Skip to main content

DNMT3B promotes the progression of pheochromocytoma by mediating the hypermethylation of LRP1B promoter

Abstract

Background

Pheochromocytoma (Pheo) represents a potential metastatic neuroendocrine tumor. As a tumor suppressor gene, LRP1B is involved in the regulation of tumor progression. However, the precise regulatory mechanism of LRP1B in Pheo remains elusive.

Methods

RT-QPCR, western blot and immunohistochemistry (IHC) were used to identify the expression levels of DNMT3B and LRP1B. Biochemistry assays including luciferase and ChIP were utilized to detect the interaction between the methyltransferase DNMT3B and LRP1B promoter. LRP1B or DNMT3B were knock-down in Pheo cell line by shRNAs. Functional experiments including clonal formation, migration, and in vivo transplantation were performed to evaluate the regulation of LRP1B or DNMT3B on tumor growth.

Results

LRP1B was down-regulated, while DNMT3B was up-regulated in Pheo.Overexpression of LRP1B or inhibition of DNMT3B inhibited the progress of Pheo. DNMT3B was responsible for the hypermethylation of LRP1B promoter in Pheo. At the same time, overexpression of DNMT3B reversed the inhibitory effect of overexpression of LRP1B on Pheo progression.

Conclusion

DNMT3B mediated the hypermethylation of the tumor suppressive gene LRP1B and promotes Pheo progression.

Highlights

  • LRP1B is down-regulated in pheochromocytoma.

  • The promoter of LRP1B is hypermethylated in pheochromocytoma cells.

  • LRP1B inhibits pheochromocytoma progression and metastasis.

  • DNMT3B mediates the DNA methylation of LRP1B promoter in pheochromocytoma.

Introduction

Pheochromocytoma (Pheo) is a rare neuroendocrine tumor mainly developing in an adrenal gland [1]. While most subtypes of Pheo are benign, it has been shown that 2-26% of Pheo subtypes were highly metastasis [2]. The occurrence and development of Pheo are often related to gene mutation, including gene point mutation, gene fusion and, copy number change [2]. Pheo tumor cells secrete catecholamines, causing headache, hypertension, palpitation, and even shock and stroke [3]. At present, surgery and radiopharmaceuticals are the main treatments for Pheo [4]. Nevertheless, all of the above treatments show limitations in the prevention of Pheo metastasis. Therefore, developing a precision therapy for Pheo by understanding the molecular mechanisms driving tumor progression and metastasis is of considerable importance.

As a member of LDL receptor family, tumor suppressor gene LRP1B (low density lipoprotein receptor-associated protein 1B) is located on chromosome 2q, containing more than 500 kilobases and more than 90 exons [5], [6]. Many findings have indicated the regulatory role of LRP1B in tumor progression, especially tumor metastasis. For example, downregulation of LRP1B in colorectal cancer cells can inhibit cell growth, migration and metastasis [7]. LRP1B is significantly associated with poor prognosis in breast cancer, especially with lymph node metastasis [8]. Given that whether LRP1B is involved in the progression of Pheo remains unknown, in this study we first analyzed the expression level of LRP1B in Pheo based on The Cancer Genome Atlas (TCGA) database. In addition, we showed that two CpG islands in the promoter region of LRP1B in Pheo cells were highly methylated, which could be a regulatory mechanism underlying the reduced expression of LRP1B in Pheo. Indeed, previous studies have shown that the expression of LRP1B is modulated by epigenetic modifications [9]. Moreover, we reported that the DNA methyltransferases DNMT3B could be responsible for the hypermethylation of LRP1B promoter in Pheo.

Overall, we indicated that the hypermethylation of LRP1B promoter mediated by DNMT3B in Pheo cells potentially promoted tumor progression and metastasis.

Materials and methods

Collection of clinical samples

Pheochromocytoma tissue (n = 34) and its adjacent tissues were taken from patients who underwent surgical resection in the People’s Hospital of Xinjiang Uygur Autonomous Region. All Pheo patients have no other medical history (including other tumors, immune-related diseases, diabetes, etc.), and they did not undergo radiotherapy or chemotherapy before surgical resection. The written informed consent of all patients was obtained in this study. The experiment was approved by the Ethics Committee of the Hospital (No. KY2023060156).

Cell culture and transfection

The rat pheochromocytoma (PC12) were obtained from American Type Culture Collection (ATCC) that have high norepinephrine transporter expression [10]. The control group was mouse adrenal medulla cells purchased from Shanghai LMAI Bio (LM-011, Shanghai, China). PC12 and adrenal medulla cells were cultured in DMEM complete medium (Gibco, USA) containing 10% fetal bovine serum (FBS, Gibco) and 1% penicillin/streptomycin. The cells were placed in an incubator with 5% CO2 at 37℃.

The LRP1B overexpression vector (oe-LRP1B-1, oe-LRP1B-2, and oe-LRP1B-3), shRNA containing DNMT3B target (sh-DNMT3B-1, sh-DNMT3B-2, and sh-DNMT3B-3), sh-DNMT1, sh-DNMT3A, and its negative control (NC) were all designed and synthesized by GenePharma (Shanghai, China). Lipofectamine 3000 (Thermo Fisher) was used for all cell transfections. ShRNAs sequences were listed in Table 1.

Table 1 ShRNA sequence information

Immunohistochemistry (IHC)

Clinical Pheo tissues or Pheo transplanted tumor mouse tissues were collected. The tissues were fixed with 10% formalin and then made into paraffin sections. After dewaxing and rehydration, 30% hydrogen peroxide was used to block the activity of oxidase. Next, the slices were incubated with normal goat serum to reduce nonspecific binding. The slices were incubated with primary antibody at 4℃ overnight. Next, a biotin-coupled secondary antibody was used to treat with slices at room temperature for 2 h. After that, 3,3-diaminobenzidine and hematoxylin were used to dye the slices for color reaction. Finally, the staining results were observed under an optical microscope (Olympus Corporation, Japan). The primary antibodies used were: anti-LRP1B (1:200, # PA5-115522, Thermo Fisher Scientific, MA, USA) and anti-Ki67 (1:200, ab15580, Abcam, Cambridge, UK). The staining was scored as the intensity of the positive staining (0 - negative, 1 - weak, 2 - moderate, 3 - strong) multiplied by the staining areas (0 = negative, 1 = 1–9%, 2 = 10–39%, 3 = 40–69%, and 4 = 70–100%). These scores were independently determined by two pathologists.

Real-time fluorescence quantitative polymerase chain reaction (RT-qPCR)

The PC12 cells or tissues to be detected were collected. Total RNA was obtained from PC12 cells or tissues using TRIzol reagent (Invitrogen, USA). Subsequently, cDNA was synthesized by FastKing cDNA First Chain Synthesis Kit (TIANGEN, Beijing). The mRNA expression levels of LRP1B, DNMT1, DNMT3A and DNMT3B were determined by SYBR Green qPCR super mix (Invitrogen). GAPDH was selected as reference gene and the 2−ΔΔCT method was used to analyze the difference of gene transcription level. The primer sequences of related genes are shown in Table 2.

Table 2 RT-qPCR primer sequence

Western blot

The Pheo tissues or cells were treated with RIPA lysis buffer (Sigma-Aldrich) to obtain the total protein. The total protein was separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to polyvinylidene fluoride membrane (Millipore, MA, USA). After the transferred membrane was sealed with 5% skim milk, it was incubated with primary antibody at 4℃ overnight. Then, the membrane was incubated with goat anti-rabbit IgG (1: 2000; Ab205718, Abcam) at room temperature for 2 h. Finally, the protein banding reaction was carried out by Western chemiluminescence HRP substrate (Abcam). Primary antibodies: DNMT3A (1:1000, ab307503, Abcam), DNMT1 (1:1000, ab188453, Abcam), DNMT3B (1:1000, ab2851, Abcam), LRP1B (1: 2000, # PA5-115522, Thermo Fisher Scientific) and β -actin (1: 2000, ab8226, Abcam). Secondary antibodies: Rabbit Anti-Mouse IgG H&L (HRP) (1:2000, ab6728, Abcam).

Methylation specific PCR (MSP)

The whole genome DNA of PC12 cells, cancer tissues and adjacent tissues were extracted, and then the DNA was modified and transformed by bisulfite by the EZ-DNA Methylation-Gold Kit (Zymo Research). The PCR primers of LRP1B promoter region were designed and synthesized to amplify the gDNA fragment transformed by bisulfite. Primers for methylated (M) and unmethylated (U) promoter regions of LRP1B gene were designed through the Methprimer online program website (http://www.urogene.org/cgi-bin/methprimer/methprimer.cgi). We screened the GAPDH gene without methylation modification as a negative control group. The unmethylated DNA was modified using methylase, and the C base of CG in the DNA was methylated as a positive control. The related primers were listed in Table 3.

Table 3 MSP PCR primer sequence

Analysis of double luciferase reporter gene

The promoter region (2000 bp) of LRP1B was inserted in the promoter-less pmirGLO-empty vector (Promega) to construct wild-type vector (WT-LRP1B). The complete sequence of multiple G mutations to C in the 1000–1500 bp sequence of the LRP1B promoter by Sangon Biotech. The mutation sequence inserted into pmirGLO vector to construct mutant vector (MUT-LRP1B). PC12 cells transfected with oe-NC or oe-DNMT3B were inoculated into 16-well plates. Then, WT-LRP1B or MUT-LRP1B and the control empty plasmid pmirGLO were co-transfected into the above cells. After 48 h’ culture, luciferase activity was detected by double luciferase assay kit (Promega).

Chromatin Immunoprecipitation (ChIP)

The PC12 cells to be detected were cross-linked with formaldehyde solution. The CHIP kit (26157, Thermo Fisher) was used for relevant treatment. In short, the genomic DNA of PC12 was extracted, and DNA fragments were obtained by mechanical ultrasonic wave or microbial nuclease (MNase) digestion. Then, the lysate was immunoprecipitated with anti-DNMT3B (ab2851, Abcam) or negative control IgG (ab172730, Abcam). The enrichment of DNMT3B protein of specific DNA fragment with LRP1B promoter was measured by PCR, the related primers were in Table 4. Input was used as the control homogenization IP product (CT). The negative antibody was used to enrich the IgG product, and Input was also used to homogenize the product (CT). Finally, the difference in the number of amplification cycles after homogenization between IP and IgG (CT) was calculated, and the enrichment fold was calculated by 2CT.

Table 4 CHIP-qPCR primer sequence
Table 5 Relationship between LRP1B expression and clinicopathological features of pheochromocytoma

Colony formation assay

Cells were plated into a 12-well plate (500 cells/well) and cultured at 37℃ for one week. Then, it was fixed with paraformaldehyde and dyed with 0.5% crystal violet solution for 20 min. Finally, the number of colonies was counted.

Transwell assays

Migration experiment: PC12 cells (1 × 105) were cultured in serum-free medium and plated in the upper chamber of a 12-wells Transwell system (with a pore size of 0.4 μm). The culture medium containing 10% FBS was added to the lower chamber. After 24 h of culture, the cells in the lower chamber were fixed with 4% paraformaldehyde and stained with 0.1% crystal violet solution. Finally, the stained cells were observed under an optical microscope and photographed. In the invasion experiment, the bottom of the upper chamber was coated with matrix glue (BD Biosciences, USA), and the other operation steps were consistent with the migration experiment.

In vivo experiment

10 male BALB/c nude mice (4 weeks) were purchased from Guangdong Medical Laboratory Animal Center (Foshan, China). PC12 cells stably transfected with oe-NC or oe-LRP1B were collected. After washing twice with PBS solution, the cells were resuspended in serum-free DMEM (1 × 107/mL) and subcutaneously injected into the armpit of nude mice (0.2 mL).,. After 28 days, mice were killed by dislocation of spine and tumors were collected. Hematoxylin-eosin staining (H&E) was used to detect the pathological changes of the tumor. Metastatic nodules in the lung specimens were counted under a light microscope.

Statistical analysis

All experimental results were expressed as mean ± standard deviation (SD). Graphpad 8.0 was used for statistical analysis. t test was used for statistical analysis between the two groups, and one-way analysis of variance and Tukey’s multiple comparison test were used for comparisons between groups. X2-test was applied to analyze the correlations between LRP1B expression and clinicopathological characterization of Pheo patients. P value < 0.05 indicates significant difference.

Results

LRP1B was lowly-expressed in Pheo

To evaluate the expression level of LRP1B in Pheo tissues, we collected the samples derived from clinical interventions of 34 patients with Pheo at the early stage, and the clinical information was shown in Table 5. We showed that both the mRNA and protein levels of LRP1B were decreased in Pheo (Fig. 1A and B, P < 0.05). Moreover, the difference of LRP1B expression was related to Ki67 proliferation index of patients (Table 5). Consistently, IHC suggested that compared with the tissues adjacent to cancer, LRP1B was lowly expressed in Pheo, while Ki67 protein, a proliferation marker, was high expressed in Pheo (Fig. 1C, P < 0.05).

Fig. 1
figure 1

LRP1B was lowly-expressed in pheochromocytoma. (A) RT-qPCR showing the expression of LRP1B in Pheo and adjacent tissues. (B) Western blot showing the expression of LRP1B in Pheo and adjacent tissues (C) IHC showing the protein levels of LRP1B and Ki67 in Pheo and adjacent tissues. All results were analyzed by t test. n=34, *P<0.05, **P<0.01, ***P<0.001. PPGLs: Phaeochromocytomas and paragangliomas.

LRP1B promoter was hypermethylated in Pheo

In line with the results from patient samples, the mRNA and protein levels of LRP1B were also reduced in Pheo cell line PC12 (Fig. 2A and B, P < 0.05). Since it has been already reported that LRP1B could be regulated at epigenetic level [9], we reasoned that the inhibited expression of LRP1B in Pheo cells might be related to an altered DNA methylation of LRP1B promoter in Pheo. Methprimer predicted that there were two CpG islands in the promoter region of LRP1B (Fig. 2C), which could be the potential methylated sites in Pheo cells. As expected, MSP assay showed that there was a high DNA methylation level in PC12 (Fig. 2D). Interestingly, we further analyzed the methylation level of LRP1B in cancer tissues and adjacent tissues of Pheo patients, and the results showed that LRP1B in cancer tissues had high methylation level (Fig. 2E). Considering the above observations, we hypothesized that the down-regulation of LRP1B in Pheo could result from the hypermethylation of LRP1B promoter.

Fig. 2
figure 2

The promoter of LRP1B was hypermethylated in pheochromocytoma. (A) RT-qCPR showing the mRNA level of LRP1B in adrenal medullary cells and pheochromocytoma cells (PC12), n=34. (B) Western blot showing the expression of LRP1B in cells. (C) Methprimer showing the predicted CpG islands in the promoter region of LRP1B; (D) MSP showing the methylation of LRP1B in pheochromocytoma cells. n=3. (A) MSP showing the methylation of LRP1B in Pheo and adjacent tissues. All results were analyzed by t test. n=4*P<0.05, **P<0.01, ***P<0.001.

DNMT3B could mediate the methylation of LRP1B promoter in Pheo

DNMT1, DNMT3A and DNMT3B are the predominate writers for DNA methylation [11]. To identify the writer catalyzing DNA methylation of LRP1B in Pheo, we first measured the expression levels of all three writers in Pheo tissues. While DNMT1 was similarly expressed by Pheo and cancer adjacent tissues, DNMT3A and DNMT3B were up-regulated in Pheo (Fig. 3A, P < 0.05). Compared with DNMT3A, DNMT3B was more significantly up-regulated in Pheo (Fig. 3A, P < 0.05). Moreover, after knocking down DNMT1, DNMT3A or DNMT3B in PC12 cells, we found that LRP1B was up-regulated most significantly after knocking down DNMT3B (Fig. 3B, P < 0.05). Therefore, we knocked down DNMT3B in PC12 cells using DNMT3B shRNAs. We observed that methylation of LRP1B promoter was decreased in the PC12 transfected with DNMT3B shRNAs (Fig. 3C, P < 0.05). The direct interaction between DNMT3B and LRP1B promoter was further confirmed by luciferase assay and ChIP (Fig. 3E and F, P < 0.05). The above results indicated that the methyltransferase DNMT3B inhibited LRP1B by mediating the methylation of LRP1B promoter in Pheo.

Fig. 3
figure 3

DNMT3B could directly bind to the promoter region of LRP1B. (A) RT-qPCR showing the expression levels of DNMT1, DNMT3A, and DNMT3B in pheochromocytoma. (B) RT-qPCR showing the expression of DNMT1, DNMT3A, and DNMT3B in PC12 cells. (C) Western blot was used to detect the changes of LRP1B protein expression after knocking down DNMT3A, DNMT3B or DNMT1, respectively. (D) MSP showing the methylation of LRP1B promoter was mediated by DNMT3B. (E) The interaction between DNMT3B and LRP1B promoter region was detected by dual luciferase reporter gene assay. (F) ChIP showing the interaction between DNMT3B and LRP1B promoter region. All results were analyzed by t test. n=3, *P<0.05, **P<0.01, ***P<0.001.

DNMT3B promoted the proliferation, migration and invasion of Pheo

To reveal how DNMT3B regulated the progression of Pheo, we identified cell proliferation, migration, and invasion of PC12 transfected with DNMT3B shRNAs. DNMT3B was efficiently silenced by all shRNAs, among which sh- DNMT3B -2 showed the highest efficiency (Fig. 4A and B, P < 0.05). More important, when PC12 was transfected with sh- DNMT3B-2, cell proliferation, migration, and invasion were significantly inhibited (Fig. 4D and E, P < 0.05). Therefore, DNMT3B promoted Pheo progression by enhancing the proliferation, migration, and invasion of tumor cells.

Fig. 4
figure 4

DNMT3B promoted the proliferation, migration and invasion of pheochromocytoma cells. (A-B) RT-qPCR and western blot showing the knockdown efficiency of sh-DNMT3B in PC12 cells. (C) Western blot showing LRP1B protein in PC12 cells. (D) Clonal formation showing the proliferation of PC12 cells. (E) Transwell showing the migration and invasion of PC12 cells. All results were analyzed by one-way ANOVA and Tukey multiple comparison test. n = 3, *P < 0.05, **P < 0.01, ***P < 0.001

DNMT3B promoted Pheo progression by repressing LRP1B

Since we have revealed the potential regulatory correlation between DNMT3B and LRP1B in Pheo, next we utilized rescue experiment to demonstrate that DNMT3B promoted Pheo by repressing the tumor repressive gene LRP1B. We simultaneously transfected PC12 with DNMT3B and (or) LRP1B overexpression vectors. The overexpression of LRP1B alone did not change the mRNA or protein of DNMT3B, whereas overexpression of DNMT3B alone reduced LRP1B protein levels. Co-overexpression of DNMT3B and LRP1B reversed the inhibitory effect of DNMT3B overexpression alone on LRP1B (Fig. 5A and B, P < 0.05). However, the overexpression of DNMT3B inhibited the exogenous expression of LRP1B in PC12 transfected with both oe-DNMT3B and oe-LRP1B (Fig. 5A and B). We also confirmed that the overexpression of LRP1B repressed the proliferation, migration and invasion of Pheo cells, which was compromised by DNMT3B overexpression (Fig. 5C and D, P < 0.05). The above results indicated that DNMT3B could promote Pheo progression by repressing LRP1B.

Fig. 5
figure 5

DNMT3B promoted the proliferation, migration and invasion of pheochromocytoma cells by regulating LRP1B. (A) RT-qPCR showing the mRNA levels of DNMT3B and LRP1B in PC12 cells. (B) Western blot showing the protein levels of DNMT3B and LRP1B. (C) Clonal formation showing the proliferation of PC12 cells. (D) Transwell showing the migration and invasion of PC12 cells. All results were analyzed by one-way ANOVA and Tukey multiple comparison test.n = 3, *P < 0.05, **P < 0.01, ***P < 0.001

Overexpression of LRP1B inhibited Pheo tumor growth in vivo

Finally, we verified the repressive effect of LRP1B on Pheo in vivo using mouse model. In vivo transplantation showed that the Pheo tumor overexpressing LRP1B was apparently smaller than the control tumor (Fig. 6A and B, P < 0.05), suggesting the tumor suppressive role of LRP1B. H&E staining showed that tumor growth was inhibited in LRP1B-OE group (Fig. 6C, P < 0.05). Moreover, LRP1B inhibited the proliferation marker Ki67 in mice Pheo transplanted tumor (Fig. 6D, P < 0.05), further supporting that LRP1B inhibited Pheo.

Fig. 6
figure 6

LRP1B inhibits tumor growth of pheochromocytoma in vivo. Transplanted tumor was formed by transfection of OE-NC or OE-LRP1B into Pheo cells, followed by subcutaneous injection into mice. (A) Image showing the tumor engraftments. (B) Isolation and display diagram of mouse tumor. (C) H&E staining of the tumor engraftments. (D) IHC showing the expression of Ki67 in transplanted tumor of mice in each group. All results were analyzed by t test. n=5, *P<0.05, **P<0.01, ***P<0.001.

Discussion

Due to the heterogeneity feature of Pheo, it’s challenging to diagnose or treat this kind of neuroendocrine tumor [12]. Therefore, understanding the molecular mechanisms regulating tumor progression could shed new light on the diagnosis and treatments for Pheo [13]. In this study, we screened out LRP1B with significant differences in Pheo through TCGA database. It was revealed that the hypermethylation of the tumor suppressive gene LRP1B promoter, which was catalyzed by DNMT3B, was a potential cause of tumor progression and metastasis of Pheo.

LRP1B has been demonstrated to be a tumor suppressor gene [14] in several types of cancer. For instances, LRP1B had potential impacts on the efficacy of the immunotherapy for non-small cell lung cancer (NSCLC) [15]. The mutation of LRP1B was highly correlated with the incidence of ovarian cancer [16]. The copy number of LRP1B has found to be lost in thyroid carcinoma [17]. And to complement previous studies, here we reported that the abnormal epigenetic regulation of LRP1B was a driver of Pheo. We demonstrated that compared with adjacent noncancerous tissue, LRP1B had a lower expression level in Pheo. And our in vivo and in vitro experiments suggested the tumor suppressive role of LRP1B in Pheo, as evidenced by the repressed proliferation, migration and invasion presented by LRP1B-overexpressed Pheo cells. It can be seen that LRP1B is also involved in regulating the progress of Pheo. Interestingly, our study found that low levels of LRP1B could increase the migration and invasion ability of PC12 cells. Metastatic pheochromocytoma and paraganglioma are usually difficult to treat and resistant to most conventional therapies [18]. Whether this regulatory role of LRP1B is also present in highly invasive murine Pheo MTT cells is worthy of investigation.

Using Methprimer, we predicted that within the promoter region of LRP1B, there were two CpG islands, which were preferred by DNA methylation [19]. Further screening revealed that the methyltransferase responsible for the hypermethylation of LRP1B promoter in Pheo was DNMT3B, which has been widely studied in the progression of thyroid cancer, renal cancer, colorectal cancer, and other tumors [25,26,27]. We observed a reversed expression pattern of LRP1B and DNMT3B in the clinical samples as well as the cell line PC12 of Pheo. Specifically, LRP1B was lowly expressed while DNMT3B was highly expressed in Pheo. Leveraging biochemistry assays including luciferase and ChIP, we demonstrated the direct interaction between DNMT3B and LRP1B promoter. More importantly, our functional assays, especially the rescue experiment, suggested the progression of Pheo was regulated by the DNMT3B-LRP1B axis.

In recent years, besides the tumor cells themselves, the stromal cells and immune cells of Pheo are also of great significance for understanding tumor biology and treatment [23]. At present, our research on Pheo is mainly concentrated in Pheo cells, and the regulation of DNMT3B-LRP1B axis is also mainly in Pheo cells. It was found that the expression of LRP1B in gastric cancer was related to the infiltration of CD4+ T cells and macrophages [24]. DNMT3B can regulate tumor microenvironment and affect cancer chemotherapy sensitivity [25]. Recent studies have found that some cell types (including fibroblasts and macrophages) in tumor microenvironment can also regulate the characteristics of Pheo cells [26], 27]. Therefore, our research can continue to explore whether the DNMT3B-LRP1B axis affects other cells in the Pheo microenvironment.

It’s noteworthy that the dysregulation of LRP1B in a variety of tumor cells has been attributed to the abnormal epigenetic modification, including DNA methylation and histone acetylation [28]. In esophageal carcinoma, LRP1B loss-of-function mutation is related to transcriptional silencing caused by hypermethylation of CpG island [29]. DNA copy number loss and CpG island methylation of LRP1B, which inhibit LRP1B gene expression, are frequently present in cancer tissues, which led to low mRNA expression [30,31,32]. Here we have only identified one potential epigenetic modification contributing to the repression of LRP1B in Pheo. Whether other epigenetic modifications, such as histone modifications and even RNA methylations, were also involved in regulating LRP1B in Pheo is worth to be studied in the future. Taken together, our study has improved the understanding towards Pheo progression by firstly reporting an epigenetic regulatory mechanism controlling the proliferation, migration, and invasion of Pheo tumor cells. And this study would provide new insights into the diagnosis and treatments for Pheo.

Data availability

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

References

  1. Calsina B, Piñeiro-Yáñez E, Martínez-Montes AM, Caleiras E, Fernández-Sanromán A, Monteagudo M, Torres-Pérez R, Fustero-Torre C, Pulgarín-Alfaro M, Gil E et al. Genomic and immune landscape of metastatic pheochromocytoma and paraganglioma. Nat Commun. 2023;14(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-023-36769-6. PMID: WOS:000955827800007. ARTN 1122.

  2. Carrasquillo JA, Chen CC, Jha A, Ling A, Lin FI, Pryma DA, Pacak K. Imaging of pheochromocytoma and paraganglioma. J Nucl Med. 2021;62(8):1033–42. https://doiorg.publicaciones.saludcastillayleon.es/10.2967/jnumed.120.259689. PMID: WOS:000711570000009.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Martinelli S, Amore F, Canu L, Maggi M, Rapizzi E. Tumour microenvironment in pheochromocytoma and paraganglioma. Front Endocrinol. 2023;14. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fendo.2023.1137456. PMID: WOS:000963005200001. ARTN 1137456.

  4. Lima JV, Kater CE. The pheochromocytoma/paraganglioma syndrome: an overview on mechanisms, diagnosis and management. Int Braz J Urol. 2023;49(3):307–19. https://doiorg.publicaciones.saludcastillayleon.es/10.1590/S1677-5538.Ibju.2023.0038. PMID: WOS:001021771100004.

    Article  PubMed  Google Scholar 

  5. Ye C, Chong W, Liu Y, Zhu X, Ren H, Xu K, Xie X, Du F, Zhang Z, Wang M, et al. Suppression of tumorigenesis in LUAD by LRP1B through regulation of the IL-6-JAK-STAT3 pathway. Am J Cancer Res. 2023;13(7):2886–905. PMID: 37560001.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Brown LC, Tucker MD, Sedhom R, Schwartz EB, Zhu J, Kao C, Labriola MK, Gupta RT, Marin D, Wu Y, et al. LRP1B mutations are associated with favorable outcomes to immune checkpoint inhibitors across multiple cancer types. J Immunother Cancer. 2021;9(3). https://doiorg.publicaciones.saludcastillayleon.es/10.1136/jitc-2020-001792. PMID: 33653800.

  7. Wang Z, Sun P, Gao C, Chen J, Li J, Chen Z, Xu M, Shao J, Zhang Y, Xie J. Down-regulation of LRP1B in colon cancer promoted the growth and migration of cancer cells. Exp Cell Res. 2017;357(1):1–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.yexcr.2017.04.010. PMID: 28408316.

    Article  CAS  PubMed  Google Scholar 

  8. Asano Y, Takeuchi T, Okubo H, Saigo C, Kito Y, Iwata Y, Futamura M, Yoshida K. Nuclear localization of LDL receptor-related protein 1B in mammary gland carcinogenesis. J Mol Med (Berl). 2019;97(2):257–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00109-018-01732-2. PMID: 30607440

    Article  CAS  PubMed  Google Scholar 

  9. Príncipe C, de Sousa IJD, Prazeres H, Soares P, Lima RT. LRP1B: A giant lost in Cancer translation. Pharmaceuticals-Base. 2021;14(9). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ph14090836. PMID: WOS:000701275400001 ARTN 836.

  10. Ohshima Y, Sudo H, Watanabe S, Nagatsu K, Tsuji AB, Sakashita T, Ito YM, Yoshinaga K, Higashi T, Ishioka NS. Antitumor effects of radionuclide treatment using alpha-emitting meta-(211)At-astato-benzylguanidine in a PC12 pheochromocytoma model. Eur J Nucl Med Mol Imaging. 2018;45(6):999–1010. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00259-017-3919-6. PMID: 29350258

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Li JS, Su XL, Dai L, Chen N, Fang C, Dong ZX, Fu JM, Yu Y, Wang WS, Zhang HT, et al. Temporal DNA methylation pattern and targeted therapy in colitis-associated cancer. Carcinogenesis. 2020;41(2):235–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/carcin/bgz199. PMID: WOS:000537366000012.

    Article  CAS  PubMed  Google Scholar 

  12. Leung K, Stamm M, Raja A, Low G, Pheochromocytoma. The range of appearances on ultrasound, CT, MRI, and functional imaging. Am J Roentgenol. 2013;200(2):370–8. https://doiorg.publicaciones.saludcastillayleon.es/10.2214/Ajr.12.9126 PMID: WOS:000313957400037.

    Article  Google Scholar 

  13. Ullrich M, Richter S, Liers J, Drukewitz S, Friedemann M, Kotzerke J, Ziegler CG, Nölting S, Kopka K, Pietzsch J. Epigenetic drugs in somatostatin type 2 receptor radionuclide theranostics and radiation transcriptomics in mouse pheochromocytoma models. Theranostics. 2023;13(1):278–94. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/thno.77918. PMID: WOS:000894576600003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rui Wang GZ, Zhu X, Xu Y, Cao N, Li Z, Han C, Shen MQY, Dong J, Zhao FMA. Prognostic Implications of LRP1B and Its Relationship with the Tumor-Infiltrating Immune Cells in Gastric Cancer. Cancers. 2023;15(24):5759. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers15245759

  15. Hao F, Ma Q, Zhong DS. Potential predictive value of comutant LRP1B and FAT for immune response in non-small cell lung cancer LRP1B and FAT comutation enhance immune response. Transl Oncol. 2022;24.https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tranon.2022.101493 PMID: WOS:000835517900004. ARTN 101493.

  16. de Sousa IDJ, Cunha AI, Saraiva IA, Portugal RV, Gimba ERP, Guimaraes M, Prazeres H, Lopes JM, Soares P, Lima RT. LRP1B expression as a putative predictor of response to pegylated liposomal doxorubicin treatment in ovarian Cancer. Pathobiology. 2021;88(6):400–11.https://doiorg.publicaciones.saludcastillayleon.es/10.1159/000517372 PMID: WOS:000713908500001.

    Article  Google Scholar 

  17. Prazeres H, Torres J, Rodrigues F, Pinto M, Pastoriza MC, Gomes D, Cameselle-Teijeiro J, Vidal A, Martins TC, Sobrinho-Simoes M, et al. Chromosomal, epigenetic and microRNA-mediated inactivation of LRP1B, a modulator of the extracellular environment of thyroid cancer cells (30, Pg 1302, 2010). Oncogene. 2017;36(1):146. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2016.143. PMID: WOS:000394069100013.

    Article  CAS  PubMed  Google Scholar 

  18. Nolting S, Giubellino A, Tayem Y, Young K, Lauseker M, Bullova P, Schovanek J, Anver M, Fliedner S, Korbonits M, et al. Combination of 13-Cis retinoic acid and Lovastatin: marked antitumor potential in vivo in a pheochromocytoma allograft model in female athymic nude mice. Endocrinology. 2014;155(7):2377–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1210/en.2014-1027 PMID: 24762141 10.1210/en.2014– 1027.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Sergeeva A, Davydova K, Perenkov A, Vedunova M. Mechanisms of human DNA methylation, alteration of methylation patterns in physiological processes and oncology. Gene. 2023;875. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cellsig.2017.01.017 PMID: WOS:001011456200001. ARTN 147487.

  20. Zhu X, Xue CY, Kang XF, Jia XM, Wang L, Younis MH, Liu DH, Huo N, Han YC, Chen Z et al. DNMT3B-mediated FAM111B methylation promotes papillary thyroid tumor glycolysis, growth and metastasis. Int J Biol Sci. 2022;18(11):4372-87. https://doiorg.publicaciones.saludcastillayleon.es/10.7150/ijbs.72397. PMID: WOS:000832879400008.

  21. Quan Z, He YF, Luo CL, Xia Y, Zhao Y, Liu NJ, Wu XH. Interleukin 6 induces cell proliferation of clear cell renal cell carcinoma by suppressing HepaCAM via the STAT3-dependent up-regulation of DNMTI or DNMT3B. Cell Signal. 2017;32:48–58. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cellsig.2017.01.017. PMID: WOS:000395953400005.

    Article  CAS  PubMed  Google Scholar 

  22. Ren GL, Li HY, Hong D, Hu FY, Jin RJ, Wu S, Sun WH, Jin HL, Zhao LL, Zhang XD et al. LINC00955 suppresses colorectal cancer growth by acting as a molecular scaffold of TRIM25 and Sp1 to inhibit DNMT3B-mediated methylation of the PHIP promoter. BMC Cancer. 2023;23(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12885-023-11403-2. PMID: WOS:001075729700003 ARTN 898.

  23. Zethoven M, Martelotto L, Pattison A, Bowen B, Balachander S, Flynn A, Rossello FJ, Hogg A, Miller JA, Frysak Z, et al. Single-nuclei and bulk-tissue gene-expression analysis of pheochromocytoma and paraganglioma links disease subtypes with tumor microenvironment. Nat Commun. 2022;13(1):6262. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-022-34011-3. PMID: 36271074.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wang R, Zhang G, Zhu X, Xu Y, Cao N, Li Z, Han C, Qin M, Shen Y, Dong J, et al. Prognostic implications of LRP1B and its relationship with the Tumor-Infiltrating immune cells in gastric Cancer. Cancers (Basel). 2023;15(24). https://doiorg.publicaciones.saludcastillayleon.es/10.3390/cancers15245759. PMID: 38136305.

  25. Song Y, Kim N, Heo J, Shum D, Heo T, Seo HR. Inhibition of DNMT3B expression in activated hepatic stellate cells overcomes chemoresistance in the tumor microenvironment of hepatocellular carcinoma. Sci Rep. 2024;14(1):115. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-023-50680-6. PMID: 38168140.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Martinelli S, Riverso M, Mello T, Amore F, Parri M, Simeone I, Mannelli M, Maggi M, Rapizzi E. SDHB and SDHD silenced pheochromocytoma spheroids respond differently to tumour microenvironment and their aggressiveness is inhibited by impairing stroma metabolism. Mol Cell Endocrinol. 2022;547:111594. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mce.2022.111594. PMID: 35149119.

    Article  CAS  PubMed  Google Scholar 

  27. Martinelli S, Amore F, Canu L, Maggi M, Rapizzi E. Tumour microenvironment in pheochromocytoma and paraganglioma. Front Endocrinol (Lausanne). 2023;14:1137456. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fendo.2023.1137456. PMID: 37033265.

    Article  PubMed  Google Scholar 

  28. Ni SB, Hu JR, Duan YS, Shi SL, Li R, Wu HJ, Qu YP, Li Y. Down expression of promotes cell migration via RhoA/Cdc42 pathway and actin cytoskeleton remodeling in renal cell cancer. Cancer Sci. 2013;104(7):817–25. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cas.12157. PMID: WOS:000325680000006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sonoda I, Imoto I, Inoue J, Shibata T, Shimada Y, Chin K, Imamura M, Amagasa T, Gray JW, Hirohashi S, et al. Frequent Silencing of low density lipoprotein receptor-related protein 1B (LRP1B) expression by genetic and epigenetic mechanisms in esophageal squamous cell carcinoma. Cancer Res. 2004;64(11):3741–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-04-0172. PMID: 15172977.

    Article  CAS  PubMed  Google Scholar 

  30. Prazeres H, Torres J, Rodrigues F, Pinto M, Pastoriza MC, Gomes D, Cameselle-Teijeiro J, Vidal A, Martins TC, Sobrinho-Simoes M, et al. Chromosomal, epigenetic and microRNA-mediated inactivation of LRP1B, a modulator of the extracellular environment of thyroid cancer cells. Oncogene. 2011;30(11):1302–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/onc.2010.512. PMID: 21057533.

    Article  CAS  PubMed  Google Scholar 

  31. Sadee W, Wang D, Hartmann K, Toland AE. Pharmacogenomics: driving personalized medicine. Pharmacol Rev. 2023;75(4):789–814. https://doiorg.publicaciones.saludcastillayleon.es/10.1124/pharmrev.122.000810. PMID: 36927888.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Li SY, Sun R, Wang HX, Shen S, Liu Y, Du XJ, Zhu YH, Jun W. Combination therapy with epigenetic-targeted and chemotherapeutic drugs delivered by nanoparticles to enhance the chemotherapy response and overcome resistance by breast cancer stem cells. J Control Release. 2015;205:7–14. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jconrel.2014.11.011. PMID: 25445694.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by Xinjiang Natural Science Foundation General Project (Grant No. 2022D01C630), National Natural Science Foundation of China Regional Project (Grant No. 82360520), Xinjiang Natural Science Foundation for Distinguished Young Scholars (Grant No. 2022D01E84) and Xinjiang Tianshan Talent Training Program (Grant No. 2023TSYCCX0061).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Min Sun, Jiuzhi Li. Methodology: Jing Wan, Bingli Zheng. Software: Zhenfeng Shi, Jiuzhi Li. Validation: Zhenfeng Shi, Yanrong Ma. Formal analysis: Jing Wan, Bingli ZhengInvestigation: Min Sun, Zhenfeng Shi, Jiuzhi Li. Resources: Min Sun, Yanrong Ma. Data Curation: Jing Wan, Bingli Zheng. Writing - Original Draft: Min Sun. Writing - Review & Editing: Zhenfeng Shi, Yanrong Ma. Visualization: Yanrong Ma, Jiuzhi Li. Supervision: Min Sun. Project administration: Min Sun. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Zhenfeng Shi or Jiuzhi Li.

Ethics declarations

Ethical approval

The collection of tissue samples obtained approval from the Ethics Committee of People’s Hospital of Xinjiang Uygur Autonomous Region (No. KY2023060156).

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Supplementary Material 2

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, M., Ma, Y., Wan, J. et al. DNMT3B promotes the progression of pheochromocytoma by mediating the hypermethylation of LRP1B promoter. Epigenetics & Chromatin 18, 29 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13072-025-00592-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13072-025-00592-8

Keywords