-test (two tailed) or, in case of unequal variances, a Mann-Whitney nonparametric test. Statistically significant was defined as a P-value < 0.05 (displayed as * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001). Results Transcriptome analysis revealed abnormal lipid metabolism in prolactinoma We conducted an analysis of the transcriptomic sequencing data of 14 samples, including 5 peritumoral gland tissues and 9 prolactinoma tissues. A total of 1277 upregulated and 643 downregulated DEGs were identified and visualized via a volcano plot (Fig. [91]1A). A weighted gene coexpression network was constructed (Supplementary Fig. 1A), and a heatmap of module-trait relationships (Fig. [92]1B) in peritumoral gland tissue and prolactinoma was constructed based on the Pearson correlation coefficient. The significantly related gene modules, including the blue module (r = 0.7, p = 0.005), magenta module (r = 0.64, p = 0.01) and turquoise module (r = 0.76, p = 0.002), were selected together with the DEGs to construct a Venn diagram. Finally, 862 overlapping genes were defined as core DEGs between prolactinoma and peritumoral gland tissue (Fig. [93]1C). We further performed functional annotation analysis on the core DEGs (Fig. [94]1D). Multiple extracellular matrix-related terms, such as extracellular matrix structural constituent (term ID: GO0005201, Z-score =-2.710687) were repressed. Pathway enrichment analysis of the core DEGs revealed several lipid metabolism pathways (Fig. [95]1E). We subsequently analyzed public pituitary hyperplasia (the anterior pituitary already being enlarged) sequencing data from wild-type (WT) and prolactin receptor (Prlr) knockout (KO) female mice at different ages (with this knockout potentially leading to prolactinoma), and found that the DEGs observed between Prlr-KO and WT mice changed over time in relation to lipid metabolism (Fig. [96]1F) [[97]26]. These results indicate that abnormal lipid metabolism is involved in the tumorigenesis of prolactinoma. Fig. 1. [98]Fig. 1 [99]Open in a new tab Transcriptome analysis revealed abnormal lipid metabolism in prolactinoma. (A) Volcano plot of DEGs between peritumor gland tissue and prolactinoma samples. (B) Heatmap of correlation between gene modules and clinical traits in peritumor gland tissue and prolactinoma samples. (C) Venn analysis of gene modules and DEGs to identify core DEGs between prolactinoma and peritumor gland tissue. (D) GO function analysis of core DEGs between prolactinoma and peritumor gland tissue. (E) Pathway enrichment of core DEGs between prolactinoma and peritumor gland tissue. (F) DEGs of public pituitary hyperplasia (anterior pituitary were already enlarged) sequencing data between prolactin receptor knockout and wild-type mice with different months, and their pathway enrichment. (G) Heatmap of DEGs between DA resistant and sensitive prolactinoma samples. (H) Pathway enrichment analysis of resistance related DEGs. (I) The intersected genes between resistance related DEGs and genes in the term metabolism of lipids in Figure F. (J) Clinical features of 14 DA sensitive and 15 DA resistant prolactinoma patients. (K-L) Blood triglyceride (K) and total cholesterol (L) levels of sensitive and resistant prolactinoma patients. * (P) < 0.05 To further investigate the molecular mechanism of DA resistance, we identified 341 upregulated and 383 downregulated DEGs between 5 DA-sensitive prolactinomas and 4 DA-resistant prolactinomas (Fig. [100]1G). Functional annotation analysis of these resistance-related DEGs revealed lipid metabolism pathways (Fig. [101]1H). We extracted the genes related to lipid metabolism, as shown in Figure F, and intersected these genes with the drug resistance-related DEGs. Five overlapping genes (UGT8, ACACB, SLC44A3, PLIN2 and PON1), which are predominantly involved in cholesterol and fatty acid metabolism, were obtained (Fig. [102]1I) [[103]33–[104]35]. Therefore, we further reviewed the clinical features of 14 DA-sensitive prolactinoma patients and 15 DA-resistant prolactinoma patients. We found that DA-resistant prolactinoma patients were more likely to exhibit elevated total cholesterol levels (p = 0.0384) (Fig. [105]1L), whereas triglyceride levels were not significantly different between the two groups (Fig. [106]1K). There was no significant difference in BMI between the two groups (Fig. [107]1J). Cholesterol promoted MMQ cell resistance to Cabergoline treatment We subsequently investigated whether cholesterol and fatty acids influence the effect of cabergoline on MMQ cells in vitro. We selected palmitic acid for analysis, which is the most abundant saturated fatty acid in human serum, and the CCK-8 results revealed that exogenous addition of palmitic acid did not alleviate the degree of cabergoline-induced inhibition of MMQ cells (Supplementary Fig. 1B). However, the presence of 20 µg/ml Mβ-CD balanced cholesterol (CHO/Mβ-CD) significantly mitigated the inhibitory effect of cabergoline on MMQ cells without impacting cell viability (Fig. [108]2A). The results of flow cytometry revealed that 20 µg/ml CHO/Mβ-CD reduced the apoptosis of MMQ cells induced by cabergoline treatment for 48 h (Fig. [109]2B-C). Fig. 2. [110]Fig. 2 [111]Open in a new tab Cholesterol promoted the resistance of MMQ cells to DAs. (A) Cell viability assays of MMQ cells after treatment with CHO/Mβ-CD and cabergoline alone or in combination. (B)-(C) Flow cytometry analysis of MMQ cell apoptosis after treatment with CHO/Mβ-CD and cabergoline alone or in combination. (D)-(E) Detection of cholesterol and triglyceride levels in the peripheral blood of nude mice after they were fed a high-fat diet or control diet for six weeks. (F) Weights of the nude mice prior to tumor resection. (G) Images of tumor xenografts in nude mice after being exposed to different diets and drugs (H-(I). Weights and volumes of the tumor xenografts. * P < 0.05, *** P < 0.001, **** P < 0.0001 In vivo experiments revealed that a high-cholesterol diet for six weeks resulted in elevated levels of cholesterol and triglycerides in nude mice (Fig. [112]2D-E), and the weights of the nude mice did not significantly change during the course of the experiment (Fig. [113]2F). In the absence of cabergoline, a high-fat diet did not affect the weights or volumes of tumor xenografts compared to a control diet. However, when cabergoline was administered, the high-fat diet attenuated the ability of cabergoline to reduce the weights and volumes of tumor xenografts (Fig. [114]2G-I). These results suggest that abnormal cholesterol metabolism may be involved in the process of DA resistance in prolactinoma. DRD2 membrane localization was decreased by cholesterol To explore the potential mechanism of cholesterol-induced drug resistance in prolactinoma, we performed transcriptomic sequencing on CHO/Mβ-CD- or Mβ-CD-treated MMQ cells. The results revealed that CHO/Mβ-CD treatment decreased the expression of genes involved in cholesterol synthesis, such as Hmgcr, Sqle and Hmgcs1 (Fig. [115]3A). Moreover, the expression of cholesterol efflux-related genes and sterol-responsive transcription factors, such as Abca1 and Srebf1, was increased (Fig. [116]3A). These results indicated that the MMQ cells did response to cholesterol stimulation. Functional analysis revealed that cholesterol synthesis and the GPCR signaling pathway were downregulated (Fig. [117]3B), whereas the nuclear receptor signaling pathway was activated (Fig. [118]3C). Given that the target of cabergoline (the DRD2 receptor) belongs to the GPCR family, we further focused on potential alterations in DRD2 caused by cholesterol. The sequencing results revealed that the transcription of long subtype of Drd2 was significantly increased after CHO/Mβ-CD treatment, which was also confirmed via qPCR experiments (Supplementary Fig. 1C). However, the total cellular protein expression level of DRD2 was not significantly altered (Fig. [119]3D). DRD2 receptor availability at the cell surface has been proposed as being a molecular marker of prolactinoma pharmacological responsiveness in the absence of genetic defects [[120]6]. Therefore, we further explored whether the spatial distribution of DRD2 was changed due to CHO/Mβ-CD treatment. The results demonstrated that CHO/Mβ-CD treatment reduced the protein abundance of DRD2 on the cell membrane and increased its content in the cytoplasm (Fig. [121]3E-F). The immunofluorescence results also revealed decreased membrane localization and increased cytoplasmic localization of DRD2 in CHO/Mβ-CD-treated MMQ and GH3 cells (Fig. [122]3G-H). Finally, we isolated the cell membrane proteins from the xenografts and found that the membrane localization of DRD2 was lower in the high-fat diet group compared to the control diet group (Fig. [123]3I). Fig. 3. [124]Fig. 3 [125]Open in a new tab Cholesterol treatment reduced the membrane localization of DRD2. (A) Volcano plot of DEGs sequenced from MMQ cells after CHO/Mβ-CD or Mβ-CD treatment. (B-C) Pathway enrichment results of downregulated and upregulated DEGs. (D) Total DRD2 protein levels in MMQ cells after CHO/Mβ-CD or Mβ-CD treatment. (E-F) Membrane and cytoplasmic protein levels of DRD2 in MMQ cells after CHO/Mβ-CD or Mβ-CD treatment. (G-H) Immunofluorescence results of DRD2 in MMQ and GH3 cells after CHO/Mβ-CD or Mβ-CD treatment. (I) The expression level of DRD2 in the cell membranes of xenografts. Scale bar: 5 μm. Western Blot experiments had three biological replicates and two technical replicates Cholesterol promoted the formation of stress granules We performed immunoprecipitation and mass spectrometry sequencing of endogenous DRD2 in MMQ cells to investigate the potential mechanism of decreased DRD2 abundance on the cell membrane. The binding affinity of several proteins (such as UBAP2L, G3BP1 and RAB7A) for DRD2 was altered following CHO/Mβ-CD treatment (Fig. [126]4A). Functional annotation analysis revealed that proteins with decreased binding affinity were mainly involved in protein localization to the membrane (Fig. [127]4B), whereas proteins with increased binding affinity were mainly related to stress granule assembly (Fig. [128]4C). Due to the fact that stress granules are dynamic assemblies of RNA and proteins that form in response to various cellular stresses, we focused on proteins with increased binding affinity after cholesterol treatment. The key marker of stress granule formation (G3BP1) was identified as the predominant protein among the increased-binding proteins via the construction of a protein interaction network (Fig. [129]4D). We observed a significant negative correlation between G3BP1 and DRD2 expression in the pituitary gland according to the GTEx database (Fig. [130]4E). Fig. 4. [131]Fig. 4 [132]Open in a new tab Cholesterol promoted the formation of stress granules. (A) Plot of proteins identified via immunoprecipitation combined with mass spectrometry sequencing. (B-C) Functional annotation analysis of proteins with decreased or increased binding affinity to DRD2 after CHO/Mβ-CD treatment in MMQ cells. (D) Protein interaction network of proteins with increased binding affinity to DRD2 and the predominant ranks. (E) Correlation plot of G3BP1 and DRD2 expression in the pituitary gland according to the GTEx database. (F) Protein level of G3BP1 in whole-cell lysates (wcl) and [133]P18000 pellets from MMQ cells. (G) Fluorescence staining of stress granules using TASG in MMQ cells. (H) Immunohistochemical staining of G3BP1 in tumor xenografts. (I) Western blot analysis of G3BP1 in tumor xenografts. (J) Immunohistochemical staining of G3BP1 in sensitive and resistant human prolactinomas. Scale bar: 5 μm We further examined the formation of stress granules after CHO/Mβ-CD treatment. The protein abundance of G3BP1 significantly increased in the [134]P18000 protein, which represents the abundant proteins of stress granules (Fig. [135]4F) [[136]15]. In addition, the staining of stress granules with the fluorescent probe TASG revealed significant accumulation of stress granules after CHO/Mβ-CD treatment (p = 0.0193) (Fig. [137]4G). The immunohistochemistry results revealed that the expression of G3BP1 was greater in the xenografts of the high-fat diet compared to the control diet group (Fig. [138]4H). Western blot analysis revealed similar results (Fig. [139]4I). Finally, the immunohistochemistry results revealed increased expression of G3BP1 in resistant human prolactinomas compared with sensitive prolactinomas (Fig. [140]4J). Targeting of stress granules could attenuate the cholesterol-mediated reduction in DRD2 membrane localization Subsequently, we explored whether stress granules mediate the regulatory effect of cholesterol on DRD2 membrane localization. We overexpressed USP10 in MMQ cells, and the Western blot results revealed that the total protein level of DRD2 was similar between USP10^Mock and USP10^OE MMQ cells (Fig. [141]5B). Compared to the USP10^Mock group, the assembly of stress granules in the USP10^OE group treated with CHO/Mβ-CD was decreased, as shown via fluorescence staining of TASG (Fig. [142]5C). The membrane abundance of DRD2 was decreased after CHO/Mβ-CD treatment in USP10^Mock MMQ cells but was not significantly altered in USP10^OE MMQ cells (Fig. [143]5D-E). Immunofluorescence staining revealed decreased membrane localization of DRD2 in CHO/Mβ-CD-treated USP10^Mock MMQ cells and constant membrane localization in USP10^OE MMQ cells (Fig. [144]5F-G). The results of the cell viability assay revealed that the inhibition rate of cabergoline was attenuated upon the addition of CHO/Mβ-CD to USP10^Mock cells, whereas cholesterol had a diminished effect on the inhibition rate of cabergoline in USP10^OE MMQ cells (Fig. [145]5H-I). Furthermore, we used ISRIB (Integrated Stress Response Inhibitor) to suppress the formation of stress granules, as previously reported [[146]36, [147]37]. We found that 1 µM ISRIB effectively inhibited the induction of stress granules by cholesterol (Fig. [148]5J). Western blot experiments revealed that the membrane abundance of DRD2 was decreased upon the addition of ISRIB after CHO/Mβ-CD treatment (Fig. [149]5K). Cell proliferation assays revealed that ISRIB treatment suppressed the ability of cholesterol to reduce cabergoline-induced MMQ-induced cell death (Fig. [150]5L). Fig. 5. [151]Fig. 5 [152]Open in a new tab Stress granules mediated the cholesterol-regulated reduction in DRD2 localization on the cell membrane. (A) USP10 can inhibit the assembly of stress granules, according to a previous study. (B) Overexpression of USP10 in MMQ cells and its effect on DRD2 protein levels. (C) Fluorescence staining of stress granules with TASG in USP10^Mock and USP10^OE MMQ cells. (D-E) Western blot analysis of the cell membrane proteins of USP10^Mock and USP10^OE MMQ cells. (F-G) Fluorescence staining of DRD2 in USP10^Mock and USP10^OE MMQ cells. (H-I) CCK-8 results of USP10^Mock and USP10^OE MMQ cells treated with CHO/Mβ-CD and cabergoline alone or in combination. (J) Fluorescence staining of stress granules with TASG in MMQ cells. (K) Western blot analysis of the levels of cell membrane proteins in MMQ cells. (L) CCK-8 results for MMQ-treated cells. *** P < 0.001, **** P < 0.0001. Scale bar: 5 μm Finally, we conducted a drug repositioning analysis to identify pharmaceuticals that are capable of targeting stress granules to overcome cabergoline resistance. The DEGs observed between sensitive and resistant prolactinoma samples were divided into 3 coexpressed gene sets by using the “NbClust” R package (Supplementary Fig. 1D-E). We mapped the gene cluster onto the Cmap database and identified the natural compound anisomycin, which has been reported to possess the ability to inhibit stress granule assembly (Supplementary Fig. 1F) [[153]38, [154]39]. Fluorescence staining with TASG revealed that 2 µM anisomycin was able to inhibit the stress granule assembly after treatment with CHO/Mβ-CD (Supplementary Fig. 1G). A cell viability assay revealed that CHO/Mβ-CD had a weaker effect on the inhibition rate of cabergoline in MMQ cells after the addition of 2 µM anisomycin (Supplementary Fig. 1H-I). Discussion Cabergoline, which is a preferred dopamine agonist, has high efficacy in decreasing serum prolactin levels, ameliorating clinical manifestations and reducing prolactinoma size [[155]1]. However, approximately 10% of prolactinoma patients develop resistance, and the underlying molecular mechanism of this resistance is not currently known [[156]1–[157]4]. This study identified dysregulated cholesterol metabolism in prolactinoma, especially in dopamine agonist-resistant tumors, by transcriptomic sequencing analysis of peritumoral gland tissue and sensitive and resistant prolactinoma samples. Further in vitro and in vivo experiments revealed that cholesterol promoted MMQ cell resistance to cabergoline treatment by reducing the membrane localization of DRD2. The interference of the formation of stress granules was able to attenuate the effect of cholesterol on the membrane localization of DRD2. Abnormal lipid metabolism has been implicated in oncogenesis, as well as in conferring resistance to therapeutic drugs. Clinical data revealed that patients with resistant prolactinomas were more likely to exhibit elevated blood cholesterol. In particular, 5 lipid metabolism-related genes (UGT8, ACACB, SLC44A3, PLIN2 and PON1) were identified as DEGs between resistant and sensitive prolactinomas. UGT8-mediated sulfatide synthesis can regulate BAX localization and influence the sensitivity of colorectal cancer cells to apoptosis [[158]40]. ACACB-encoded acetyl-CoA carboxylase elicits a shift from glycolysis-dependent to lipogenesis-dependent activities, thereby endowing head and neck squamous cell carcinoma with resistance to cetuximab [[159]41]. The lipid droplet-coating protein PLIN2 mediates the accumulation of lipid droplets, which act as functional acentriolar microtubule organizing centers, thereby interfering with mitotic spindle polarity and endowing tumors with resistance to paclitaxel [[160]42]. The stability of PON1 has been shown to support the expansion of resistant clones in a tumor regression model [[161]43]. In vivo and in vitro experiments revealed that cholesterol promoted MMQ resistance in cells without affecting their proliferation ability. However, palmitic acid, which is the most abundant saturated fatty acid in the human serum, did not influence the resistance of MMQ cells, thus indicating specific underlying resistance effects among lipid metabolites in MMQ cells. The first and most obvious explanation for dopamine agonist resistance in prolactinomas is the loss of receptor expression on the tumor cell surface, and many studies have attempted to validate this hypothesis [[162]6]. The results of sequencing and qPCR experiments revealed increased transcription of total Drd2 after cholesterol treatment, whereas the total protein level of DRD2 remained unchanged, as confirmed via Western blotting. Due to the fact that the GPCR signaling pathway was downregulated according to the functional analysis results, we further focused on the protein function of DRD2. Interestingly, we found that cholesterol was able to reduce the membrane localization of DRD2, which may explain the acquired resistance of MMQ cells. This finding expands the comprehension of the role of cholesterol in prolactinoma resistance and indicates that cholesterol metabolism may influence the surface abundance of other drug receptors, such as EGFR and PD-L1 [[163]44, [164]45]. Currently, the regulatory mechanism of DRD2 cellular localization in prolactinomas is poorly understood. Previous study reported that leucine-rich repeat kinase-2 (LRRK2) and tetraspanin-7 (TSPAN7) regulated the transport and membrane localization of DRD2 [[165]46, [166]47]. G protein-coupled receptor kinase-2 (GRK2), β-arrestins and FLNA reportedly control the internalization and recycling of membrane DRD2 [[167]6, [168]48]. We performed immunoprecipitation to explore the potential mechanism of cholesterol-induced changes in DRD2 membrane localization, and functional annotation analysis showed that these proteins with increased binding affinity to DRD2 were mainly related to stress granules. We subsequently overexpressed USP10 in MMQ cells (which can inhibit SG formation) and found that the membrane abundance of DRD2 remained unchanged in USP10^OE MMQ cells after cholesterol treatment [[169]11]. Similar results were obtained via the use of ISRIB, which inhibits SG formation. In addition, anisomycin, a non-specificity inhibitor of SGs, was identified through drug reposition analysis, and showed attenuate effect on MMQ resistance induced by cholesterol in vitro. These findings indicate a potential association between SGs and DRD2 membrane localization, and USP10 may serve as a tool for modulating DRD2 membrane localization and enhancing prolactinoma sensitivity to cabergoline. This is not only significant for treating prolactinoma drug resistance, but also potentially provides insights for diseases such as Parkinson’s disease dopamine resistance. Some limitations remain to be addressed from this study. First, the manner in which SGs regulate DRD2 membrane localization is still unclear, and it is likely that this relationship is not straightforward. Another concern is whether the formation of SGs induced by other stressors can lead to resistance to cabergoline in prolactinomas. In summary, our results indicate that abnormal cholesterol metabolism plays an important role in the resistance of prolactinoma to DAs. Cholesterol was able to reduce the membrane localization of DRD2 in MMQ cells, and interference with the formation of SGs can attenuate the effect of cholesterol on the membrane localization of DRD2. Future research and clinical practice may consider the role of stress granules in resistant prolactinomas. Conclusions Our findings revealed dysregulated cholesterol metabolism in prolactinoma, especially in dopamine agonist-resistant tumors. Cholesterol can reduce the membrane localization of DRD2, which may be an important reason for the resistance of prolactinomas to DAs. Electronic supplementary material Below is the link to the electronic supplementary material. [170]Supplementary Material 1^ (4.4MB, tif) [171]Supplementary Material 2^ (2.9MB, tif) [172]Supplementary Material 3^ (370.9KB, docx) [173]Supplementary Material 4^ (84MB, zip) Abbreviations DRD2 Dopamine receptor D2 DA Dopamine agonist G3BP1 G3BP stress granule assembly factor 1 PitNETs Pituitary neuroendocrine tumors GRK2 G protein-coupled receptor kinase 2 DEGs Differentially expressed genes Wcl Whole cell lysate IP Immunoprecipitation PPI Protein–protein interaction Mβ-CD Mβ-CD balanced cholesterol USP10 Ubiquitin specific peptidase 10 PD-L1 Programmed-death ligand 1 Author contributions Hui Yang and Song Li designed this work. Yuyang Peng and Chengcheng Wang carried out experiments. Yuyang Peng and Huachun Yin carried out data analysis. Xiangdong Pei and Yuan Zhang conducted clinical data collection. Yuyang Peng written the manuscript. Hong Liang provided suggestions. Xin Zheng, Song Li and Hui Yang revised the manuscript. Funding This work was supported by Natural Science Foundation of Chongqing (CSTB2024NSCQ-MSX0021, CSTB2022NSCQ-MSX1534) and Young Doctor Incubation Program of Xinqiao Hospital (2022YQB046, 2023YQB0384, 2023YQB055). Data availability The data covered in this article are available on reasonable request from the corresponding author. Declarations Ethical approval This study was conducted in according to the principles of the Helsinki declaration and approval of the Ethics Committee of Xinqiao Hospital, Army Medical University (authorization ID: 2022-356-01). All animal experimental procedures were conducted under review and approval by the Internal Animal Care and Use Committee of the Army Medical University (authorization ID: AMUWEC20224532). Competing interests The authors declare no competing interests. Footnotes Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. Yuyang Peng, Huachun Yin, Xiangdong Pei and Yuan Zhang contributed equally to this work. Contributor Information Hui Yang, Email: huiyangtg2018@aliyun.com. Song Li, Email: tmmuls@tmmu.edu.cn. References